1
|
Zheng S, Feng W, Sun Z, Xu P, Dong S, Pan L, Shen H, He J, Chen P, Shu C. HSD17B1-mediated trophoblast differentiation lowers estrogen levels in early-onset preeclampsia. Sci Rep 2025; 15:17448. [PMID: 40394177 PMCID: PMC12092795 DOI: 10.1038/s41598-025-02490-1] [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: 02/14/2025] [Accepted: 05/13/2025] [Indexed: 05/22/2025] Open
Abstract
Early-onset preeclampsia (EOPE) with fetal growth restriction (FGR) is a severe hypertensive disorder of pregnancy characterized by placental dysfunction and estrogen deficiency. Based on single-cell RNA sequencing (scRNA-seq) profiling of specific placental trophoblast subtypes from EOPE-FGR and normotensive pregnancies, we identified HSD17B1, which encodes a key enzyme mediating estradiol conversion, as the central dysregulated node in EOPE pathogenesis. Multi-modal computational analysis (cluster annotation, cellular proportion calculation, comparison of differentially expressed genes, and characterization of cellular developmental trajectories) revealed key expression dynamics during syncytiotrophoblast (SCT) differentiation, with substantial suppression in EOPE specimens. Further validation using clinical placental samples confirmed the downregulation of HSD17B1 at the protein level in patients with EOPE, as demonstrated by immunohistochemistry and western blotting. Mechanistically, HSD17B1 knockdown in BeWo trophoblast models recapitulated the core EOPE phenotypes of impaired SCT differentiation and estrogen biosynthesis blockade. These findings reveal that HSD17B1 is a master coordinator of trophoblast-endocrine crosstalk, the impairment of which in placental trophoblasts may contribute to EOPE pathogenesis. Our findings provide a mechanistic basis for developing HSD17B1-targeted interventions that could contribute to the concurrent restoration of placental competence and hormonal regulation, improving the perinatal outcomes of patients with EOPE.
Collapse
Affiliation(s)
- Shu Zheng
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Wei Feng
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Zewen Sun
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Peng Xu
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Shuai Dong
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Lin Pan
- The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Huimin Shen
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Jin He
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Peng Chen
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, China.
| | - Chang Shu
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China.
| |
Collapse
|
2
|
Liu Y, Du Z, Li L, Huang J, Liu S, Lu B, Duan Y, Cheng Y, Li T, Zhang J, Mo J, Yang Y, Wang W, Zou H, Liang T, Jiang M, Yang M, Chen Y, Ouyang C, Chen C. scRNA-seq reveals an immune microenvironment and JUN-mediated NK cell exhaustion in relapsed T-ALL. Cell Rep Med 2025; 6:102098. [PMID: 40306275 DOI: 10.1016/j.xcrm.2025.102098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/21/2025] [Accepted: 04/08/2025] [Indexed: 05/02/2025]
Abstract
T cell acute lymphoblastic leukemia (T-ALL) is a heterogeneous disease characterized by a high relapse rate. By single-cell transcriptome analysis, we characterize the bone marrow immune microenvironment in patients with T-ALL, identifying 13 major cell clusters. These patients exhibited abnormally expanded hematopoietic stem cells (HSCs) and granulocyte-monocyte progenitors (GMPs), immunosuppressive traits in CD4+ T, CD8+ T, and natural killer (NK) cells. Subdividing CD4+ T cells reveal two subsets transitioning between T helper (Th)1/Th2, Annexin-A1 (ANXA1)-GATA3-CD4+ T, and ANXA1+GATA3+CD4+ T. Additionally, NK cells demonstrate exhaustion in the tumor microenvironment of patients with relapsed T-ALL, with JUN identified as a critical factor. Additionally, JUN is also highly expressed in T-ALL and is crucial for maintaining its proliferation. The JUN inhibitor exhibited successful lethality toward leukemia cells and ameliorated NK cell exhaustion in relapsed T-ALL cell line, as well as in cell-derived tumor xenograft (CDX), patient-derived tumor xenograft (PDX), and NOTCH1-mutant mouse models. In summary, our findings enhance the understanding of T-ALL relapse mechanisms and support the development of innovative immunotherapies for patients with relapsed T-ALL.
Collapse
Affiliation(s)
- Yong Liu
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Zefan Du
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Lindi Li
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Junbin Huang
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Su Liu
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Bo Lu
- Department of Haematology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Yifei Duan
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Yucai Cheng
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Tianwen Li
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Jing Zhang
- Department of Thyroid and Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Jiani Mo
- Department of Hematology, Affiliated Hospital of Guangdong Medical University (GDMU), Zhanjiang 524001, Guangdong, China
| | - Yalin Yang
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Wengqing Wang
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Hailin Zou
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Tianqi Liang
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Meng Jiang
- School of Medicine, Sun Yat-sen University, Shenzhen 518107, Guangdong, China
| | - Mo Yang
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China; Department of Thyroid and Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China.
| | - Yun Chen
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China.
| | - Cheng Ouyang
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China.
| | - Chun Chen
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, Guangdong, China.
| |
Collapse
|
3
|
Gholamin S, Natri HM, Zhao Y, Xu S, Aftabizadeh M, Comin-Anduix B, Saravanakumar S, Masia C, Wong RA, Peter L, Chung MI, Mee ED, Aguilar B, Starr R, Torrejon DY, Alizadeh D, Wu X, Kalbasi A, Ribas A, Forman S, Badie B, Banovich N, Brown C. Overcoming myeloid-driven resistance to CAR T therapy by targeting SPP1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646202. [PMID: 40236117 PMCID: PMC11996542 DOI: 10.1101/2025.04.01.646202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Chimeric antigen receptor CAR T cell therapy faces notable limitations in treatment of solid tumors. The suppressive tumor microenvironment TME, characterized by complex interactions among immune and stromal cells, is gaining recognition in conferring resistance to CAR T cell therapy. Despite the abundance and diversity of macrophages in the TME, their intricate involvement in modulating responses to CAR T cell therapies remains poorly understood. Here, we conducted single-cell RNA sequencing scRNA seq on tumors from 41 glioma patients undergoing IL13Ra2-targeted CAR T cell therapy, identifying elevated suppressive SPP1 signatures predominantly in macrophages from patients who were resistant to treatment. Further integrative scRNA seq analysis of high-grade gliomas as well as an interferon-signaling deficient syngeneic mouse model both resistant to CAR T therapy demonstrated the role of congruent suppressive pathways in mediating resistance to CAR T cells and a dominant role for SPP1+ macrophages. SPP1 blockade with an anti-SPP1 antibody abrogates the suppressive TME effects and substantially prolongs survival in IFN signaling-deficient and glioma syngeneic mouse models resistant to CAR T cell therapy. These findings illuminate the role of SPP1+ macrophages in fueling a suppressive TME and driving solid tumor resistance to CAR cell therapies. Targeting SPP1 may serve as a universal strategy to reprogram immune dynamics in solid tumors mitigating resistance to CAR T therapies.
Collapse
|
4
|
Zou L, Chen K, Hong X, Ye B. Single-cell RNA sequencing reveals immunological link between house dust mite allergy and childhood asthma. Sci Rep 2025; 15:16812. [PMID: 40368964 PMCID: PMC12078649 DOI: 10.1038/s41598-025-01538-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 05/06/2025] [Indexed: 05/16/2025] Open
Abstract
Allergic asthma in children is typically associated with house dust mites (HDM) as the key allergen. Nevertheless, the diagnostic rate remains below 60% due to the absence of specific symptoms and diagnostic markers, which hinders the implementation of targeted personalized therapies. This study investigates immunological features of asthma with house dust mite (HDM) sensitisation in children, aiming to uncover diagnostic markers at single-cell resolution. The cohort comprised 8 children with physician-diagnosed asthma (age range: 4-11 years), stratified into groups based on HDM sensitization status. Single-cell RNA sequencing of peripheral blood mononuclear cells (PBMCs) was conducted, employing Seurat for cell identification and differential gene expression analysis. Enrichment analyses and LASSO regression identified signature genes related to cellular origin, with protein-protein interaction networks elucidating cellular communication differences between groups. A total of 11 distinct cell types were identified, with classical monocytes and monocytes being the predominant cell types that differentiated the two groups. Among these, 12 genes were up-regulated, and 40 down-regulated, mainly involving MHC-II complex and antigen presentation pathways, as validated by Gene Ontology and Gene Set Enrichment Analysis. The machine learning model accurately predicted cellular groupings, evidenced by an area under the curve of 0.83. Enhanced communication signals in HDM allergy cases involved monocytes, contrasting with reduced interactions in naive CD8 + cells. HLA-DR and HLA-DP were identified as the primary hallmark receptors, and the innate immunity differences with non-dust mite allergic asthma were characterized by 18 genes including top candidates MT-ND4 and RPS3A. Individuals with HDM-sensitized asthma exhibited altered expression of MHC-II complex genes in their PBMCs and distinct gene expression patterns in antigen-presenting cells, highlighting the critical role of HLA-DR and HLA-DP in the HDM allergen presentation.
Collapse
Affiliation(s)
- Lingyun Zou
- Department of Clinical Data Research, Chongqing Emergency Medical Center, Chongqing Key Laboratory of Emergency Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China.
| | - Kang Chen
- Department of Nuclear Medicine, First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xianou Hong
- Shenzhen Baoan Women's and Children's Hospital, Jinan University, Guangdong, China
| | - Bo Ye
- Department of Clinical Data Research, Chongqing Emergency Medical Center, Chongqing Key Laboratory of Emergency Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China.
| |
Collapse
|
5
|
Ali M, Garcia P, Lunkes LP, Sciortino A, Thomas MH, Heurtaux T, Grzyb K, Halder R, Skupin A, Buée L, Blum D, Buttini M, Glaab E. Temporal transcriptomic changes in the THY-Tau22 mouse model of tauopathy display cell type- and sex-specific differences. Acta Neuropathol Commun 2025; 13:93. [PMID: 40336141 PMCID: PMC12060421 DOI: 10.1186/s40478-025-02013-z] [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: 04/22/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Tauopathies, including Alzheimer's disease (AD) and frontotemporal dementia (FTD), display sex-specific differences in prevalence and progression, but the underlying molecular mechanisms remain unclear. Single-cell transcriptomic analysis of animal models can reveal how AD pathology affects different cell types across sex and age. OBJECTIVE To understand sex-specific and sex-dimorphic transcriptomic changes in different cell types and their age-dependence in the THY-Tau22 mouse model of AD-linked tauopathy. METHODS We applied single-cell RNA sequencing (scRNA-seq) to cortical tissue from male and female THY-Tau22 and wild-type mice at 17 months of age, when they had prominent tau inclusion pathology, and compared the results with corresponding data previously obtained at 7 months of age. Using differential statistical analysis for individual genes, pathways, and gene regulatory networks, we identified sex-specific, sex-dimorphic, and sex-neutral changes, and looked at how they evolved over age. To validate the most robust findings across distinct mouse models and species, the results were compared with cortical scRNA-seq data from the transgenic hAPP-based Tg2576 mouse model and human AD. RESULTS We identified several significant sex-specific and sex-dimorphic differentially expressed genes in neurons, microglia, astrocytes and oligodendrocytes, including both cross-sectional changes and alterations from 7 months to 17 months of age. Key pathways affected in a sex-dependent manner across age included neurotransmitter signaling, RNA processing and splicing, stress response pathways, and protein degradation pathways. In addition, network analysis revealed the AD-associated genes Clu, Mbp, Fos and Junb as relevant regulatory hubs. Analysis of age-dependent changes highlighted genes and pathways associated with inflammatory response (Malat1, Cx3cr1), protein homeostasis (Cst3), and myelin maintenance (Plp1, Cldn11, Mal) that showed consistent sex-dependent changes as the THY-Tau22 mice aged. Multiple genes with established implications in AD, including the long non-coding RNA gene Malat1, displayed concordant sex-specific changes in mouse models and human AD. CONCLUSIONS This study provides a comprehensive single-cell transcriptomic characterization of sex-linked and age-dependent changes in the THY-Tau22 tauopathy model, revealing new insights into the interplay between age-dependent AD-like pathologies and sex. The identified sex-specific changes and their conservation across models and human AD highlight molecular targets for further preclinical investigation of sex-specific therapeutic strategies in AD.
Collapse
Affiliation(s)
- Muhammad Ali
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg
| | - Pierre Garcia
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg
| | - Laetitia P Lunkes
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg
| | - Alessia Sciortino
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg
| | - Melanie H Thomas
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg
| | - Tony Heurtaux
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, 6 Avenue du Swing, Belvaux, L-4367, Luxembourg
| | - Kamil Grzyb
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg
| | - Rashi Halder
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg
| | - Luc Buée
- Lille Neuroscience & Cognition, University of Lille, Inserm, CHU Lille, Alzheimer & Tauopathies, LabEx DISTALZ, Lille, UMR-S1172, France
| | - David Blum
- Lille Neuroscience & Cognition, University of Lille, Inserm, CHU Lille, Alzheimer & Tauopathies, LabEx DISTALZ, Lille, UMR-S1172, France
| | - Manuel Buttini
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg.
| |
Collapse
|
6
|
Li S, Liu Y, Wu X, Pan M, Zhao H, Hong Y, Zhang Q, Hu S, Ouyang A, Li G, Wu M, Fan S, Jia Z, Zhao S, Wu G, Gao X, Yang Z, Chen Z. The m 5C methyltransferase NSUN2 promotes progression of acute myeloid leukemia by regulating serine metabolism. Cell Rep 2025; 44:115661. [PMID: 40343793 DOI: 10.1016/j.celrep.2025.115661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/22/2025] [Accepted: 04/15/2025] [Indexed: 05/11/2025] Open
Abstract
Acute myeloid leukemia (AML) is one of the most prevalent heterogeneous hematologic malignancies with a complicated etiology. RNA post-transcriptional modifications have been linked to the incidence and progression of AML, while the detailed mechanism remains to be elucidated. In this study, we find that NOP2/Sun domain family member 2 (NSUN2), a methyltransferase of 5-methylcytosine (m5C) RNA methylation, is upregulated in AML and predicts a poor prognosis for patients with AML. Knockdown of NSUN2 in AML cells inhibits proliferation and colony formation and promotes apoptosis. Depletion of NSUN2 in AML mice reduces the tumor burden and prolongs survival. Mechanistically, NSUN2 promotes the expression of phosphoglycerate dehydrogenase (PHGDH) and serine hydroxymethyltransferase 2 (SHMT2), two key enzymes in the serine/glycine biosynthesis pathway, by stabilizing the corresponding mRNAs through regulation of m5C modifications. Overall, our findings demonstrate a critical role of NSUN2 in AML development and highlight the therapeutic potential of targeting the NSUN2/m5C axis for the treatment of this cancer.
Collapse
Affiliation(s)
- Songyu Li
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang 524000, China; Department of Hematology, Central People's Hospital of Zhanjiang, Zhanjiang 524000, China; Zhanjiang Key Laboratory of Leukemia Pathogenesis and Targeted Therapy Research, Zhanjiang 524000, China
| | - Ya Liu
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang 524000, China
| | - Xiang Wu
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang 524000, China
| | - Minjia Pan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hongxia Zhao
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang 524000, China
| | - Yunguang Hong
- Department of Hematology, Central People's Hospital of Zhanjiang, Zhanjiang 524000, China; Zhanjiang Key Laboratory of Leukemia Pathogenesis and Targeted Therapy Research, Zhanjiang 524000, China
| | - Qinghua Zhang
- Department of Radiation Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Shushu Hu
- Department of Radiation Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Aorong Ouyang
- Department of Urology, Maoming People's Hospital, Maoming 525000, China
| | - Guangru Li
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang 524000, China
| | - Minhui Wu
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang 524000, China
| | - Shanshan Fan
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang 524000, China
| | - Zhirong Jia
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang 524000, China
| | - Shanchao Zhao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Guocai Wu
- Department of Hematology, Central People's Hospital of Zhanjiang, Zhanjiang 524000, China
| | - Xiangwei Gao
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhigang Yang
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang 524000, China; Department of Hematology, Central People's Hospital of Zhanjiang, Zhanjiang 524000, China; Zhanjiang Key Laboratory of Leukemia Pathogenesis and Targeted Therapy Research, Zhanjiang 524000, China
| | - Zhanghui Chen
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang 524000, China.
| |
Collapse
|
7
|
Yuan Z, Fang K, Miao X, Zhang Y, Gu M, Xu W, Li H, Zhu D, Zhou J, Sun J, Gu X. Investigating the mechanisms by which low NAT1 expression in tumor cells contributes to chemo-resistance in colorectal cancer. Clin Epigenetics 2025; 17:77. [PMID: 40329330 PMCID: PMC12053866 DOI: 10.1186/s13148-025-01882-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND In the therapeutic landscape of colorectal cancer (CRC), chemo-resistance poses a significant and prevalent obstacle that complicates treatment efficacy and patient outcomes. Over time, cancer cells can develop mechanisms to resist the toxic effects of chemo-therapy drugs, leading to reduced sensitivity or complete insensitivity to these agents. The enzyme Arylamine N-acetyltransferase 1 (NAT1) has emerged as a promising target in strategies aimed at overcoming this challenge. NAT1 is involved in the metabolism of various xenobiotics, including some chemotherapeutic agents. Understanding the complex interactions between NAT1 and chemotherapeutic agents, as well as the molecular mechanisms underlying chemo-resistance, is crucial for the development of novel therapeutic approaches. OBJECTIVE This study aimed to assess the role of NAT1 in mediating chemo-resistance in CRC, with the goal of identifying novel strategies to overcome this clinical challenge. METHODS We conducted a comprehensive analysis using various bioinformatics tools and in vitro experiments to evaluate the effect of NAT1 expression on chemo-resistance in CRC. Furthermore, we employed a multi-omics approach, including metabolomics and next-generation sequencing, to uncover the mechanisms by which NAT1 influences chemo-resistance. Additionally, we utilized single-cell RNA sequencing (scRNA-seq), the Cellchat assay, and western blot to explore the intercellular communication between tumor and endothelial cells in the context of anti-PD-1 therapy and NAT1's impact. RESULTS Our study reveals that decreased NAT1 expression in CRC tumor tissues, relative to adjacent normal tissues, is significantly associated with a poorer patient prognosis. Experimental data indicate that silencing NAT1 in CaCO2 and HCT116 cell lines results in heightened resistance to five chemotherapeutic agents: vinblastine, docetaxel, gemcitabine, vincristine, and daporinad. Additionally, NAT1 silencing increases the proportion of LGR5+ cells, which are known to be chemo-resistant. Our research further revealed that exposure to these five drugs induces a decrease in NAT1 expression within CRC cells. Mechanistic insights show that NAT1 knockdown triggers a metabolic reprogramming in CRC cells, shifting from oxidative phosphorylation and the tricarboxylic acid cycle to a preference for glycolysis. Furthermore, silencing of NAT1 in CRC cells leads to an up-regulation of VEGFA expression. Notably, the application of anti-PD-1 therapy was demonstrated to significantly disrupt the VEGFA-VEGFR axis signaling, an interaction critical between CRC cells and endothelial cells. This discovery underscores the potential of targeting the VEGFA pathway as a therapeutic approach to mitigate the adverse effects associated with NAT1 down-regulation in CRC. CONCLUSION Our study underscores the multifaceted role of NAT1 in modulating chemo-sensitivity, cellular metabolism, and angiogenesis in CRC. These findings position NAT1 as a compelling candidate for a biomarker and a potential therapeutic target, offering new avenues for CRC management.
Collapse
Affiliation(s)
- Zheng Yuan
- Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China
| | - Kai Fang
- College of Basic Medical Sciences, Suzhou University, Suzhou, China
| | - Xinsheng Miao
- Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China
| | - Yan Zhang
- Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China
| | - Menghui Gu
- Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China
| | - Wei Xu
- Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China
| | - Hao Li
- Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China
| | - Dawei Zhu
- Nanjing Medical University, Suzhou, China
| | - Jiahui Zhou
- Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China.
| | - Jian Sun
- Affiliated Suzhou Hospital of Nanjing Medical University, Center for Reproduction and Genetics of Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China.
| | - Xinhua Gu
- Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China.
| |
Collapse
|
8
|
Yuan Z, Fang K, Miao X, Zhang Y, Gu M, Xu W, Li H, Zhu D, Zhou J, Sun J, Gu X. Investigating the mechanisms by which low NAT1 expression in tumor cells contributes to chemo-resistance in colorectal cancer. Clin Epigenetics 2025; 17:77. [DOI: doi.org/10.1186/s13148-025-01882-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 04/11/2025] [Indexed: 05/20/2025] Open
|
9
|
Liu JG, Yu L, Guo XL, He XM, Li M, Gao RY, Zhao BH, Li QY, Zhu WJ, Xu P, Gu XH, Chen YA, Yin XL, Shang Y, Guo ZH, Mao JH, Hu YX, Lu LM, Hua J, Zhang H, Li Y. Characterizing the immune landscape of tumor-infiltrating lymphocytes in non-small cell lung cancer. Genes Immun 2025:10.1038/s41435-025-00330-w. [PMID: 40325180 DOI: 10.1038/s41435-025-00330-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/11/2025] [Accepted: 04/25/2025] [Indexed: 05/07/2025]
Abstract
Tumor-Infiltrating Lymphocytes (TILs) immunotherapy is a highly promising treatment for Non-small Cell Lung Cancer (NSCLC), which is responsible for 18% of all cancer-related deaths. The heterogeneity of TILs remains poorly understood. Here, we utilized combined single-cell RNA (scRNA)/T cell receptor sequencing (scTCR-seq) data from lung adenocarcinoma (LUAD) patients. Naïve CD4+ and effector memory CD8+ T cells were increased in tumor tissue compared with circulating blood samples. Activated signaling pathways were detected, and GZMA was identified as a potential novel diagnostic biomarker. During the transitional phase, macrophages (FTL) and dendritic (AIF1) cells transported the most CD3 TCR clones to T cells, while cytotoxicity CD8+ T (NKG7) cells transported to terminal exhausted CD8+ T cells. In both transition and expansion phases, T helper cells (CXCL13) are transported to regulatory T cells (Tregs). Additionally, we investigated the expression profiles of key cytokines, checkpoint receptors, and their ligands. Cytotoxicity CD8+ T cells (CCL5 and IFNG), T helper cells (FTL, TNFRSF4, and TIGIT), and regulatory T cells (CTLA4, TIGIT and FTL) exhibited functional roles in both primary and metastatic tumor stages. Taken together, our study provides a single-cell resolution of the TIL immune landscape and suggests potential treatment strategies to overcome drug resistance.
Collapse
Affiliation(s)
- Jin-Guo Liu
- Department of Oncology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Lin Yu
- Department of Pathology, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Xian-Ling Guo
- Department of Oncology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Xue-Min He
- Department of Oncology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Man Li
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Ren-Yuan Gao
- Department of Abdominal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Bing-Hui Zhao
- Department of Radiology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Qian-Yu Li
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Wen-Jing Zhu
- Department of Orthopedics, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Ping Xu
- Standard BioTools, Shanghai, China
| | - Xiao-Hua Gu
- Department of Interventional Therapy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yong-An Chen
- Department of Oncology, No. 455 Hospital of Chinese People's Liberation Army, The Navy Medical University, Shanghai, China
| | - Xiao-Lan Yin
- Department of Oncology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Yan Shang
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Zhen-Hong Guo
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, China
| | - Jia-Hao Mao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Yang-Xi Hu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Li-Ming Lu
- Central Laboratory, Shanghai Chest Hospital and Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Hua
- SPH Biotherapeutics (Shanghai) Limited, Cellular Therapeutics Center for Cancers, Shanghai, China
| | - Hua Zhang
- SPH Biotherapeutics (Shanghai) Limited, Cellular Therapeutics Center for Cancers, Shanghai, China
| | - Yue Li
- SPH Biotherapeutics (Shanghai) Limited, Cellular Therapeutics Center for Cancers, Shanghai, China.
| |
Collapse
|
10
|
Azim S, Rousselle T, Zubair H, Shetty AC, Archer KJ, Marshall JN, Rajabi A, Lara CM, Mustofa S, Drachenberg C, Bromberg J, Menon M, Maluf DG, Akalin E, Mas VR. Epithelial-Immune-Stromal Interactions Define Divergent Repair and Fibrosis Pathways After Acute Kidney Injury in Human Renal Transplants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.30.651080. [PMID: 40364910 PMCID: PMC12073942 DOI: 10.1101/2025.04.30.651080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Acute kidney injury (AKI) is a major cause of early graft dysfunction after kidney transplantation, particularly in recipients of high-risk donor kidneys prone to ischemia-reperfusion injury. However, the cellular mechanisms dictating whether injury resolves or progresses to fibrosis remain unclear. This study combines single-nucleus RNA sequencing and imaging mass cytometry (IMC) analysis of human kidney allograft biopsies collected within eight weeks posttransplant, stratified by long-term functional outcomes. Grafts that recovered function were enriched in regenerative proximal tubular (PT) cells co-expressing PROM1, CD24, and injury markers, consistent with scattered tubular cells (STCs). In contrast, non-recovering grafts contained a unique subpopulation of transitional proximal tubule cells (tPT4) characterized by dedifferentiation, loss of epithelial identity, and acquisition of fibroblast-like features. Fibroblast trajectory analysis revealed a profibrotic lineage, progressing from stromal progenitors to myofibroblasts, exclusive to nonrecovery grafts. Immune profiling showed divergent macrophage (MΦ) polarization, with reparative MΦ2 cells and regulatory dendritic cell (DC)-like signatures in recovering grafts, versus inflammatory MΦ1 and pro-fibrotic DCs in non-recovery. IMC confirmed spatial colocalization of injured tubules, activated fibroblasts, and immune cells in fibrotic regions, validated in an independent cohort. Functional assays demonstrated that ischemic epithelial injury activated monocyte-derived MΦs with mixed inflammatory/reparative profiles and induced fibroblast-related gene expression, while PAX8 knockdown impaired epithelial proliferation and promoted pro-inflammatory signaling. These findings reveal epithelial cell plasticity as a central driver of divergent repair outcomes following renal transplant AKI and highlight epithelial-immune-stromal crosstalk as a therapeutic target to promote recovery and prevent chronic graft injury. One Sentence Summary Single-cell and spatial mapping of human kidney transplants reveal regenerative and fibrotic cell programs across tubular, immune, and stromal compartments that determine whether acute injury resolves or progresses to chronic allograft injury.
Collapse
|
11
|
Guo Y, Ma J, Qi R, Ma R, Ma X, Xu J, Ye K, Huang Y, Yang X, Zhang J, Wang G, Zhao X. snCED-seq: high-fidelity cryogenic enzymatic dissociation of nuclei for single-nucleus RNA-seq of FFPE tissues. Nat Commun 2025; 16:4101. [PMID: 40316516 PMCID: PMC12048618 DOI: 10.1038/s41467-025-59464-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 04/22/2025] [Indexed: 05/04/2025] Open
Abstract
Recent advances have shown that single-nucleus RNA sequencing (snRNA-seq) can be applied to formalin-fixed, paraffin-embedded (FFPE) tissues, opening avenues for transcriptomic analysis of archived specimens. Yet, isolating intact nuclei remains difficult due to RNA cross-linking. Here, we introduce a cryogenic enzymatic dissociation (CED) strategy for rapid, high-yield and fidelity nuclei extraction from FFPE samples and validate its utility with snRandom-seq (snCED-seq) using male C57/BL6 mice. Compared with conventional approaches, CED delivers a tenfold increase in nuclei yield with significantly reduced hands-on time, while minimizing secondary RNA degradation and preserving intranuclear transcripts. snCED-seq enhances gene detection sensitivity, lowers mitochondrial and ribosomal contamination, and increases overall gene expression quantification. In Alzheimer's disease studies, it distinguished two astrocyte subpopulations, microglia, and oligodendrocytes, revealing cellular heterogeneity. Additionally, snCED-seq identify major cell types in a single 50 μm FFPE human lung section. Our results demonstrate that snCED-seq is robust for FFPE specimens and poised to enable multi-omics analyses of clinical samples.
Collapse
Affiliation(s)
- Yunxia Guo
- State Key Laboratory of Digital Medical Engineering, School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
- Department of Anesthesiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Junjie Ma
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruicheng Qi
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rongrong Ma
- Department of Anesthesiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Xiaoying Ma
- State Key Laboratory of Digital Medical Engineering, School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Jitao Xu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Kaiqiang Ye
- State Key Laboratory of Digital Medical Engineering, School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Yan Huang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Xi Yang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Jianyou Zhang
- Department of Anesthesiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China.
| | - Guangzhong Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Xiangwei Zhao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science & Medical Engineering, Southeast University, Nanjing, China.
| |
Collapse
|
12
|
Liu Y, Li C, Shen LC, Yan H, Wei G, Gasser RB, Hu X, Song J, Yu DJ. scRCA: A Siamese network-based pipeline for annotating cell types using noisy single-cell RNA-seq reference data. Comput Biol Med 2025; 190:110068. [PMID: 40158457 DOI: 10.1016/j.compbiomed.2025.110068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
Accurate cell type annotation is fundamentally critical for single-cell sequencing (scRNA-seq) data analysis to provide insightful knowledge of tissue-specific cell heterogeneity and cell state transition tracking. Cell type annotation is usually conducted by comparative analysis with known data (i.e., reference) - which contains a presumably accurate representation of cell types. However, this assumption is often problematic, as factors such as human errors in wet-lab experiments and methodological limitations can introduce annotation errors in the reference dataset. As current pipelines for single-cell transcriptomic analysis do not adequately consider this challenge, there is a major demand for constructing a computational pipeline that achieves high-quality cell type annotation using reference datasets containing inherent errors (referred to as "noise" in this study). Here, we built a Siamese network-based pipeline, termed scRCA, to accurately annotate cell types based on noisy reference data. To help users evaluate the reliability of scRCA annotations, an interpreter was also developed to explore the factors underlying the model's predictions. Our experiments demonstrate that, across 14 datasets, scRCA outperformed other widely adopted reference-based methods for cell type annotation. Using an independent dataset of four multiple myeloma patients, we further illustrated that scRCA can distinguish cancerous cells based on gene expression levels and identify genes closely associated with multiple myeloma through scRCA's interpretable module, providing significant information for subsequent clinical treatments. With these advancements, we anticipate that scRCA will serve as a practical reference-based approach for accurate annotating cell type annotation.
Collapse
Affiliation(s)
- Yan Liu
- Department of Computer Science, Yangzhou University, Yangzhou, 225100, China
| | - Chen Li
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, 3800, Australia
| | - Long-Chen Shen
- School of Computer Science and Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei, Nanjing, 210094, China
| | - He Yan
- School of Computer Science and Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei, Nanjing, 210094, China
| | - Guo Wei
- School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Robin B Gasser
- Monash Data Futures Institute, Monash University, Melbourne, Victoria, 3800, Australia
| | - Xiaohua Hu
- Information Department, The First Affiliated Hospital of Naval Military Medical University, Changhai Road 168, Shanghai, 200433, China
| | - Jiangning Song
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, 3800, Australia; Monash Data Futures Institute, Monash University, Melbourne, Victoria, 3800, Australia.
| | - Dong-Jun Yu
- School of Computer Science and Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei, Nanjing, 210094, China.
| |
Collapse
|
13
|
Ye W, Shi M, Cheng Y, Liu Y, Ren K, Fang Y, Younas W, Zhang W, Wang Y, Xia XQ. Integrated single-cell transcriptome and comparative genome analysis reveals the origin of intermuscular bones in zebrafish. Int J Biol Macromol 2025; 308:142397. [PMID: 40127795 DOI: 10.1016/j.ijbiomac.2025.142397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 03/26/2025]
Abstract
The evolutionary process of intermuscular bones (IBs) is complex, the molecular regulatory mechanisms of their development are not clear, and even the genes involved in the evolution and development of IBs are poorly understood. In this study, comparative genomic analysis of four fish species with IBs and eleven fish species without IBs identified 106 genes that are more conservatively evolved in fish species with IBs, but highly variable in fish species without IBs. These genes are mainly involved in swimming behavior and BMP signaling pathways. We performed single-cell transcriptome sequencing of IBs origin tissues in zebrafish before and after IBs formation and found that osteoblasts and mesenchymal stem cells (MSCs) increased significantly after IBs formation. RNA velocity analysis showed that osteoblasts in IBs differentiate from MSCs, and the differentiation trajectory of MSCs into osteoblasts was successfully constructed by pseudo-time analysis. Combined with the results of multi-omics analysis, seven candidate genes associated with IBs development were screened and knocked out in zebrafish. It was found that foxn3 mutation resulted in a delay in IB development, whereas bmp6 mutation resulted in a total loss of IB. By comparing the transcriptome of IBs tissues between bmp6+/+ zebrafish and bmp6-/- zebrafish, we found that bmp6 deletion may inhibit the differentiation of MSCs into osteoblasts while promoting the formation of osteoclasts and ultimately inhibiting the formation of IBs. This study provides new insights into the molecular regulatory mechanisms and evolutionary processes of IB development.
Collapse
Affiliation(s)
- Weidong Ye
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture and Rural Affairs, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; Department of Vascular Surgery, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China
| | - Mijuan Shi
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture and Rural Affairs, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yingyin Cheng
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture and Rural Affairs, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuhang Liu
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture and Rural Affairs, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
| | - Keyi Ren
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture and Rural Affairs, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
| | - Yutong Fang
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture and Rural Affairs, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Waqar Younas
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture and Rural Affairs, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wanting Zhang
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture and Rural Affairs, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaping Wang
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture and Rural Affairs, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao-Qin Xia
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture and Rural Affairs, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
14
|
Luo X, Deng H, Li Q, Zhao M, Zhang Y, Guo J, Wen Y, Chen G, Li J. Bulk transcriptome and single-nucleus RNA sequencing analyses highlight the role of recombination activating 1 in non-alcoholic fatty liver disease. Int J Biol Macromol 2025; 307:141919. [PMID: 40074128 DOI: 10.1016/j.ijbiomac.2025.141919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 03/07/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a prevalent chronic condition with an incompletely understood pathogenesis. In this study, five candidate genes-RAG1, CKAP2, CENPK, TYMS, and BUB1-were identified as being associated with NAFLD progression through integrative bioinformatics analyses. A predictive model incorporating these genes demonstrated strong robustness and diagnostic accuracy. Single-nucleus RNA sequencing analysis further revealed that RAG1 plays a potential role in hepatocytes of NAFLD patients. Functional experiments using RNA interference to suppress RAG1 expression in HepG2 cells treated with oleic and palmitic acids showed reduced total glyceride and cholesterol levels, mitigated lipid accumulation, and alterations in pathways related to lipid metabolism, inflammation, and fibrosis. Furthermore, adeno-associated virus-specific knockdown of RAG1 in hepatocytes attenuated hepatic steatosis in high-fat diet-fed mice. These findings suggest that investigating the molecular mechanisms of hub genes like RAG1 may advance our understanding of NAFLD pathogenesis and inform therapeutic development.
Collapse
Affiliation(s)
- Xiaohua Luo
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Hongbo Deng
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Qiang Li
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Miao Zhao
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Science, Central South University, 410078 Changsha, China
| | - Yu Zhang
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Junjie Guo
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Yifan Wen
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Guangshun Chen
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China.
| | - Jiequn Li
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China.
| |
Collapse
|
15
|
Li T, Wang Z, Liu Y, He S, Zou Q, Zhang Y. An overview of computational methods in single-cell transcriptomic cell type annotation. Brief Bioinform 2025; 26:bbaf207. [PMID: 40347979 PMCID: PMC12065632 DOI: 10.1093/bib/bbaf207] [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: 12/11/2024] [Revised: 03/14/2025] [Accepted: 04/01/2025] [Indexed: 05/14/2025] Open
Abstract
The rapid accumulation of single-cell RNA sequencing data has provided unprecedented computational resources for cell type annotation, significantly advancing our understanding of cellular heterogeneity. Leveraging gene expression profiles derived from transcriptomic data, researchers can accurately infer cell types, sparking the development of numerous innovative annotation methods. These methods utilize a range of strategies, including marker genes, correlation-based matching, and supervised learning, to classify cell types. In this review, we systematically examine these annotation approaches based on transcriptomics-specific gene expression profiles and provide a comprehensive comparison and categorization of these methods. Furthermore, we focus on the main challenges in the annotation process, especially the long-tail distribution problem arising from data imbalance in rare cell types. We discuss the potential of deep learning techniques to address these issues and enhance model capability in recognizing novel cell types within an open-world framework.
Collapse
Affiliation(s)
- Tianhao Li
- School of Computer Science, Chengdu University of Information Technology, No. 24 Block 1, Xuefu Road, 610225 Chengdu, China
| | - Zixuan Wang
- College of Electronics and Information Engineering, Sichuan University, No. 24 South Section 1, 1st Ring Road, 610065 Chengdu, China
| | - Yuhang Liu
- Faculty of Applied Sciences, Macao Polytechnic University, 999078 Macao, China
| | - Sihan He
- School of Computer Science, Chengdu University of Information Technology, No. 24 Block 1, Xuefu Road, 610225 Chengdu, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Shahe Campus: No. 4, Section 2, North Jianshe Road, 611731 Chengdu, China
| | - Yongqing Zhang
- School of Computer Science, Chengdu University of Information Technology, No. 24 Block 1, Xuefu Road, 610225 Chengdu, China
| |
Collapse
|
16
|
Ushakumary MG, Feng S, Bandyopadhyay G, Olson H, Weitz KK, Huyck HL, Poole C, Purkerson JM, Bhattacharya S, Ljungberg MC, Mariani TJ, Deutsch GH, Misra RS, Carson JP, Adkins JN, Pryhuber GS, Clair G. Cell Population-resolved Multiomics Atlas of the Developing Lung. Am J Respir Cell Mol Biol 2025; 72:484-495. [PMID: 39447176 PMCID: PMC12051933 DOI: 10.1165/rcmb.2024-0105oc] [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] [Accepted: 10/24/2024] [Indexed: 10/26/2024] Open
Abstract
The lung is a vital organ that undergoes extensive morphological and functional changes during postnatal development. To disambiguate how different cell populations contribute to organ development, we performed proteomic and transcriptomic analyses of four sorted cell populations from the lung of human subjects 0-8 years of age with a focus on early life. The cell populations analyzed included epithelial, endothelial, mesenchymal, and immune cells. Our results revealed distinct molecular signatures for each of the sorted cell populations that enable the description of molecular shifts occurring in these populations during postnatal development. We confirmed that the proteome of the different cell populations was distinct regardless of age and identified functions specific to each population. We identified a series of cell population protein markers, including those located at the cell surface, that show differential expression and distribution on RNA in situ hybridization and immunofluorescence imaging. We validated the spatial distribution of alveolar type 1 and endothelial cell surface markers. Temporal analyses of the proteomes of the four populations revealed processes modulated during postnatal development and clarified the findings obtained from whole-tissue proteome studies. Finally, the proteome was compared with a transcriptomics survey performed on the same lung samples to evaluate processes under post-transcriptional control.
Collapse
Affiliation(s)
- Mereena G. Ushakumary
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Song Feng
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Gautam Bandyopadhyay
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Heather Olson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Karl K. Weitz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Heidi L. Huyck
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Cory Poole
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Jeffrey M. Purkerson
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Soumyaroop Bhattacharya
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - M. Cecilia Ljungberg
- Department of Pediatrics, College of Medicine, Baylor University, Houston, Texas
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas
| | - Thomas J. Mariani
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Gail H. Deutsch
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Ravi S. Misra
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - James P. Carson
- Texas Advanced Computing Center, University of Texas at Austin, Austin, Texas; and
| | - Joshua N. Adkins
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - Gloria S. Pryhuber
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Geremy Clair
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| |
Collapse
|
17
|
Xie R, Li C, Yun J, Zhang S, Zhong A, Cen Y, Li Z, Chen J. Identifying the Pattern Characteristics of Anoikis-Related Genes in Keloid. Adv Wound Care (New Rochelle) 2025; 14:223-237. [PMID: 38775414 DOI: 10.1089/wound.2024.0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024] Open
Abstract
Objective: Anoikis is a kind of programmed cell death that is triggered when cells lose contact with each other or with the matrix. However, the potential value of anoikis-related genes (ARGs) in keloid (KD) has not been investigated. Approach: We downloaded three keloid fibroblast (KF) RNA sequencing (RNA-seq) datasets from the Gene Expression Omnibus (GEO) and obtained 338 ARGs from a search of the GeneCards database and PubMed articles. Weighted correlation network analysis was used to construct the coexpression network and obtain the KF-related ARGs. The LASSO-Cox method was used to screen the hub ARGs and construct the best prediction model. Then, GEO single-cell sequencing datasets were used to verify the expression of hub genes. We used whole RNA-seq for gene-level validation and the correlation between KD immune infiltration and anoikis. Results: Our study comprehensively analyzed the role of ARGs in KD for the first time. The least absolute shrinkage and selection operator (LASSO) regression analysis identified six hub ARGs (HIF1A, SEMA7A, SESN1, CASP3, LAMA3, and SIK2). A large number of miRNAs participate in the regulation of hub ARGs. In addition, correlation analysis revealed that ARGs were significantly correlated with the infiltration levels of multiple immune cells in patients with KD. Innovation: We explored the expression characteristics of ARGs in KD, which is extremely important for determining the molecular pathways and mechanisms underlying KD. Conclusions: This study provides a useful reference for revealing the characteristics of ARGs in the pathogenesis of KD. The identified hub genes may provide potential therapeutic targets for patients. This study provides new ideas for individualized therapy and immunotherapy.
Collapse
Affiliation(s)
- Ruxin Xie
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Chenyu Li
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Jiao Yun
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Shiwei Zhang
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Ai Zhong
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Ying Cen
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Zhengyong Li
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Junjie Chen
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Jiang Y, Ye D, Zhou Y. An integrated analysis of scRNA-seq and RNA-seq data revealed metastasis-related regulators as prognostic indicators in lung adenocarcinoma. J Thorac Dis 2025; 17:2473-2491. [PMID: 40400936 PMCID: PMC12090110 DOI: 10.21037/jtd-2025-482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 04/16/2025] [Indexed: 05/23/2025]
Abstract
Background The incidence and mortality rates of lung cancer are exceptionally high. Many patients are diagnosed with early stage lung cancer but experience rapid recurrence post-surgery. Many research studies have shown that the unfavorable prognosis of patients may be associated with micro-metastasis in the lymph nodes. Our research aimed to develop a nomogram to predict the prognosis of lung adenocarcinoma (LUAD). Methods Single-cell RNA sequencing (scRNA-seq) data were analyzed to identify 11 cell clusters. Patterns of incoming and outgoing signals were identified across the entire cell population. A weighted gene co-expression network analysis (WGCNA) was conducted to uncover critical genes in LUAD. The intersecting marker genes were used to construct the prognostic model. Results scRNA-seq data were analyzed to identify 19 cell clusters. We identified 3,464 marker genes from the scRNA-seq dataset, 1,994 differentially expressed genes from the bulk RNA sequencing (RNA-seq) dataset, and 1,863 genes associated with a key module identified by the WGCNA. After performing the intersection, univariate Cox, and least absolute shrinkage and selection operator analyses, a prognostic model was established based on the expression levels of 13 signature genes. Subsequent functional experiments confirmed the role of selected regulated genes. Conclusions Through the integration of scRNA-seq data and bulk RNA-seq data, we developed an innovative model to predict the prognosis of patients. The risk score was found to be a significant independent predictor and clinical-pathological features of LUAD.
Collapse
Affiliation(s)
- Yang Jiang
- Department of Thoracic Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Danrong Ye
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yongxin Zhou
- Department of Thoracic Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
19
|
Ying Y, Lin J, Gao W, Yue L, Zeng Q, Bartas K, Cheong D, Jiang H, Zheng Z, Shi L, Ping A, Fang Y, Yan F, Guo T, Zhang J, Wu H, Beier K, Zhu J, Zhu Z. Proteomic profiling in cerebrospinal fluid reveal biomarkers for shunt outcome in idiopathic normal-pressure hydrocephalus. J Adv Res 2025:S2090-1232(25)00287-5. [PMID: 40311753 DOI: 10.1016/j.jare.2025.04.043] [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: 09/25/2024] [Revised: 04/08/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND The pathophysiology of idiopathic normal pressure hydrocephalus (iNPH) remains unclear, and the treatment strategy remains suboptimal. This study aims to identify biomarkers for shunt prognosis by cerebrospinal fluid (CSF) proteomic profiling. METHODS CSF samples collected from 37 iNPH patients from the discovery cohort and 12 iNPH patients from an independent validation cohort (71.9 ± 6.1 years (mean ± SD)), and 16 age-balanced controls (69.9 ± 7.6 years (mean ± SD)) were collected from September 2020 to December 2023. 53 CSF samples were analyzed using a mass spectrometry-based proteomic workflow. Clinical evaluations were performed on all iNPH patients, and 44 patients underwent ventriculoperitoneal shunting. Postoperative CSF were also collected from 10 iNPH patients who underwent shunting surgery. Bioinformatics, machine learning, and enzyme-linked immunosorbent assay (ELISA) were performed to identify CSF proteome changes related to pathophysiology in iNPH, and screen for biomarkers associated with shunt response. RESULTS 39 and 285 proteins significantly increased and decreased in iNPH CSF compared to the control group. Gene ontology analysis revealed that the noticeably increased proteins were mainly associated with myeloid leukocyte migration and extracellular matrix organization, and significantly decreased proteins were primarily associated with axon development and synapse organization. Machine learning identified 6 candidate biomarkers that potentially predicted the response to shunt surgery. Among these, QPCT levels were found to be elevated in non-responders, while RBP4 levels were decreased, and both of these changes were validated through ELISA. CONCLUSIONS Our findings provide support for the hypothesis that the pathophysiology of iNPH is characterized by a state of neuroinflammation, extracellular matrix remodeling, and neurodegeneration, and CSF shunting can reverse such pathological state. Machine learning using preoperative proteomic profiles satisfactorily predicted the clinical outcome of the shunt procedure in iNPH. Future research targeting specific proteins in iNPH may be warranted to better comprehend the disease mechanism and design patient-tailored treatments.
Collapse
Affiliation(s)
- Yuqi Ying
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou 310009, China; Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; Department of Neurosurgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu 322000, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Jingquan Lin
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou 310009, China; Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Wei Gao
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou 310009, China; Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Liang Yue
- Affiliated Hangzhou First People's Hospital, State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, Zhejiang Province, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
| | - Qingze Zeng
- Department of Radiology, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou 310009, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Katrina Bartas
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA
| | - Dayeon Cheong
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA
| | - Hongjie Jiang
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou 310009, China; Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Zhe Zheng
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou 310009, China; Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Ligen Shi
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou 310009, China; Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - An Ping
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou 310009, China; Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Yuanjian Fang
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou 310009, China; Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Feng Yan
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou 310009, China; Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Tiannan Guo
- Affiliated Hangzhou First People's Hospital, State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, Zhejiang Province, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
| | - Jianmin Zhang
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou 310009, China; Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; Department of Neurosurgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu 322000, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China.
| | - Hemmings Wu
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou 310009, China; Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China.
| | - Kevin Beier
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, CA 92617, USA; Department of Biomedical Engineering, University of California, Irvine, CA 92617, USA; Department of Neurobiology and Behavior, University of California, Irvine, CA 92617, USA; Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA 92617, USA; UCI Mind, University of California, Irvine, CA 92617, USA.
| | - Junming Zhu
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou 310009, China; Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China.
| | - Zhoule Zhu
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou 310009, China; Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases of Zhejiang Province, Hangzhou 310009, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China.
| |
Collapse
|
20
|
Pan B, Chen S, Wu H, Zhang X, Zhang Z, Ye D, Yao Y, Luo Y, Zhang X, Wang X, Tang N. Short-term starvation inhibits CD36 N-glycosylation and downregulates USP7 UFMylation to alleviate RBPJ-maintained T cell exhaustion in liver cancer. Theranostics 2025; 15:5931-5952. [PMID: 40365281 PMCID: PMC12068301 DOI: 10.7150/thno.110567] [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: 01/16/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
Rationale: Short-term starvation (STS) has been shown to enhance the sensitivity of tumors to chemotherapy while concurrently safeguarding normal cells from its detrimental side effects. Nonetheless, the extent to which STS relies on the anti-tumor immune response to impede the progression of hepatocellular carcinoma (HCC) remains uncertain. Methods: In this study, we employed mass cytometry, flow cytometry, immunoprecipitation, immunoblotting, CUT&Tag, RT-qPCR, and DNA pull-down assays to evaluate the relationship between STS and T-cell antitumor immunity in HCC. Results: We demonstrated that STS alleviated T cell exhaustion in HCC. This study elucidated the mechanism by which STS blocked CD36 N-glycosylation, leading to the upregulation of AMPK phosphorylation and the downregulation of USP7 UFMylation, thus enhancing ubiquitination and destabilized USP7. Consequently, diminished USP7 levels facilitated the ubiquitination and subsequent degradation of RBPJ, thereby inhibiting T cell exhaustion through the IRF4/TNFRSF1B axis. From a therapeutic standpoint, STS not only suppressed the growth of patient-derived orthotopic xenografts but also enhanced their sensitivity to immunotherapy. Conclusions: These findings uncovered a novel mechanism by which N-glycosylation participated in UFMylation/ubiquitination to regulate T cell exhaustion, and we underscored the potential of targeting USP7 and RBPJ in anti-tumor immunotherapy strategies.
Collapse
Affiliation(s)
- Banglun Pan
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Siyan Chen
- Department of Laboratory Medicine, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Hao Wu
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Xiaoxia Zhang
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Zhu Zhang
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Dongjie Ye
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Yuxin Yao
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Yue Luo
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Xinyu Zhang
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Xiaoqian Wang
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Cancer Center of Fujian Medical University, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Nanhong Tang
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Cancer Center of Fujian Medical University, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Key Laboratory of Clinical Laboratory Technology for Precision Medicine (Fujian Medical University), Fujian Province University; Fuzhou 350122, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou 350122, China
| |
Collapse
|
21
|
Tao Z, Zou P, Yang Z, Xiong T, Deng Z, Chen Q. Single-cell multi-omics elucidates the role of RPS27-RPS24 fusion gene in osteosarcoma chemoresistance and metabolic regulation. Cell Death Discov 2025; 11:197. [PMID: 40280903 PMCID: PMC12032165 DOI: 10.1038/s41420-025-02487-9] [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: 06/08/2024] [Revised: 03/03/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Osteosarcoma (OS) presents significant treatment challenges due to chemoresistance. This study explores the molecular mechanisms underlying chemoresistance in OS, focusing on the novel fusion gene RPS27-RPS24. Using single-cell multi-omics techniques, we identified a significant upregulation of RPS27-RPS24 in chemoresistant OS cells. Our analyses revealed that RPS27-RPS24 enhances glutaminase (GLS)-mediated glutamine metabolism and inhibits copper-induced cell death, thereby promoting chemoresistance. In vitro experiments with adriamycin-resistant (ADMR) OS cells confirmed that overexpression of RPS27-RPS24 leads to increased cell viability and proliferation under chemotherapy. In vivo studies further validated these findings, demonstrating that targeting glutamine metabolism can reverse chemoresistance. Our results suggest that the RPS27-RPS24 fusion gene plays a critical role in OS chemoresistance through metabolic reprogramming, providing a potential therapeutic target for improving OS treatment outcomes. The application of multiple analytical techniques in this study (as shown in the upper image) and the hypothesized mechanism (as shown in the lower image).
Collapse
Affiliation(s)
- Zhiwei Tao
- Bone and Soft Tissue Sarcoma Department, Jiangxi Cancer Hospital, 330029, Nanchang, P.R. China.
| | - Pingan Zou
- Bone and Soft Tissue Sarcoma Department, Jiangxi Cancer Hospital, 330029, Nanchang, P.R. China
| | - Zhengxu Yang
- Bone and Soft Tissue Sarcoma Department, Jiangxi Cancer Hospital, 330029, Nanchang, P.R. China
| | - Tao Xiong
- Bone and Soft Tissue Sarcoma Department, Jiangxi Cancer Hospital, 330029, Nanchang, P.R. China
| | - Zhi Deng
- Bone and Soft Tissue Sarcoma Department, Jiangxi Cancer Hospital, 330029, Nanchang, P.R. China
| | - Qincan Chen
- Bone and Soft Tissue Sarcoma Department, Jiangxi Cancer Hospital, 330029, Nanchang, P.R. China
| |
Collapse
|
22
|
Xia J, Zheng L, Zhang H, Fan Q, Liu H, Wang O, Yan H. Drug Resistance Analysis of Pancreatic Cancer Based on Universally Differentially Expressed Genes. Int J Mol Sci 2025; 26:3936. [PMID: 40362181 PMCID: PMC12071644 DOI: 10.3390/ijms26093936] [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: 02/14/2025] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
The high heterogeneity between patients can complicate the diagnosis and treatment of pancreatic ductal adenocarcinoma (PDAC). Here, we explored the association of universally differentially expressed genes (UDEGs) with resistance to chemotherapy and immunotherapy in the context of pancreatic cancer. In this work, sixteen up-regulated and three down-regulated genes that were dysregulated in more than 85% of 102 paired and 5% of 521 unpaired PDAC samples were identified and defined as UDEGs. A single-cell level analysis further validated the high expression levels of the up-UDEGs and the low levels of the down-UDEGs in cancer-related ductal cells, which could represent the malignant changes seen in pancreatic cancer. Based on a drug sensitivity analysis, we found that ANLN, GPRC5A and SERPINB5 are closely related to the resistance mechanism of PDAC, and their high expression predicted worse survival for PDAC patients. This suggests that targeting these genes could be a potential way to reduce drug resistance and improve survival. Based on the immune infiltration analysis, the abnormal expression of the UDEGs was found to be related to the formation of an immunosuppressive tumor microenvironment. In conclusion, these UDEGs are common features of PDAC and could be involved in the resistance of pancreatic cancer and might serve as novel drug targets to guide research into drug repurposing.
Collapse
Affiliation(s)
- Jie Xia
- School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, China;
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350100, China; (L.Z.); (H.Z.); (Q.F.); (H.L.); (O.W.)
| | - Linyong Zheng
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350100, China; (L.Z.); (H.Z.); (Q.F.); (H.L.); (O.W.)
| | - Huarong Zhang
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350100, China; (L.Z.); (H.Z.); (Q.F.); (H.L.); (O.W.)
| | - Qi Fan
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350100, China; (L.Z.); (H.Z.); (Q.F.); (H.L.); (O.W.)
| | - Hui Liu
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350100, China; (L.Z.); (H.Z.); (Q.F.); (H.L.); (O.W.)
| | - Ouxi Wang
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350100, China; (L.Z.); (H.Z.); (Q.F.); (H.L.); (O.W.)
| | - Haidan Yan
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou 350100, China; (L.Z.); (H.Z.); (Q.F.); (H.L.); (O.W.)
| |
Collapse
|
23
|
Wang S, Li C, Fan W, Chen T, Xu W, Hu X, Wu Z, Xiao Z, Lin G, Ma B, Cheng L. Neurotrophin-3/chitosan inhibits cuproptosis-related genes to enable functional recovery after spinal cord injury. Int J Biol Macromol 2025; 310:143403. [PMID: 40268016 DOI: 10.1016/j.ijbiomac.2025.143403] [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: 11/17/2024] [Revised: 04/10/2025] [Accepted: 04/19/2025] [Indexed: 04/25/2025]
Abstract
OBJECTIVES This study investigated the regulatory mechanisms of cuproptosis-related genes (CRGs) in spinal cord injury (SCI) and explored the therapeutic potential of neurotrophin-3 (NT3)-loaded chitosan in promoting functional recovery. METHODS We conducted integrated bulk RNA-seq and single-cell RNA-seq (scRNA-seq) analyses of mouse spinal cord tissue at various time points after SCI. The key CRGs were identified using differential expression analysis, weighted gene co-expression network analysis, and machine learning. The therapeutic effects of NT3-loaded chitosan were evaluated using animal models and molecular docking analysis. RESULTS We identified four key CRGs (Atp7a, Cp, Loxl2, and Pde3b) and three key transcription factors (C/EBPα, Stat6, and Runx1) that were upregulated post-SCI, promoting cuproptosis and neuroinflammation. NT3-loaded chitosan treatment significantly inhibited CRG expression and enhanced functional recovery in the animal models. Molecular docking analysis demonstrated binding interactions between chitosan and key CRGs, suggesting a potential mechanism for their therapeutic effects. CONCLUSIONS Our findings highlight the critical role of CRGs in SCI progression and the potential of NT3-loaded chitosan as a therapeutic strategy for inhibiting cuproptosis and promoting functional recovery. Future studies should focus on validating these findings in larger cohorts and exploring the detailed mechanisms by which NT3-loaded chitosan modulates CRG expression.
Collapse
Affiliation(s)
- Siqiao Wang
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai 200065, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China
| | - Chen Li
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai 200065, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Institute of Spinal and Spinal Cord Injury, Tongji University School of Medicine, Shanghai 200065, China; Shanghai Research Center for Spine and Spinal Cord Diseases, Tongji University School of Medicine, Shanghai 200065, China; Clinical Center for Brain and Spinal Cord Research, Tongji University School of Medicine, Shanghai 200065, China
| | - Wenyong Fan
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai 200065, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Institute of Spinal and Spinal Cord Injury, Tongji University School of Medicine, Shanghai 200065, China; Shanghai Research Center for Spine and Spinal Cord Diseases, Tongji University School of Medicine, Shanghai 200065, China; Clinical Center for Brain and Spinal Cord Research, Tongji University School of Medicine, Shanghai 200065, China
| | - Tao Chen
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai 200065, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Institute of Spinal and Spinal Cord Injury, Tongji University School of Medicine, Shanghai 200065, China; Shanghai Research Center for Spine and Spinal Cord Diseases, Tongji University School of Medicine, Shanghai 200065, China; Clinical Center for Brain and Spinal Cord Research, Tongji University School of Medicine, Shanghai 200065, China
| | - Wei Xu
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai 200065, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Institute of Spinal and Spinal Cord Injury, Tongji University School of Medicine, Shanghai 200065, China; Shanghai Research Center for Spine and Spinal Cord Diseases, Tongji University School of Medicine, Shanghai 200065, China; Clinical Center for Brain and Spinal Cord Research, Tongji University School of Medicine, Shanghai 200065, China
| | - Xiao Hu
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai 200065, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Institute of Spinal and Spinal Cord Injury, Tongji University School of Medicine, Shanghai 200065, China; Shanghai Research Center for Spine and Spinal Cord Diseases, Tongji University School of Medicine, Shanghai 200065, China; Clinical Center for Brain and Spinal Cord Research, Tongji University School of Medicine, Shanghai 200065, China
| | - Zhourui Wu
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai 200065, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Institute of Spinal and Spinal Cord Injury, Tongji University School of Medicine, Shanghai 200065, China; Shanghai Research Center for Spine and Spinal Cord Diseases, Tongji University School of Medicine, Shanghai 200065, China; Clinical Center for Brain and Spinal Cord Research, Tongji University School of Medicine, Shanghai 200065, China
| | - Zhihui Xiao
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai 200065, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China
| | - Gufa Lin
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai 200065, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Institute of Spinal and Spinal Cord Injury, Tongji University School of Medicine, Shanghai 200065, China; Shanghai Research Center for Spine and Spinal Cord Diseases, Tongji University School of Medicine, Shanghai 200065, China; Clinical Center for Brain and Spinal Cord Research, Tongji University School of Medicine, Shanghai 200065, China.
| | - Bei Ma
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai 200065, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Institute of Spinal and Spinal Cord Injury, Tongji University School of Medicine, Shanghai 200065, China; Shanghai Research Center for Spine and Spinal Cord Diseases, Tongji University School of Medicine, Shanghai 200065, China; Clinical Center for Brain and Spinal Cord Research, Tongji University School of Medicine, Shanghai 200065, China.
| | - Liming Cheng
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai 200065, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Institute of Spinal and Spinal Cord Injury, Tongji University School of Medicine, Shanghai 200065, China; Shanghai Research Center for Spine and Spinal Cord Diseases, Tongji University School of Medicine, Shanghai 200065, China; Clinical Center for Brain and Spinal Cord Research, Tongji University School of Medicine, Shanghai 200065, China.
| |
Collapse
|
24
|
Martelossi Cebinelli GC, de Oliveira Leandro M, Rocha Oliveira AE, Alves de Lima K, Donate PB, da Cruz Oliveira Barros C, Ramos ADS, Costa V, Bernardo Nascimento DC, Alves Damasceno LE, Tavares AC, Aquime Gonçalves AN, Imoto Nakaya HT, Cunha TM, Alves-Filho JC, Cunha FQ. CXCR4 + PD-L1 + neutrophils are increased in non-survived septic mice. iScience 2025; 28:112083. [PMID: 40241761 PMCID: PMC12003019 DOI: 10.1016/j.isci.2025.112083] [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: 09/05/2024] [Revised: 12/21/2024] [Accepted: 02/18/2025] [Indexed: 04/18/2025] Open
Abstract
The dysregulated host response to infections can lead to sepsis, a complex disease characterized by a spectrum of clinical phenotypes. Using scRNA-seq, we analyzed the immune cell of survived and non-survived CLP-septic mice to gain insights into the immunological mechanisms by which neutrophils contribute to the hyperinflammatory phenotype. Our findings reveal that non-survived mice exhibit increased frequencies of immature CXCR4+ PD-L1+ neutrophils in the bloodstream, accompanied by an accumulation of trafficking-specific CXCR4+ PD-L1+ neutrophils into the lungs. The IFN-gamma and LPS promote the PD-L1 expression on neutrophils and an activation profile associated with inflammation and organ damage. Notably, abrogating the IFN-gamma reduced susceptibility to CLP-sepsis and diminished CXCR4+ PD-L1+ neutrophils frequency. This study provides insights into the immune cell activation profiles associated with the worsening of the CLP-sepsis, and the CXCR4+ PD-L1+ neutrophils population highlighted here represents a promising target for therapeutic modulation in clinical sepsis hyperinflammatory phenotype.
Collapse
Affiliation(s)
- Guilherme Cesar Martelossi Cebinelli
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Maísa de Oliveira Leandro
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | | | - Kalil Alves de Lima
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Paula Barbim Donate
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Cleyson da Cruz Oliveira Barros
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Núcleo de Biologia Experimental, Universidade de Fortaleza (UNIFOR), Fortaleza, CE, Brazil
| | - Anderson dos Santos Ramos
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Victor Costa
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Daniele Carvalho Bernardo Nascimento
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Luis Eduardo Alves Damasceno
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Amanda Curto Tavares
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - André Nicolau Aquime Gonçalves
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Helder Takashi Imoto Nakaya
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Thiago Mattar Cunha
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - José Carlos Alves-Filho
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Fernando Queiroz Cunha
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School – University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| |
Collapse
|
25
|
Wang Y, Gao H, Li X, Li D, Huang F, Sun Y, Liu X, Yang J, Sun F. PRC1 as an independent adverse prognostic factor in Wilms tumor via integrated bioinformatics and experimental validation. Sci Rep 2025; 15:13282. [PMID: 40247060 PMCID: PMC12006549 DOI: 10.1038/s41598-025-98030-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: 12/16/2024] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
Wilms Tumor (WT), a prevalent pediatric renal malignancy, exhibits marked heterogeneity and variable clinical outcomes. Epithelial-mesenchymal transition (EMT), a biological process enabling epithelial cells to acquire mesenchymal traits associated with enhanced migratory and invasive capacities, plays a crucial role in cancer progression. Protein Regulator of Cytokinesis 1 (PRC1) is a critical protein in cell division, whose overexpression is linked to poor prognosis in various cancers. This study investigates the role of PRC1 as a key prognostic factor in WT and explore the mechanism through comprehensive bioinformatic and experimental approaches. Through bulk RNA-seq data from the TARGET database, we identified PRC1 as significantly up-regulated in WT and associated with poor overall survival. Functional enrichment analyses (GO, KEGG, GSEA) demonstrated PRC1's involvement in cell division, chromatin dynamics, and activation of oncogenic pathways including Wnt/β-catenin, PI3K/AKT/mTOR, and Hedgehog signaling. Immunological analysis showed that elevated PRC1 expression correlates with diminished immune cell activity, particularly in NK cells, suggesting potential immune evasion mechanisms. Single-cell RNA-seq analysis (GSE200256) confirmed PRC1's elevated expression in anaplastic Wilms tumor (AWT) compared to favorable Wilms tumor (FWT), and highlighted its involvement in intercellular communication and metastasis via the EMT process. Genomic analyses identified copy number variations (CNVs) and downregulated PRC1-targeting microRNAs as drivers of its overexpression. In vitro, PRC1 knockdown in WIT-49 cells significantly impaired migratory capacity, invasive potential, EMT progression, and glycolytic metabolism. These findings collectively position PRC1 as a promising therapeutic target and prognostic biomarker in WT.
Collapse
Affiliation(s)
- Yanping Wang
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Hongjie Gao
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Xuetian Li
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Ding Li
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Fan Huang
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Yuqiang Sun
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xingjian Liu
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Junli Yang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China.
| | - Fengyin Sun
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
26
|
Wu C, Ren Y, Li Y, Cui Y, Zhang L, Zhang P, Zhang X, Kan S, Zhang C, Xiong Y. Identification and Experimental Validation of NETosis-Mediated Abdominal Aortic Aneurysm Gene Signature Using Multi-omics, Machine Learning, and Mendelian Randomization. J Chem Inf Model 2025; 65:3771-3788. [PMID: 40105795 DOI: 10.1021/acs.jcim.4c02318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening disorder with limited therapeutic options. Neutrophil extracellular traps (NETs) are formed by a process known as "NETosis" that has been implicated in AAA pathogenesis, yet the roles and prognostic significance of NET-related genes in AAA remain poorly understood. This study aimed to identify key AAA- and NET-related genes (AAA-NETs-RGs), elucidate their potential mechanisms in contributing to AAA, and explore potential therapeutic compounds for AAA therapy. Through bioinformatics analysis of multiomics and machine learning, we identified six AAA-NETs-RGs: DUSP26, FCN1, MTHFD2, GPRC5C, SEMA4A, and CCR7, which exhibited strong diagnostic potential for predicting AAA progression, were significantly enriched in pathways related to cytokine-cytokine receptor interaction and chemokine signaling. Immune infiltration analysis revealed a causal association between AAA-NETs-RGs and immune cell infiltration. Cell-cell communication analysis indicated that AAA-NETs-RGs predominantly function in smooth muscle cells, B cells, T cells, and NK cells, primarily through cytokine and chemokine signaling. Gene profiling revealed that CCR7 and MTHFD2 exhibited the most significant upregulation in AAA patients compared to non-AAA controls, as well as in in vitro AAA models. Notably, genetic depletion of CCR7 and MTHFD2 strongly inhibited Ang II-induced phenotypic switching, functional impairment, and senescence in vascular smooth muscle cells (VSMCs). Based on AAA-NETs-RGs, molecular docking analysis combined with the Connectivity Map (CMap) database identified mirdametinib as a potential therapeutic agent for AAA. Mirdametinib effectively alleviated Ang II-induced phenotypic switching, biological dysfunction, and senescence. These findings provide valuable insights into understanding the pathophysiology of AAA and highlight promising therapeutic strategies targeting AAA-NETs-RGs.
Collapse
Affiliation(s)
- Chengsong Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 710069 Xi'an, Shaanxi, P. R. China
| | - Yuanyuan Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 710069 Xi'an, Shaanxi, P. R. China
| | - Yang Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 710069 Xi'an, Shaanxi, P. R. China
| | - Yue Cui
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 710069 Xi'an, Shaanxi, P. R. China
| | - Liyao Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 710069 Xi'an, Shaanxi, P. R. China
| | - Pan Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 710069 Xi'an, Shaanxi, P. R. China
| | - Xuejiao Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 710069 Xi'an, Shaanxi, P. R. China
| | - Shangguang Kan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 710069 Xi'an, Shaanxi, P. R. China
| | - Chan Zhang
- Department of Blood Transfusion, the First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, 650032 Kunming, Yunnan, China
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 710069 Xi'an, Shaanxi, P. R. China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, 710018 Xi'an, Shaanxi, P. R. China
| |
Collapse
|
27
|
Lei M, Zhang Y, Yu Y, Wang G, Hu N, Xie J. ITGB2 and ICAM3 predict increased survival of sepsis with decreased intercellular communication in cytotoxic CD8+ T cells. Sci Rep 2025; 15:12635. [PMID: 40221459 PMCID: PMC11993633 DOI: 10.1038/s41598-025-93685-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 03/10/2025] [Indexed: 04/14/2025] Open
Abstract
Sepsis is closely linked to immunity. Our research aimed to identify key genes associated with sepsis immunity utilizing single-cell RNA sequencing (scRNA-seq) data. This study obtained the GSE167363 and GSE54514 datasets from the Gene Expression Omnibus (GEO). The GSE167363 dataset was subjected to cluster analysis, cell proportion analysis, cell interaction analysis, and gene set enrichment analysis (GSEA). The differentially expressed genes (DEGs) of CD8+ T cells were intersected with the DEGs in the GSE54514 dataset, and key genes related to immunity in sepsis patients were identified through Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Finally, we validated the gene expression levels in a mouse model of sepsis caused by cecum ligation and puncture (CLP). Findings indicated that intercellular communication of Cytotoxic CD8+ T cells was reduced in the sepsis survivors compared to non-survivors. The expression of 3 down-regulated key DEGs (ITGB2, SELL and ICAM3) was negatively correlated with the abundance of CD8+ T cells. Moreover, Cytotoxic CD8+ T cells with low expression of ITGB2, SELL and ICAM3 were more adverse to the survival of sepsis as compared to those with high expression of the above genes. These genes may predict increased survival in sepsis by regulating intercellular communication in cytotoxic CD8+ T cells, suggesting that they are potential therapeutic targets for improving sepsis prognosis.
Collapse
Affiliation(s)
- Min Lei
- Department of Anesthesiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 East Qing Chun Road, Hangzhou, 310016, Zhejiang, China
| | - Yaping Zhang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 East Qing Chun Road, Hangzhou, 310016, Zhejiang, China
| | - Yijin Yu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 East Qing Chun Road, Hangzhou, 310016, Zhejiang, China
| | - Gaojian Wang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 East Qing Chun Road, Hangzhou, 310016, Zhejiang, China
| | - Nianqiang Hu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 East Qing Chun Road, Hangzhou, 310016, Zhejiang, China
| | - Junran Xie
- Department of Anesthesiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 East Qing Chun Road, Hangzhou, 310016, Zhejiang, China.
| |
Collapse
|
28
|
Huang J, Zhang H, Lin X, Wu X, Chen X, Chen W, Liang S, Chen Y, Luo Q, Xu C, Liu S, Liu X, Zhang S. Regulatory T Cell Infiltration-Driven Single-Cell Transcriptomic Analysis Identifies SAP18 as a Prognostic Marker for Esophageal Squamous Cell Carcinoma. J Gastrointest Cancer 2025; 56:97. [PMID: 40208395 DOI: 10.1007/s12029-025-01174-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Advanced esophageal squamous cell carcinoma (ESCC) is characterized by molecular heterogeneity and distinct patterns of immune cell infiltration. Regulatory T cells (Tregs), in particular, play a critical role in shaping an immunosuppressive tumor microenvironment (TME), which is associated with poor clinical outcomes. METHODS We developed a prognostic model by integrating GEO-derived bulk RNA sequencing data and single-cell transcriptome. Model predictions were confirmed through RT-qPCR, Western blot, and immunohistochemistry on clinical specimens, while in vitro assays (CCK8, transwell invasion, scratch, colony formation, and immunofluorescence) validated the function of SAP18 in cell proliferation, invasion, and ECM remodeling. RESULTS Expression patterns of the 5 Tregs-associated genes in clinical specimens aligned with model predictions, underscoring the model's robustness. The high-risk subgroup was associated with upregulated extracellular matrix (ECM) remodeling, an abundance of immune-suppressive cells, higher TP53 mutation rate, and limited benefit from immunotherapy. In contrast, the low-risk subgroup exhibited anti-tumor immunity. Cell-cell communication analysis also implicated the collagen pathway in Tregs-mediated immune evasion in ESCC. Functional assays indicated that SAP18 in the prognostic model significantly promotes proliferation, invasion, and ECM reconstruction, further highlighting its potential as a therapeutic target. CONCLUSION Our findings elucidate the role of Tregs in the TME, underscoring significant potential of SAP18, which is essential for assessing patient prognosis and may facilitate the development of personalized therapies for ESCC.
Collapse
Affiliation(s)
- Jianxiang Huang
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
- College of Pharmacy, Jinan University, Guangzhou, 510220, PR China
| | - Hanshuo Zhang
- Gastrointestinal Anorectal Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
| | - Xinyue Lin
- Department of Pharmacology, Medical College of Shantou University, Shantou, 515063, PR China
| | - Xiaolong Wu
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
- College of Pharmacy, Jinan University, Guangzhou, 510220, PR China
| | - Xiaoshan Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
| | - Wang Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
| | - Shanshan Liang
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
| | - Yun Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
| | - Qianhua Luo
- Department of Pharmacy, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, PR China
| | - Chengcheng Xu
- College of Pharmacy, Jinan University, Guangzhou, 510220, PR China
| | - Shaojie Liu
- Gastrointestinal Anorectal Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
| | - Xingmei Liu
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China.
- Department of Nursing, Guangzhou Red Cross Hospital of Jinan University, Haizhu District, No. 396, Tongfuzhong Road, Guangzhou, 510220, PR China.
| | - Shuyao Zhang
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China.
- College of Pharmacy, Jinan University, Guangzhou, 510220, PR China.
| |
Collapse
|
29
|
Su A, Lee H, Tran M, Dela Cruz RC, Sathe A, Bai X, Wichmann I, Pflieger L, Moulton B, Barker T, Haslem D, Jones D, Nadauld L, Nguyen Q, Ji HP, Rhodes T. The single-cell spatial landscape of stage III colorectal cancers. NPJ Precis Oncol 2025; 9:101. [PMID: 40189697 PMCID: PMC11973205 DOI: 10.1038/s41698-025-00853-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
We conducted a spatial analysis of stage III colorectal adenocarcinomas using Hyperion Imaging Mass Cytometry, examining 52 tumors to assess the tumor microenvironment at the single-cell level. This approach identified 10 distinct cell phenotypes in the tumor microenvironment, including stromal and immune cells, with a subset showing a proliferative phenotype. By focusing on spatial neighborhood interactions and tissue niches, particularly regions with tumor-infiltrating lymphocytes, we investigated how cellular organization relates to clinicopathological and molecular features such as microsatellite instability (MSI) and recurrence. We determined that microsatellite stable (MSS) colorectal cancers had an increased risk of recurrence if they had the following features: 1) a low level of stromal tumor-infiltrating lymphocytes, and 2) low interactions between CD4 + T cells and stromal cells. Our results point to the utility of spatial single-cell interaction analysis in defining novel features of the tumor immune microenvironments and providing useful clinical cell-related spatial biomarkers.
Collapse
Affiliation(s)
- Andrew Su
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - HoJoon Lee
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Minh Tran
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | | | - Anuja Sathe
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Xiangqi Bai
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ignacio Wichmann
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Division of Obstetrics and Gynecology, Department of Obstetrics, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | | | - Bryce Moulton
- Intermountain Healthcare, Saint George, UT, 84770, USA
| | - Tyler Barker
- Intermountain Healthcare, Saint George, UT, 84770, USA
| | | | - David Jones
- Intermountain Healthcare, Saint George, UT, 84770, USA
| | | | - Quan Nguyen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Hanlee P Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | | |
Collapse
|
30
|
Tang Y, Cao L, Jin J, Li T, Chen Y, Lu Y, Li T, Weiss LM, Pan G, Bao J, Zhou Z. Single-cell transcriptional responses of T cells during microsporidia infection. Commun Biol 2025; 8:567. [PMID: 40185986 PMCID: PMC11971339 DOI: 10.1038/s42003-025-07990-4] [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: 05/08/2024] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
T cells have been reported to play critical roles in preventing of microsporidia dissemination. However, there roles and functions of each subset remain unclear. Here in the study, we performed a thorough analysis of murine splenic T-cell response analysis via single-cell RNA sequencing during microsporidia E. cuniculi infection. We demonstrated that Type I T helper (Th1) cells, T follicular helper (Tfh) cells, effector CD8 + T cells and proliferating CD8 + T cells were activated and expanded after infection. Activated Th1 cells and Tfh cells presented significantly upregulated gene expression of Ifng and Il21, respectively. A subcluster of Th1 cells with high Csf1 expression was detected after infection. Subsets of activated CD8 + T cells were markedly enriched with high expression of cytotoxic-function related genes such as Gzma and Gzmb, whereas some active CD8 T cells were enriched with proliferation-function related genes Mki67 and Stmn1. Other subsets of T cells including NK T cells, Myb+ T cells, γδ T cells and Cxcr6+ T cells, were also analyzed in this study yet no expansion was observed. In summary, our findings provide in-depth and comprehensive insights into T-cell responses during microsporidia infection, which will be valuable for further investigations.
Collapse
Affiliation(s)
- Yunlin Tang
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, China
| | - Lu Cao
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, China
| | - Jiangyan Jin
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, China
| | - Tangxin Li
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, China
| | - Yebo Chen
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, China
| | - Yishan Lu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, China
| | - Tian Li
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, China
| | - Louis M Weiss
- Department of Pathology, Albert Einstein College of Medicine, New York, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
| | - Guoqing Pan
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, China
| | - Jialing Bao
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China.
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, China.
| | - Zeyang Zhou
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China.
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, China.
- College of Life Sciences, Chongqing Normal University, Chongqing, China.
| |
Collapse
|
31
|
Xu Q, Ma L, Streuer A, Altrock E, Schmitt N, Rapp F, Klär A, Nowak V, Obländer J, Weimer N, Palme I, Göl M, Zhu HH, Hofmann WK, Nowak D, Riabov V. Machine learning-based in-silico analysis identifies signatures of lysyl oxidases for prognostic and therapeutic response prediction in cancer. Cell Commun Signal 2025; 23:169. [PMID: 40186284 PMCID: PMC11971788 DOI: 10.1186/s12964-025-02176-1] [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: 09/25/2024] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Lysyl oxidases (LOX/LOXL1-4) are crucial for cancer progression, yet their transcriptional regulation, potential therapeutic targeting, prognostic value and involvement in immune regulation remain poorly understood. This study comprehensively evaluates LOX/LOXL expression in cancer and highlights cancer types where targeting these enzymes and developing LOX/LOXL-based prognostic models could have significant clinical relevance. METHODS We assessed the association of LOX/LOXL expression with survival and drug sensitivity via analyzing public datasets (including bulk and single-cell RNA sequencing data of six datasets from Gene Expression Omnibus (GEO), Chinese Glioma Genome Atlas (CGGA) and Cancer Genome Atlas Program (TCGA)). We performed comprehensive machine learning-based bioinformatics analyses, including unsupervised consensus clustering, a total of 10 machine-learning algorithms for prognostic prediction and the Connectivity map tool for drug sensitivity prediction. RESULTS The clinical significance of the LOX/LOXL family was evaluated across 33 cancer types. Overexpression of LOX/LOXL showed a strong correlation with tumor progression and poor survival, particularly in glioma. Therefore, we developed a novel prognostic model for glioma by integrating LOX/LOXL expression and its co-expressed genes. This model was highly predictive for overall survival in glioma patients, indicating significant clinical utility in prognostic assessment. Furthermore, our analysis uncovered a distinct LOXL2-overexpressing malignant cell population in recurrent glioma, characterized by activation of collagen, laminin, and semaphorin-3 pathways, along with enhanced epithelial-mesenchymal transition. Apart from glioma, our data revealed the role of LOXL3 overexpression in macrophages and in predicting the response to immune checkpoint blockade in bladder and renal cancers. Given the pro-tumor role of LOX/LOXL genes in most analyzed cancers, we identified potential therapeutic compounds, such as the VEGFR inhibitor cediranib, to target pan-LOX/LOXL overexpression in cancer. CONCLUSIONS Our study provides novel insights into the potential value of LOX/LOXL in cancer pathogenesis and treatment, and particularly its prognostic significance in glioma.
Collapse
Affiliation(s)
- Qingyu Xu
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany.
- Department of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Ling Ma
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Alexander Streuer
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Eva Altrock
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Nanni Schmitt
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Felicitas Rapp
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Alessa Klär
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Verena Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Julia Obländer
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Nadine Weimer
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Iris Palme
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Melda Göl
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Hong-Hu Zhu
- Department of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Chinese Institutes for Medical Research, Beijing, China
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Daniel Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Vladimir Riabov
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| |
Collapse
|
32
|
Tang Q, Tang L, Wang X, Zhang Y, Liu W, Yang T, Wu Y, Ma Y, Lei T, Song W. Comprehensive Analyses of Single-Cell and Bulk RNA Sequencing Data From M2 Macrophages to Elucidate the Immune Prognostic Signature in Patients with Gastric Cancer Peritoneal Metastasis. Immunotargets Ther 2025; 14:383-402. [PMID: 40201390 PMCID: PMC11977558 DOI: 10.2147/itt.s506143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/04/2025] [Indexed: 04/10/2025] Open
Abstract
Purpose The peritoneum is a common site of metastasis in gastric cancer (GC), associated with poor prognosis and significant morbidity. The proclivity of GCs to metastasize to the peritoneum has been hypothesized to occur due the latter's immunosuppressive microenvironment, such as stromal infiltration and M2 macrophage enrichment, which are associated with increased risk of PM. As far as we know, a model that can effectively predict the prognosis of patients with GCPM is still lacking. Consequently, we constructed a prognostic risk model based on M2 macrophages associated with gastric cancer peritoneal metastasis, aiming to enhance predictive precision and guide tailored therapeutic interventions. Methods M2 macrophage-associated genes were identified in combination with marker genes from single-cell RNA sequencing (scRNA-seq) and modular genes from weighted gene coexpression network analysis (WGCNA). A prognostic model was constructed via LASSO analysis and validated in internal and external cohorts. We further compared the immune microenvironment, immune checkpoints, and chemotherapeutic drug sensitivity between patient groups stratified by risk to clarify the immune landscape in the GCPM. Results Our study identified 38 M2 macrophage-related genes via single-cell and bulk RNA sequencing. We developed a prognostic model based on the expression levels of 4 signature genes: DAB2, SPARC, PLTP, and FOLR2. The feasibility of the model was validated with internal and external validation sets (TCGA, GSE62254 and IMvigor210). The model also supported the prediction results of prognosis on the basis of the immunohistochemical results. Notably, patients with higher risk scores had a lower proportion of MSI-H and TMB, a higher prevalence of stages III-IV, and a lower likelihood of responding favorably to immunotherapy. Conclusion Our prognostic risk model could effectively predict the prognosis and response to chemo-immune therapy in patients with GCPM. The risk score is a promising independent prognostic factor that is closely correlated with the immune microenvironment and clinicopathological characteristics.
Collapse
Affiliation(s)
- Qiao Tang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-SenYou University, Guangzhou, Guangdong, People’s Republic of China
- Laboratory of Surgery, The First Affiliated Hospital, Sun Yat-SenYou University, Guangzhou, Guangdong, People’s Republic of China
| | - Liang Tang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-SenYou University, Guangzhou, Guangdong, People’s Republic of China
- Laboratory of Surgery, The First Affiliated Hospital, Sun Yat-SenYou University, Guangzhou, Guangdong, People’s Republic of China
| | - Xiaofeng Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-SenYou University, Guangzhou, Guangdong, People’s Republic of China
| | - Yongxin Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-SenYou University, Guangzhou, Guangdong, People’s Republic of China
| | - Wenwei Liu
- Center for Stem Cell Biology and Tissue Engineering, Sun Yat-SenYou University, Guangzhou, Guangdong, People’s Republic of China
| | - Ting Yang
- Center for Stem Cell Biology and Tissue Engineering, Sun Yat-SenYou University, Guangzhou, Guangdong, People’s Republic of China
| | - Yuxin Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-SenYou University, Guangzhou, Guangdong, People’s Republic of China
| | - Yuanchen Ma
- Center for Stem Cell Biology and Tissue Engineering, Sun Yat-SenYou University, Guangzhou, Guangdong, People’s Republic of China
| | - Tianxiang Lei
- Department of Thyroid and Hernia Surgery, Guangdong Provincial People’s Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Wu Song
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-SenYou University, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
33
|
Xie R, Li C, Zhao T, Zhang S, Zhong A, Chen N, Li Z, Chen J. Integration of Flow Cytometry and Single-Cell RNA Sequencing Analysis to Explore the Fibroblast Subpopulations in Keloid that Correlate with Recurrence. Adv Wound Care (New Rochelle) 2025. [PMID: 40177712 DOI: 10.1089/wound.2024.0262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
Objective: Fibroblasts (FBs) are the cytological basis of keloid (KD) formation. This study aimed to identify the key pathogenic target cell subpopulation involved in KD recurrence. Approach: Single-cell RNA sequencing data were retrieved from public databases, revealing distinct gene expression patterns in FB subpopulations. Flow cytometry (FCM) was used to identify the surface molecular phenotypes of FBs that affect KD recurrence. Simultaneously, logistic regression analysis was performed to assess the predictive value of changes in FB subpopulation percentages for clinical KD recurrence. Results: The percentage of keloid fibroblasts was significantly greater than that in normal tissues. Through further clustering analysis of the FB population, we obtained four subpopulations, FB1-FB4, in which the percentages of FB1 subpopulation were increased, and functional enrichment analysis suggested that the FB1 subpopulation may play a greater role in extracellular matrix collagen oversynthesis in KD. In addition, the gene expression of CD26 (DPP4), CD117 (c-KIT), and CD34 in the FB1 subpopulation was significantly higher than that in FB2-4 subpopulations. Moreover, the percentage of CD26+/CD117+/CD34+ cell subpopulations in the FCM data of patients with KD recurrence was significantly increased. Regression analysis confirmed that the CD26+/CD117+/CD34+ FB subpopulation was a risk factor for relapse. Innovation: We demonstrated that the molecular phenotypic and functional heterogeneity of FBs influences KD recurrence. Conclusion: We identified key pathogenic FB subpopulations that may affect KD development, which can be used as potential markers to predict recurrence and provide potential target cell populations for future clinical treatment.
Collapse
Affiliation(s)
- Ruxin Xie
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Chenyu Li
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Tian Zhao
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Shiwei Zhang
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Ai Zhong
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Nengbin Chen
- Cosmetic Burn and Plastic Surgery, The People's Hospital of Leshan, Leshan, China
| | - Zhengyong Li
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Junjie Chen
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Priam P, Krasteva V, Polsinelli A, Côté L, Dilauro F, Poinsignon TM, Thibault P, Lessard JA. Bcl7b and Bcl7c subunits of BAF chromatin remodeling complexes are largely dispensable for hematopoiesis. Exp Hematol 2025; 146:104769. [PMID: 40187480 DOI: 10.1016/j.exphem.2025.104769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 03/16/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
Chromatin remodelers have emerged as prominent regulators of hematopoietic cell development and potential drivers of various human hematological malignancies. ATP-dependent BAF chromatin remodeling complexes, related to yeast SWI/SNF, determine gene expression programs and consequently contribute to the self-renewal, commitment, and lineage-specific differentiation of hematopoietic stem cells (HSCs) and progenitors. Here, we investigated the elusive biological function of the core Bcl7b and Bcl7c subunits of BAF complexes in hematopoietic tissue. Our analysis of mouse constitutive knockout alleles revealed that both Bcl7b and Bcl7c are dispensable for animal survival and steady-state adult hematopoiesis. Bcl7b and Bcl7c double knockout (dKO) mice can maintain long-term hematopoiesis with no observable effect on the HSC compartment. Moreover, we show that Bcl7b/Bcl7c dKO HSCs are capable of normal multilineage hematopoietic reconstitution after competitive serial transplantation. Collectively, these studies suggest that the Bcl7b and Bcl7c subunits of BAF complexes are dispensable for normal hematopoiesis.
Collapse
Affiliation(s)
- Pierre Priam
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, Quebec, Canada
| | - Veneta Krasteva
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, Quebec, Canada
| | - Alexandre Polsinelli
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, Quebec, Canada
| | - Laurence Côté
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, Quebec, Canada
| | - Francis Dilauro
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, Quebec, Canada
| | - Thérèse-Marie Poinsignon
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, Quebec, Canada
| | - Pierre Thibault
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, Quebec, Canada
| | - Julie A Lessard
- Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, Quebec, Canada; Department of Pathology and Cellular Biology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada.
| |
Collapse
|
35
|
Saxena P, Sinha A, Singh SK. Computer-assisted interpretation, in-depth exploration and single cell type annotation of RNA sequence data using k-means clustering algorithm. Comput Methods Biomech Biomed Engin 2025; 28:668-678. [PMID: 38235728 DOI: 10.1080/10255842.2023.2300685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/25/2023] [Accepted: 12/24/2023] [Indexed: 01/19/2024]
Abstract
At now, the majority of approaches rely on manual techniques for annotating cell types subsequent to clustering the data obtained from single-cell RNA sequencing (scRNA-seq). These approaches require a significant amount of physical exertion and depend substantially on the user's skill, perhaps resulting in uneven outcomes and inconsistency in treatment. In this paper, we provide a computer-assisted interpretation of every single cell of a tissue sample, along with an in-depth exploration of an individual cell's molecular, phenotypic and functional attributes. The paper will also perform k-means clustering followed by silhouette validation based on similar phenotype and functional attributes, and also, cell type annotation is performed, where we match a cell's gene profile against some known database by applying certain statistical conditions. Finally, all the genes are mapped spatially on the tissue sample. This paper is an aid to medicine to know which cells are expressed/not expressed in a tissue sample and their spatial location on the tissue sample.
Collapse
Affiliation(s)
- Pranshu Saxena
- Department of Information Technology, ABES Engineering College, Ghaziabad, India
| | - Amit Sinha
- Department of Information Technology, ABES Engineering College, Ghaziabad, India
| | - Sanjay Kumar Singh
- University School of Automation and Robotics, Guru Gobind Singh Indraprastha University, Surajmal Vihar, Delhi, India
| |
Collapse
|
36
|
Hadizadeh M, Askari N, Jafarinejad-Farsangi S. A single-cell approach to analyzing vascular endothelial cell contributions in VEGF-driven angiogenesis and LINC02313 in gastric cancer. Comput Biol Chem 2025; 115:108361. [PMID: 39914073 DOI: 10.1016/j.compbiolchem.2025.108361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/11/2025] [Accepted: 01/21/2025] [Indexed: 02/26/2025]
Abstract
Gastric cancer (GC) heterogeneity and lack of suitable molecular markers remain major challenges for this disease. The critical role of long non-coding RNAs (lncRNAs) in cancer biological processes has been increasingly recognized. A novel lncRNA, LINC02313, was identified in GC in this study, and its function was examined bioinformatically. The differential expression of LINC02313 was examined, and its target genes were predicted using RNA-Seq data from TCGA. LINC02313 showed correlation with 272 significant DEGs in GC. The analysis of single-cell transcriptomes revealed 11 unique clusters of cell types, but vascular endothelial cells have the most targets (30 genes). Receiver Operating Characteristic (ROC) analysis illustrated the diagnostic capabilities of LINC02313 and its targets across most cellular clusters, achieving the highest levels of accuracy. Functionally related signaling pathways were classified through cell-cell communication analysis; in the tumorous state, emphasizing the more prominent role of vascular endothelial cells in the Vascular Endothelial Growth Factor (VEGF) signaling pathway compared to the normal state. Trajectory analysis showed vascular endothelial cells are at the start of pseudotime in a normal state, but in a tumorous state, they shift to the middle of pseudotime. The results of this study highlight the critical role of endothelial cells in the advancement of GC and propose novel therapeutic approaches that focus on modulating angiogenic signaling pathways and lncRNA function to enhance treatment efficacy.
Collapse
Affiliation(s)
- Morteza Hadizadeh
- Department of Biotechnology, Institute of Sciences and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Nahid Askari
- Department of Biotechnology, Institute of Sciences and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran.
| | | |
Collapse
|
37
|
Zhao C, Qi W, Lv X, Gao X, Liu C, Zheng S. Elucidating the Role of Trem2 in Lipid Metabolism and Neuroinflammation. CNS Neurosci Ther 2025; 31:e70338. [PMID: 40205810 PMCID: PMC11982525 DOI: 10.1111/cns.70338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive impairment and neuroinflammation. Astrocytes play a key role in the neuroinflammatory environment of AD, especially through lipid metabolism regulation. However, the mechanisms by which astrocytes, particularly through the triggering receptor expressed on myeloid cells 2 (Trem2) receptor, contribute to lipid dysregulation and neuroinflammation in AD remain inadequately understood. METHODS We employed an AD mouse model and integrated single-cell RNA sequencing (scRNA-seq), transcriptomics, and high-throughput metabolomics to analyze lipid metabolism and inflammatory profiles in astrocytes. Differential gene expression was further validated with the GEO database, and in vitro and in vivo experiments were conducted to assess the impact of Trem2 modulation on astrocytic inflammation and lipid composition. RESULTS Our findings demonstrate that Trem2 modulates lipid metabolism in astrocytes, affecting fatty acid and phospholipid pathways. In the AD model, Trem2 expression was suppressed, enhancing nuclear factor-κB (NF-κB) signaling and promoting the secretion of pro-inflammatory factors such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Trem2 overexpression reduced astrocytic inflammation and altered lipid composition, attenuating neuroinflammation both in vitro and in vivo. These results underscore Trem2's regulatory role in lipid metabolism and its significant impact on neuroinflammation in AD. CONCLUSIONS This study identifies Trem2 as a pivotal regulator of astrocytic lipid metabolism and neuroinflammation in AD, providing potential molecular targets for early intervention and therapeutic strategies aimed at mitigating AD progression.
Collapse
Affiliation(s)
- Chenhui Zhao
- College of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
- Heilongjiang Key Laboratory of Laboratory Animals and Comparative MedicineHarbinChina
| | - Wei Qi
- Suzhou Frontage New Drug Development Co., Ltd.SuzhouChina
| | - Xiaoping Lv
- College of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
- Heilongjiang Key Laboratory of Laboratory Animals and Comparative MedicineHarbinChina
| | - Xueli Gao
- College of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
- Heilongjiang Key Laboratory of Laboratory Animals and Comparative MedicineHarbinChina
| | - Chaonan Liu
- College of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
- Heilongjiang Key Laboratory of Laboratory Animals and Comparative MedicineHarbinChina
| | - Shimin Zheng
- College of Veterinary MedicineNortheast Agricultural UniversityHarbinChina
- Heilongjiang Key Laboratory of Laboratory Animals and Comparative MedicineHarbinChina
| |
Collapse
|
38
|
Jia G, He P, Dai T, Goh D, Wang J, Sun M, Wee F, Li F, Lim JCT, Hao S, Liu Y, Lim TKH, Ngo NT, Tao Q, Wang W, Umar A, Nashan B, Zhang Y, Ding C, Yeong J, Liu L, Sun C. Spatial immune scoring system predicts hepatocellular carcinoma recurrence. Nature 2025; 640:1031-1041. [PMID: 40074893 DOI: 10.1038/s41586-025-08668-x] [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: 03/18/2023] [Accepted: 01/17/2025] [Indexed: 03/14/2025]
Abstract
Given the high recurrence rates of hepatocellular carcinoma (HCC) post-resection1-3, improved early identification of patients at high risk for post-resection recurrence would help to improve patient outcomes and prioritize healthcare resources4-6. Here we observed a spatial and HCC recurrence-associated distribution of natural killer (NK) cells in the invasive front and tumour centre from 61 patients. Using extreme gradient boosting and inverse-variance weighting, we developed the tumour immune microenvironment spatial (TIMES) score based on the spatial expression patterns of five biomarkers (SPON2, ZFP36L2, ZFP36, VIM and HLA-DRB1) to predict HCC recurrence risk. The TIMES score (hazard ratio = 88.2, P < 0.001) outperformed current standard tools for patient risk stratification including the TNM and BCLC systems. We validated the model in 231 patients from five multicentred cohorts, achieving a real-world accuracy of 82.2% and specificity of 85.7%. The predictive power of these biomarkers emerged through the integration of their spatial distributions, rather than individual marker expression levels alone. In vivo models, including NK cell-specific Spon2-knockout mice, revealed that SPON2 enhances IFNγ secretion and NK cell infiltration at the invasive front. Our study introduces TIMES, a publicly accessible tool for predicting HCC recurrence risk, offering insights into its potential to inform treatment decisions for early-stage HCC.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Middle Aged
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Carcinoma, Hepatocellular/diagnosis
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/surgery
- Cohort Studies
- Extracellular Matrix Proteins/genetics
- Extracellular Matrix Proteins/deficiency
- Extracellular Matrix Proteins/metabolism
- Interferon-gamma/metabolism
- Killer Cells, Natural/immunology
- Killer Cells, Natural/cytology
- Liver Neoplasms/diagnosis
- Liver Neoplasms/immunology
- Liver Neoplasms/pathology
- Liver Neoplasms/surgery
- Mice, Knockout
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/diagnosis
- Neoplasm Recurrence, Local/pathology
- Reproducibility of Results
- Tumor Microenvironment
Collapse
Affiliation(s)
- Gengjie Jia
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Peiqi He
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Tianli Dai
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Denise Goh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Jiabei Wang
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Mengyuan Sun
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Felicia Wee
- Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Fuling Li
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Jeffrey Chun Tatt Lim
- Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Shuxia Hao
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yao Liu
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Tony Kiat Hon Lim
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Duke-NUS Medical School, Singapore, Singapore
| | | | - Qingping Tao
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Wei Wang
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Ahitsham Umar
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Björn Nashan
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
| | - Yongchang Zhang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Central South University, Changsha, China
| | - Chen Ding
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Joe Yeong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore.
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Singapore, Singapore.
- Cancer Science Institute, National University of Singapore, Singapore, Singapore.
| | - Lianxin Liu
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China.
| | - Cheng Sun
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China.
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
39
|
Gao X, Fan Y, Wang G, Xu J, Deng R, Song J, Sun B, Wang Y, Wu Z, Jia R, Huang J, He H, Gao L, Zhang Y, Sun N, Wu B. Combined analysis of single-cell and bulk transcriptome sequencing data identifies critical glycolysis genes in idiopathic pulmonary arterial hypertension. J Transl Med 2025; 23:373. [PMID: 40140873 PMCID: PMC11948795 DOI: 10.1186/s12967-025-06373-x] [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: 02/04/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Abnormal glycolytic metabolism plays a significant role in pulmonary vascular remodeling in idiopathic pulmonary arterial hypertension (IPAH), yet the specific mechanisms remain unclear. The primary objective of this study is to investigate the key regulatory mechanisms of glycolysis in IPAH. METHODS Bulk and single-cell sequencing data obtained from IPAH patient tissue samples were downloaded from the GEO database. scMetabolism and AUCcell analyses of the IPAH single-cell sequencing data were carried out to quantify the glycolytic metabolic activity and identify the main cell types regulating glycolysis, respectively. The ssGSEA method was used to assess the glycolytic activity in each bulk sample within the bulk sequencing data. Differential analysis, weighted gene co-expression network analysis (WGCNA), and protein-protein interaction (PPI) network analysis were conducted to identify key genes associated with glycolysis in IPAH samples. Single-cell sequencing and a monocrotaline (MCT)-induced model of PH in rats were utilized to validate the expression of these key genes. RESULTS Single-cell sequencing data indicated that IPAH patients displayed increased glycolytic activity, which was primarily regulated by fibroblasts. Similarly, bulk transcriptomic data revealed a significant increase in glycolytic activity in IPAH patients. Differential analysis, WGCNA, PPI network analysis, and integrated single-cell analysis further identified insulin-like growth factor-1 (IGF1), lysyl-tRNA synthetase (KARS), caspase-3 (CASP3), and cyclin-dependent kinase inhibitor 2 A (CDKN2A) as key genes associated with fibroblast-mediated glycolysis in IPAH patients. Differential expression of IGF1, KARS, CASP3, and CDKN2A was also observed in our in vivo model of PH. CONCLUSION Our study identifies IGF1, KARS, CASP3, and CDKN2A as key regulatory genes in glycolysis in IPAH, which provides the basis for the development of targeted therapies.
Collapse
Affiliation(s)
- Xuan Gao
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China
| | - Youli Fan
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China
| | - Guijia Wang
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
| | - Jiangjiang Xu
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
| | - Runwei Deng
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China
| | - Jiangwei Song
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China
| | - Binfeng Sun
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
| | - Yongbing Wang
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China
| | - Zixuan Wu
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China
| | - Ruyi Jia
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China
| | - Jing Huang
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China
| | - Huiyu He
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China
| | - Lei Gao
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
| | - Yihao Zhang
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
| | - Na Sun
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China.
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China.
| | - Bingxiang Wu
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China.
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China.
| |
Collapse
|
40
|
Pan S, Tang H, Yao B, Tian G, Sun B, Hu Y, Chen Y, Li J, Xu X, Zhang C, Ying S. Decoding the ontogeny of myeloid lineage diversity by cross-species and developmental analyses of hematopoietic progenitor atlases. Cell Rep 2025; 44:115406. [PMID: 40057952 DOI: 10.1016/j.celrep.2025.115406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 12/23/2024] [Accepted: 02/15/2025] [Indexed: 03/29/2025] Open
Abstract
Myeloid cells play vital roles in homeostasis and immune responses in vertebrates, but the developmental pathway underlying their lineage diversity remains elusive. Here, we construct a single-cell transcriptional map of myeloid progenitors from mouse bone marrow and conduct cross-species and developmental analyses across human, monkey, mouse, and zebrafish. We uncover a conserved specification program separating the eosinophil-basophil-mast cell (EBM) lineage and neutrophil-monocyte (NM) lineage, reclassifying myeloid cells beyond the conventional granulocytic and monocytic framework. By generating Ikzf2-EGFP reporter mice, we identify IKZF2 as a priming marker for EBM lineage specification. Ikzf2-EGFP+ and Ikzf2-EGFP- granulocyte-monocyte progenitors (GMPs) exhibit distinct potential to generate EBM and NM lineages, and Ikzf2-EGFP expression robustly distinguishes their progenies. Additionally, we demonstrate that lineage specification emerges early during myelopoiesis. These findings provide a redefined perspective on myeloid lineage ontogeny, highlighting the conservation of lineage specification and offering insights into the understanding and therapeutic development of myelopoiesis.
Collapse
Affiliation(s)
- Sheng Pan
- Department of Pharmacy, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu 322000, China; Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Haoyu Tang
- Department of Pharmacy, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu 322000, China; Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Bingpeng Yao
- Department of Pharmacy, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu 322000, China
| | - Guoxiong Tian
- Department of Pharmacy, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu 322000, China
| | - Beibei Sun
- Department of Pharmacy, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu 322000, China; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yangmingzi Hu
- Department of Pharmacy, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu 322000, China
| | - Yan Chen
- Department of Pharmacy, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu 322000, China
| | - Jiaqian Li
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiao Xu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310053, China.
| | - Chao Zhang
- Department of Pharmacy, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu 322000, China; Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Anatomy, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Songmin Ying
- Department of Pharmacy, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu 322000, China; Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
41
|
Wu W, Wang S, Zhang K, Li H, Qiao S, Zhang Y, Pang S. scMDCL: A Deep Collaborative Contrastive Learning Framework for Matched Single-Cell Multiomics Data Clustering. J Chem Inf Model 2025; 65:3048-3063. [PMID: 40068854 DOI: 10.1021/acs.jcim.4c02114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Single-cell multiomics clustering integrates multiple omics data to analyze cellular heterogeneity and is crucial for uncovering complex biological processes and disease mechanisms. However, existing matched single-cell multiomics clustering methods often neglect the full utilization of intercellular relationships and the interactions and synergy between features from different omics, leading to suboptimal clustering performance. In this paper, we propose a deep collaborative contrastive learning framework for matched single-cell multiomics data clustering, named scMDCL. This framework fully leverages intercell relationships while enhancing feature interactions among identical cells across different omics data, thereby facilitating efficient clustering of multiomics data. Specifically, to fully utilize the topological information between cells, a graph autoencoder and a feature information enhancement module are designed for different omics, enabling the extraction and augmentation of cell features. Additionally, contrastive learning techniques are employed to strengthen the interactions among the different omics features of the same cell. Ultimately, multiomics deep collaborative clustering modules are utilized to achieve single-cell multiomics clustering. Extensive experiments conducted on nine publicly available single-cell multiomics datasets demonstrate the superior performance of the proposed framework in integrating multiomics data for clustering tasks.
Collapse
Affiliation(s)
- Wenhao Wu
- Qingdao Institute of Software, College of Computer Science and Technology, State Key Laboratory of Chemical Safety, Shandong Key Laboratory of Intelligent Oil & Gas Industrial Software, China University of Petroleum (East China), Qingdao 266580, China
| | - Shudong Wang
- Qingdao Institute of Software, College of Computer Science and Technology, State Key Laboratory of Chemical Safety, Shandong Key Laboratory of Intelligent Oil & Gas Industrial Software, China University of Petroleum (East China), Qingdao 266580, China
| | - Kuijie Zhang
- Qingdao Institute of Software, College of Computer Science and Technology, State Key Laboratory of Chemical Safety, Shandong Key Laboratory of Intelligent Oil & Gas Industrial Software, China University of Petroleum (East China), Qingdao 266580, China
| | - Hengxiao Li
- Qingdao Institute of Software, College of Computer Science and Technology, State Key Laboratory of Chemical Safety, Shandong Key Laboratory of Intelligent Oil & Gas Industrial Software, China University of Petroleum (East China), Qingdao 266580, China
| | - Sibo Qiao
- School of software, Tiangong university, Tianjin 300387, China
| | - Yuanyuan Zhang
- The College of Information and Control Engineering, Qingdao University of Technology, Qingdao, Shandong 266520, China
| | - Shanchen Pang
- Qingdao Institute of Software, College of Computer Science and Technology, State Key Laboratory of Chemical Safety, Shandong Key Laboratory of Intelligent Oil & Gas Industrial Software, China University of Petroleum (East China), Qingdao 266580, China
| |
Collapse
|
42
|
Zhou J, He M, Zhao Q, Shi E, Wang H, Ponkshe V, Song J, Wu Z, Ji D, Kranz G, Tscherne A, Schwenk-Zieger S, Razak NA, Hess J, Belka C, Zitzelsberger H, Ourailidis I, Stögbauer F, Boxberg M, Budczies J, Reichel CA, Canis M, Baumeister P, Wang H, Unger K, Mock A, Gires O. EGFR-mediated local invasiveness and response to Cetuximab in head and neck cancer. Mol Cancer 2025; 24:94. [PMID: 40121428 PMCID: PMC11929204 DOI: 10.1186/s12943-025-02290-1] [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: 10/09/2024] [Accepted: 03/04/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Recurrent/metastatic head and neck squamous cell carcinoma (R/M-HNSCC) is a severe, frequently lethal condition. Oncogene addiction to epidermal growth factor receptor (EGFR) is a hallmark of HNSCC, but the clinical efficacy of EGFR-targeted therapies remains low. Understanding molecular networks governing EGFR-driven progression is paramount to the exploration of (co)-treatment targets and predictive markers. METHODS We performed function-based mapping of differentially expressed genes in EGFR-mediated local invasion (fDEGs) using photoconvertible tracers and RNA-sequencing (RNA-seq) in a cellular 3D-model. RESULTS Upon alignment with public single-cell RNA-seq (scRNA-seq) datasets and HNSCC-specific regulons, a gene regulatory network of local invasion (invGRN) was inferred from gene expression data, which was overrepresented in budding tumors. InvGRN comprises the central hubs inhibin subunit beta alpha (INHBA) and snail family transcriptional repressor 2 (SNAI2), and druggable fDEGs integrin subunit beta 4 (ITGB4), laminin 5 (LAMB3/LAMC2), and sphingosine kinase 1 (SPHK1). Blockade of INHBA repressed local invasion and was reverted by activin A, laminin 5, and sphingosine-1-phosphate, demonstrating a functional interconnectivity of the invGRN. Epithelial-to-mesenchymal transition (EMT) of malignant cells and the invGRN are induced by newly defined EGFR-activity subtypes with prognostic value that are promoted by amphiregulin (AREG) and epiregulin (EREG). Importantly, co-inhibition of SPHK1 showed synthetic effects on Cetuximab-mediated invasion blockade and high expression of selected fDEGs was associated with response to Cetuximab in patient-derived xenotransplantation (PDX) and R/M-HNSCC patients. CONCLUSIONS We describe an actionable network of EGFR-mediated local invasion and define druggable effectors with predictive potential regarding the response of R/M-HNSCC to Cetuximab.
Collapse
Affiliation(s)
- Jiefu Zhou
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Sports Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha, 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
| | - Min He
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Qiong Zhao
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Enxian Shi
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Dermatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, Guangdong, China
| | - Hairong Wang
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Vaidehi Ponkshe
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Jiahang Song
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Zhengquan Wu
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Dongmei Ji
- Department of Medical Oncology, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Gisela Kranz
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Anna Tscherne
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sabina Schwenk-Zieger
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Nilofer Abdul Razak
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Julia Hess
- Research Unit Translational Metabolic Oncology, Institute for Diabetes and Cancer, Helmholtz Zentrum München, Deutsches Forschungszentrum Für Gesundheit Und Umwelt (GmbH), Neuherberg, Germany
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- German Cancer Consortium (DKTK), Partner Site, Munich, Germany
- Comprehensive Cancer Center (CCC), Munich, Germany
| | - Horst Zitzelsberger
- Research Unit Translational Metabolic Oncology, Institute for Diabetes and Cancer, Helmholtz Zentrum München, Deutsches Forschungszentrum Für Gesundheit Und Umwelt (GmbH), Neuherberg, Germany
| | - Iordanis Ourailidis
- Institute of Pathology, University of Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Fabian Stögbauer
- Technical University of Munich, TUM School of Medicine and Health, Institute of General and Surgical Pathology, Munich, Germany
| | - Melanie Boxberg
- German Cancer Consortium (DKTK), Partner Site, Munich, Germany
| | - Jan Budczies
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Christoph A Reichel
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Martin Canis
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Philipp Baumeister
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Hongxia Wang
- Department of Medical Oncology, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Kristian Unger
- Research Unit Translational Metabolic Oncology, Institute for Diabetes and Cancer, Helmholtz Zentrum München, Deutsches Forschungszentrum Für Gesundheit Und Umwelt (GmbH), Neuherberg, Germany
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- German Cancer Consortium (DKTK), Partner Site, Munich, Germany
- Comprehensive Cancer Center (CCC), Munich, Germany
| | - Andreas Mock
- Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Olivier Gires
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
43
|
Wang B, Zhou X, Wu D, Gao L, Wan Z, Wu R. Development and validation of M2 macrophage-related genes in a prognostic model of lung adenocarcinoma based on bulk RNA and ScRNA datasets. Discov Oncol 2025; 16:352. [PMID: 40100580 PMCID: PMC11920479 DOI: 10.1007/s12672-025-02123-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 03/11/2025] [Indexed: 03/20/2025] Open
Abstract
OBJECTIVE This study aimed to investigate the correlation between M2 macrophages activity with the prognosis of lung adenocarcinoma (LUAD). We sought to identify key genes associated with M2 macrophage activity and examine their relationship with clinicopathological features to elucidate the underlying mechanism. METHODS Published datases were analyzed for differentially expressed genes. After quality control, batch effect removal, and annotation, the scRNA dataset identified M2 macrophage-associated differentially expressed genes in the LUAD group, which were cross-analyzed and referred to as M2 macrophage-linked genes. A risk model was generated using machine learing for these genes. Thereafter, two bulk RNA-seq datasets were used to evaluate the model. We computed risk scores for all samples and grouped them into low and high risk, aiding in the comparison of clinical characteristics, immune and stromal infiltration, and drug sensitivity. Finally, key genes were validated through immunohistochemistry in IPA samples. RESULTS We identified four key M2 macrophage-linked genes: TIMP1, CAV2, MIF, and SELENBP1. Survival durations in the high-riskscore cluster were lower across the TCGA-LUAD (P = 1.2 × 10-4), GSE14814 (P = 0.02), and GSE37745 (P = 0.01) data sets. The stromal score, fibroblast infiltration, and cytokinesis activation were increased in the high-risk subgroup. Neutrophil and endothelial cell infiltration and activation of the linolenic acid pathway occurred in the low-risk group. IHC confirmed that CAV2 and SELENBP1 expression was significantly reduced, while TIMP1 and MIF were significantly increased in LUAD, which was consistent with the bioinformatics findings. CONCLUSION The role of M2 macrophages in tumor progression could anticipate the prognosis of LUAD and develop novel immunotherapy strategies.
Collapse
Affiliation(s)
- Bolin Wang
- Graduate School of Chengde Medical College, Chengde, Hebei, China
- Department of Chest Surgery, Baoding First Central Hospital, Baoding, Hebei, China
| | - Xiaofeng Zhou
- Department of Chest Surgery, Baoding First Central Hospital, Baoding, Hebei, China
| | - Di Wu
- Department of Chest Surgery, Baoding First Central Hospital, Baoding, Hebei, China
| | - Lu Gao
- Department of Chest Surgery, Baoding First Central Hospital, Baoding, Hebei, China
| | - Zhihua Wan
- Department of Chest Surgery, Baoding First Central Hospital, Baoding, Hebei, China
| | - Ruifeng Wu
- Department of Chest Surgery, Baoding First Central Hospital, Baoding, Hebei, China.
| |
Collapse
|
44
|
Zhao X, Zhang Y, Jia H, Lv L, Ahsan M, Fu X, Hu R, Shen Z, Shen N. Diversities of African swine fever virus host-virus dynamics revealed by single-cell profiling. J Virol 2025; 99:e0203524. [PMID: 39932318 PMCID: PMC11917525 DOI: 10.1128/jvi.02035-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/08/2025] [Indexed: 03/19/2025] Open
Abstract
African swine fever virus (ASFV) causes epidemics with high mortality; however, effective vaccines and therapies remain missing. Here, we depict a temporal single-cell landscape of primary porcine alveolar macrophages (PAMs) exposed to three different virulent ASFV strains in vitro. We found that attenuated and low-virulence ASFV strains tend to exhibit higher viral loads than highly virulent strain, which may result from upregulated RNA polymerase subunit genes expression. On the host side, our study highlights the IRF7-mediated positive feedback loop to the activation of the interferon signaling pathway in cells exposed to attenuated and low virulent ASFV strains. Moreover, we unraveled the PAMs populations marked by expressions of the IFI16 and CD163, respectively, which produce high levels of interferon-stimulated genes (ISGs) and IL18 to regulate the host response to different virulent ASFV strains. Collectively, our data provide insights into the complex host-virus interactions with various ASFV strain infections, which may shed light on the development of effective antiviral strategies.IMPORTANCEThere is still no available research on the temporal transcriptional profile of host cells exposed to different virulent ASFV strains at the single-cell level. Here, we first profiled the temporal viral and host transcriptomes in PAMs exposed to high virulent, attenuated virulent, and low virulent ASFV strains. Our analysis revealed that attenuated and low-virulence ASFV strains tend to exhibit higher viral loads than highly virulent strains, which may result from upregulated RNA polymerase subunit genes expression. We also found a positive feedback loop of the interferon signaling pathway mediated through IRF7 and identified the populations of PAMs marked by IFI6 and CD163, respectively, which produce high levels of ISGs and IL18 to regulate host response to different virulent ASFV strains. Our study delineated a comprehensive single-cell landscape of host-virus dynamics across ASFV strains with different virulences and would provide an important resource for future research.
Collapse
MESH Headings
- African Swine Fever Virus/pathogenicity
- African Swine Fever Virus/physiology
- African Swine Fever Virus/genetics
- Animals
- Swine
- Single-Cell Analysis
- African Swine Fever/virology
- African Swine Fever/immunology
- African Swine Fever/genetics
- Macrophages, Alveolar/virology
- Macrophages, Alveolar/metabolism
- Macrophages, Alveolar/immunology
- Host-Pathogen Interactions
- Interferon Regulatory Factor-7/metabolism
- Interferon Regulatory Factor-7/genetics
- Viral Load
- CD163 Antigen
- Signal Transduction
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Interferons/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Virulence
Collapse
Affiliation(s)
- Xiaoyang Zhao
- Department of
Obstetrics and Gynecology of Sir Run Run Shaw Hospital & Liangzhu
Laboratory, Zhejiang University School of
Medicine, Hangzhou,
China
| | - Yanyan Zhang
- Changchun Veterinary
Research Institute, Chinese Academy of Agricultural
Sciences, Changchun,
China
| | - Hanying Jia
- Liangzhu Laboratory,
Zhejiang University School of Medicine, Hangzhou,
China
| | - Lin Lv
- Department of
Obstetrics and Gynecology of Sir Run Run Shaw Hospital & Liangzhu
Laboratory, Zhejiang University School of
Medicine, Hangzhou,
China
| | - Md.Asif Ahsan
- Department of
Obstetrics and Gynecology of Sir Run Run Shaw Hospital & Liangzhu
Laboratory, Zhejiang University School of
Medicine, Hangzhou,
China
| | - Xudong Fu
- Department of
Obstetrics and Gynecology of Sir Run Run Shaw Hospital & Liangzhu
Laboratory, Zhejiang University School of
Medicine, Hangzhou,
China
| | - Rongliang Hu
- Changchun Veterinary
Research Institute, Chinese Academy of Agricultural
Sciences, Changchun,
China
| | - Zhiqiang Shen
- Shandong Lvdu
Bio-Sciences and Technology Co., Ltd.,
Binzhou, Shandong,
China
- Shandong Binzhou
Academy of Animal Science and Veterinary Medicine, Shandong Academy of
Agricultural Sciences, Binzhou,
Shandong, China
| | - Ning Shen
- Department of
Obstetrics and Gynecology of Sir Run Run Shaw Hospital & Liangzhu
Laboratory, Zhejiang University School of
Medicine, Hangzhou,
China
| |
Collapse
|
45
|
Zhou W, Ruan H, Zhu L, Chen S, Yang M. Unveiling a Novel Glioblastoma Deep Molecular Profiling: Insight into the Cancer Cell Differentiation-Related Mechanisms. ACS OMEGA 2025; 10:10230-10250. [PMID: 40124014 PMCID: PMC11923693 DOI: 10.1021/acsomega.4c09586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/27/2025] [Accepted: 02/19/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND the sophisticated cellular heterogeneity of cell populations in glioblastoma (GBM) has been a key factor influencing tumor progression and response to therapy. The lack of more precise stratification based on cellular differentiation status poses a great challenge to therapeutic strategies. MATERIALS AND METHODS harnessing the bulk multiomics and single-nucleus RNA sequencing data available from the National Center for Biotechnology Information (NCBI) and The Cancer Genome Atlas (TCGA) Program repositories, we developed a novel and accurate GBM risk classification using an ensemble consensus clustering approach based on the junction of prognosis and trajectory analysis. Comprehensive cluster labeling and multiomics data characterization were also performed. RESULTS a novel GBM stratification model was constructed using 45 malignant cell fate genes: (a) energy metabolism-enhanced-type GBM; (b) invasion-enhanced-type GBM; (c) invasion-attenuated-type GBM; and (d) glycolysis-dominant energy metabolism-enhanced-type GBM. The biological plausibility of the model was verified through a range of comprehensive analyses of multiomics data, showing that cases with invasion-attenuated-type were the best prognosis and energy metabolism-enhanced-type the poorest. CONCLUSIONS the study has uncovered GBM complex cellular heterogeneity and a differentiated hierarchy of cell populations underlying tumorigenesis. This precise stratification system provided implications for further studies of individual therapies.
Collapse
Affiliation(s)
- Weili Zhou
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Hongtao Ruan
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Lihua Zhu
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Shunqiang Chen
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Muyi Yang
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| |
Collapse
|
46
|
Li X, Zhang X, Liu T, Zhang G, Chen D, Lin S. Identification of immune characteristic biomarkers and therapeutic targets in cuproptosis for rheumatoid arthritis by integrated bioinformatics analysis and single-cell RNA sequencing analysis. Front Med (Lausanne) 2025; 12:1520400. [PMID: 40166070 PMCID: PMC11955502 DOI: 10.3389/fmed.2025.1520400] [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: 11/01/2024] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Rheumatoid arthritis (RA) is a chronic autoimmune disorder intricately liked with inflammation. Cuproptosis, an emerging type of cell death, has been implicated in the initiation and development of RA. However, the exact alterations in the expression and biological function of cuproptosis-related genes (CRGs) in RA remain poorly understood. Therefore, our study aims to elucidate the potential association between CRGs and RA, with the goal of identifying novel biomarkers for the treatment and prognosis of RA. Methods In this study, we identified ten differentially expressed cuproptosis-related genes (DE-CRGs) between patients with RA and controls. Through comprehensive functional enrichment and protein-protein interaction (PPI) network analysis, we explored the functional roles of the DE-CRGs. Additionally, we investigated the correlation between DE-CRGs and immune infiltration, immune factors, diagnostic efficacy, and potential therapeutic drugs. Results Leveraging single-cell RNA sequencing data, we conducted a detailed analysis to elucidate alterations in various cell clusters associated with RA. Our study unveiled a significant association between DE-CRGs and diverse biological functions, as well as potential drug candidates. Discussion These findings provide crucial insights into the involvement of DE-CRGs in the pathogenesis of RA and shed light on potential therapeutic strategies.
Collapse
Affiliation(s)
- Xianbin Li
- School of Computer and Big Data Science, Jiujiang University, Jiujiang, China
- Department of Digital Media Technology, Hangzhou Dianzi University, Hangzhou, China
- Jiujiang Key Laboratory of Digital Technology, Jiujiang, China
| | - Xueli Zhang
- Department of Medical Technology, Zhengzhou Railway Vocational and Technical College, Zhengzhou, China
| | - Tao Liu
- School of Computer and Big Data Science, Jiujiang University, Jiujiang, China
| | - Guodao Zhang
- Department of Digital Media Technology, Hangzhou Dianzi University, Hangzhou, China
| | - Dan Chen
- Department of Rheumatology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Suxian Lin
- Department of Rheumatology, Wenzhou People’s Hospital, Wenzhou, China
| |
Collapse
|
47
|
Kamel M, Song Y, Solbas A, Villordo S, Sarangi A, Senin P, Sunaal M, Ayestas LC, Levin C, Wang S, Classe M, Bar-Joseph Z, Pla Planas A. ENACT: End-to-End Analysis of Visium High Definition (HD) Data. Bioinformatics 2025; 41:btaf094. [PMID: 40053700 PMCID: PMC11925495 DOI: 10.1093/bioinformatics/btaf094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/28/2025] [Accepted: 02/25/2025] [Indexed: 03/09/2025] Open
Abstract
MOTIVATION Spatial transcriptomics (ST) enables the study of gene expression within its spatial context in histopathology samples. To date, a limiting factor has been the resolution of sequencing based ST products. The introduction of the Visium High Definition (HD) technology opens the door to cell resolution ST studies. However, challenges remain in the ability to accurately map transcripts to cells and in assigning cell types based on the transcript data. RESULTS We developed ENACT, a self-contained pipeline that integrates advanced cell segmentation with Visium HD transcriptomics data to infer cell types across whole tissue sections. Our pipeline incorporates novel bin-to-cell assignment methods, enhancing the accuracy of single-cell transcript estimates. Validated on diverse synthetic and real datasets, our approach is both scalable to samples with hundreds of thousands of cells and effective, offering a robust solution for spatially resolved transcriptomics analysis. AVAILABILITY AND IMPLEMENTATION ENACT source code is available at https://github.com/Sanofi-Public/enact-pipeline. Experimental data are available at https://zenodo.org/records/14748859.
Collapse
Affiliation(s)
- Mena Kamel
- Digital R&D, Sanofi, Toronto, ON M5V 1V6, Canada
| | - Yiwen Song
- Digital R&D, Sanofi, Toronto, ON M5V 1V6, Canada
| | - Ana Solbas
- Digital R&D, Sanofi, Barcelona 08016, Spain
| | | | | | - Pavel Senin
- Digital R&D, Sanofi, Toronto, ON M5V 1V6, Canada
| | | | - Luis Cano Ayestas
- Precision Medicine & Computational Biology, Sanofi, Paris 94400, France
| | - Clement Levin
- Precision Medicine & Computational Biology, Sanofi, Paris 94400, France
| | - Seqian Wang
- Digital R&D, Sanofi, Toronto, ON M5V 1V6, Canada
| | - Marion Classe
- Precision Medicine & Computational Biology, Sanofi, Paris 94400, France
| | | | | |
Collapse
|
48
|
Ge X, Zhu X, Liu W, Li M, Zhang Z, Zou M, Deng M, Cui H, Chen Z, Wang L, Hu X, Ju R, Tang X, Ding X, Gong L. cGAMP promotes inner blood-retinal barrier breakdown through P2RX7-mediated transportation into microglia. J Neuroinflammation 2025; 22:58. [PMID: 40025497 PMCID: PMC11871612 DOI: 10.1186/s12974-025-03391-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/20/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Impairment of the inner blood-retinal barrier (iBRB) leads to various blinding diseases including diabetic retinopathy (DR). The cGAS-STING pathway has emerged as a driving force of cardiovascular destruction, but its impact on the neurovascular system is unclear. Here, we show that cGAMP, the endogenous STING agonist, causes iBRB breakdown and retinal degeneration thorough P2RX7-mediated transport into microglia. METHODS Extracellular cGAMP and STING pathway were determined in tissue samples from patients with proliferative DR (PDR) and db/db diabetic mice. Histological, molecular, bioinformatic and behavioral analysis accessed effects of cGAMP on iBRB. Single-cell RNA sequencing identified the primary retinal cell type responsive to cGAMP. Specific inhibitors and P2RX7-deficienct mice were used to evaluate P2RX7' role as a cGAMP transporter. The therapeutic effects of P2RX7 inhibitor were tested in db/db mice. RESULTS cGAMP was detected in the aqueous humor of patients with PDR and elevated in the vitreous humor with STING activation in db/db mouse retinas. cGAMP administration led to STING-dependent iBRB breakdown and neuron degeneration. Microglia were the primary cells responding to cGAMP, essential for cGAMP-induced iBRB breakdown and visual impairment. The ATP-gated P2RX7 transporter was required for cGAMP import and STING activation in retinal microglia. Contrary to previous thought that mouse P2RX7 nonselectively transports cGAMP only at extremely high ATP concentrations, human P2RX7 directly binds to cGAMP and activates STING under physiological conditions. Clinically, cGAMP-induced microglial signature was recapitulated in fibrovascular membranes from patients with PDR, with P2RX7 being predominantly expressed in microglia. Inhibiting P2RX7 reduced cGAMP-STING activation, protected iBRB and improved neuron survival in diabetic mouse retinas. CONCLUSIONS Our study reveals a mechanism for cGAMP-mediated iBRB breakdown and suggests that targeting microglia and P2RX7 may mitigate the deleterious effects of STING activation in retinal diseases linked to iBRB impairment.
Collapse
Affiliation(s)
- Xiangyu Ge
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xingfei Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Wei Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Mingsen Li
- Interdisciplinary Eye Research Institute (EYE-X Institute), Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, China
| | - Zhaotian Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Ming Zou
- Health Science Center, Peking University International Cancer Institute, Peking University, Beijing, China
| | - Mi Deng
- Health Science Center, Peking University International Cancer Institute, Peking University, Beijing, China
- Peking University Cancer Hospital and Institute, Peking University, Beijing, China
| | - Haifeng Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Ziqing Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Li Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xuebin Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Rong Ju
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xiangcheng Tang
- Shenzhen Eye Hospital, Shenzhen Eye Medical Center, Southern Medical University, Shenzhen, 518040, Guangdong, China.
| | - Xiaoyan Ding
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China.
| | - Lili Gong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
49
|
Cheng B, Wen Y, Wei W, Cheng S, Pan C, Meng P, Liu L, Yang X, Liu H, Jia Y, Zhang F. Polygenic enrichment analysis in multi-omics levels identifies cell/tissue specific associations with schizophrenia based on single-cell RNA sequencing data. Schizophr Res 2025; 277:93-101. [PMID: 40036903 DOI: 10.1016/j.schres.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 01/24/2025] [Accepted: 02/22/2025] [Indexed: 03/06/2025]
Abstract
OBJECTIVE Understanding the specific cellular origin and tissue heterogeneity in schizophrenia is critically important for exploring the disease etiology. This study aims to investigate these aspects by performing multiple analyses based on omics data. METHOD We performed single-cell disease relevance score (scDRS) algorithm to link brain single-cell RNA sequencing (scRNA-seq) with schizophrenia risk across multi-omics scales at single-cell resolution. This approach identified cell types with overexpression of schizophrenia-related genes implicated by multi-omics panels (ATAC-seq, RNA-seq, TWAS, and GWAS). Schizophrenia-related genes from these multi-omics panels were extracted and combined with scRNA-seq data to calculate scDRS. Subsequently, the cell-type vs. disease association and tissue heterogeneity were assessed using scDRS for each omics panel. RESULTS We identified two novel cell subpopulations in the brain that differentially express SCUBE3 (59 cells, 7.0 %) and FN1 (21 cells, 2.5 %). At the individual cell level, schizophrenia-associated cell subpopulations included microglial cell associated with ATAC-seq panel (Passociation = 0.002, Pheterogeneity = 0.009) and deep layer neuron suggestively associated with GWAS panel (Passociation = 0.033, Pheterogeneity = 0.017). At the brain tissue level, microglial cell was significantly associated with cortical plate in ATAC-seq panel (Passociation = 0.002, Pheterogeneity = 0.011). Gene level analysis identified several genes associated with schizophrenia across multi-omics panels. CONCLUSIONS Our study outlines the signature of cell subpopulations, brain regions, and disease risk genes in schizophrenia at single-cell resolution across multi-omics scales. These findings provide a reference for future precision medicine approaches targeting specific cell types and brain regions in schizophrenia.
Collapse
Affiliation(s)
- Bolun Cheng
- NHC Key Laboratory of Environment and Endemic Diseases (Xi'an Jiaotong University), Xi'an 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China; Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 710061, China
| | - Yan Wen
- NHC Key Laboratory of Environment and Endemic Diseases (Xi'an Jiaotong University), Xi'an 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China; Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 710061, China
| | - Wenming Wei
- NHC Key Laboratory of Environment and Endemic Diseases (Xi'an Jiaotong University), Xi'an 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China; Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 710061, China
| | - Shiqiang Cheng
- NHC Key Laboratory of Environment and Endemic Diseases (Xi'an Jiaotong University), Xi'an 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China; Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 710061, China
| | - Chuyu Pan
- NHC Key Laboratory of Environment and Endemic Diseases (Xi'an Jiaotong University), Xi'an 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China; Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 710061, China
| | - Peilin Meng
- NHC Key Laboratory of Environment and Endemic Diseases (Xi'an Jiaotong University), Xi'an 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China; Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 710061, China
| | - Li Liu
- NHC Key Laboratory of Environment and Endemic Diseases (Xi'an Jiaotong University), Xi'an 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China; Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 710061, China
| | - Xuena Yang
- NHC Key Laboratory of Environment and Endemic Diseases (Xi'an Jiaotong University), Xi'an 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China; Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 710061, China
| | - Huan Liu
- NHC Key Laboratory of Environment and Endemic Diseases (Xi'an Jiaotong University), Xi'an 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China; Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 710061, China
| | - Yumeng Jia
- NHC Key Laboratory of Environment and Endemic Diseases (Xi'an Jiaotong University), Xi'an 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China; Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 710061, China.
| | - Feng Zhang
- NHC Key Laboratory of Environment and Endemic Diseases (Xi'an Jiaotong University), Xi'an 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China; Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 710061, China.
| |
Collapse
|
50
|
Puertas-Umbert L, Alonso J, Blanco-Casoliva L, Almendra-Pegueros R, Camacho M, Rodríguez-Sinovas A, Galán M, Roglans N, Laguna JC, Martínez-González J, Rodríguez C. Inhibition of ATP-citrate lyase by bempedoic acid protects against abdominal aortic aneurysm formation in mice. Biomed Pharmacother 2025; 184:117876. [PMID: 39889383 DOI: 10.1016/j.biopha.2025.117876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025] Open
Abstract
Abdominal aortic aneurysm (AAA) is a prevalent degenerative disease characterized by an exacerbated inflammation and destructive vascular remodeling. Unfortunately, effective pharmacological tools for the treatment of this disease remain a challenge. ATP-citrate lyase (ACLY), the primary enzyme responsible for acetyl-CoA biosynthesis, is a key regulator of inflammatory signaling in macrophages and lymphocytes. Here, we found increased levels of the active (phosphorylated) form of ACLY (p-ACLY) in the inflammatory infiltrate of AAA from patients and in aneurysmal lesions from angiotensin II (Ang II)-infused apolipoprotein E-deficient mice (ApoE-/-). Furthermore, plasma ACLY levels positively correlates with IL6 and IFNγ levels in patients with AAA, while inflammatory stimuli strongly upregulated ACLY expression in macrophages and Jurkat cells. The administration of the ACLY inhibitor bempedoic acid (BemA) protected against Ang II-induced AAA formation in ApoE-/- mice, limiting the progression of aortic dilatation and reducing mortality due to aortic rupture. BMS-303141, another ACLY inhibitor, also ameliorated AAA formation, although to a lesser extent. BemA attenuated vascular remodeling and the disorganization and rupture of elastic fibers induced by Ang II, as well as vascular inflammation, decreasing the recruitment of macrophages (CD68 +) and neutrophils (Ly-6G+) into the aortic wall. Moreover, BemA shifted splenic monocytes toward a functionally anti-inflammatory phenotype, and increased the percentage of CD4+CD69+ cells. Taken together, these results support the contribution of ACLY to AAA and point to BemA as a promising tool to be considered for future clinical trials addressing the management of this disease which is quite often associated with disorders of lipoprotein metabolism.
Collapse
Affiliation(s)
- Lídia Puertas-Umbert
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona 08036, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Judith Alonso
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona 08036, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Laia Blanco-Casoliva
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona 08036, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain
| | | | - Mercedes Camacho
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Antonio Rodríguez-Sinovas
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain; Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Barcelona 08035, Spain
| | - María Galán
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain; Facultad de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, Madrid 28922, Spain
| | - Nuria Roglans
- Dept. Farmacologia, Toxicologia i Química Terapèutica. Facultat de Farmàcia i Ciències de l'Alimentació, Institut de Biomedicina, Universitat de Barcelona, Barcelona 08028, Spain; CIBER de Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Juan Carlos Laguna
- Dept. Farmacologia, Toxicologia i Química Terapèutica. Facultat de Farmàcia i Ciències de l'Alimentació, Institut de Biomedicina, Universitat de Barcelona, Barcelona 08028, Spain; CIBER de Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - José Martínez-González
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona 08036, Spain; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain.
| | - Cristina Rodríguez
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid 28029, Spain.
| |
Collapse
|