1
|
Chen C, Ma Y, Gao Y, Ge H, Zhang X. Prognostic significance of neutrophil extracellular trap-related genes in childhood acute lymphoblastic leukemia: insights from multi-omics and in vitro experiment. Hematology 2025; 30:2452701. [PMID: 39829399 DOI: 10.1080/16078454.2025.2452701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND This study aimed to develop a prognostic model based on extracellular trap-related genes (NETRGs) for patients with cALL. METHODS Data from the TARGET-ALL-P2 and TARGET-ALL-P3 cohorts in the Genomic Data Commons database, the transcriptome dataset GSE26713, the single-cell transcriptome dataset GSE130116 from the Gene Expression Omnibus database and 306 NETRGs identified were analysed. Differentially expressed genes (DEGs) were identified from GSE26713 and differentially expressed NETRGs (DE-NETRGs) were obtained by overlapping DEGs with NETRGs. Functional analyses were conducted. Key feature genes were identified through univariate and least absolute shrinkage and selection operator (LASSO) regression. Prognostic genes were determined via multivariate Cox regression analysis, followed by the construction and validation of a risk model and nomogram. Additional analyses included immune profiling, drug sensitivity, functional differences, cell-type-specific expression, enrichment analysis and RT-qPCR. RESULTS A total of 1,270 DEGs were identified in GSE26713, of which 74 overlapped with NETRGs. Seven prognostic genes were identified using univariate, LASSO and multivariate Cox regression analyses. Survival analysis revealed lower survival rates in the high-risk group. Independent prognostic analysis identified risk scores and primary diagnosis as independent predictors of prognosis. Immune cell profiling showed significant differences in cell populations such as aDCs, eosinophils and Th2 cells between risk groups. Six cell subtypes were annotated, with prognostic genes predominantly expressed in myeloid cells. RT-qPCR revealed that PTAFR, FCGR2A, RETN and CAT were significantly downregulated, while TLR2 and S100A12 were upregulated in cALL. CONCLUSION TLR2, PTAFR, FCGR2A, RETN, S100A12 and CAT may serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Pediatrics, Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), Yinchuan, People's Republic of China
| | - Yu Ma
- Department of Pediatrics, Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), Yinchuan, People's Republic of China
| | - Yadai Gao
- Department of Pediatrics, Yinchuan Women and Children Healthcare Hospital, Yinchuan, People's Republic of China
| | - Huiqing Ge
- Department of Pediatrics, Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), Yinchuan, People's Republic of China
| | - Xiaochun Zhang
- Department of Pediatrics, Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), Yinchuan, People's Republic of China
| |
Collapse
|
2
|
Li X, Zhu L. Comprehensive profiling of cell type-specific expression and distribution of complement genes in mouse and human kidneys: insights into normal physiology and response to kidney transplantations. Ren Fail 2025; 47:2471568. [PMID: 40015727 PMCID: PMC11869339 DOI: 10.1080/0886022x.2025.2471568] [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: 07/24/2024] [Revised: 01/24/2025] [Accepted: 02/15/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Recent studies innovatively revealed the localized expression of complement genes in kidneys and shed light on the vital roles of the intracellular complement system in the physiologic function and pathological conditions. However, a comprehensive analysis of the expression of complement genes in the context of the evolving cellular landscape of the kidney is not available. METHODS We analyzed single-cell RNA sequencing data from healthy human subjects, C57BL/6 mice, and kidney transplant-rejected mice. The data were sourced from the NCBI Gene Expression Omnibus and processed using quality control measures and unsupervised clustering. Differential gene analyses were based on expression levels. RESULTS In total, 50 complement genes were categorized into pattern recognition molecules, proteases, complement components, receptors, and regulators. In normal mice kidneys, complement genes were expressed at relatively low levels. Among different complement gene categories, receptor genes were most widely expressed in kidney cells. Comparatively, macrophages and mesangial cells are the most abundant immune and nonimmune cell types for complement gene expression. A comparison of human and mouse data showed similar expression patterns, but human kidney complement gene expression was more abundant. Comparative analysis between mouse transplant-rejected and normal kidneys demonstrated stronger complement gene expression in transplant-rejected kidneys. CONCLUSIONS This study illustrated significant similarities in complement gene expression between murine and human kidneys and highlighted the responsive nature of complement genes to kidney injury, underscoring the dynamic nature of local complement regulation. These findings enhance our understanding of the complex regulation of the complement system within the kidney, offering insights into its role in renal disease pathogenesis.
Collapse
Affiliation(s)
- Xianzhi Li
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease (Peking University), National Health Commission, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Li Zhu
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease (Peking University), National Health Commission, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| |
Collapse
|
3
|
Xu JX, Su YX, Chen YY, Huang YY, Chen ZS, Peng YC, Qi LN. Immune infiltration landscape and potential drug-targeted implications for hepatocellular carcinoma with 'progression/hyper-progression' recurrence. Ann Med 2025; 57:2456113. [PMID: 39865865 PMCID: PMC11774162 DOI: 10.1080/07853890.2025.2456113] [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: 01/03/2024] [Revised: 12/20/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND AND AIMS Hepatocellular carcinoma (HCC) recurrence was previously characterized into four types, and patients with progression/hyper-progression recurrence (type III-IV) have an extremely poor prognosis. However, the immune background of resectable HCC, particularly in patients who experience recurrence, remains underexplored. Therefore, this study aimed to describe the immune landscape of resectable HCC, especially postoperative type III-IV recurrent HCC, and explore potential immune-targeted anti-relapse strategies for treated populations. METHODS The differences in gene expression in patients with recurrent HCC (type I-II (solitary or multi-intrahepatic oligo recurrence) vs. type III-IV) were investigated using bulk sequencing. Multiple immune infiltration methods (single-sample gene set enrichment analysis (GSEA), Microenvironment Cell Populations-counter and ESTIMATE) were used, and patients were divided into four groups to identify four distinct immune subtypes: immune-enrichment/matrix-poor (IE1), immune-enrichment/matrix-rich (IE2), immune intermediate/matrix-rich (ITM) and immune desert/matrix-poor (ID). Co-expression and protein interaction analyses were used to identify characteristic genes in ITM closely associated with type III-IV recurrence, which was matched with drug targets for Huaier granules (HG) and lenvatinib. Virtual docking was used to identify potential therapeutic targets, and the results were verified using single-nuclei RNA sequencing and histological analysis. RESULTS ITM was closely related to type III-IV recurrence and exhibited immunotherapy potential. The potential efficacy of inhibiting CCNA2, VEGFA, CXCL8, PLK2, TIMP1, ITGB2, ALDOA, ANXA5 and CSK in ITM reversal was determined. Molecular docking demonstrated that the proteins of these genes could bind to HG or lenvatinib. The immunohistochemical findings demonstrated differential VEGFA (p < .01) and PLK2 (p < .001) expression in ITM type and ID in type III-IV recurrent HCC. CONCLUSIONS Three primary immunotypes of resectable HCC (IE2, ITM and ID) were identified, and HG and lenvatinib could potentially overcome immune checkpoint blockade (ICB) resistance in ITM patients with HCC, particularly those classified as type III-IV.
Collapse
Affiliation(s)
- Jing-Xuan Xu
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumour, Ministry of Education, Nanning, China
| | - Yue-Xiang Su
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumour, Ministry of Education, Nanning, China
| | - Yuan-Yuan Chen
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yi-Yue Huang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumour, Ministry of Education, Nanning, China
| | - Zu-Shun Chen
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yu-Chong Peng
- Department of General Surgery, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Lu-Nan Qi
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumour, Ministry of Education, Nanning, China
- Guangxi Liver Cancer Diagnosis and Treatment Engineering and Technology Research Center, Nanning, China
| |
Collapse
|
4
|
Lo Presti E, Cupaioli F, Scimeca D, Unti E, Di Martino V, Daidone R, Amata M, Scibetta N, Soucie E, Meraviglia S, Iovanna J, Dusetti N, De Gaetano A, Merelli I, Di Mitri R. The pancreatic tumor microenvironment of treatment-naïve patients causes a functional shift in γδ T cells, impairing their anti-tumoral defense. Oncoimmunology 2025; 14:2466301. [PMID: 39945298 PMCID: PMC11834455 DOI: 10.1080/2162402x.2025.2466301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 01/14/2025] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents a unique challenge for researchers due to its late diagnosis caused by vague symptoms and lack of early detection markers. Additionally, PDAC is characterized by an immunosuppressive microenvironment (TME), making it a difficult tumor to treat. While γδ T cells have shown potential for anti-tumor activity, conflicting studies exist regarding their effectiveness in pancreatic cancer. This study aims to explore the hypothesis that the PDAC TME hinders the anti-tumor capabilities of γδ T cells through blockade of cytotoxic functions. For this reason, we chose to enroll PDAC treatment-naive patients to avoid the possibility of therapy modifying the TME. By flow cytometry, our research findings indicate that the presence of γδ T cells among CD45+ cells in tumor tissue is lower compared to CD66+ cells, but higher than in blood. Circulating Vδ1 T cells exhibit a terminal effector memory phenotype (TEMRA) more than Vδ2 T cells. Interestingly, Vδ1 and Vδ2 T cells appear to be more prevalent at different stages of tumor development. In our in vitro culture using conditioned medium derived from Patient-derived organoids ;(PDOs), we observed a shift in expression markers in γδ T cells of healthy individuals toward an activation and exhaustion phenotype, as confirmed by scRNA-seq analysis extracted from a public database. A deeper understanding of γδ T cells in PDAC could be valuable for developing novel therapies aimed at mitigating the impact of the pancreatic tumor microenvironment on this cell population.
Collapse
Affiliation(s)
- Elena Lo Presti
- National Research Council of Italy (CNR), Institute for Biomedical Research and Innovation (IRIB), Palermo, Italy
| | - Francesca Cupaioli
- National Research Council of Italy, Bioinformatics Research Unit, Institute for Biomedical Technologies Segrate, Milan, Italy
| | - Daniela Scimeca
- Gastroenterology and Endoscopy Unit, Arnas Civico Di Cristina Benfratelli Hospital, Palermo, Italy
| | - Elettra Unti
- ‘Anatomic-pathology Unit, Arnas Civico Di Cristina Benfratelli Hospital, Palermo, Italy
| | - Vincenzo Di Martino
- Immunohaematology and Transfusion Medicine Unit, Imperia Hospital ASL1 Imperiese, Imperia, Italy
| | - Rossella Daidone
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Marseille, France
| | - Michele Amata
- Gastroenterology and Endoscopy Unit, Arnas Civico Di Cristina Benfratelli Hospital, Palermo, Italy
| | - Nunzia Scibetta
- ‘Anatomic-pathology Unit, Arnas Civico Di Cristina Benfratelli Hospital, Palermo, Italy
| | - Erinn Soucie
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Marseille, France
| | - Serena Meraviglia
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy
| | - Juan Iovanna
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Marseille, France
| | - Nelson Dusetti
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Marseille, France
| | - Andrea De Gaetano
- National Research Council of Italy (CNR), Institute for Biomedical Research and Innovation (IRIB), Palermo, Italy
- National Research Council of Italy, Institute for Systems Analysis and Computer Science “A. Ruberti, ” BioMatLab (Biomathematics Laboratory), Rome, Italy
- Department of Mathematics, Mahidol University, Bangkok, Thailand
| | - Ivan Merelli
- National Research Council of Italy, Bioinformatics Research Unit, Institute for Biomedical Technologies Segrate, Milan, Italy
| | - Roberto Di Mitri
- Gastroenterology and Endoscopy Unit, Arnas Civico Di Cristina Benfratelli Hospital, Palermo, Italy
| |
Collapse
|
5
|
Zhou HY, Wang X, Li Y, Wang D, Zhou XZ, Xiao N, Li GX, Li G. Dynamic development of microglia and macrophages after spinal cord injury. Neural Regen Res 2025; 20:3606-3619. [PMID: 39101644 PMCID: PMC11974661 DOI: 10.4103/nrr.nrr-d-24-00063] [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: 01/17/2024] [Revised: 04/09/2024] [Accepted: 05/28/2024] [Indexed: 08/06/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202512000-00029/figure1/v/2025-01-31T122243Z/r/image-tiff Secondary injury following spinal cord injury is primarily characterized by a complex inflammatory response, with resident microglia and infiltrating macrophages playing pivotal roles. While previous studies have grouped these two cell types together based on similarities in structure and function, an increasing number of studies have demonstrated that microglia and macrophages exhibit differences in structure and function and have different effects on disease processes. In this study, we used single-cell RNA sequencing and spatial transcriptomics to identify the distinct evolutionary paths of microglia and macrophages following spinal cord injury. Our results showed that microglia were activated to a pro-inflammatory phenotype immediately after spinal cord injury, gradually transforming to an anti-inflammatory steady state phenotype as the disease progressed. Regarding macrophages, our findings highlighted abundant communication with other cells, including fibroblasts and neurons. Both pro-inflammatory and neuroprotective effects of macrophages were also identified; the pro-inflammatory effect may be related to integrin β2 ( Itgb2 ) and the neuroprotective effect may be related to the oncostatin M pathway. These findings were validated by in vivo experiments. This research underscores differences in the cellular dynamics of microglia and macrophages following spinal cord injury, and may offer new perspectives on inflammatory mechanisms and potential therapeutic targets.
Collapse
Affiliation(s)
- Hu-Yao Zhou
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Xia Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Yi Li
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Duan Wang
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Xuan-Zi Zhou
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Nong Xiao
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Guo-Xing Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Gang Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Li H, Liu H, Wu H, Guo C, Zuo W, Zheng Y, Deng X, Xu J, Wang Y, Wang Z, Lu B, Hou B, Cao B. Reading of human acute immune dynamics in omicron SARS-CoV-2 breakthrough infection. Emerg Microbes Infect 2025; 14:2494705. [PMID: 40231451 PMCID: PMC12064115 DOI: 10.1080/22221751.2025.2494705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/19/2025] [Accepted: 04/13/2025] [Indexed: 04/16/2025]
Abstract
The dynamics of the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) breakthrough infections remain unclear, particularly when compared to responses in naive individuals. In this longitudinal prospective cohort study, 13 participants were recruited. Peripheral blood samples were collected every other day until day 7 after symptom onset. Transcriptome sequencing, single-cell sequencing, T-cell receptor (TCR) sequencing, B-cell receptor (BCR) sequencing, Olink proteomics, and antigen-antibody binding experiments were then performed. During the incubation periods of breakthrough infections, peripheral blood exhibited type 2 cytokine response, which shifted to type 1 cytokine response upon symptom onset. Plasma cytokine levels of C-X-C motif chemokine ligand 10, monocyte chemoattractant protein-1, interferon-γ, and interleukin-6 show larger changes in breakthrough infections than naïve infections. The inflammatory response in breakthrough infections rapidly subsided, returning to homeostasis by day 5 after symptom onset. Notably, the levels of monocyte-derived S100A8/A9, previously considered a marker of severe disease, physiologically significantly increased in the early stages of mild cases and persisted until day 7, suggesting a specific biological function. Longitudinal tracking also revealed that antibodies anti-Receptor Binding Domain (anti-RBD) in breakthrough infections significantly increased by day 7 after symptom onset, whereas cytotoxic T lymphocytes appeared by day 5. This study presents a reference for interpreting the immunological response to breakthrough infectious disease in humans.
Collapse
Affiliation(s)
- Haibo Li
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- New Cornerstone Science Laboratory, Beijing, People’s Republic of China
| | - Hongyu Liu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- Department of Respiratory Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Hongping Wu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Chang Guo
- Changping National Laboratory (CPNL), Beijing, People’s Republic of China
| | - Wenting Zuo
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Ying Zheng
- Department of Respiratory Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Xiaoyan Deng
- Tsinghua University-Peking University Joint Center for Life Sciences, Beijing, People’s Republic of China
| | - Jiuyang Xu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Yeming Wang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Zai Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Binghuai Lu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Baidong Hou
- State Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Bin Cao
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- New Cornerstone Science Laboratory, Beijing, People’s Republic of China
- Department of Respiratory Medicine, Capital Medical University, Beijing, People’s Republic of China
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| |
Collapse
|
7
|
Lu Y, Fang YX, Ou-Yang ZM, Wu T, Zhang Q, Zou YW, Zheng HW, Jing J, Lin LH, Ma JD, Liang Z, Dai L. Deficiency of FUN14 domain-containing 1 enhances the migration and invasion of fibroblast-like synoviocytes in rheumatoid arthritis through mitochondrial dysregulation. Cell Signal 2025; 132:111829. [PMID: 40274085 DOI: 10.1016/j.cellsig.2025.111829] [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: 11/22/2024] [Revised: 04/01/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Fibroblast-like synoviocytes (FLS) display aggressive phenotypes contributing to synovitis and joint destruction in rheumatoid arthritis (RA). Disrupted mitochondrial homeostasis has been proposed to aggravate the RA pathogenesis, however, the underlying mechanism remains to be elucidated. This study aimed to elucidate the role of mitophagy receptor FUN14 domain-containing 1 (FUNDC1) on RA-FLS migration and invasion. METHODS We analyzed the correlation of synovial FUNDC1 expression with joint destruction and disease activity in RA patients. Single cell sequencing data analysis combined with immunofluorescence indicated the specific expression and localization of FUNDC1 in synovial tissue and RA-FLS. The roles of FUNDC1 in the migration, invasion, and cytokine secretion of RA-FLS were examined by patient-derived primary culture as well as SCID mouse models. We investigated the effects and mechanism of FUNDC1 on mitophagy and mitochondrial quality control network in primary RA-FLS. RESULTS We found that the FUNDC1 was mainly expressed in FLS and exhibited a decreased level in RA synovium, which was correlated with severe joint destruction. Deficiency of FUNDC1 enhanced migration, invasion as well as secretion of matrix metalloproteinases in RA-FLS. On the contrary, overexpression of FUNDC1 in RA-FLS with low FUNDC1 inhibited the migration, invasion and secretion capacity of RA-FLS. Mechanistically, repressed FUNDC1 level in RA-FLS impaired mitophagy, imbalanced mitochondrial quality control, and increased mitochondrial reactive oxygen species (mtROS) production, leading to the overactivation of the MAPK pathway. Treatment with mtROS scavenger mtTEMPO can reverse this process and diminish the invasiveness of RA-FLS. CONCLUSIONS Deficiency of FUNDC1 dysregulates mitochondrial quality-control system and induces aggressive phenotype of RA-FLS, resulting in joint destruction during RA progression.
Collapse
Affiliation(s)
- Ye Lu
- Department of Rheumatology and Immunology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Ya-Xiong Fang
- Bioscience and Biomedical Engineering Thrust, Brain and Intelligence Research Institute, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou 511453, PR China
| | - Zhi-Ming Ou-Yang
- Department of Rheumatology and Immunology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Tao Wu
- Department of Rheumatology and Immunology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Qian Zhang
- Department of Rheumatology and Immunology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Yao-Wei Zou
- Department of Rheumatology and Immunology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Hu-Wei Zheng
- Department of Rheumatology and Immunology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Jun Jing
- Department of Rheumatology and Immunology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Le-Hang Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Jian-Da Ma
- Department of Rheumatology and Immunology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China.
| | - Zhuoyi Liang
- Bioscience and Biomedical Engineering Thrust, Brain and Intelligence Research Institute, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou 511453, PR China.
| | - Lie Dai
- Department of Rheumatology and Immunology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China.
| |
Collapse
|
8
|
Song C. Single-cell transcriptomic reveals network topology changes of cancer at the individual level. Comput Biol Chem 2025; 117:108401. [PMID: 40037020 DOI: 10.1016/j.compbiolchem.2025.108401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 02/15/2025] [Accepted: 02/21/2025] [Indexed: 03/06/2025]
Abstract
Network biology facilitates a better understanding of complex diseases. Single-sample networks retain individual information and have the potential to distinguish disease status. Previous studies mainly used bulk RNA sequencing data to construct single-sample networks, but different cell types in the tissue microenvironment perform significantly different functions. In this study, we investigated whether network topology features of cell-type-specific networks varied in different pathological states at the individual level. Protein-protein interaction network (PPI) and co-expression network of cancer and ulcerative colitis were established using four publicly single-cell RNA sequencing (scRNA-seq) datasets. We analyzed cell-cell interactions of epithelial cells and immune cells using CellChat R package. Network topology changes between normal tissues and pathological tissues were analyzed using Cytoscape software and QUACN R package. Results showed cell-cell interactions of epithelial cells were enhanced in carcinoma and adenoma. The average number of neighbors and graphindex of co-expression network increased in epithelial cells of adenoma, carcinoma and paracancer compared with normal tissues. The co-expression network density of T cells in tumors was significantly higher than that in normal tissues. The co-expression network complexity of epithelial cells in the benign tissues was associated with the grade group of paired tumors. This study suggests topological properties of cell-type-specific individual network vary in different pathological states, providing an insight into understanding complex diseases.
Collapse
Affiliation(s)
- Chenhui Song
- Chongqing Kingbiotech Corporation, Beijing, China.
| |
Collapse
|
9
|
Fortmann SD, Frey BF, Rosencrans RF, Adu-Rutledge Y, Ready V E, Kilchrist KV, Welner RS, Boulton ME, Saban DR, Grant MB. Prenatally derived macrophages support choroidal health and decline in age-related macular degeneration. J Exp Med 2025; 222:e20242007. [PMID: 40261298 PMCID: PMC12013653 DOI: 10.1084/jem.20242007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/02/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Hallmark findings in age-related macular degeneration (AMD) include the accumulation of extracellular lipid and vasodegeneration of the choriocapillaris. Choroidal inflammation has long been associated with AMD, but little is known about the immune landscape of the human choroid. Using 3D multiplex immunofluorescence, single-cell RNA sequencing, and flow cytometry, we unravel the cellular composition and spatial organization of the human choroid and the immune cells within it. We identify two populations of choroidal macrophages with distinct FOLR2 expression that account for the majority of myeloid cells. FOLR2+ macrophages predominate in the nondiseased eye, express lipid-handling machinery, uptake lipoprotein particles, and contain high amounts of lipid. In AMD, FOLR2+ macrophages are decreased in number and exhibit dysfunctional lipoprotein metabolism. In mice, FOLR2+ macrophages are negative for the postnatal fate-reporter Ms4a3, and their depletion causes an accelerated AMD-like phenotype. Our results show that prenatally derived resident macrophages decline in AMD and are implicated in multiple hallmark functions known to be compromised in the disease.
Collapse
Affiliation(s)
- Seth D. Fortmann
- Medical Scientist Training Program (MSTP), University of Alabama at Birmingham (UAB), Birmingham, AL, USA
- Department of Ophthalmology, UAB, Birmingham, AL, USA
| | - Blake F. Frey
- Medical Scientist Training Program (MSTP), University of Alabama at Birmingham (UAB), Birmingham, AL, USA
- Department of Pathology, UAB, Birmingham, AL, USA
| | - Robert F. Rosencrans
- Medical Scientist Training Program (MSTP), University of Alabama at Birmingham (UAB), Birmingham, AL, USA
- Department of Ophthalmology, UAB, Birmingham, AL, USA
| | | | - Edgar Ready V
- Department of Ophthalmology, UAB, Birmingham, AL, USA
| | | | - Robert S. Welner
- Division of Hematology/Oncology, Department of Medicine, UAB, Birmingham, AL, USA
| | | | - Daniel R. Saban
- Department of Ophthalmology, Duke University, Durham, NC, USA
| | | |
Collapse
|
10
|
Muñoz Sandoval D, Bach FA, Ivens A, Harding AC, Smith NL, Mazurczyk M, Themistocleous Y, Edwards NJ, Silk SE, Barrett JR, Cowan GJ, Napolitani G, Savill NJ, Draper SJ, Minassian AM, Nahrendorf W, Spence PJ. Plasmodium falciparum infection induces T cell tolerance that is associated with decreased disease severity upon re-infection. J Exp Med 2025; 222:e20241667. [PMID: 40214640 PMCID: PMC11987708 DOI: 10.1084/jem.20241667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/18/2024] [Accepted: 03/12/2025] [Indexed: 04/14/2025] Open
Abstract
Immunity to severe malaria is acquired quickly, operates independently of pathogen load, and represents a highly effective form of disease tolerance. The mechanism that underpins tolerance remains unknown. We used a human rechallenge model of falciparum malaria in which healthy adult volunteers were infected three times over a 12 mo period to track the development of disease tolerance in real-time. We found that parasitemia triggered a hardwired innate immune response that led to systemic inflammation, pyrexia, and hallmark symptoms of clinical malaria across the first three infections of life. In contrast, a single infection was sufficient to reprogram T cell activation and reduce the number and diversity of effector cells upon rechallenge. Crucially, this did not silence stem-like memory cells but instead prevented the generation of cytotoxic effectors associated with autoinflammatory disease. Tolerized hosts were thus able to prevent collateral tissue damage in the absence of antiparasite immunity.
Collapse
Affiliation(s)
- Diana Muñoz Sandoval
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
- Instituto de Microbiologia, Universidad San Francisco de Quito, Quito, Ecuador
| | - Florian A. Bach
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Alasdair Ivens
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Adam C. Harding
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Natasha L. Smith
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Michalina Mazurczyk
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | | | | | - Sarah E. Silk
- The Jenner Institute, University of Oxford, Oxford, UK
- Department of Biochemistry and Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Jordan R. Barrett
- The Jenner Institute, University of Oxford, Oxford, UK
- Department of Biochemistry and Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Graeme J.M. Cowan
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Giorgio Napolitani
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Nicholas J. Savill
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Simon J. Draper
- The Jenner Institute, University of Oxford, Oxford, UK
- Department of Biochemistry and Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Angela M. Minassian
- The Jenner Institute, University of Oxford, Oxford, UK
- Department of Biochemistry and Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Wiebke Nahrendorf
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Philip J. Spence
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
11
|
Chen J, Li F, Luo WS, Zhu MF, Zhao NJ, Zhang ZH, Chen YF, Feng DX, Yang SY, Sun WJ. Therapeutic potential of Da Cheng Qi Decoction and its ingredients in regulating ferroptosis via the NOX2-GPX4 signaling pathway to alleviate and predict severe acute pancreatitis. Cell Signal 2025; 131:111733. [PMID: 40081545 DOI: 10.1016/j.cellsig.2025.111733] [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: 08/26/2024] [Revised: 01/27/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
OBJECTIVE This study aimed to elucidate the protective effects of Da Cheng Qi Decoction (DCQD) on severe acute pancreatitis (SAP) by targeting ferroptosis in pancreatic acinar cells and to establish a predictive signature and nomogram for acute pancreatitis (AP) risk assessment. METHODS We utilized microarray analysis to delineate gene expression patterns among 32 healthy controls and 87 AP patients stratified by severity. Employing SAP models and NOX2-deficient cells, we investigated the molecular underpinnings of ferroptosis. The impact of DCQD and the ferroptosis inhibitor Fer-1 on gene expression, oxidative stress, and inflammation was assessed. Machine learning algorithms identified differentially expressed genes (DEGs) sensitive to DCQD, SAP, and ferroptosis (DSNFGs), which were validated across multiple datasets. A predictive nomogram integrating DSNFGs was developed, and single-cell analysis provided a comprehensive view of the cellular dynamics. RESULTS The microarray analysis revealed upregulation of NOX2 and downregulation of GPX4 in AP, with expression patterns correlating with disease severity. DCQD ameliorated SAP-induced pancreatic acinar cell damage and ferroptosis by reducing inflammatory markers and enhancing GPX4 expression. NOX2 knockout mitigated ferroptosis in SAP models, suggesting a key role in the disease process. DCQD and Fer-1 differentially regulated the expression of ferroptosis-related genes, reduced reactive oxygen species (ROS) and high-mobility group box 1 (HMGB1) levels, and suppressed the inflammatory response in a SAP mouse model. The HPLC analysis of DCQD constituents indicated eight components (aloe-emodin, rhein, emodin, chrysophanol, naringin, hesperidin, magnolol, and honokiol) with the capacity to modulate ferroptosis. Venn analysis identified 48 DSNFGs, with a subset of five genes demonstrating significant predictive value. The developed nomogram, based on LASSO regression, showed high accuracy in validation cohorts. Single-cell RNA sequencing (scRNA-seq) and CellChat analysis uncovered heterogeneity and cell-cell communication networks in the pancreas during recovery from pancreatitis, implicating several signaling pathways. CONCLUSION DCQD and its eight ingredients exert its protective effect in SAP by inhibiting ferroptosis through the NOX2/GPX4 pathway. The DCQD-SAP-ferroptosis-related signature and nomogram offer a novel tool for AP risk assessment, prognosis prediction, and personalized therapeutic strategies in SAP management.
Collapse
Affiliation(s)
- Jian Chen
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Department of Public Health, International College, Krirk University, Bangkok, Thailand
| | - Fu Li
- Department of Hepatopancreatobiliary Surgery, Shuguang Hospital affliated to Shanghai University of Traditional Chinese Medincine, Shanghai 201203, China
| | - Wang-Sheng Luo
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421200, China
| | - Mei-Fang Zhu
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Neng-Jiang Zhao
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Zhi-Hai Zhang
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Ya-Feng Chen
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Dian-Xu Feng
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China.
| | - Shu-Yu Yang
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China.
| | - Wen-Jie Sun
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China; Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China; Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
12
|
Yang Y, Li L, Dai F, Deng L, Yang K, He C, Chen Y, Yang X, Song L. Fibroblast-derived versican exacerbates periodontitis progression by regulating macrophage migration and inflammatory cytokine secretion. Cell Signal 2025; 131:111755. [PMID: 40112905 DOI: 10.1016/j.cellsig.2025.111755] [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: 02/11/2025] [Revised: 03/05/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
OBJECTIVE Versican (VCAN), a prominent extracellular matrix component upregulated in inflammatory diseases, demonstrates context-specific regulatory mechanisms. Periodontitis, a chronic inflammatory disease leading to periodontal tissue destruction and tooth loss, the pathological role of it remains poorly defined. Our study aims to examine VCAN-mediated mechanisms in periodontitis. METHODS We conducted a comprehensive analysis of bulk RNA sequencing and single-cell RNA sequencing data to examine VCAN expression level and source in periodontitis. Functional and correlation analyses were used to explore its biological functions. We then validated VCAN expression using quantitative real-time polymerase chain reaction, immunohistochemical staining, and immunofluorescence staining in animal models and investigated its biological functions in inflammation through in vitro experiments. RESULTS Our findings reveal that VCAN is mainly generated by fibroblast in periodontitis, and its expression significantly upregulated at both mRNA and protein levels. Using VCAN-overexpressing L929 cells, we demonstrated enhanced proliferative capacity and inflammatory potential. Co-culture experiments with RAW264.7 cells showed promoted migration, adhesion, M1 polarization, and mitogen-activated protein kinase (MAPK) pathway activation. CONCLUSION VCAN enhances fibroblast proliferation and migration, and upregulates inflammatory cytokines expression. Furthermore, fibroblast-derived VCAN not only induces macrophage chemotaxis, migration, adhesion, and polarization toward the proinflammatory M1 phenotype, but also activates MAPK signaling of macrophage, which may amplify inflammatory cascades to exacerbate periodontal tissue destruction. Targeted regulation of VCAN expression may become a promising precision treatment strategy for periodontitis.
Collapse
Affiliation(s)
- Yuting Yang
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China; The Second Clinical Medical School, NanchangUniversity, Nanchang, China
| | - Li Li
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China; The Second Clinical Medical School, NanchangUniversity, Nanchang, China
| | - Fang Dai
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China
| | - Libin Deng
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang, China
| | - Kaiqiang Yang
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China; The Second Clinical Medical School, NanchangUniversity, Nanchang, China
| | - Chenjiang He
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China; The Second Clinical Medical School, NanchangUniversity, Nanchang, China
| | - Yeke Chen
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China; The Second Clinical Medical School, NanchangUniversity, Nanchang, China
| | - Xinbo Yang
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China; The Second Clinical Medical School, NanchangUniversity, Nanchang, China
| | - Li Song
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital of Nanchang University, Nanchang, China; The institute of Periodontal Disease, Nanchang University, Nanchang, China.
| |
Collapse
|
13
|
Zhou M, Zhao W, Zhang X, Cheng Y, Wang M, Chen Y, Zhao L. Nicotinamide metabolism affects the prognosis of hepatocellular carcinoma by influencing the tumor microenvironment. Cytokine 2025; 191:156939. [PMID: 40228405 DOI: 10.1016/j.cyto.2025.156939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/15/2025] [Accepted: 04/05/2025] [Indexed: 04/16/2025]
Abstract
In this study, we utilized the public database along with single-cell genomics techniques to systematically analyze the expression patterns and clinical significance of key genes in the nicotinamide metabolism pathway in liver cancer samples. The findings indicate that differential nicotinamide metabolism-related key genes are expressed in liver cancer samples. The liver cancer samples were put into separate subgroups using consistency clustering analysis based on differential gene expression levels observed. Additionally, immune infiltration and drug sensitivity analysis also revealed differences between the two subgroups. Survival analysis suggested that the key genes were associated with prognosis. Finally, a prognostic model was established using the key genes, offering a fresh viewpoint on the molecular mechanism investigating liver cancer. This study demonstrated the significant correlation between key genes in the nicotinamide metabolism pathway and the occurrence and progression of liver cancer and indicated that these key genes could serve as prognostic markers and tailored treatment targets for liver cancer.
Collapse
Affiliation(s)
- Min Zhou
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210000, China
| | - Wenhui Zhao
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210000, China
| | - Xiaobo Zhang
- School of Life Sciences, Westlake University, Hangzhou, 310024, China
| | - Ye Cheng
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210000, China
| | - Mengxiang Wang
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210000, China
| | - Yan Chen
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210000, China.
| | - Lingrui Zhao
- School of Life Sciences, Westlake University, Hangzhou, 310024, China.
| |
Collapse
|
14
|
Zhao S, Wu D, Lu Y, Zhu L, Wang S, Li Z, Peng X, Li H, Xu X, Su W. Single-cell RNA sequencing indicates AP-1 as a potential therapeutic target for autoimmune uveitis. Biochem Pharmacol 2025; 237:116945. [PMID: 40228638 DOI: 10.1016/j.bcp.2025.116945] [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: 12/06/2024] [Revised: 03/06/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
Autoimmune uveitis (AU) is a sight-threatening eye disease, marked by a complex pathogenesis and limited treatment options. Herein, we conducted single-cell RNA sequencing (scRNA-seq) on the spleen and cervical draining lymph nodes (CDLNs) of both normal and experimental autoimmune uveitis (EAU) mice and found common alterations in celluar composition and transcriptional regulation occurred throughout the EAU process. Moreover, we identified activator protein-1 (AP-1) as a pivotal disease-related molecule in the pathogenesis of EAU. Inhibiting AP-1 alleviated symptoms of EAU and reduced the retina infiltration of T helper 17 cells (Th17) and Th1 cells. Additionally, following treatment with the AP-1 inhibitor, both the spleen and CDLNs showed decreased Th17 and Th1 cell proportions. Meanwhile, in vitro studies revealed that treatment with AP-1 inhibitor reduced the level of granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-23 (IL-23), two pivotal molecules implicated in the Th17 cell pathogenicity, during EAU. The adoptive transfer experiment also showed that inhibiting AP-1 in CD4+ T cells suppressed their ability to elicit EAU. Altogether, our study demonstrates that AP-1 might involved in EAU pathogenesis by supporting Th17 cell pathogenicity via the GM-CSF/IL-23 feedback loop. Thus, AP-1 inhibition might be a novel treatment strategy for uveitis.
Collapse
Affiliation(s)
- Sichen Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou 510060, China
| | - Dongting Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou 510060, China
| | - Yao Lu
- National Clinical Research Center for Eye Diseases, Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Lei Zhu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | | | - Zhaohuai Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou 510060, China
| | - Xuening Peng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou 510060, China
| | - He Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Xiaofang Xu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China.
| | - Wenru Su
- 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
|
15
|
Xia F, Santacruz A, Wu D, Bertho S, Fritz E, Morales-Sosa P, McKinney S, Nowotarski SH, Rohner N. Reproductive adaptation of Astyanax mexicanus under nutrient limitation. Dev Biol 2025; 523:82-98. [PMID: 40222642 PMCID: PMC12068995 DOI: 10.1016/j.ydbio.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/15/2025]
Abstract
Reproduction is a fundamental biological process for the survival and continuity of species. Examining changes in reproductive strategies offers valuable insights into how animals have adapted their life histories to different environments. Since reproduction is one of the most energy-intensive processes in female animals, nutrient scarcity is expected to interfere with the ability to invest in gametes. Lately, a new model to study adaptation to nutrient limitation has emerged; the Mexican tetra Astyanax mexicanus. This fish species exists as two different morphs, a surface river morph and a cave-dwelling morph. The cave-dwelling morph has adapted to the dark, lower biodiversity, and nutrient-limited cave environment and consequently evolved an impressive starvation resistance. However, how reproductive strategies have adapted to nutrient limitations in this species remains poorly understood. Here, we compared breeding activities and maternal contributions between laboratory-raised surface fish and cavefish. We found that cavefish produce different clutch sizes of eggs with larger yolk compared to surface fish, indicating a greater maternal nutrient deposition in cavefish embryos. To systematically characterize yolk compositions, we used untargeted proteomics and lipidomics approaches to analyze protein and lipid profiles in 2-cell stage embryos and found an increased proportion of sphingolipids in cavefish compared to surface fish. Additionally, we generated transcriptomic profiles of surface fish and cavefish ovaries using a combination of single cell and bulk RNA sequencing to examine differences in maternal contribution. We found that genes essential for hormone regulation were upregulated in cavefish follicular somatic cells compared to surface fish. To evaluate whether these differences contribute to their reproductive abilities under natural-occurring stress, we induced breeding in starved female fish. Remarkably, cavefish maintained their ability to breed under starvation, whereas surface fish largely lost this ability. We identified insulin-like growth factor 1a receptor (igf1ra) as a potential candidate gene mediating the downregulation of ovarian development genes, potentially contributing to the starvation-resistant fertility of cavefish. Taken together, we investigated the female reproductive strategies in Astyanax mexicanus, which will provide fundamental insights into the adaptations of animals to environments with extreme nutrient deficit.
Collapse
Affiliation(s)
- Fanning Xia
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Ana Santacruz
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Di Wu
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Sylvain Bertho
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Elizabeth Fritz
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Sean McKinney
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Nicolas Rohner
- Stowers Institute for Medical Research, Kansas City, MO, USA.
| |
Collapse
|
16
|
Gerakopoulos V, Ramos C, Müller C, Walterskirchen N, Vintila S, Zotter C, Ilg M, Pap A, Riss S, Bergmann M, Unger LW, Vogt AB, Oehler R, Lukowski SW. Single-cell transcriptomic analysis identifies tissue-specific fibroblasts as the main modulators of myeloid cells in peritoneal metastasis of different origin. Cancer Lett 2025; 620:217678. [PMID: 40154914 DOI: 10.1016/j.canlet.2025.217678] [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: 12/20/2024] [Revised: 03/12/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Colorectal cancer (CRC) peritoneal metastasis (CPM) is related to limited therapy options and poor prognosis. Although stromal cells heavily infiltrate most CPMs, interactions between different cell types in their microenvironment remain unclear. Here, we investigated tumor and distant normal tissue from CPM and CRC patients using single-cell RNA sequencing. Investigating the incoming and outgoing signals between cells revealed that fibroblasts dominate the CPM signaling landscape with myeloid cells as their strongest interaction partner. Using immunohistochemistry, we confirmed that fibroblasts co-localize with macrophages in the CPM microenvironment. A fibroblast sub-population detected only in CPM and normal peritoneum demonstrated immunoregulatory properties in co-culture experiments, and was further detected in additional peritoneal malignancies derived from ovarian and gastric origin. This novel fibroblast type and its communication with macrophages could be attractive targets for therapeutic interventions in CPM and potentially peritoneal surface malignancies in general.
Collapse
Affiliation(s)
- Vasileios Gerakopoulos
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Cristiano Ramos
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Catharina Müller
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Natalie Walterskirchen
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Stefania Vintila
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Chiara Zotter
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Mathias Ilg
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim RCV GmBH & Co KG., Dr. Boehringer Gasse 5-11, 1120, Vienna, Austria
| | - Anna Pap
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim RCV GmBH & Co KG., Dr. Boehringer Gasse 5-11, 1120, Vienna, Austria
| | - Stefan Riss
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Michael Bergmann
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Lukas W Unger
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria; Dept. of Colorectal Surgery, Oxford University Hospitals, Old Rd, Headington, Oxford, OX3 7LE, United Kingdom
| | - Anne B Vogt
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim RCV GmBH & Co KG., Dr. Boehringer Gasse 5-11, 1120, Vienna, Austria
| | - Rudolf Oehler
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria.
| | - Samuel W Lukowski
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim RCV GmBH & Co KG., Dr. Boehringer Gasse 5-11, 1120, Vienna, Austria
| |
Collapse
|
17
|
Ren H, Liu S, Ji D, Li X, Sun X, Wang W, Liu T, Li Y. Transcriptome analysis reveals the potential role of neural factor EN1 for long-terms survival in estrogen receptor-independent breast cancer. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200965. [PMID: 40207200 PMCID: PMC11981748 DOI: 10.1016/j.omton.2025.200965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/27/2025] [Accepted: 03/05/2025] [Indexed: 04/11/2025]
Abstract
Breast cancer patients with estrogen receptor-negative (ERneg) status, encompassing triple negative breast cancer (TNBC) and human epidermal growth factor receptor 2 positive breast cancer, are confronted with a heightened risk of drug resistance, often leading to early recurrence; the biomarkers and biological processes associated with recurrence is still unclear. In this study, we analyzed bulk RNA sequencing (RNA-seq) data from 285 cancer and paracancerous samples from 155 TNBC patients, along with transcriptome data from 11 independent public cohorts comprising 7,449 breast cancer patients and 26 single-cell RNA-seq datasets. Our results revealed differential enrichment of nerve-related pathways between TNBC patients with and without 10-year recurrence-free survival. We developed an early recurrence index (ERI) using a machine learning model and constructed a nomogram that accurately predicts the 10-year survival of ERneg patients (area under the curve [AUC]Training = 0.79; AUCTest = 0.796). Further analysis linked ERI to enhanced neural function and immunosuppression. Additionally, we identified EN1, the most significant ERI gene, as a potential biomarker that may regulate the tumor microenvironment and sensitize patients to immunotherapy.
Collapse
Affiliation(s)
- He Ren
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Shan Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Dongchen Ji
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Xue Li
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Xue Sun
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Wenzheng Wang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Tong Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, China
| | - Yingpu Li
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, China
- Genomics Research Center (Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province 150081, China
| |
Collapse
|
18
|
Xu X, Lin W, Liu T, Yuan C, Yan Y, Diao Y, Xiong J, Shao Y, Ni B. The upregulation of TNKS1 drives the phenotypic switching of vascular smooth muscle cells in aortic dissection through the activation of ferroptosis. Int Immunopharmacol 2025; 158:114722. [PMID: 40359887 DOI: 10.1016/j.intimp.2025.114722] [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: 10/28/2024] [Revised: 03/31/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025]
Abstract
OBJECTIVE Aortic dissection (AD) is a life-threatening disease. Tankyrase1 (TNKS1), a PARylating ADP-ribosyl transferase, plays a major role in myogenesis, a vital process known to drive muscle fiber formation and regeneration.This study explores the impact of TNKS1 on the transformation of human aortic smooth muscle cells (HASMCs) in AD. METHODS AND RESULTS Single-cell RNA sequencing was performed and clusters were used for between-disease differential gene expression analyses. In the AD aorta, WB, immunofluorescence and RT-q-PCR revealed that TNKS1 expression was elevated, accompanied by a disorganized cell phenotype. Further examination like WB,immunofluorescence,Scratch-Wound Assay confirmed the upregulation of TNKS1 triggers phenotypic switching.Subsequent studies revealed that ferroptosis played a key role in TNKS1-induced phenotypic switching. Increased ferroptosis markers, such as elevated iron content,ROS and lipid peroxidation, were observed in HASMCs overexpressing TNKS1, while inhibition of ferroptosis restored the contractile phenotype.Co-IP assay demonstrated a direct protein-protein interaction between TNKS1 and SLC7A11 at the molecular level. In vivo, the upregulation of TNKS1 not only activated ferroptosis but also triggered phenotypic transformation. CONCLUSION This study demonstrates that TNKS1 is a key regulator of AD pathogenesis, driving HASMC phenotypic switching through ferroptosis activation, ultimately leading to aortic wall destabilization and dissection. Targeting TNKS1 or the ferroptosis pathway may offer novel therapeutic strategies for AD prevention and treatment.
Collapse
Affiliation(s)
- Xinyang Xu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenfeng Lin
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tianyu Liu
- Department of General Surgery, Sir Run Run Hospital of Nanjing Medical University, Nanjing, China
| | - Chunze Yuan
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuhan Yan
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yifei Diao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiaqi Xiong
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yongfeng Shao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Buqing Ni
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
19
|
Wu Y, Xie L, Sun J, Wang Q, Xia W, Cai Q, Lu X, Gou X. Response of astrocytes and their interaction with surrounding brain cells after acute ischemia-reperfusion analyzed by single-cell transcriptome sequencing. Brain Res Bull 2025; 226:111355. [PMID: 40286940 DOI: 10.1016/j.brainresbull.2025.111355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/10/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Astrocytes play a key role in the occurrence and development of ischemic stroke. However, reactive astrocytes have both detrimental and protective roles in ischemic stroke. Regrettably, the stimulation signals associated with the transformation of astrocytes into different subclusters lack systemic analysis, and the mechanism by which astrocytes produce multiple effects is not entirely clear. We investigated the heterogeneity of mouse astrocytes 12 h after cerebral ischemia-reperfusion via Single-cell RNA sequencing and verified gene expressions by reverse transcription-polymerase chain reaction. We acquired astrocyte subclusters' transcriptional characteristics involved in diversified functions. To explore what stimulus signals cause astrocyte heterogeneity, we present a blueprint for cellular communication between astrocyte subclusters and other surrounding brain cells 12 h after ischemia-reperfusion, and identified 9 genes which are potential and promising for being therapeutic targets and 6 genes were specific to astrocyte subcluster 2 that tend to resist ischemia-reperfusion injury. At 12 h after ischemia-reperfusion, each subcluster of astrocytes is characteristic in terms of function and communication with surrounding cells, which is based on the activation genes and transcription molecules that we have revealed with subcluster characteristics. Our results provide a basis for revealing the anti-injury response of astrocytes to cerebral ischemia-reperfusion, which involves coordination of different subclusters and the coordination of astrocytes with surrounding brain cells.
Collapse
Affiliation(s)
- YongHong Wu
- School of Medical Technology & Institute of Basic Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi Province 710021, China; Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shanxi Province 710049, China
| | - Lei Xie
- Department of Radiology, Cancer Hospital of Shantou University Medical College, Shantou City, Guangdong Province 515041, China
| | - Jing Sun
- School of Medical Technology & Institute of Basic Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi Province 710021, China
| | - Qing Wang
- School of Medical Technology & Institute of Basic Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi Province 710021, China
| | - WangXiao Xia
- School of Medical Technology & Institute of Basic Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi Province 710021, China
| | - Qiang Cai
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan City, Hubei Province 430060, China.
| | - XiaoYun Lu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shanxi Province 710049, China.
| | - XingChun Gou
- School of Medical Technology & Institute of Basic Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi Province 710021, China.
| |
Collapse
|
20
|
Guo X, Yuan J, Zhang Y, Wu J, Wang X. Developmental landscape and asymmetric gene expression in the leaf vasculature of Brassica rapa revealed by single-cell transcriptome. HORTICULTURE RESEARCH 2025; 12:uhaf060. [PMID: 40271455 PMCID: PMC12017798 DOI: 10.1093/hr/uhaf060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/16/2025] [Indexed: 04/25/2025]
Abstract
Leaf vasculature not only acts as a channel for nutrients and signaling information but also influences leaf morphology. It consists of several distinct cell types with specialized functions. Cell type-specific characterizations based on single-cell RNA sequencing technology could aid in understanding the identities of vascular tissues and their roles in leaf morphogenesis in Brassica rapa. Here, we generated a single-cell transcriptome landscape of the Chinese cabbage leaf vasculature. A total of 12 cell clusters covering seven known cell types were identified. Different vascular cell types were characterized by distinct identities. The xylem parenchyma and companion cells exhibited an active expression pattern of amino acid metabolism genes. Tracheary elements and sieve elements were enriched in many genes related to cell wall biosynthesis, and the phloem parenchyma was enriched in many sugar transporter-encoding genes. Pseudo-time analyses revealed the developmental trajectories of the xylem and phloem and the potential roles of auxin and ethylene in xylem development. Furthermore, we identified key candidate regulators along the differentiation trajectory of the sieve elements and companion cells. Most of the homoeologous genes in the syntenic triads from the three subgenomes showed asymmetric gene expression patterns in different vascular cell types. Collectively, our study revealed that Chinese cabbage leaf vasculature cells had highly heterogeneous transcriptomes, providing new insights into the complex processes of leaf vasculature development in B. rapa leafy vegetables and other Brassica crops.
Collapse
Affiliation(s)
- Xinlei Guo
- Henan Engineering Research Center of the Development and Utilization of Characteristic Horticultural Plants, School of Horticulture and Landscape Architecture, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Jingping Yuan
- Henan Engineering Research Center of the Development and Utilization of Characteristic Horticultural Plants, School of Horticulture and Landscape Architecture, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Yuanyuan Zhang
- Henan Engineering Research Center of the Development and Utilization of Characteristic Horticultural Plants, School of Horticulture and Landscape Architecture, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Jian Wu
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xiaowu Wang
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
21
|
McGrail K, Granado-Martínez P, Orsenigo R, Caratù G, Nieto P, Heyn H, Ferrer B, Hernández-Losa J, Muñoz-Couselo E, García-Patos V, Recio JA. Transcriptional reprogramming triggered by neonatal UV radiation or Lkb1 loss prevents BRAF V600E-induced growth arrest in melanocytes. Oncogene 2025; 44:1592-1608. [PMID: 40057604 PMCID: PMC12095085 DOI: 10.1038/s41388-025-03339-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 02/12/2025] [Accepted: 02/28/2025] [Indexed: 05/23/2025]
Abstract
The mechanisms behind UVB-initiated, neonatal-specific melanoma linked to BRAFV600E are not well understood, particularly regarding its role in growth arrest. We found that, beyond mutations, neonatal UV irradiation or Lkb1 loss promotes a cell-autonomous transcriptional reprogramming that prevents BRAFV600E-induced growth arrest, leading to melanoma development. Using UVB-dependent and independent mouse models, genomic analyses, clinical data, and single-cell transcriptomics, we identified a transcriptional program that bypasses growth arrest, promoting melanoma. In humans, many of these genes are linked to poor survival and are upregulated in melanoma progression and other RAS pathway-driven tumors. Reconstitution experiments showed these genes cooperate with BRAFV600E in melanocyte transformation, dedifferentiation, and drug resistance. Depleting gene products like UPP1 highlights their potential as therapeutic targets. Our findings reveal that BRAFV600E-mutated melanomas can develop independently of nevus progression and identify novel targets for treatment.
Collapse
Affiliation(s)
- Kimberley McGrail
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory-Vall d´Hebron Research Institute VHIR-Vall d'Hebron Hospital-UAB, Barcelona, Spain
| | - Paula Granado-Martínez
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory-Vall d´Hebron Research Institute VHIR-Vall d'Hebron Hospital-UAB, Barcelona, Spain
| | - Roberto Orsenigo
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory-Vall d´Hebron Research Institute VHIR-Vall d'Hebron Hospital-UAB, Barcelona, Spain
| | - Ginevra Caratù
- Single Cell Genomics Group at the Spanish National Centre for Genomic Analysis (CNAG), Barcelona, Spain
| | - Paula Nieto
- Single Cell Genomics Group at the Spanish National Centre for Genomic Analysis (CNAG), Barcelona, Spain
| | - Holger Heyn
- Single Cell Genomics Group at the Spanish National Centre for Genomic Analysis (CNAG), Barcelona, Spain
| | - Berta Ferrer
- Anatomy Pathology Department, Vall d'Hebron Hospital-UAB, Barcelona, Spain
| | | | - Eva Muñoz-Couselo
- Clinical Oncology Program, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Hospital-UAB, Barcelona, Spain
| | | | - Juan A Recio
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory-Vall d´Hebron Research Institute VHIR-Vall d'Hebron Hospital-UAB, Barcelona, Spain.
| |
Collapse
|
22
|
Zhan Y, Deng Q, Jia Y, Chen Z, Zhao X, Ling Y, Qiu Y, Wang X, Wang F, He M, Huang W, Shen J, Wen S. Pdia3 deficiency exacerbates intestinal injury by disrupting goblet and Paneth cell function during ischemia/reperfusion. Cell Signal 2025; 130:111682. [PMID: 39988288 DOI: 10.1016/j.cellsig.2025.111682] [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: 12/24/2024] [Revised: 01/27/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025]
Abstract
Intestinal ischemia/reperfusion (I/R) injury is a severe medical condition associated with high mortality rates due to its disruption of intestinal homeostasis and impairment of mucosal defenses. The intestinal epithelium, particularly goblet and Paneth cells, plays a critical role in maintaining gut barrier integrity. Protein disulfide isomerase A3 (PDIA3) is involved in protein folding within intestinal epithelial cells (IECs) and has been linked to the stress response during I/R injury. This study aims to explore the role of PDIA3 in preserving intestinal integrity and immune function during I/R injury. Our study employed both human and mouse models to investigate PDIA3's expression and function. The correlation between PDIA3 expression and disease severity was analyzed using statistical tests, including Pearson's correlation coefficient. An intestinal I/R model was established in intestinal epithelium-specific conditional knockout mice lacking the Pdia3 gene. Single-cell RNA sequencing, immunohistochemistry, and transcriptomic analysis were used to assess PDIA3 expression in various intestinal cell types and to evaluate its role in epithelial differentiation and immune responses. PDIA3 was found to be highly expressed in healthy IECs, especially in goblet and Paneth cells. Its expression was reduced in patients with mesenteric artery ischemia and Pdia3-deficient mice, leading to severe intestinal damage, including impaired goblet and Paneth cell function, reduced antimicrobial peptide production, and altered gut microbiota. Treatment with recombinant defensin α1, an antimicrobial peptide secreted by Paneth cells, significantly alleviated the adverse effects of Pdia3 deficiency, restoring gut microbiota balance and reducing inflammation in the intestinal I/R injury mice. Taken together, our findings suggest that Pdia3 plays a vital role in maintaining intestinal barrier function and immune defense. Its deficiency exacerbates I/R-induced intestinal damage by impairing epithelial differentiation, mucus production, and antimicrobial peptide secretion. Targeting Pdia3 and associated pathways offers promising therapeutic strategies for mitigating I/R injury and restoring intestinal homeostasis.
Collapse
Affiliation(s)
- Yaqing Zhan
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Qiwen Deng
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yifan Jia
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhaorong Chen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Xu Zhao
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yihong Ling
- State Key Laboratory of Oncology in South, China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuxin Qiu
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiwen Wang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Fan Wang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Muchen He
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wenqi Huang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Jiantong Shen
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Shihong Wen
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Department of Anesthesiology, Guangxi Hospital Division of the First Affiliated Hospital of Sun Yat-sen University, Nanning, China.
| |
Collapse
|
23
|
Ye W, Clark E, Talatala E, Davis R, Ramirez‐Solano M, Sheng Q, Yang J, Collins S, Hillel A, Gelbard A. Characterizing the Cellular Constituents of Proximal Airway Disease in Granulomatosis With Polyangiitis. Otolaryngol Head Neck Surg 2025; 172:2009-2017. [PMID: 40062629 PMCID: PMC12120036 DOI: 10.1002/ohn.1197] [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: 10/31/2024] [Revised: 01/07/2025] [Accepted: 02/11/2025] [Indexed: 05/31/2025]
Abstract
OBJECTIVE Granulomatosis with polyangiitis (GPA) is a rare multisystem autoimmune vasculitis. 10-20% of patients suffer life-threatening obstruction of their proximal airways. Although progress has been made in the treatment of systemic disease, ameliorating airway disease in GPA remains an unmet need arising from limited understanding of disease pathogenesis. We sought to characterize the cellular constituents of the affected proximal airway mucosa in GPA airway scar. STUDY DESIGN Basic/translational study. SETTING Single tertiary care center. METHODS Using single-cell RNA sequencing, we profiled the cellular constituents of proximal airway samples from GPA and disease comparators (GPA; n = 9, idiopathic subglottic stenosis: iSGS; n = 7, post-intubation proximal stenosis: PIPS; n = 5, and control; n = 10). We report transcriptomes for subglottic epithelial, immune, endothelial, and stromal cell types and map expression of GPA risk genes to tissue types present in the proximal airway. We compared differential gene expression across immune cell populations and performed pseudotime analysis using Monocle 3. RESULTS Similar to iSGS and PIPS, the subglottic mucosa of GPA patients demonstrated an abundant immune infiltrate. 71% of the established GPA risk genes (10 of 14) localized to T cells and macrophages. Differential gene expression and pseudotime analysis revealed a sub-population of CD4-/CD8- inflammatory T cells that only originated from GPA. CONCLUSION We characterized the cellular composition of GPA airway disease and demonstrated that the expression of GPA risk alleles is predominantly localized to immune cell populations. We also identified a subset of inflammatory T cells that is unique to GPA.
Collapse
Affiliation(s)
- Wenda Ye
- Department of Otolaryngology–Head and Neck SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Evan Clark
- Department of Otolaryngology–Head and Neck SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Edward Talatala
- Department of Otolaryngology–Head and Neck SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Ruth Davis
- Department of Otolaryngology–Head and Neck SurgeryUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | | | - Quanhu Sheng
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jing Yang
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Sam Collins
- Department of Otolaryngology–Head and Neck SurgeryJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Alexander Hillel
- Department of Otolaryngology–Head and Neck SurgeryJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Alexander Gelbard
- Department of Otolaryngology–Head and Neck SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
24
|
Melkonian AL, Cheung MD, Erman EN, Moore KH, Lever JMP, Jiang Y, Yang Z, Lasseigne BN, Agarwal A, George JF. Single-cell RNA sequencing and spatial transcriptomics reveal unique subpopulations of infiltrating macrophages and dendritic cells following AKI. Am J Physiol Renal Physiol 2025; 328:F907-F920. [PMID: 40331777 DOI: 10.1152/ajprenal.00059.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/12/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025] Open
Abstract
Kidney infiltrating macrophages (KIMs) and kidney dendritic cells (KDCs) are strongly associated with inflammation and fibrosis in acute kidney injury (AKI) and chronic kidney disease (CKD). Contrary to kidney resident macrophages (KRMs), which are self-renewing and present in the kidney prior to injury, KIMs are bone-marrow derived F4/80int, CD11bhigh macrophages that infiltrate the kidney during AKI. Here, we combined single-cell RNA sequencing (scRNAseq), spatial transcriptomics, and cellular indexing of transcriptomes and epitopes (CITE)-sequencing to elucidate temporal, spatial, and transcriptional characteristics of unique subpopulations of KIMs and KDCs in ischemia-induced AKI. scRNAseq revealed three KIM, two KDC, and one proliferative macrophage subpopulation. All six clusters were localized in unique, spatially constrained microenvironments and their locations were dynamically regulated following bilateral ischemia reperfusion injury. We showed that a specific Arginase 1-expressing KIM cluster infiltrates the kidney cortex at day 1 after ischemia. We also identified a macrophage subpopulation that expresses genes specific to cell proliferation that resides in the cortex in uninjured states and in the medulla at day 6 during the reparative phase of AKI. Gene ontology analysis revealed functional characteristics that distinguish each KIM and KDC population. By day 28 after ischemia, the transcriptional profiles of KIMs upregulate C1q, Cd81, and Cd74, markers normally limited to KRMs in quiescence and early AKI. Since KIMs and KDCs are profoundly involved in AKI, it is paramount that we understand their dynamics-temporally and spatially-and identify their key genes and surface protein markers to develop macrophage-specific therapeutics aimed toward targeting kidney disease.NEW & NOTEWORTHY In this work, we fully characterized both single cell and spatial transcriptomes of kidney infiltrating macrophages (KIMs) and kidney dendritic cells (KDCs) following bilateral ischemia reperfusion injury. We also discovered distinct markers that differentiate KIMs from one another and kidney resident macrophages (KRMs). Finally, we show evidence suggesting that KIMs may reprogram and express genes previously limited to KRMs by day 28 following injury resolution.
Collapse
Affiliation(s)
- Arin L Melkonian
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Matthew D Cheung
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Elise N Erman
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Kyle H Moore
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Jeremie M P Lever
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Yanlin Jiang
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Zhengqin Yang
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Brittany N Lasseigne
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Anupam Agarwal
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - James F George
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
25
|
Peng G, Mosleh E, Yuhas A, Katada K, Kasinathan D, Cherry C, Golson ML. FOXM1 cooperates with ERα to regulate functional β-cell mass. Am J Physiol Endocrinol Metab 2025; 328:E804-E821. [PMID: 40261794 DOI: 10.1152/ajpendo.00438.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/02/2024] [Accepted: 04/11/2025] [Indexed: 04/24/2025]
Abstract
The transcription factor forkhead box (FOX)M1 regulates β-cell proliferation and insulin secretion. Our previous work demonstrates that expressing a constitutively active form of FOXM1 (FOXM1*) in β-cells increases β-cell function, proliferation, and mass in male mice. However, in contrast to what is observed in males, we demonstrate here that in female mice expression of FOXM1* in β-cells does not affect β-cell proliferation or glucose tolerance. Similarly, FOXM1* transduction of male but not female human islets enhances insulin secretion in response to elevated glucose. We therefore examined the mechanism behind this sexual dimorphism. Estrogen contributes to diabetes susceptibility differences between males and females, and estrogen receptor (ER)α is the primary mediator of β-cell estrogen signaling. Moreover, in breast cancer cells, ERα and FOXM1 work together to drive gene expression. We therefore examined whether FOXM1 and ERα functionally interact in β-cells. FOXM1* rescued elevated fasting glucose, glucose intolerance, and homeostatic model assessment of β-cell function (HOMA-B) in female mice with a β-cell-specific ERα deletion. Furthermore, in the presence of estrogen, the FOXM1 and ERα cistromes exhibit significant overlap in βTC6 β-cells. In addition, FOXM1 and ERα binding sites frequently occur in complex enhancers co-occupied by other islet transcription factors. These data indicate that FOXM1 and nuclear ERα cooperate to regulate β-cell function and suggest a general mechanism contributing to the lower incidence of diabetes observed in women.NEW & NOTEWORTHY Here we investigate why the effects of increasing FOXM1 activity in β-cells observed in male mice are not seen in female mice. ERα likely collaborates with FOXM1 and other transcription factors to enhance gene expression related to β-cell function. Higher estrogen levels in females may contribute to their increased insulin secretion and the more severe consequences of losing transcription factors like FOXM1 in males. Overall, these findings shed light on sex differences in diabetes susceptibility.
Collapse
Affiliation(s)
- Guihong Peng
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Elham Mosleh
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Andrew Yuhas
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Kay Katada
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Devi Kasinathan
- Department of Physiology, Johns Hopkins University, Baltimore, Maryland, United States
| | | | - Maria L Golson
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
26
|
Yang Q, Zhu PW, Wen YJ, Zhang R, Chen WW, Huang X, Chang Q. MAP4K4 aggravates microvascular anomalies in diabetic retinopathy in a YTHDF2-dependent manner. Diabetologia 2025; 68:1335-1351. [PMID: 40072537 DOI: 10.1007/s00125-025-06398-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025]
Abstract
AIMS/HYPOTHESIS Signalling pathways that regulate endothelial cell (EC) dysfunction, ischaemia and inflammation play a crucial role in retinal microangiopathy such as diabetic retinopathy. MAP4K4 is highly expressed in ECs. However, the involvement of MAP4K4 in retinal vasculopathy of diabetic retinopathy remains unclear. METHODS We analysed publicly available single-cell RNA sequencing (scRNA-seq) data from fibrovascular membranes (FVMs) from eight individuals with proliferative diabetic retinopathy (PDR) and normal retinas from 11 individuals without diabetes. Using db/db mice and human primary retinal endothelial cells (HRMECs), we further investigated the effects of MAP4K4 on retinal microangiopathy and endothelial dysfunction to explore the underlying regulatory mechanisms. RESULTS The scRNA-seq analysis revealed that MAP4K4 was predominantly expressed in retinal ECs, with elevated expression in FVMs from individuals with PDR compared with normal retinas from individuals without diabetes. This finding was confirmed at the protein level, with MAP4K4 expression and activity being upregulated in both the FVMs of individuals with PDR and the retinas of db/db mice. Inhibition of MAP4K4 using DMX-5804 alleviated retinal microvascular leakage by enhancing the expression and integrity of junctional proteins in both ECs from db/db mice and HRMECs. Additionally, DMX-5804 reduced retinal angiogenesis by inhibiting EC migration and vascular sprouting. Mechanistically, MAP4K4 regulated EC characteristics through NF-κB signalling pathway activity. The exacerbating effect of recombinant MAP4K4 on diabetic retinopathy in db/db mice was mitigated by a p65 inhibitor, confirming the involvement of NF-κB. Moreover, MAP4K4 expression was regulated by YTH N6-methyladenosine RNA-binding protein 2 (YTHDF2), which modulates the stability of MAP4K4 mRNA. CONCLUSIONS/INTERPRETATION Our study highlights the critical role of MAP4K4 in EC dysfunction and diabetic retinal microangiopathy, providing new insights into its molecular pathogenesis. Targeting MAP4K4, particularly through modulation of the YTHDF2/MAP4K4/NF-κB axis, may provide a novel therapeutic strategy for diabetic retinopathy.
Collapse
Affiliation(s)
- Qian Yang
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Pei-Wen Zhu
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Yan-Jun Wen
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Ran Zhang
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Wen-Wen Chen
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China.
| | - Xin Huang
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China.
| | - Qing Chang
- Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China.
| |
Collapse
|
27
|
Song L, Wang Y, Wang C, Yu Z, Wang L, He W, Zhang H, Li X, Zhong S. Integration of Bulk RNA and Single-Cell Analyses Reveal Distinct Expression Patterns of Anoikis-Related Genes and the Immunosuppressive Role of NQO1 + Macrophages in Hepatocellular Carcinoma. FASEB J 2025; 39:e70654. [PMID: 40386974 DOI: 10.1096/fj.202501310r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2025] [Revised: 05/01/2025] [Accepted: 05/09/2025] [Indexed: 05/20/2025]
Abstract
Anoikis resistance plays a crucial role in the proliferation, metastasis, and invasion of hepatocellular carcinoma (HCC). However, the key genes involved remain to be identified. This study aimed to investigate the prognostic value and impact of anoikis-related genes (ARGs) on the immunosuppressive microenvironment in HCC patients through the integration of bulk RNA and single-cell RNA sequencing (scRNA-seq) bioinformatic analysis. An anoikis-related gene risk score model (ARGRS) comprising 11 ARGs was established via machine learning. scRNA-seq was performed to assess the heterogeneity of ARGs in HCC. In vitro experiments were conducted to investigate the effects of NAD(P)H: quinone oxidoreductase 1 (NQO1) on the polarization, phenotype, and function of macrophages. Bioinformatics analysis demonstrated that ARGRS had perfect efficiency in predicting the prognosis of HCC patients and that ARGs potentially play a role in maintaining the invasion and metastasis of malignant cells. Notably, NQO1+ macrophages presented features consistent with alternatively activated macrophages (M2) and displayed a powerful immunosuppressive effect, particularly in close interaction with T cells within the tumor immune microenvironment. Moreover, inhibition of NQO1 expression via dicoumarol resulted in reduced expression of the M2-associated markers CD206 and CD163, as well as the immunosuppressive cytokines IL-10 and TGF-β. Strikingly, this treatment effectively mitigated the immunosuppressive impact of macrophages on T cells. Collectively, ARGs are closely associated with the poor prognosis of HCC patients, and NQO1+ macrophages may have an immunosuppressive effect on HCC, suggesting that intervention in anoikis may represent a potential strategy for HCC treatment.
Collapse
Affiliation(s)
- Linnan Song
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, China
| | - Yuhao Wang
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, China
| | - Chen Wang
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, China
| | - Ziqian Yu
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, China
| | - Liping Wang
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, China
| | - Weixin He
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, China
| | - Hui Zhang
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, China
| | - Xiaoyi Li
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, China
| | - Shihong Zhong
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, China
| |
Collapse
|
28
|
Shirazi SP, Negretti NM, Jetter CS, Sharkey AL, Garg S, Kapp ME, Wilkins D, Fortier G, Mallapragada S, Banovich NE, Eldredge LC, Deutsch GH, Wright CVE, Frank DB, Kropski JA, Sucre JMS. Bronchopulmonary dysplasia with pulmonary hypertension associates with semaphorin signaling loss and functionally decreased FOXF1 expression. Nat Commun 2025; 16:5004. [PMID: 40442177 PMCID: PMC12122835 DOI: 10.1038/s41467-025-60371-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 05/20/2025] [Indexed: 06/02/2025] Open
Abstract
Lung injury in preterm infants leads to structural and functional respiratory deficits, with a risk for bronchopulmonary dysplasia (BPD) that in its most severe form is accompanied by pulmonary hypertension (PH). To identify potential cellular and molecular drivers of BPD in humans, we performed single-cell RNA sequencing of preterm infant lungs with evolving BPD and BPD + PH compared to term infants. Examination of endothelial cells reveals a unique, aberrant capillary cell-state in BPD + PH defined by ANKRD1 expression. Within the alveolar parenchyma in infants with BPD/BPD + PH, predictive signaling analysis identifies surprising deficits in the semaphorin guidance-cue pathway, with decreased expression of pro-angiogenic transcription factor FOXF1. Loss of semaphorin signaling is replicated in a murine BPD model and in humans with causal FOXF1 mutations for alveolar capillary dysplasia (ACDMPV), suggesting a mechanistic link between developmental programs underlying BPD and ACDMPV and uncovering a critical role for semaphorin signaling in normal lung development.
Collapse
Affiliation(s)
- Shawyon P Shirazi
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Nicholas M Negretti
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Christopher S Jetter
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexandria L Sharkey
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shriya Garg
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Meghan E Kapp
- Department of Pathology, Case Western Reserve University Hospitals, Cleveland, OH, USA
| | - Devan Wilkins
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gabrielle Fortier
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Saahithi Mallapragada
- Division of Bioinnovation and Genome Science, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Nicholas E Banovich
- Division of Bioinnovation and Genome Science, Translational Genomics Research Institute, Phoenix, AZ, USA
- Biodevelopmental Origins of Lung Disease (BOLD) Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Laurie C Eldredge
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, USA
| | - Gail H Deutsch
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, USA
| | - Christopher V E Wright
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Biodevelopmental Origins of Lung Disease (BOLD) Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Program in Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - David B Frank
- Department of Pediatrics, Division of Cardiology, Children's Hospital of Philadelphia, Penn Cardiovascular Institute, Penn-CHOP Lung Biology Institute, Philadelphia, PA, USA
| | - Jonathan A Kropski
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.
- Biodevelopmental Origins of Lung Disease (BOLD) Center, Vanderbilt University Medical Center, Nashville, TN, USA.
- Program in Developmental Biology, Vanderbilt University, Nashville, TN, USA.
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Veterans Affairs Medical Center, Nashville, TN, USA.
| | - Jennifer M S Sucre
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.
- Biodevelopmental Origins of Lung Disease (BOLD) Center, Vanderbilt University Medical Center, Nashville, TN, USA.
- Program in Developmental Biology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
29
|
Su W, Ye Z, Liu J, Deng K, Liu J, Zhu H, Duan L, Shi C, Wang L, Zhao Y, Gong F, Zhang Y, Hou B, You H, Feng F, Ling Q, Xiao Y, Guo Y, Fan W, Zhang S, Zhang Z, Hu X, Yao Y, Zheng C, Lu L. Single-cell and spatial transcriptome analyses reveal tumor heterogeneity and immune remodeling involved in pituitary neuroendocrine tumor progression. Nat Commun 2025; 16:5007. [PMID: 40442104 PMCID: PMC12122724 DOI: 10.1038/s41467-025-60028-5] [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: 10/10/2024] [Accepted: 05/12/2025] [Indexed: 06/02/2025] Open
Abstract
Pituitary neuroendocrine tumors (PitNETs) can be invasive or aggressive, yet the mechanisms behind these behaviors remain poorly understood, impeding treatment advancements. Here, we integrat single-cell RNA sequencing and spatial transcriptomics, analyzing over 177,000 cells and 35,000 spots across 57 tissue samples. This comprehensive approach facilitates the identification of PitNETs tumor populations and characterizes the reconfiguration of the tumor microenvironment (TME) as PitNETs progress and invade. We trace the trajectory of TPIT-lineage PitNETs and identify an aggressive tumor cluster marked by elevated p53-mediated proliferation and a higher Trouillas classification, both associated with tumor progression. Additionally, we document the heterogeneity of immune stromal cells within PitNETs, particularly noting the enrichment of SPP1+ tumor associated macrophages (TAMs) in invasive tumors. These TAMs facilitate tumor invasion through the SPP1-ITGAV/ITGB1 signaling pathway. Our in-depth single-cell and spatial analysis of PitNETs uncovers the molecular dynamics within the TME, suggesting potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Wan Su
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhang Ye
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jifang Liu
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Kan Deng
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jinghua Liu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Huijuan Zhu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lian Duan
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chen Shi
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Linjie Wang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuxing Zhao
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fengying Gong
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yi Zhang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Bo Hou
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hui You
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Feng Feng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Qing Ling
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yu Xiao
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yongdong Guo
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Wenyi Fan
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Peking University Cancer Hospital & Institute, Beijing, China
- Frontiers Science Center for Cancer Integrative Omics, Peking University International Cancer Institute, Peking University, Beijing, China
| | - Sumei Zhang
- Clinical Biobank, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zixin Zhang
- Clinical Biobank, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaomin Hu
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Yong Yao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Chunhong Zheng
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Peking University Cancer Hospital & Institute, Beijing, China.
- Frontiers Science Center for Cancer Integrative Omics, Peking University International Cancer Institute, Peking University, Beijing, China.
| | - Lin Lu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
30
|
Guo X, Wang Y, Zhao C, Tan C, Yan W, Xiang S, Zhang D, Zhang H, Zhang M, Yang L, Yan M, Xie P, Wang Y, Li L, Fang D, Guang X, Shao W, Wang F, Wang H, Sahu SK, Liu M, Wei T, Peng Y, Qiu Y, Peng T, Zhang Y, Ni X, Xu Z, Lu H, Li Z, Yang H, Wang E, Lisby M, Liu H, Guo H, Xu X. An Arabidopsis single-nucleus atlas decodes leaf senescence and nutrient allocation. Cell 2025; 188:2856-2871.e16. [PMID: 40220755 DOI: 10.1016/j.cell.2025.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 07/30/2024] [Accepted: 03/12/2025] [Indexed: 04/14/2025]
Abstract
With rapid advancements in single-cell RNA sequencing (scRNA-seq) technologies, exploration of the systemic coordination of critical physiological processes has entered a new era. Here, we generated a comprehensive Arabidopsis single-nucleus transcriptomic atlas using over 1 million nuclei from 20 tissues encompassing multiple developmental stages. Our analyses identified cell types that have not been characterized in previous single-protoplast studies and revealed cell-type conservation and specificity across different organs. Through time-resolved sampling, we revealed highly coordinated onset and progression of senescence among the major leaf cell types. We originally formulated two molecular indexes to quantify the aging state of leaf cells at single-cell resolution. Additionally, facilitated by weighted gene co-expression network analysis, we identified hundreds of promising hub genes that may integratively regulate leaf senescence. Inspired by the functional validation of identified hub genes, we built a systemic scenario of carbon and nitrogen allocation among different cell types from source leaves to sink organs.
Collapse
Affiliation(s)
- Xing Guo
- BGI Research, Wuhan 430047, China
| | - Yichuan Wang
- New Cornerstone Science Laboratory, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | | | - Cong Tan
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
| | - Wei Yan
- New Cornerstone Science Laboratory, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Sunhuan Xiang
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
| | - Dan Zhang
- New Cornerstone Science Laboratory, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hui Zhang
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
| | - Mengting Zhang
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China; BGI Life Science Joint Research Center, Northeast Forestry University, Harbin, Heilongjiang 150040, China
| | - Liujing Yang
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meng Yan
- School of Agriculture, Yunnan University, Kunming 650500, China
| | - Pingli Xie
- School of Agriculture, Yunnan University, Kunming 650500, China
| | - Yi Wang
- School of Agriculture, Yunnan University, Kunming 650500, China
| | - Li Li
- BGI Research, Wuhan 430047, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dongming Fang
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
| | - Xuanmin Guang
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
| | - Wenwen Shao
- BGI Research, Wuhan 430047, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fang Wang
- BGI Research, Wuhan 430047, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haoxuan Wang
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sunil Kumar Sahu
- BGI Research, Wuhan 430047, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
| | - Min Liu
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
| | - Tong Wei
- BGI Research, Wuhan 430047, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
| | - Yang Peng
- New Cornerstone Science Laboratory, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yuping Qiu
- New Cornerstone Science Laboratory, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Tao Peng
- New Cornerstone Science Laboratory, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yi Zhang
- New Cornerstone Science Laboratory, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xuemei Ni
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
| | - Zhicheng Xu
- China National GeneBank, Shenzhen 518083, China
| | - Haorong Lu
- China National GeneBank, Shenzhen 518083, China
| | - Zhonghai Li
- State Key Laboratory of Tree Genetics and Breeding, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Huanming Yang
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
| | - Ertao Wang
- New Cornerstone Science Laboratory, Key Laboratory of Plant Carbon Capture, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, SIBS, Chinese Academy of Sciences, Shanghai 200032, China
| | - Michael Lisby
- Department of Biology, University of Copenhagen, Copenhagen 2100, Denmark
| | - Huan Liu
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China; BGI Life Science Joint Research Center, Northeast Forestry University, Harbin, Heilongjiang 150040, China.
| | - Hongwei Guo
- New Cornerstone Science Laboratory, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Xun Xu
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China; Guangdong Provincial Key Laboratory of Genome Read and Write, Shenzhen 518083, China.
| |
Collapse
|
31
|
Ball BK, Park JH, Bergendorf AM, Proctor EA, Brubaker DK. Translational disease modeling of peripheral blood identifies type 2 diabetes biomarkers predictive of Alzheimer's disease. NPJ Syst Biol Appl 2025; 11:58. [PMID: 40442087 PMCID: PMC12122922 DOI: 10.1038/s41540-025-00539-5] [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: 01/09/2025] [Accepted: 05/16/2025] [Indexed: 06/02/2025] Open
Abstract
Type 2 diabetes (T2D) is a significant risk factor for Alzheimer's disease (AD). Despite multiple studies reporting this connection, the mechanism by which T2D exacerbates AD is poorly understood. It is challenging to design studies that address co-occurring and comorbid diseases, limiting the number of existing evidence bases. To address this challenge, we expanded the applications of a computational framework called Translatable Components Regression (TransComp-R), initially designed for cross-species translation modeling, to perform cross-disease modeling to identify biological programs of T2D that may exacerbate AD pathology. Using TransComp-R, we combined peripheral blood-derived T2D and AD human transcriptomic data to identify T2D principal components predictive of AD status. Our model revealed genes enriched for biological pathways associated with inflammation, metabolism, and signaling pathways from T2D principal components predictive of AD. The same T2D PC predictive of AD outcomes unveiled sex-based differences across the AD datasets. We performed a gene expression correlational analysis to identify therapeutic hypotheses tailored to the T2D-AD axis. We identified six T2D and two dementia medications that induced gene expression profiles associated with a non-T2D or non-AD state. We next assessed our blood-based T2DxAD biomarker signature in post-mortem human AD and control brain gene expression data from the hippocampus, entorhinal cortex, superior frontal gyrus, and postcentral gyrus. Using partial least squares discriminant analysis, we identified a subset of genes from our cross-disease blood-based biomarker panel that significantly separated AD and control brain samples. Finally, we validated our findings using single cell RNA-sequencing blood data of AD and healthy individuals and found erythroid cells contained the most gene expression signatures to the T2D PC. Our methodological advance in cross-disease modeling identified biological programs in T2D that may predict the future onset of AD in this population. This, paired with our therapeutic gene expression correlational analysis, also revealed alogliptin, a T2D medication that may help prevent the onset of AD in T2D patients.
Collapse
Affiliation(s)
- Brendan K Ball
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Jee Hyun Park
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Alexander M Bergendorf
- Center for Global Health & Diseases, Department of Pathology, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Elizabeth A Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Penn State University, State College, PA, USA
- Penn State Neuroscience Institute, Penn State University, State College, PA, USA
- Department of Engineering Science & Mechanics, Penn State University, State College, PA, USA
| | - Douglas K Brubaker
- Center for Global Health & Diseases, Department of Pathology, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Blood Heart Lung Immunology Research Center, University Hospitals, Cleveland, OH, USA.
| |
Collapse
|
32
|
Marshall JL, Satti I, Surakhy M, Harris SA, Morrison H, Wittenberg RE, Peralta Alvarez MP, Li S, Lopez Ramon R, Hoogkamer E, Salguero FJ, Ramos Lopez F, Mitton C, Cabrera Puig I, Powell Doherty R, Tanner R, Hinks TSC, Bettinson H, McShane H. Early mucosal responses following a randomised controlled human inhaled infection with attenuated Mycobacterium bovis BCG. Nat Commun 2025; 16:4989. [PMID: 40442144 PMCID: PMC12122720 DOI: 10.1038/s41467-025-60285-4] [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: 08/01/2024] [Accepted: 05/20/2025] [Indexed: 06/02/2025] Open
Abstract
The development of an effective vaccine against Mycobacterium tuberculosis is hampered by an incomplete understanding of immunoprotective mechanisms. We utilise an aerosol human challenge model using attenuated Mycobacterium bovis BCG, in BCG-naïve UK adults. The primary endpoint of this study (NCT03912207) was to characterise the early immune responses induced by aerosol BCG infection, the secondary endpoint was to identify immune markers associated with in-vitro protection. Blinded volunteers were randomised to inhale 1 × 107 CFU aerosolised BCG or 0.9% saline (20:6); and sequentially allocated to bronchoscopy at day 2 or 7 post-inhalation (10 BCG, 3 saline each timepoint). In the bronchoalveolar lavage post-aerosol BCG infection, there was an increase in frequency of eosinophils, neutrophils, NK cells and Donor-Unrestricted T cells at day 7, and the frequency of antigen presenting cells decreased at day 7 compared with day 2. The frequency of interferon-gamma+ BCG-specific CD4+ T cells increased in the BAL and peaked in the blood at day 7 post-BCG infection compared to day 2. BAL cells at day 2 and day 7 upregulated gene pathways related to phagocytosis, MHC-II antigen loading, T cell activation and proliferation. BCG's lack of key virulence factors and its failure to induce granulomas, may mean the observed immune responses do not fully recapitulate Mycobacterium tuberculosis infection. However, human infection models can provide unique insights into early immune mechanisms, informing vaccine design for complex pathogens.
Collapse
Affiliation(s)
- Julia L Marshall
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, United Kingdom
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Australia
| | - Iman Satti
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Mirvat Surakhy
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Stephanie A Harris
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Hazel Morrison
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Rachel E Wittenberg
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Marco Polo Peralta Alvarez
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Shuailin Li
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Raquel Lopez Ramon
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Emily Hoogkamer
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | | | - Fernando Ramos Lopez
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Celia Mitton
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Ingrid Cabrera Puig
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Rebecca Powell Doherty
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Rachel Tanner
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Timothy S C Hinks
- Oxford Centre for Respiratory Medicine, Nuffield Department of Clinical Medicine, The University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Henry Bettinson
- Oxford Centre for Respiratory Medicine, Nuffield Department of Clinical Medicine, The University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - Helen McShane
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, United Kingdom.
| |
Collapse
|
33
|
Wang G, Zhao J, Lin Y, Liu T, Zhao Y, Zhao H. scMODAL: a general deep learning framework for comprehensive single-cell multi-omics data alignment with feature links. Nat Commun 2025; 16:4994. [PMID: 40442129 PMCID: PMC12122792 DOI: 10.1038/s41467-025-60333-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: 03/30/2025] [Accepted: 05/16/2025] [Indexed: 06/02/2025] Open
Abstract
Recent advancements in single-cell technologies have enabled comprehensive characterization of cellular states through transcriptomic, epigenomic, and proteomic profiling at single-cell resolution. These technologies have significantly deepened our understanding of cell functions and disease mechanisms from various omics perspectives. As these technologies evolve rapidly and data resources expand, there is a growing need for computational methods that can integrate information from different modalities to facilitate joint analysis of single-cell multi-omics data. However, integrating single-cell omics datasets presents unique challenges due to varied feature correlations and technology-specific limitations. To address these challenges, we introduce scMODAL, a deep learning framework tailored for single-cell multi-omics data alignment using feature links. scMODAL integrates datasets with limited known positively correlated features, leveraging neural networks and generative adversarial networks to align cell embeddings and preserve feature topology. Our experiments demonstrate scMODAL's effectiveness in removing unwanted variation, preserving biological information, and accurately identifying cell subpopulations across diverse datasets. scMODAL not only advances integration tasks but also supports downstream analyses such as feature imputation and feature relationship inference, offering a robust solution for advancing single-cell multi-omics research.
Collapse
Affiliation(s)
- Gefei Wang
- Department of Biostatistics, Yale University, New Haven, CT, USA
| | - Jia Zhao
- Department of Biostatistics, Yale University, New Haven, CT, USA
| | - Yingxin Lin
- Department of Biostatistics, Yale University, New Haven, CT, USA
| | - Tianyu Liu
- Department of Biostatistics, Yale University, New Haven, CT, USA
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Yize Zhao
- Department of Biostatistics, Yale University, New Haven, CT, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale University, New Haven, CT, USA.
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA.
| |
Collapse
|
34
|
Du H, Si G, Si J, Song X, Si F. Single-cell RNA sequencing reveals the role of GTF2F2 in ovarian cancer oncogenesis and progression. J Ovarian Res 2025; 18:114. [PMID: 40442848 PMCID: PMC12121145 DOI: 10.1186/s13048-025-01686-3] [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: 02/18/2025] [Accepted: 05/05/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Ovarian cancer is one of the most common malignancies of the female reproductive system and is associated with poor prognosis. This study aimed to utilize single-cell RNA sequencing to investigate the heterogeneity of malignant epithelial cells in ovarian cancer, focusing on their potential functions and the implications for treatment and prognosis. METHODS Single-cell RNA sequencing data were clustered using a single-cell transcriptome clustering method, and malignant epithelial cells were identified through copy number variation analysis. The interaction patterns between different malignant subpopulations and immune/stromal cells were analyzed using cell-to-cell communication analysis. A risk score (URS) model based on the UBE2C + epithelial subpopulation was then constructed through LASSO and multivariable Cox regression. High and low URS groups were compared in terms of tumor mutational burden (TMB), survival outcomes, and drug sensitivity. Finally, the role of GTF2F2 in ovarian cancer progression was validated through gene knockdown experiments in an ovarian cancer cell line (ES-2). RESULTS Three major malignant epithelial cell subpopulations were identified (TMSB4X + Epi, TSC22D1 + Epi, and UBE2C + Epi). The UBE2C + Epi subpopulation exhibited higher stemness and greater invasive potential. The constructed URS model effectively stratified patients into high- and low-risk groups, with the high-risk group displaying a higher TMB level (p = 0.00011). Drug sensitivity predictions indicated that osimertinib, rapamycin, and dihydrorotenone might have stronger inhibitory effects in the high-risk group, whereas ERK inhibitors were more effective in the low-risk group. Functional assays demonstrated that GTF2F2 knockdown significantly suppressed ovarian cancer cell migration and invasion. Western blot analyses further showed elevated E-cadherin and reduced N-cadherin expression, suggesting that GTF2F2 may promote epithelial-mesenchymal transition (EMT). CONCLUSION The risk score model established in this study offers a novel framework for patient stratification and personalized therapy. Notably, the identification of the UBE2C + Epi subpopulation and key genes such as GTF2F2 highlights potential diagnostic and therapeutic targets, shedding light on the pathogenesis of ovarian cancer and paving the way for precision medicine approaches.
Collapse
Affiliation(s)
- Haiyang Du
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, 450046, China
- Henan Key Laboratory of TCM Syndrome and Prescription Signaling, Henan International Joint Laboratory of TCM Syndrome and Prescription Signaling, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Gao Si
- Department of Orthopedic, The Third Hospital of Peking University, Beijing, 100029, China
| | - Jiqing Si
- Henan Hospital of TCM, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450046, China
| | - Xuejie Song
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, 450046, China
- Henan Key Laboratory of TCM Syndrome and Prescription Signaling, Henan International Joint Laboratory of TCM Syndrome and Prescription Signaling, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Fuchun Si
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
- Henan Key Laboratory of TCM Syndrome and Prescription Signaling, Henan International Joint Laboratory of TCM Syndrome and Prescription Signaling, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| |
Collapse
|
35
|
Xia P, Zhou J, Shen R, Wang D. Deciphering the cellular and molecular landscape of cervical cancer progression through single-cell and spatial transcriptomics. NPJ Precis Oncol 2025; 9:158. [PMID: 40437003 PMCID: PMC12120119 DOI: 10.1038/s41698-025-00948-z] [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: 08/27/2024] [Accepted: 05/12/2025] [Indexed: 06/01/2025] Open
Abstract
Cervical cancer represents a significant global health challenge, with complex cellular and molecular mechanisms driving its progression from HPV infection to invasive malignancy. This study employed an integrated approach combining single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (stRNA-seq) to comprehensively characterize the tumor microenvironment (TME) across different stages of cervical cancer development. Through analysis of samples from normal cervix, HPV-infected normal cervix, high-grade squamous intraepithelial lesions (HSIL), and invasive cervical cancer, we identified distinct cellular populations and their dynamic changes during disease progression. Our findings revealed significant heterogeneity in immune cell populations, particularly highlighting the role of SPP1+ macrophages that were substantially enriched in cervical cancer compared to precancerous and normal tissues. Cell-cell communication networks and spatial mapping demonstrated that SPP1+ macrophages interact extensively with immune cells through the SPP1-CD44 signaling axis. This interaction contributes to an immunosuppressive microenvironment through modulation of T cell function and promotion of tumor cell survival. Furthermore, high expression of SPP1 correlated with advanced tumor stages and poor overall survival in cervical cancer patients, highlighting its potential as a prognostic biomarker. Our comprehensive characterization of the cellular landscape and intercellular communication networks in cervical cancer progression provides valuable insights for the development of targeted therapeutic strategies aimed at modulating the TME, particularly through disruption of the SPP1-CD44 axis. These findings establish a foundation for more effective personalized approaches to improve clinical outcomes in cervical cancer patients.
Collapse
Affiliation(s)
- Peng Xia
- Department of Anatomy and Histology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Juanhong Zhou
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, China
| | - Rong Shen
- Department of Anatomy and Histology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| | - Degui Wang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| |
Collapse
|
36
|
Wang P, Liu J, Zhang M, Yang J, Lian P, Cheng X, Qin J. Radiation Exposure Induced Blood-Brain Barrier Injury via Mitochondria-Mediated Sterile Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e02356. [PMID: 40433769 DOI: 10.1002/advs.202502356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 05/07/2025] [Indexed: 05/29/2025]
Abstract
Radiation-induced brain injury (RIBI) is caused by exposure to high doses of ionizing radiation and characterized by severe cognitive dysfunction and brain necrosis. However, the pathogenesis of RIBI is not fully understood, and no effective intervention is available. This work describes a blood-brain barrier (BBB) microphysiological system (MPS), that allowed to explore the responses of BBB and distinct brain cells to radiation exposure. Following acute exposure to radiation of X-ray or γ-ray, characteristic RIBI-associated pathological responses are observed, including BBB compromise, DNA breaks, inhibited cell proliferation, cell hypertrophy, and proinflammatory cytokine release. Among the distinctive types of cells, brain endothelial cells show the highest radiosensitivity as compared to other cells in the MPS. Intriguingly, X-ray and γ-ray radiation consistently induce prominent sterile inflammation responses, especially type I interferon response, in the BBB MPS. These responses are mediated by radiation-induced mitochondrial DNA release and subsequent activation of cGAS-STING signaling pathway. Furthermore, it is found abrocitinib (JAK1 inhibitor) and idebenone (mitochondrial protectant) can attenuate radiation-induced inflammation and ameliorate injuries in the BBB MPS. These findings reveal the involvement of mitochondria-mediated sterile inflammation in RIBI pathogenesis, identifying mitochondria as a potential target for new radioprotective measures.
Collapse
Affiliation(s)
- Peng Wang
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
| | - Jiayue Liu
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
| | - Min Zhang
- Disvision of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P. R. China
| | - Juan Yang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, P. R. China
| | - Peihan Lian
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
| | - Xiu Cheng
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
| | - Jianhua Qin
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
- Disvision of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P. R. China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100000, China
| |
Collapse
|
37
|
Zhang Y, Zuo A, Ba Y, Liu S, Chen J, Yang S, Weng S, Chen Y, Xu H, Luo P, Cheng Q, Tang B, Liu B, Zhang C, Yang J, Han X, Liu Z. Cancer-associated fibroblast-derived SEMA3C facilitates colorectal cancer liver metastasis via NRP2-mediated MAPK activation. Proc Natl Acad Sci U S A 2025; 122:e2423077122. [PMID: 40402249 DOI: 10.1073/pnas.2423077122] [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: 11/06/2024] [Accepted: 04/09/2025] [Indexed: 05/23/2025] Open
Abstract
Liver metastasis remains the predominant cause of mortality in patients with colorectal cancer (CRC). Nevertheless, the mechanisms underlying the initiation of colorectal cancer liver metastasis remain poorly elucidated. During the metastatic process of CRC cells from the primary site to the liver, we performed time-resolved analyses and identified a subset of tumor cells spatially located in the primary tumor and temporally distributed in the early stages of liver metastasis. These cells were termed liver metastasis-initiating cells (LMICs). LMICs exhibit high stemness, low proliferation, active interaction with surrounding stromal components, and a close association with liver metastasis. Notably, we found significant interactions between cancer-associated fibroblasts (CAFs) and LMICs via the SEMA3C-NRP2 receptor-ligand pair. Further in vivo and in vitro experiments confirmed that CAF-secreted SEMA3C could bind to the NRP2 receptor, which activates the MAPK pathway and promotes colorectal cancer liver metastasis. Our findings suggest potential therapeutic strategies for the early prevention of colorectal cancer liver metastasis.
Collapse
Affiliation(s)
- Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
- Interventional Institute of Zhengzhou University, Zhengzhou 450052, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou 450052, Henan, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jingqi Chen
- Department of Clinical Medicine, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Shuaixi Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yukang Chen
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410000, China
| | - Bufu Tang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Benyu Liu
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Chuhan Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jingkuan Yang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
- Interventional Institute of Zhengzhou University, Zhengzhou 450052, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou 450052, Henan, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
- Interventional Institute of Zhengzhou University, Zhengzhou 450052, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou 450052, Henan, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
38
|
Rahman J, Bibby JA, Singh P, Merle NS, West EE, Bohrer A, Mayer-Barber K, Liu C, Brinster LR, Afzali B, Briones AM, Alehashemi S, Bhuyan F, Ge J, Chen X, Zhou Y, Clarke MCH, Liu B, Goldbach-Mansky R, Serezani CH, Kemper C. A CD4 + T cell-intrinsic complement C5aR2-prostacyclin-IL-1R2 axis orchestrates Th1 cell contraction. Immunity 2025:S1074-7613(25)00222-5. [PMID: 40449486 DOI: 10.1016/j.immuni.2025.05.003] [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: 09/05/2024] [Revised: 02/06/2025] [Accepted: 05/06/2025] [Indexed: 06/03/2025]
Abstract
T helper 1 (Th1) cell initiation pathways are well characterized; however, those regulating their contraction are less understood. Here, we define a CD4+ T cell-autonomous pathway in which complement C5 orchestrated a shift from prostaglandin E2 (PGE2) dominance to enhanced prostacyclin (PGI2) production via activation of C5a receptor 2 (C5aR2). This pivot in lipid mediators induced autocrine signaling through the PGI2 receptor and expression of the interleukin-1 (IL-1) decoy IL-1 receptor type 2 (IL-1R2), which sequestered Th1 cell-driving intrinsic IL-1β, facilitating Th1 cell contraction. Disruption of this C5aR2-PGI2-R axis was a hallmark of pathologically persistent Th1 cell activity in inflammatory conditions, including cryopyrin-associated periodic syndromes (CAPS), Crohn's disease, and rheumatoid arthritis. Rebalancing this axis through selective PGE2 synthase inhibition rectified the hyperactive Th1 cell phenotype in vitro in T cells from individuals with CAPS. Therefore, complement is a key controller of prostanoid metabolism, and the latter is an intrinsic-and potentially druggable-checkpoint for the cessation of Th1 cell effector responses.
Collapse
Affiliation(s)
- Jubayer Rahman
- Complement and Inflammation Research Section, NHLBI, NIH, Bethesda, MD 20892, USA.
| | - Jack A Bibby
- Complement and Inflammation Research Section, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Parul Singh
- Complement and Inflammation Research Section, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Nicolas S Merle
- Complement and Inflammation Research Section, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Erin E West
- Complement and Inflammation Research Section, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Andrea Bohrer
- Laboratory of Clinical Infectious Diseases, Inflammation & Innate Immunity Unit, NIAID, NIH, Bethesda, MD 20892, USA
| | - Katrin Mayer-Barber
- Laboratory of Clinical Infectious Diseases, Inflammation & Innate Immunity Unit, NIAID, NIH, Bethesda, MD 20892, USA
| | - Chengyu Liu
- Transgenic Core, NHLBI/NIH, Bethesda, MD 20892, USA
| | - Lauren R Brinster
- Office of Research Services, Division of Veterinary Resources, NIH, Bethesda, MD 20892, USA
| | - Behdad Afzali
- Immunoregulation Section, Kidney Diseases Branch, NIDDK, NIH, Bethesda, MD 20892, USA
| | - Ana M Briones
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid, Hospital La Paz Institute for Health Research (IdiPaz), CIBER Cardiovascular (CIBERCV), Madrid, Spain
| | - Sara Alehashemi
- Laboratory of Clinical Immunology & Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Farzana Bhuyan
- Laboratory of Clinical Immunology & Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Jiahui Ge
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Xijian Chen
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Yingbi Zhou
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Murray C H Clarke
- The University of Cambridge, Heart & Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Bin Liu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China; Chaoshan Branch of State Key Laboratory for Esophageal Cancer Prevention and Treatment, Shantou University Medical College, Shantou, China
| | | | - C Henrique Serezani
- Vanderbilt Center for Immunobiology and Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Claudia Kemper
- Complement and Inflammation Research Section, NHLBI, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
39
|
Guo G, Zhang L, Liu X, Deng Y, Wu P, Zhao R, Wang W. Fibroblast reprogramming in the dura mater of NTG-induced migraine-related chronic hypersensitivity model drives monocyte infiltration via Angptl1-dependent stromal signaling. J Headache Pain 2025; 26:130. [PMID: 40419944 DOI: 10.1186/s10194-025-02058-4] [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: 03/30/2025] [Accepted: 04/30/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Migraine, characterized by recurrent episodes of severe headache, remains mechanistically enigmatic. While traditional theories emphasize trigeminovascular activation, the role of meningeal stromal-immune crosstalk in disease chronicity is poorly understood. METHODS A migraine-related chronic hypersensitivity model was utilized via intermittent intraperitoneal nitroglycerin (NTG, 10 mg/kg, every other day for 9 days) and peripheral mechanical hypersensitivity was assessed using von Frey filaments. Single-cell RNA sequencing (scRNA-seq) was performed on dura tissues to construct a cellular atlas of NTG-induced remodeling. These data were then integrated with migraine genome-wide association study (GWAS) risk genes, cell-cell interaction networks, and transcriptional regulation analysis to dissect NTG-driven meningeal remodeling. RESULTS The NTG-induced migraine-related chronic hypersensitivity model demonstrated sustained mechanical allodynia, as evidenced by significantly decreased paw withdrawal thresholds (p < 0.0001). Single-cell profiling of the dura mater revealed a 2.4-fold expansion of a pro-inflammatory fibroblast subpopulation (Fibro_c5: 1.9% in Vehicle vs. 4.6% in NTG group), which exhibited marked activation of TNF-α/NF-κB signaling pathways (normalized enrichment score [NES] = 1.83). Concomitantly, we observed an 82% increase in meningeal monocytes (5.7-10.4%) that showed preferential interaction with Fibro_c5 fibroblasts through Angptl1-mediated stromal-immune crosstalk (log2 fold change = 1.41). Regulatory network analysis identified Mafk as the upstream transcriptional regulator orchestrating Angptl1 expression in this pathological communication axis. CONCLUSION Our study reveals that NTG reprograms meningeal fibroblasts to expand a pro-inflammatory fibroblast subtype, which drives migraine-related chronic hypersensitivity through TNF-α/NF-κB signaling and Angptl1-mediated monocyte crosstalk. The identified Mafk-Angptl1 axis presents a potential therapeutic target, though human validation remains essential.
Collapse
Affiliation(s)
- Guangyu Guo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lei Zhang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuyang Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Yiping Deng
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Peiyu Wu
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Ruofan Zhao
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Wei Wang
- Headache Center, Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
40
|
Kawasaki K, Salehi S, Zhan YA, Chen K, Lee JH, Salataj E, Zhong H, Manoj P, Kinyua D, Mello BP, Sridhar H, Tischfield SE, Linkov I, Ceglia N, Zatzman M, Havasov E, Shah NJ, Meng F, Loomis B, Bhanot UK, Redin E, de Stanchina E, Hamard PJ, Koche RP, McPherson A, Quintanal-Villalonga Á, Shah SP, Massagué J, Rudin CM. FOXA2 promotes metastatic competence in small cell lung cancer. Nat Commun 2025; 16:4865. [PMID: 40419484 PMCID: PMC12106783 DOI: 10.1038/s41467-025-60141-5] [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: 10/22/2024] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
Small cell lung cancer (SCLC) is known for its high metastatic potential, with most patients demonstrating clinically evident metastases in multiple organs at diagnosis. The factors contributing to this exceptional metastatic capacity have not been defined. To bridge this gap, we compare gene expression in SCLC patient samples who never experienced metastasis or relapse throughout their clinical course, versus primary SCLC patient samples from more typical patients who had metastatic disease at diagnosis. This analysis identifies FOXA2 as a transcription factor strongly associated with SCLC metastasis. Subsequent analyses in experimental models demonstrates that FOXA2 induces a fetal neuroendocrine gene expression program and promotes multi-site metastasis. Moreover, we identify ASCL1, a transcription factor known for its initiating role in SCLC tumorigenesis, as a direct binder of the FOXA2 promoter and regulator of FOXA2 expression. Taken together, these data define the ASCL1-FOXA2 axis as a critical driver of multiorgan SCLC metastasis.
Collapse
Affiliation(s)
- Kenta Kawasaki
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sohrab Salehi
- Computational Oncology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yingqian A Zhan
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kevin Chen
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jun Ho Lee
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eralda Salataj
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hong Zhong
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Parvathy Manoj
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dennis Kinyua
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Barbara P Mello
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Harsha Sridhar
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sam E Tischfield
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Irina Linkov
- Pathology Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicholas Ceglia
- Computational Oncology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew Zatzman
- Computational Oncology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eliyahu Havasov
- Computational Oncology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Neil J Shah
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fanli Meng
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brian Loomis
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Umesh K Bhanot
- Pathology Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Esther Redin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pierre-Jacques Hamard
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard P Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew McPherson
- Computational Oncology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Sohrab P Shah
- Computational Oncology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joan Massagué
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY, USA.
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
41
|
Beck A, Gabler-Pamer L, Alencastro Veiga Cruzeiro G, Lambo S, Englinger B, Shaw ML, Hack OA, Liu I, Haase RD, de Biagi CAO, Baumgartner A, Nascimento Silva AD, Klenner M, Freidel PS, Herms J, von Baumgarten L, Tonn JC, Thon N, Bruckner K, Madlener S, Mayr L, Senfter D, Peyrl A, Slavc I, Lötsch D, Dorfer C, Geyregger R, Amberg N, Haberler C, Mack N, Schwalm B, Pfister SM, Korshunov A, Baird LC, Yang E, Chi SN, Alexandrescu S, Gojo J, Kool M, Hovestadt V, Filbin MG. Cellular hierarchies of embryonal tumors with multilayered rosettes are shaped by oncogenic microRNAs and receptor-ligand interactions. NATURE CANCER 2025:10.1038/s43018-025-00964-9. [PMID: 40419763 DOI: 10.1038/s43018-025-00964-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 04/01/2025] [Indexed: 05/28/2025]
Abstract
Embryonal tumor with multilayered rosettes (ETMR) is a pediatric brain tumor with dismal prognosis. Characteristic alterations of the chromosome 19 microRNA cluster (C19MC) are observed in most ETMR; however, the ramifications of C19MC activation and the complex cellular architecture of ETMR remain understudied. Here we analyze 11 ETMR samples from patients using single-cell transcriptomics and multiplexed spatial imaging. We reveal a spatially distinct cellular hierarchy that spans highly proliferative neural stem-like cells and more differentiated neuron-like cells. C19MC is predominantly expressed in stem-like cells and controls a transcriptional network governing stemness and lineage commitment, as resolved by genome-wide analysis of microRNA-mRNA binding. Systematic analysis of receptor-ligand interactions between malignant cell types reveals fibroblast growth factor receptor and Notch signaling as oncogenic pathways that can be successfully targeted in preclinical models and in one patient with ETMR. Our study provides fundamental insights into ETMR pathobiology and a powerful rationale for more effective targeted therapies.
Collapse
Affiliation(s)
- Alexander Beck
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Neuropathology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Lisa Gabler-Pamer
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Neurosurgery and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Gustavo Alencastro Veiga Cruzeiro
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sander Lambo
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Bernhard Englinger
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - McKenzie L Shaw
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Olivia A Hack
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ilon Liu
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rebecca D Haase
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carlos A O de Biagi
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alicia Baumgartner
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Andrezza Do Nascimento Silva
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marbod Klenner
- Center for Neuropathology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Pia S Freidel
- Center for Neuropathology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jochen Herms
- Center for Neuropathology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Louisa von Baumgarten
- Department of Neurosurgery, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Joerg C Tonn
- Department of Neurosurgery, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Niklas Thon
- Department of Neurosurgery, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Katharina Bruckner
- Department of Pediatrics and Adolescent Medicine, Comprehensive Cancer Center and Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Sibylle Madlener
- Department of Pediatrics and Adolescent Medicine, Comprehensive Cancer Center and Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Lisa Mayr
- Department of Pediatrics and Adolescent Medicine, Comprehensive Cancer Center and Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Daniel Senfter
- Department of Pediatrics and Adolescent Medicine, Comprehensive Cancer Center and Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Andreas Peyrl
- Department of Pediatrics and Adolescent Medicine, Comprehensive Cancer Center and Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Irene Slavc
- Department of Pediatrics and Adolescent Medicine, Comprehensive Cancer Center and Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Daniela Lötsch
- Department of Neurosurgery and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christian Dorfer
- Department of Neurosurgery and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Rene Geyregger
- Clinical Cell Biology and FACS Core Unit, St. Anna Children's Cancer Research Institute, Vienna, Austria
| | - Nicole Amberg
- Department of Neurology, Division of Neuropathology and Neurochemistry and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christine Haberler
- Department of Neurology, Division of Neuropathology and Neurochemistry and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Norman Mack
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
| | - Benjamin Schwalm
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
| | - Stefan M Pfister
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Department of Paediatric Haematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Andrey Korshunov
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Lissa C Baird
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA
| | - Edward Yang
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA
| | - Susan N Chi
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | | | - Johannes Gojo
- Department of Pediatrics and Adolescent Medicine, Comprehensive Cancer Center and Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Marcel Kool
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- University Medical Center Utrecht (UMCU), Utrecht, the Netherlands
| | - Volker Hovestadt
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Mariella G Filbin
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
42
|
Kim JP, Cho M, Kim C, Lee H, Jang B, Jung SH, Kim Y, Koh IG, Kim S, Shin D, Lee EH, Lee JY, Park Y, Jang H, Kim BH, Ham H, Kim B, Kim Y, Cho AH, Raj T, Kim HJ, Na DL, Seo SW, An JY, Won HH. Whole-genome sequencing analyses suggest novel genetic factors associated with Alzheimer's disease and a cumulative effects model for risk liability. Nat Commun 2025; 16:4870. [PMID: 40419521 DOI: 10.1038/s41467-025-59949-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 05/08/2025] [Indexed: 05/28/2025] Open
Abstract
Genome-wide association studies (GWAS) on Alzheimer's disease (AD) have predominantly focused on identifying common variants in Europeans. Here, we performed whole-genome sequencing (WGS) of 1,559 individuals from a Korean AD cohort to identify various genetic variants and biomarkers associated with AD. Our GWAS analysis identified a previously unreported locus for common variants (APCDD1) associated with AD. Our WGS analysis was extended to explore the less-characterized genetic factors contributing to AD risk. We identified rare noncoding variants located in cis-regulatory elements specific to excitatory neurons associated with cognitive impairment. Moreover, structural variation analysis showed that short tandem repeat expansion was associated with an increased risk of AD, and copy number variant at the HPSE2 locus showed borderline statistical significance. APOE ε4 carriers with high polygenic burden or structural variants exhibited severe cognitive impairment and increased amyloid beta levels, suggesting a cumulative effects model of AD risk.
Collapse
Affiliation(s)
- Jun Pyo Kim
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Minyoung Cho
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Chanhee Kim
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, Republic of Korea
- L-HOPE Program for Community-Based Total Learning Health Systems, Korea University, Seoul, Republic of Korea
| | - Hyunwoo Lee
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Beomjin Jang
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sang-Hyuk Jung
- Department of Medical Informatics, Kangwon National University College of Medicine, Chuncheon, Republic of Korea
| | - Yujin Kim
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, Republic of Korea
- L-HOPE Program for Community-Based Total Learning Health Systems, Korea University, Seoul, Republic of Korea
| | - In Gyeong Koh
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, Republic of Korea
- L-HOPE Program for Community-Based Total Learning Health Systems, Korea University, Seoul, Republic of Korea
| | - Seoyeon Kim
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, Republic of Korea
- L-HOPE Program for Community-Based Total Learning Health Systems, Korea University, Seoul, Republic of Korea
| | - Daeun Shin
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Eun Hye Lee
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - YoungChan Park
- Division of Bio Bigdata, Department of Precision Medicine, Korea National Institution of Health, Cheongju, Republic of Korea
| | - Hyemin Jang
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Seoul National University Hospital, Seoul National University School of Medicine, Seoul, Republic of Korea
| | - Bo-Hyun Kim
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Hongki Ham
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Beomsu Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Yujin Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - A-Hyun Cho
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Towfique Raj
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hee Jin Kim
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Duk L Na
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Sang Won Seo
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea.
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea.
| | - Joon-Yong An
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, Republic of Korea.
- L-HOPE Program for Community-Based Total Learning Health Systems, Korea University, Seoul, Republic of Korea.
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, Republic of Korea.
| | - Hong-Hee Won
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea.
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
43
|
Hsiao C, Liao WC, Li JP, Chou YC, Chou YL, Lin JR, Chen CH, Liu CH. Revealing a novel Decorin-expressing tumor stromal subset in hepatocellular carcinoma via integrative analysis single-cell RNA sequencing. Cancer Cell Int 2025; 25:194. [PMID: 40420150 DOI: 10.1186/s12935-025-03811-0] [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: 11/20/2024] [Accepted: 05/07/2025] [Indexed: 05/28/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains a leading cause of cancer-related mortality, emphasizing the need for novel therapeutic strategies. Decorin (DCN), a chondroitin sulfate proteoglycan (CSPG), has been proposed as a tumor suppressor, yet its precise role in HCC and the tumor microenvironment (TME) remains underexplored. Through integrated analyses of bulk RNA and single-cell RNA sequencing datasets, we identified a distinct tumor stromal subset highly expressing DCN and associated chondroitin sulfate (CS) synthases. Our findings revealed that DCN expression is significantly downregulated in HCC tissue, but upregulated in peri-tumor stroma, where it correlates with better prognosis and reduced capsular invasion. Western blot analysis demonstrated that CS-DCN, the glycosylated form of DCN, plays a dominant role in this context. Single-cell clustering analysis identified a unique stromal subset in HCC characterized by elevated expression of DCN, CSPGs, and CS synthases, associated with extracellular matrix (ECM) remodeling and protective barrier functions. A six-gene DCN-associated signature derived from this subset, including DCN, BGN, SRPX, CHSY3, CHST3, and CHPF, was validated as a prognostic marker for HCC. Furthermore, functional assays demonstrated that CS-DCN significantly inhibited HCC cell proliferation and invasion. Our study highlights the critical role of DCN in HCC TME and provides insights into its therapeutic potential. Modulating CSPG pathways, particularly on CS-DCN-expressing stromal cells, may offer a promising approach for improving HCC treatment and patient outcomes.
Collapse
Affiliation(s)
- Chi Hsiao
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing University, Taichung, Republic of China
- Department of Pathology, School of Medicine, Chung Shan Medical University, Taichung, Republic of China
| | - Wen-Chieh Liao
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing University, Taichung, Republic of China
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Republic of China
| | - Ju-Pi Li
- Department of Pathology, School of Medicine, Chung Shan Medical University, Taichung, Republic of China
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Republic of China
| | - Yu-Cheng Chou
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Republic of China
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Republic of China
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Yu-Lun Chou
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing University, Taichung, Republic of China
| | - Jeng-Rong Lin
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing University, Taichung, Republic of China
| | - Chia-Hua Chen
- Department of Anatomy, School of Medicine, Chang Gung University, Taoyuan, Republic of China.
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Republic of China.
- Neuroscience Research Center, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Republic of China.
| | - Chiung-Hui Liu
- Doctoral Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing University, Taichung, Republic of China.
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Republic of China.
| |
Collapse
|
44
|
Zhang Q, Wang M, Li Y, Zhang H, Wang Y, Chen X, Yao L, Cui M, Dong H, Li X, Liu J, Zhu B, Xu Y. Efficacy, safety and exploratory analysis of neoadjuvant tislelizumab (a PD-1 inhibitor) plus nab-paclitaxel followed by epirubicin/cyclophosphamide for triple-negative breast cancer: a phase 2 TREND trial. Signal Transduct Target Ther 2025; 10:169. [PMID: 40414961 DOI: 10.1038/s41392-025-02254-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/13/2025] [Accepted: 04/28/2025] [Indexed: 05/27/2025] Open
Abstract
The optimal chemotherapy backbone and specific population of triple-negative breast cancer (TNBC) patients that benefit from neoadjuvant immunotherapy are not well established. This prospective, single-arm, phase II TREND trial assessed the efficacy and safety of tislelizumab plus nab-paclitaxel and epirubicin/cyclophosphamide-based chemotherapy as a neoadjuvant treatment for TNBC (ChiCTR2000035262). The primary endpoint was pathological complete response (pCR), with the secondary endpoints including safety assessment and objective response rate (ORR). ScRNA-seq, bulk RNA-seq, TCR-seq, cyTOF and WES were performed on pre-treatment and post-treatment samples. Among 53 total enrolled patients, 44 completed the combined neoadjuvant therapy, and 30 of 44 patients (68.18%) achieved pCR. Additionally, 14 out of 44 patients had a complete response (31.82%), with an ORR of 93.18%. The most commonly observed treatment-related adverse events (TRAEs) were alopecia, nausea and liver injury with 6 cases classified as grade 3 or higher adverse events. Immune response-related pathways, including TNF signaling pathway, T cell receptor signaling pathway, were enriched in pCR group. Pre-treatment model was identified and construct to predict response to immunotherapy. CDKN1A+ CD8 T lymphocytes were enriched in pCR group after neoadjuvant immunotherapy. Dynamic change of immune-related pathways at an early stage during the neoadjuvant immunotherapy may be associated with the treatment efficacy. In conclusion, neoadjuvant treatment of tislelizumab with nab-paclitaxel and anthracycline-based chemotherapy showed promising clinical activity and was well-tolerated among TNBC patients, without high incidence of TRAEs. These findings provide evidence supporting neoadjuvant tislelizumab with chemotherapy as an effective rational approach for treating TNBC.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, 110801, P. R. China
| | - Mozhi Wang
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, P. R. China
| | - Yumeng Li
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, P. R. China
| | - Hengjun Zhang
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, P. R. China
| | - Yusong Wang
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, P. R. China
| | - Xiuyun Chen
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, P. R. China
| | - Litong Yao
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, P. R. China
| | - Mingke Cui
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, 110801, P. R. China
| | - Haoran Dong
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, P. R. China
| | - Xiang Li
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, P. R. China
| | - Jian Liu
- Centre for Bioinformatics and Intelligent Medicine, Nankai University, Tianjin, 300071, P. R. China
- College of Computer Science, Nankai University, Tianjin, 300071, P. R. China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P. R. China.
| | - Yingying Xu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, P. R. China.
| |
Collapse
|
45
|
Zhang J, Jin X, Hou Y, Gu B, Li H, Yi L, Wu W, Hu S. Comprehensive analysis of the critical role of the epithelial mesenchymal transition subtype - TAGLN-positive fibroblasts in colorectal cancer progression and immunosuppression. Cell Biosci 2025; 15:66. [PMID: 40413514 PMCID: PMC12102804 DOI: 10.1186/s13578-025-01405-x] [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: 01/29/2025] [Accepted: 05/05/2025] [Indexed: 05/27/2025] Open
Abstract
Epithelial-mesenchymal transition (EMT) plays a pivotal role in tumor metastasis and immune suppression in colorectal cancer (CRC). However, the specific mechanisms of EMT and its relationship with the clinical prognosis and immunotherapy response in CRC patients remain unclear. In this study, we identified TAGLN-positive fibroblasts (TAGLN⁺Fib) as a cancer-associated fibroblast (CAF) subtype within the tumor microenvironment (TME) that promotes tumor metastasis and immune evasion. High EMT scores, strongly associated with TAGLN expression, were correlated with advanced tumor stages, poor prognosis, and resistance to immunotherapy. Functional experiments demonstrated that TAGLN knockdown significantly reduced CRC cell proliferation, migration, and EMT phenotypes in vitro and suppressed tumor growth in vivo. Furthermore, TAGLN⁺Fib closely interacted with MMP7-positive tumor epithelial cells and SPP1-positive macrophages, forming a pro-metastatic and immunosuppressive network. An EMT-TME risk model constructed using TAGLN⁺Fib exhibited robust predictive power for CRC prognosis and immunotherapy response. This study reveals the association of EMT scores with CRC prognosis and immunotherapy response, highlights TAGLN⁺Fib's critical role in tumor progression, and develops an EMT-TME risk model, offering insights for personalized CRC treatment and precision medicine.
Collapse
Affiliation(s)
- Junli Zhang
- Department of Blood transfusion, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, No. 38 Shengli Road, Bengshan District, Bengbu City, Anhui Province, China
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Anhui, China
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, Anhui, 233030, China
| | - Xinxin Jin
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, Anhui, 233030, China
| | - Yachao Hou
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, Anhui, 233030, China
| | - Biao Gu
- Department of Blood transfusion, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, No. 38 Shengli Road, Bengshan District, Bengbu City, Anhui Province, China
| | - Hongwei Li
- Department of Blood transfusion, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, No. 38 Shengli Road, Bengshan District, Bengbu City, Anhui Province, China
| | - Li Yi
- Department of Blood transfusion, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, No. 38 Shengli Road, Bengshan District, Bengbu City, Anhui Province, China
| | - Wenjuan Wu
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, Anhui, 233030, China.
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, 233030, China.
| | - Shangshang Hu
- Department of Blood transfusion, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, No. 38 Shengli Road, Bengshan District, Bengbu City, Anhui Province, China.
- School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China.
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui, 233030, China.
| |
Collapse
|
46
|
Tokavanich N, Chan B, Strauss K, Castro Andrade CD, Arai Y, Nagata M, Foretz M, Brooks DJ, Ono N, Ono W, Wein MN. Control of alveolar bone development, homeostasis, and socket healing by salt-inducible kinases. J Bone Miner Res 2025; 40:656-670. [PMID: 40057979 DOI: 10.1093/jbmr/zjaf038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 05/26/2025]
Abstract
Alveolar bone supports and anchors teeth. The parathyroid hormone-related protein (PTHrP) pathway plays a key role in alveolar bone biology. Salt-inducible kinases (SIKs) are important downstream regulators of PTH/PTHrP signaling in the appendicular skeleton, where SIK inhibition increases bone formation and trabecular bone mass. However, the function of these kinases in alveolar bone remains unknown. Here, we report a critical role for SIK2/SIK3 in alveolar bone development, homeostasis, and socket healing after tooth extraction. Inducible SIK2/SIK3 (Ubq-creERt;Sik2f/f;Sik3f/f) deletion led to dramatic alveolar bone defects without changes in tooth eruption. Ablating these kinases impairs alveolar bone formation due to disrupted osteoblast maturation, a finding associated with ectopic periostin expression by fibrous cells in regions of absent alveolar bone at steady state and following molar extraction. Notably, this phenotype is the opposite of the increased trabecular bone mass observed in long bones following SIK2/SIK3 deletion. Distinct phenotypic consequences of SIK2/SIK3 deletion in appendicular versus craniofacial bones prompted us to identify a specific transcriptomic signature in alveolar versus long bone osteoblasts. Thus, SIK2/SIK3 deletion illuminates a key role for these kinases in alveolar bone biology and highlights the emerging concept that different osteoblast subsets utilize unique genetic programs.
Collapse
Affiliation(s)
- Nicha Tokavanich
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
- Harvard School of Dental Medicine, Boston, MA, 02115, United States
| | - Byron Chan
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - Katelyn Strauss
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - Christian D Castro Andrade
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
- Molecular, Cellular, and Integrative Physiology, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Yuki Arai
- University of Texas Health Science Center at Houston School of Dentistry, Houston, TX 77204, United States
| | - Mizuki Nagata
- University of Texas Health Science Center at Houston School of Dentistry, Houston, TX 77204, United States
| | - Marc Foretz
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | - Daniel J Brooks
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - Noriaki Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, TX 77204, United States
| | - Wanida Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, TX 77204, United States
| | - Marc N Wein
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, United States
- Harvard Stem Cell Institute, Cambridge, MA 02138, United States
| |
Collapse
|
47
|
Jie XL, Wei JC, Wang D, Zhang XW, Lv MY, Lin YF, Tan YS, Wang Z, Alifu A, Ji L, Shen YK, Wang C, Xu BQ, Liu Z, Han SC, Wang ZH, Tong XW, Feng L, Ying JM, Zhou GB, Wang GZ. CDC34 suppresses macrophage phagocytic activity and predicts poor response to immune checkpoint inhibitor in cancers. Cancer Lett 2025:217822. [PMID: 40419082 DOI: 10.1016/j.canlet.2025.217822] [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: 01/11/2025] [Revised: 05/08/2025] [Accepted: 05/23/2025] [Indexed: 05/28/2025]
Abstract
The cell division cycle 34 (CDC34) is an E2 ubiquitin-conjugating enzyme that is required for proteasomal degradation of substrate proteins, and is able to stabilize proteins including the epidermal growth factor receptor to promote lung carcinogenesis. Here, we conducted a pan-cancer analysis of CDC34 in The Cancer Genome Atlas datasets, and found its high expression in breast cancer and negative association with patient outcomes. Analysis of single-cell RNA-sequencing data revealed a negative role of CDC34 in macrophage phagocytotic activity for cancer cells. CDC34 stabilized hypoxia-inducible factor 1α (HIF1α) and transcriptionally upregulated CD47 in cancer cells to evade phagocytosis by macrophages. Inhibition of CDC34 inhibited tumor growth and synergized with anti-PD-L1 antibody in murine models. CDC34 was positively associated with CD47 and negatively associated with CD8+ granzyme B+ T-cell infiltration in patient samples, and patients with co-overexpression of CDC34 and CD47 had markedly poorer prognosis compared to those with high expression of either marker alone. In pre-treatment tumor samples, non-responders to immunotherapy exhibited significantly higher CDC34 levels and reduced CD8+ T-cell infiltration compared to responders. These findings indicated that CDC34 is critical to immune evasion and could be a potential therapeutic target for those resistant to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Xiao-Liang Jie
- State Key Laboratory of Molecular Oncology & Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Jiangxi Key Laboratory of Oncology (2024SSY06041), Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang 330029, Jiangxi Province of China
| | - Jia-Cong Wei
- State Key Laboratory of Molecular Oncology & Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Di Wang
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xiang-Wei Zhang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250117, Shandong Province of China
| | - Meng-Yao Lv
- State Key Laboratory of Molecular Oncology & Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yong-Fang Lin
- State Key Laboratory of Molecular Oncology & Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yi-Shuai Tan
- State Key Laboratory of Molecular Oncology & Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; West China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan Province of China
| | - Zheng Wang
- State Key Laboratory of Molecular Oncology & Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Aikede Alifu
- State Key Laboratory of Molecular Oncology & Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lei Ji
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yu-Ke Shen
- State Key Laboratory of Molecular Oncology & Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Cong Wang
- State Key Laboratory of Molecular Oncology & Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Bing-Qing Xu
- State Key Laboratory of Molecular Oncology & Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zheng Liu
- State Key Laboratory of Molecular Oncology & Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Si-Chong Han
- State Key Laboratory of Molecular Oncology & Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zi-Hao Wang
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xiao-Wan Tong
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lin Feng
- Department of Pathology, The First Medical Center of PLA General Hospital, Beijing 100853, China
| | - Jian-Ming Ying
- State Key Laboratory of Molecular Oncology & Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Guang-Biao Zhou
- State Key Laboratory of Molecular Oncology & Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Institute of Cancer Research, Henan Academy of Innovations in Medical Sciences, Zhengzhou 450000, Henan Province, China.
| | - Gui-Zhen Wang
- State Key Laboratory of Molecular Oncology & Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
48
|
Li QY, Liu F, Li X, Kang M, Bai L, Tong T, Zheng C, Jin Y, Zhang X, Xie Y, Tian D, Pan Y, Wang J, Fu H, Jiao N, Wu J, Mao J. Single cell analysis identified IFN signaling activation contributes to the pathogenesis of pediatric steroid-sensitive nephrotic syndrome. Biomark Res 2025; 13:77. [PMID: 40413560 PMCID: PMC12103753 DOI: 10.1186/s40364-025-00790-2] [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: 10/02/2024] [Accepted: 05/16/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Idiopathic nephrotic syndrome (INS) is a prevalent condition whose recurrence leads to multiple adverse effects. Previous studies on INS pathogenesis primarily focused on immune dysregulation, particularly T-cell changes and their correlation with cytokine shifts. Accumulating evidence suggests that B-cell dysfunction also plays a role. Nevertheless, a comprehensive understanding of the mechanisms and effective treatment strategies remains incomplete. METHODS This study investigates changes in gene expressions and cellular interactions of immune cells at a single-cell level using peripheral blood mononuclear cells (PBMCs). And subsequently validated through quantitative PCR (qPCR), enzyme-linked immunosorbent assay (ELISA), and flow cytometry. RESULTS We identified seven main clusters using unsupervised clustering of 103,213 high-quality single cells. Through unsupervised clustering, patient-specific T cells (IFI44L + CD4 + T cells) that exhibited a pronounced elevation of interferon-stimulated genes (ISGs) is identified. Activation of ISGs and interferon (IFN)-related pathways are also observed in other clusters. Specifically, this study demonstrates that interferon-γ (IFN-γ) plays a crucial role by promoting the interaction between B-cell activating factor (BAFF) and receptors on B cells. This interaction triggers the release of autoantibodies, thereby initiating INS pathogenesis. Furthermore, telitacicept has shown efficacy in treating pediatric patients with frequent relapse NS(FRNS). CONCLUSIONS Overall, our findings underscore the role of interferon and its related pathways in INS pathogenesis, providing novel therapeutic interventions for NS.
Collapse
Affiliation(s)
- Qiu-Yu Li
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333, Binsheng Road, Binjiang District, Hangzhou, Zhejiang, 310052, China
| | - Fei Liu
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333, Binsheng Road, Binjiang District, Hangzhou, Zhejiang, 310052, China
| | - Xiaoyi Li
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333, Binsheng Road, Binjiang District, Hangzhou, Zhejiang, 310052, China
| | - Minchao Kang
- Department of Nephrology, Zhejiang University Medical College Affiliated Sir Run Run Shaw Hospital, Qingchun Road 3rd, Hangzhou, Zhejiang Province, 310016, China
| | - Linnan Bai
- Department of Nephrology, Zhejiang University Medical College Affiliated Sir Run Run Shaw Hospital, Qingchun Road 3rd, Hangzhou, Zhejiang Province, 310016, China
| | - Tong Tong
- Zhejiang University School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, China
| | - Chen Zheng
- Zhejiang University School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, China
| | - Yanyan Jin
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333, Binsheng Road, Binjiang District, Hangzhou, Zhejiang, 310052, China
| | - Xiaojing Zhang
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333, Binsheng Road, Binjiang District, Hangzhou, Zhejiang, 310052, China
| | - Yi Xie
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333, Binsheng Road, Binjiang District, Hangzhou, Zhejiang, 310052, China
| | - Dandan Tian
- Department of Clinical Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yuanqing Pan
- Department of Nephrology, Quanzhou Women's and Children's Hospital, Quanzhou, Fujian Province, 362000, China
| | - Jingjing Wang
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333, Binsheng Road, Binjiang District, Hangzhou, Zhejiang, 310052, China
| | - Haidong Fu
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333, Binsheng Road, Binjiang District, Hangzhou, Zhejiang, 310052, China
| | - Na Jiao
- State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Junnan Wu
- Department of Nephrology, Zhejiang University Medical College Affiliated Sir Run Run Shaw Hospital, Qingchun Road 3rd, Hangzhou, Zhejiang Province, 310016, China.
| | - JianHua Mao
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333, Binsheng Road, Binjiang District, Hangzhou, Zhejiang, 310052, China.
| |
Collapse
|
49
|
Xu J, Yang J, Sun Q, Chang J, Wang F. Analyses of single-cell RNA sequencing uncover the role of intratumoral Helicobacter pylori in shaping tumor progression and immunity in gastric cancer. Cancer Immunol Immunother 2025; 74:218. [PMID: 40411560 PMCID: PMC12103440 DOI: 10.1007/s00262-025-04048-6] [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: 12/04/2024] [Accepted: 04/08/2025] [Indexed: 05/26/2025]
Abstract
The intratumoral microbiota is closely associated with tumor initiation and progression in multiple solid tumors, including gastric cancer (GC). Single-cell analysis of host-microbiome interactions (SAHMI) is a pipeline used to systematically recover and denoise microbial signals in human clinical tissues and examine tumor-microbiome interactions at the single-cell transcriptome level. In a large GC cohort, we used SAHMI to detect 12 bacteria, among which Helicobacter pylori (H. pylori) was widely present in multiple tumor and normal samples. Meanwhile, we verified the presence of H. pylori in GC tissues via fluorescence in situ hybridization and immunohistochemistry. We performed single-cell RNA sequencing to analyze 11 cell populations, including B cells, T cells, and epithelial cells, and these cell types contained large numbers of H. pylori. We detected obvious enrichment of H. pylori in cancer cells and identified 13 upregulated differentially expressed genes exhibiting significantly negative correlations with patient survival in the H. pylori-positive tumor group compared with the findings in the other groups, indicating that these genes could represent prognostic biomarkers or therapeutic targets for H. pylori-infected patients with GC. Moreover, H. pylori-enriched immune cells, including T cells, B cells, and macrophages, were associated with cell-type-specific gene expression and pathway activities, including cell fate and immune signaling. In summary, tumor-microbiome interactions might reflect or influence tumorigenesis in GC, which has implications for clinical practice.
Collapse
Affiliation(s)
- Jiao Xu
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Jin Yang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China
- Phase I Clinical Trial Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Qi Sun
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Jingbo Chang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Fan Wang
- Phase I Clinical Trial Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China.
| |
Collapse
|
50
|
Kumar S, De T, Subramani J, Rangarajan A, Pal D. Combined analysis of somatic mutations and gene expression reveals nuclear speckles-associated enhanced stemness in gingivobuccal carcinoma under DNA damage response. Comput Biol Chem 2025; 119:108513. [PMID: 40424937 DOI: 10.1016/j.compbiolchem.2025.108513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 05/04/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025]
Abstract
Smokeless tobacco chewing habits in India lead to a high prevalence of Gingivobuccal oral squamous cell carcinoma (OSCC-GB). Cancer stem cells (CSCs) are a sub-population of cancer cells within a tumor with stem-like properties and are believed to contribute to tumor initiation, progression, increased resistance to drug therapy, and promote post-therapeutic cancer relapse. An RNA-sequencing data-based combined analysis of somatic mutations and gene expression was performed to explore the role of CSCs in disease progression using the novel Indian-origin OSCC-GB cell line 'IIOC019' from a patient with tobacco-chewing habit. The identified DNA damage-related known mutational signature (1 bp T/(A) nucleotide insertions and C>T mutations) indicates the impact of smokeless tobacco-related carcinogens in the IIOC019 cell line. The differentially expressed somatic variants, functional impact predictions, and survival analysis reveal the role of DNA damage response (DDR)-related genes in OSCC-GB, with the SON gene as a significant player. The study suggests that the loss-of-function in a somatic variant of the SON gene is linked to nuclear speckles-associated enhanced stemness and increased risk of disease progression in OSCC-GB under DDR conditions. The newly identified CSC-associated somatic variants appear to promote cancer spread, local recurrence, and resistance to chemotherapy or radiotherapy, contributing to the high mortality rates among Indian OSCC-GB patients.
Collapse
Affiliation(s)
- Sachendra Kumar
- IISc Mathematics Initiative, Indian Institute of Science, Bengaluru, Karnataka 560 012, India; Computational and Data Sciences, Indian Institute of Science, Bengaluru, Karnataka 560 012, India
| | - Tamasa De
- Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka 560 012, India
| | - Janavi Subramani
- Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka 560 012, India
| | - Annapoorni Rangarajan
- Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, Karnataka 560 012, India.
| | - Debnath Pal
- Computational and Data Sciences, Indian Institute of Science, Bengaluru, Karnataka 560 012, India.
| |
Collapse
|