1
|
Sinha S, Roy R, Barman N, Sarkar P, Saha A, Biswas N. IL6 mediated cFLIP downregulation increases the migratory and invasive potential of triple negative breast cancer cell. Cell Signal 2025; 130:111679. [PMID: 39988287 DOI: 10.1016/j.cellsig.2025.111679] [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: 08/14/2024] [Revised: 02/03/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
c-FLIP (cellular FLICE-Like Inhibitor of Apoptotic protein) alias CFLAR (Cellular FADD-like apoptosis regulator) is an inhibitor of Caspase 8 and thus plays a key role in the regulation of extrinsic apoptotic pathway. However, the mechanisms of cFLIP regulation during the course of progression of cancer and it's involvement in tumour cell migration and invasion is yet to be known. Our TCGA data analysis has shown that cFLIP is downregulated in many cancers, including breast cancer, especially at the later stages. Next, we have analysed the role of cFLIP in breast cancer progression in In-vitro study. In doing so, we have used luminal breast cancer cell line MCF7 as non-aggressive and non-invasive breast cancer model and triple negative breast cancer cell lines MDA-MB-231, MDA-MB-468 and MDA-MB-453 as highly aggressive and invasive breast cancer cell model. When, we analysed and compared MCF7 and triple negative cell lines, we found a negative correlation between cFLIP expression pattern and metastasis which supported our In-silico study. Moreover, we found that Il6, one of the most prominent cytokines inside tumour microenvironment, helped in cFLIP downregulation via activation of p38 in MDA-MB-231 cell line. Not only that we have shown that cFLIP negatively regulated autophagy and this autophagy down-regulation resulted in decrease in metastasis. Thus, we have shown in an In-vitro model, for the first time, a complete interconnecting pathway in which IL6 mediated p38 activation directly influences metastasis by regulating autophagy via cFLIP downregulation.
Collapse
Affiliation(s)
- Samraj Sinha
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Rajdeep Roy
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Nilesh Barman
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Purandar Sarkar
- Institute of Health Sciences, Presidency University, Kolkata, India
| | - Abhik Saha
- Institute of Health Sciences, Presidency University, Kolkata, India
| | - Nabendu Biswas
- Department of Life Sciences, Presidency University, Kolkata, India.
| |
Collapse
|
2
|
Han Y, Hu M, Wang Y, Xu S, Jiang F, Wang Y, Liu Z. A coagulation-related long non-coding RNA signature to predict prognosis and immune features of breast cancer. Discov Oncol 2025; 16:662. [PMID: 40317354 DOI: 10.1007/s12672-025-02316-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/04/2025] [Indexed: 05/07/2025] Open
Abstract
Breast cancer (BC) remains one of the most common malignancies among women worldwide, with persistently poor prognosis despite advancements in diagnostics and therapies. Long non-coding RNAs (lncRNAs) and coagulation-related genes (CRGs) are increasingly recognized for their roles in prognosis and immune modulation. Using transcriptomic data from 1,045 BC patients in TCGA, we identified CRG-associated lncRNAs via coexpression analysis (Pearson |R|> 0.4, p < 0.001) and constructed a prognostic model through univariate Cox analysis, LASSO regression with tenfold cross-validation (λ = 0.05), and multivariate Cox analysis. The model stratified patients into high- and low-risk groups with distinct overall survival (HR = 3.21, p < 0.001) and demonstrated robust predictive accuracy (AUC = 0.795 at 1 year). Functional enrichment revealed immune-related pathways (e.g., cytokine signaling, PD-L1 regulation), and high-risk patients exhibited elevated tumor mutational burden (TMB) and PD-L1 expression, suggesting enhanced immunotherapy responsiveness. Drug sensitivity analysis identified 5 targeted agents (e.g., BIBW2992) with differential efficacy between risk groups. This CRG-lncRNA signature provides a novel tool for prognosis prediction and personalized immunotherapy in BC, illuminating crosstalk between coagulation and immune pathways.
Collapse
Affiliation(s)
- Yetao Han
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Mengsi Hu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Yanzhong Wang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Shoufang Xu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Feiyu Jiang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Yingjian Wang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China
| | - Zhiwei Liu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, China.
| |
Collapse
|
3
|
Chalepaki AM, Gkoris M, Chondrou I, Kourti M, Georgakopoulos-Soares I, Zaravinos A. A multi-omics analysis of effector and resting treg cells in pan-cancer. Comput Biol Med 2025; 189:110021. [PMID: 40088713 DOI: 10.1016/j.compbiomed.2025.110021] [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: 10/05/2024] [Revised: 02/09/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025]
Abstract
Regulatory T cells (Tregs) are critical for maintaining the stability of the immune system and facilitating tumor escape through various mechanisms. Resting T cells are involved in cell-mediated immunity and remain in a resting state until stimulated, while effector T cells promote immune responses. Here, we investigated the roles of two gene signatures, one for resting Tregs (FOXP3 and IL2RA) and another for effector Tregs (FOXP3, CTLA-4, CCR8 and TNFRSF9) in pan-cancer. Using data from The Cancer Genome Atlas (TCGA), The Cancer Proteome Atlas (TCPA) and Gene Expression Omnibus (GEO), we focused on the expression profile of the two signatures, the existence of single nucleotide variants (SNVs) and copy number variants (CNVs), methylation, infiltration of immune cells in the tumor and sensitivity to different drugs. Our analysis revealed that both signatures are differentially expressed across different cancer types, and correlate with patient survival. Furthermore, both types of Tregs influence important pathways in cancer development and progression, like apoptosis, epithelial-to-mesenchymal transition (EMT) and the DNA damage pathway. Moreover, a positive correlation was highlighted between the expression of gene markers in both resting and effector Tregs and immune cell infiltration in adrenocortical carcinoma, while mutations in both signatures correlated with enrichment of specific immune cells, mainly in skin melanoma and endometrial cancer. In addition, we reveal the existence of widespread CNVs and hypomethylation affecting both Treg signatures in most cancer types. Last, we identified a few correlations between the expression of CCR8 and TNFRSF9 and sensitivity to several drugs, including COL-3, Chlorambucil and GSK1070916, in pan-cancer. Overall, these findings highlight new evidence that both Treg signatures are crucial regulators of cancer progression, providing potential clinical outcomes for cancer therapy.
Collapse
Affiliation(s)
- Anna-Maria Chalepaki
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| | - Marios Gkoris
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| | - Irene Chondrou
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus.
| | - Malamati Kourti
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus.
| | - Ilias Georgakopoulos-Soares
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA.
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| |
Collapse
|
4
|
Chen SH, Yu BY, Kuo WY, Lin YB, Su SY, Chuang WH, Lu IH, Lin CY. Unveiling the immune microenvironment of complex tissues and tumors in transcriptomics through a deconvolution approach. BMC Cancer 2025; 25:733. [PMID: 40307726 PMCID: PMC12044707 DOI: 10.1186/s12885-025-14089-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Accurately resolving the composition of tumor-infiltrating leukocytes is pivotal for advancing cancer immunotherapy strategies. Despite the success of some clinical trials, applying these strategies remains limited due to the challenges in deciphering the immune microenvironment. In this study, we developed a streamlined, two-step workflow to address the complexity of bioinformatics processes involved in analyzing immune cell composition from transcriptomics data. Our dockerized toolkit, DOCexpress_fastqc, integrates the hisat2-stringtie pipeline with customized scripts within Galaxy/Docker environments, facilitating RNA sequencing (RNA-seq) gene expression profiling. The output from DOCexpress_fastqc is seamlessly formatted with mySORT, a web application that employs a deconvolution algorithm to determine the immune content across 21 cell subclasses. We validated mySORT using synthetic pseudo-bulk data derived from single-cell RNA sequencing (scRNA-seq) datasets. Our predictions exhibit strong concordance with the ground-truth immune cell composition, achieving Pearson's correlation coefficients of 0.871 in melanoma patients and 0.775 in head and neck cancer patients. Additionally, mySORT outperforms existing methods like CIBERSORT in accuracy and provides a wide range of data visualization features, such as hierarchical clustering and cell complexity plots. The toolkit and web application are freely available for the research community, providing enhanced resolution for conventional bulk RNA sequencing data and facilitating the analysis of immune microenvironment responses in immunotherapy. The mySORT demo website and Docker image are free at https://mysort.iis.sinica.edu.tw and https://hub.docker.com/r/lsbnb/mysort_2022 .
Collapse
Affiliation(s)
- Shu-Hwa Chen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, 250 Wu-Xing Street, Taipei, Taiwan
| | - Bo-Yi Yu
- Research Center for Advanced Science and Technology, the University of Tokyo, 4-6-1 Komaba, Meguro-Ku, Tokyo, 153-8904, Japan
| | - Wen-Yu Kuo
- Institute of Information Science, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan
| | - Ya-Bo Lin
- Institute of Information Science, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan
| | - Sheng-Yao Su
- Department of Smart Computing and Applied Mathematics, Tunghai University, Taichung City, 407224, Taiwan
| | - Wei-Hsuan Chuang
- Institute of Information Science, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan
| | - I-Hsuan Lu
- Institute of Information Science, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan
| | - Chung-Yen Lin
- Institute of Information Science, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan.
- Institute of Fisheries Science, National Taiwan University, Taipei, Taiwan.
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
5
|
Lee S, Ho YY, Hao S, Ouyang Y, Liew UL, Goyal A, Li S, Barbour JA, He M, Huang Y, Wong JWH. A tumour necrosis factor-α responsive cryptic promoter drives overexpression of the human endogenous retrovirus ERVK-7. J Biol Chem 2025:108568. [PMID: 40316021 DOI: 10.1016/j.jbc.2025.108568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 04/09/2025] [Accepted: 04/28/2025] [Indexed: 05/04/2025] Open
Abstract
Endogenous retroviruses (ERVs) shape human genome functionality and influence disease pathogenesis, including cancer. ERVK-7, a significant ERV, acts as an immune modulator and prognostic marker in lung adenocarcinoma (LUAD). Although ERVK-7 overexpression has been linked to the amplification of the 1q22 locus in approximately 10% of LUAD cases, it predominantly arises from alternative regulatory mechanisms. Our findings indicate that the canonical 5' long terminal repeat (LTR) of ERVK-7 is methylated and inactive, necessitating the use of alternative upstream promoters. We identified two novel transcripts, ERVK-7.long and ERVK-7.short, arising from distinct promoters located 2.8 kb and 13.8 kb upstream of the 5'LTR of ERVK-7, respectively. ERVK-7.long is predominantly overexpressed in LUAD. Through comprehensive epigenetic mapping and single-cell transcriptomics, we demonstrate that ERVK-7.long activation is predetermined by cell lineage, specifically in small airway epithelial cells (SAECs), where its promoter displays tumor-specific H3K4me3 modifications. Single-cell RNA sequencing further reveals a distinct enrichment of ERVK-7.long in LUAD tumor cells and alveolar type 2 epithelial cells, underscoring a cell-type-specific origin. Additionally, inflammatory signaling significantly influences ERVK-7 expression; TNF-α enhances ERVK-7.long, while interferon signaling preferentially augments ERVK-7.short by differential recruitment of NF-κB/RELA and IRF to their respective promoters. This differential regulation clarifies the elevated ERVK-7 expression in LUAD compared to lung squamous cell carcinoma (LUSC). Our study elucidates the complex regulatory mechanisms governing ERVK-7 in LUAD and proposes these transcripts as potential biomarkers and therapeutic targets, offering new avenues to improve patient outcomes.
Collapse
Affiliation(s)
- Sojung Lee
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China; Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China
| | - Yin Yee Ho
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Suyu Hao
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Yingqi Ouyang
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - U Ling Liew
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Ashish Goyal
- Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephen Li
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Jayne A Barbour
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Mu He
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Yuanhua Huang
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China; Center for Translational Stem Cell Biology, Hong Kong Science and Technology Park, Hong Kong SAR, China; Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Jason W H Wong
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China; Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China.
| |
Collapse
|
6
|
He A, Huang Z, Chen X, Qi K, Zhang S, Li F, Lu H, Wang J, Peng J, Song C. Decoding the role of lipid metabolism in NSCLC: From macrophage subtype identification to prognostic model development. FASEB J 2025; 39:e70467. [PMID: 40277347 DOI: 10.1096/fj.202500124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/07/2025] [Indexed: 04/26/2025]
Abstract
Lipid metabolism plays a pivotal role in shaping the tumor microenvironment, particularly by influencing macrophage function. This study aimed to identify lipid-associated macrophage (LAM) marker genes involved in the onset and progression of non-small cell lung cancer (NSCLC) through integrated single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing (bulk RNA-seq) analyses. Mutation and RNA-seq data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were analyzed to explore the relationship between lipid metabolism pathways and NSCLC progression. scRNA-seq analysis revealed macrophage subtypes closely associated with lipid metabolism, with three key marker genes-S100A10, HLA-DMB, and CTSL-identified as predictive factors for patient prognosis. A prognostic risk scoring model was constructed and validated using survival analysis and ROC curves, demonstrating high accuracy in stratifying NSCLC patients by risk. Further in vivo experiments using subcutaneous tumor xenografts and lung metastasis models showed that S100A10 and CTSL promoted tumor growth and metastasis, while HLA-DMB inhibited these processes. Immune infiltration analysis highlighted the immunological relevance of the identified marker genes, providing insights into their functional roles. This study underscores the critical influence of LAMs in NSCLC progression and highlights a robust prognostic model that offers potential therapeutic targets for improving patient outcomes.
Collapse
Affiliation(s)
- Aoxiao He
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhihao Huang
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xianglai Chen
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Kai Qi
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Shan Zhang
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Fan Li
- Department of Hematology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Department of Gastroenterology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hongcheng Lu
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jiakun Wang
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jinhua Peng
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Chao Song
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
7
|
Zhang Z, Zhang Q, Wang Y. CAF-mediated tumor vascularization: From mechanistic insights to targeted therapies. Cell Signal 2025; 132:111827. [PMID: 40288665 DOI: 10.1016/j.cellsig.2025.111827] [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: 01/21/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Cancer-associated fibroblasts (CAFs) are a major component of the tumor microenvironment (TME) and play a crucial role in tumor progression. The biological properties of tumors, such as drug resistance, vascularization, immunosuppression, and metastasis are closely associated with CAFs. During tumor development, CAFs contribute to tumor progression by remodeling the extracellular matrix (ECM), inhibiting immune cell function, promoting angiogenesis, and facilitating tumor cell growth, invasion, and metastasis. Studies have shown that CAFs can promote endothelial cell proliferation by directly secreting cytokines such as vascular endothelial growth factor (VEGF) and fibroblast Growth Factor (FGF), as well as through exosomes. CAFs also secrete the chemokine stromal cell-derived factor 1 (SDF-1) to recruit endothelial progenitor cells (EPCs) into the peripheral blood and guide their migration to the tumor periphery. Additionally, CAFs can induce tumor cells to transform into "endothelial cells" that participate in vascular wall formation. However, the precise mechanisms remain to be further investigated. Due to their widespread presence in various solid tumors and their tumor-promoting function, CAFs are emerging as therapeutic targets. In this review, we summarize the specific mechanisms through which CAFs promote angiogenesis and outline current therapeutic strategies targeting CAF-induced vascularization, ongoing clinical trials targeting CAFs, and discuss potential future treatment approaches. We hope this will contribute to the advancement of CAF-targeted tumor treatment strategies.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Qing Zhang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Yang Wang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
8
|
Laga T, Van Rompuy AS, Busschaert P, Marquina G, Loverix L, Olbrecht S, Ottenbourgs T, Baert T, Van Gorp T, Vergote I, Lambrechts D, Van Nieuwenhuysen E. Single-cell profiling in ovarian germ cell and sex cord-stromal tumours. Br J Cancer 2025:10.1038/s41416-025-03012-6. [PMID: 40269311 DOI: 10.1038/s41416-025-03012-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/25/2025] [Accepted: 03/31/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND The tumour microenvironment of rare ovarian germ cell tumours (OGCT) and sex-cord stromal tumours (SCST) remains unexplored. To better understand their immune and stromal landscape, we constructed a blueprint using single-cell RNA sequencing (scRNA-seq). METHODS We performed scRNA-seq of 66, 919 cells from twelve fresh tumour samples: seven adult granulosa cell tumour (aGSCT), one juvenile GSCT (jGSCT), one Sertoli-Leydig (SL) tumour, two immature teratoma (IT) and one dysgerminoma (DG). We characterised immune cell subtypes and fibroblasts based on their specific marker genes. Validation included combined positive score (CPS) of 46 OGCTs and 66 SCSTs, and bulk RNA sequencing (n = 32). RESULTS Cell clustering and annotation revealed a immune-activated microenvironment in DG, driven by PD-1- exhausted T cells, reflected in high CPS (≥10) and upregulated immune pathways. IT samples displayed no immunoreactive profile, consistent with a negative CPS. aGSCTs exhibited a fibroblast-enriched, immune-desert phenotype, with low T cell infiltration and increased immunosuppressive LYVE1 and CX3CR1+ macrophages, corresponding to negative CPS. CONCLUSION We constructed a detailed blueprint of the OGCT and SCSTs microenvironment of, elucidating potential modulators that shape their immune landscape. The immune-suppressive environment in aGSCTs likely limits immunotherapy response, as immunosuppressive macrophages inhibit T cell expansion along with EMT activation and fibroblast predominance.
Collapse
Affiliation(s)
- T Laga
- Department of Gynaecological Oncology, University Hospital Leuven, Leuven, Belgium.
- Laboratory of Gynaecological Oncology, KU Leuven, Leuven, Belgium.
- VIB Centre for Cancer Biology, Lab of Translational Genetics, Leuven, Belgium.
| | - A S Van Rompuy
- Department of Pathology, University Hospital Leuven, Leuven, Belgium
- Laboratory of Translational Cell & Tissue Research, Department of Imaging and Pathology, KU Leuven-University of Leuven, Leuven, Belgium
| | - P Busschaert
- Laboratory of Gynaecological Oncology, KU Leuven, Leuven, Belgium
- VIB Centre for Cancer Biology, Lab of Translational Genetics, Leuven, Belgium
| | - G Marquina
- Department of Medical Oncology, San Carlos Hospital, Madrid, Spain
| | - L Loverix
- Department of Gynaecological Oncology, University Hospital Leuven, Leuven, Belgium
| | - S Olbrecht
- Department of Gynaecological Oncology, University Hospital Leuven, Leuven, Belgium
| | - T Ottenbourgs
- Laboratory of Gynaecological Oncology, KU Leuven, Leuven, Belgium
| | - T Baert
- Department of Gynaecological Oncology, University Hospital Leuven, Leuven, Belgium
| | - T Van Gorp
- Department of Gynaecological Oncology, University Hospital Leuven, Leuven, Belgium
- Laboratory of Gynaecological Oncology, KU Leuven, Leuven, Belgium
| | - I Vergote
- Department of Gynaecological Oncology, University Hospital Leuven, Leuven, Belgium
| | - D Lambrechts
- VIB Centre for Cancer Biology, Lab of Translational Genetics, Leuven, Belgium
| | - E Van Nieuwenhuysen
- Department of Gynaecological Oncology, University Hospital Leuven, Leuven, Belgium
- Laboratory of Gynaecological Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
9
|
Weber F, Reese KL, Pantel K, Smit DJ. Cancer-associated fibroblasts as a potential novel liquid biopsy marker in cancer patients. J Exp Clin Cancer Res 2025; 44:127. [PMID: 40259388 PMCID: PMC12010557 DOI: 10.1186/s13046-025-03387-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/07/2025] [Indexed: 04/23/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) are tissue residing cells within the tumor microenvironment (TME). Stromal CAFs have been shown to be associated with poor prognosis and tumor progression in several solid tumor entities. Although the molecular mechanisms are not fully understood yet, a critical role within the TME through direct interaction with the tumor cells as well as other cells has been proposed. While most studies on CAFs focus on stromal CAFs, recent reports highlight the possibility of detecting circulating CAFs (cCAFs) in the blood. In contrast to invasive tissue biopsies for stromal CAF characterization, liquid biopsy allows a minimally invasive isolation of cCAFs. Furthermore, liquid biopsy methods could enable continuous monitoring of cCAFs in cancer patients and therefore may present a novel biomarker for solid tumors. In this work, we present an overview of cCAF studies currently available and summarize the liquid biopsy techniques for cCAF isolation and detection. Moreover, the future research directions in the emerging field are highlighted and the potential applications of cCAFs as novel biomarkers for solid tumor patients discussed.
Collapse
Affiliation(s)
- Franziska Weber
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Kim-Lea Reese
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- European Liquid Biopsy Society (ELBS), Martinistraße 52, 20246, Hamburg, Germany
| | - Daniel J Smit
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
10
|
Ackermann J, Bernard C, Sirven P, Salmon H, Fraldi M, Ben Amar MD. Mechanistic insight for T-cell exclusion by cancer-associated fibroblasts in human lung cancer. eLife 2025; 13:RP101885. [PMID: 40208246 PMCID: PMC11984955 DOI: 10.7554/elife.101885] [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] [Indexed: 04/11/2025] Open
Abstract
The tumor stroma consists mainly of extracellular matrix, fibroblasts, immune cells, and vasculature. Its structure and functions are altered during malignancy: tumor cells transform fibroblasts into cancer-associated fibroblasts, which exhibit immunosuppressive activities on which growth and metastasis depend. These include exclusion of immune cells from the tumor nest, cancer progression, and inhibition of T-cell-based immunotherapy. To understand these complex interactions, we measure the density of different cell types in the stroma using immunohistochemistry techniques on tumor samples from lung cancer patients. We incorporate these data into a minimal dynamical system, explore the variety of outcomes, and finally establish a spatio-temporal model that explains the cell distribution. We reproduce that cancer-associated fibroblasts act as a barrier to tumor expansion, but also reduce the efficiency of the immune response. Our conclusion is that the final outcome depends on the parameter values for each patient and leads to either tumor invasion, persistence, or eradication as a result of the interplay between cancer cell growth, T-cell cytotoxicity, and fibroblast activity. However, despite the existence of a wide range of scenarios, distinct trajectories, and patterns allow quantitative predictions that may help in the selection of new therapies and personalized protocols.
Collapse
Affiliation(s)
- Joseph Ackermann
- Laboratoire Jean Perrin, Sorbonne UniversitéParisFrance
- Laboratoire de Physique de l’Ecole normale supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université Paris CitéParisFrance
| | - Chiara Bernard
- Department of Structures for Engineering and Architecture, University of Naples "Federico II"NaplesItaly
| | | | - Helene Salmon
- Institut Curie, PSL Research University, INSERMParisFrance
| | - Massimiliano Fraldi
- Laboratoire de Physique de l’Ecole normale supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université Paris CitéParisFrance
- Department of Structures for Engineering and Architecture, University of Naples "Federico II"NaplesItaly
| | - Martine D Ben Amar
- Laboratoire de Physique de l’Ecole normale supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université Paris CitéParisFrance
- Institut Universitaire de Cancérologie, Faculté de médecine, Sorbonne UniversitéParisFrance
| |
Collapse
|
11
|
Pentimalli TM, Schallenberg S, León-Periñán D, Legnini I, Theurillat I, Thomas G, Boltengagen A, Fritzsche S, Nimo J, Ruff L, Dernbach G, Jurmeister P, Murphy S, Gregory MT, Liang Y, Cordenonsi M, Piccolo S, Coscia F, Woehler A, Karaiskos N, Klauschen F, Rajewsky N. Combining spatial transcriptomics and ECM imaging in 3D for mapping cellular interactions in the tumor microenvironment. Cell Syst 2025:101261. [PMID: 40220761 DOI: 10.1016/j.cels.2025.101261] [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: 07/12/2024] [Revised: 12/13/2024] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
Tumors are complex ecosystems composed of malignant and non-malignant cells embedded in a dynamic extracellular matrix (ECM). In the tumor microenvironment, molecular phenotypes are controlled by cell-cell and ECM interactions in 3D cellular neighborhoods (CNs). While their inhibition can impede tumor progression, routine molecular tumor profiling fails to capture cellular interactions. Single-cell spatial transcriptomics (ST) maps receptor-ligand interactions but usually remains limited to 2D tissue sections and lacks ECM readouts. Here, we integrate 3D ST with ECM imaging in serial sections from one clinical lung carcinoma to systematically quantify molecular states, cell-cell interactions, and ECM remodeling in CN. Our integrative analysis pinpointed known immune escape and tumor invasion mechanisms, revealing several druggable drivers of tumor progression in the patient under study. This proof-of-principle study highlights the potential of in-depth CN profiling in routine clinical samples to inform microenvironment-directed therapies. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Tancredi Massimo Pentimalli
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin
| | - Simon Schallenberg
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Daniel León-Periñán
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ivano Legnini
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Human Technopole, Milan, Italy
| | - Ilan Theurillat
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Gwendolin Thomas
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Anastasiya Boltengagen
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Sonja Fritzsche
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany; Humboldt-Universität zu Berlin, Institute of Biology, 10099 Berlin, Germany
| | - Jose Nimo
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany; Humboldt-Universität zu Berlin, Institute of Biology, 10099 Berlin, Germany
| | | | - Gabriel Dernbach
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany; Aignostics GmbH, Berlin, Germany; BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
| | | | | | | | - Yan Liang
- NanoString® Technologies, Inc, Seattle, WA, USA
| | | | - Stefano Piccolo
- Department of Molecular Medicine, University of Padua, Padua, Italy; IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Fabian Coscia
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
| | - Andrew Woehler
- Systems Biology Imaging Platform, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA, USA
| | - Nikos Karaiskos
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Frederick Klauschen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany; Institute of Pathology, Ludwig Maximilians Universität, Munich, Germany
| | - Nikolaus Rajewsky
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; German Center for Cardiovascular Research (DZHK), Site Berlin, Berlin, Germany; NeuroCure Cluster of Excellence, Berlin, Germany; German Cancer Consortium (DKTK), Berlin, Germany; National Center for Tumor Diseases (NCT), Site Berlin, Berlin, Germany.
| |
Collapse
|
12
|
Liu K, Cui Y, Han H, Guo E, Shi X, Xiong K, Zhang N, Zhai S, Sang S, Liu M, Chen B, Gu Y. Fibroblast atlas: Shared and specific cell types across tissues. SCIENCE ADVANCES 2025; 11:eado0173. [PMID: 40173240 PMCID: PMC11963979 DOI: 10.1126/sciadv.ado0173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 02/26/2025] [Indexed: 04/04/2025]
Abstract
Understanding the heterogeneity of fibroblasts depends on decoding the complexity of cell subtypes, their origin, distribution, and interactions with other cells. Here, we integrated 249,156 fibroblasts from 73 studies across 10 tissues to present a single-cell atlas of fibroblasts. We provided a high-resolution classification of 18 fibroblast subtypes. In particular, we revealed a previously undescribed cell population, TSPAN8+ chromatin remodeling fibroblasts, characterized by high expression of genes with functions related to histone modification and chromatin remodeling. Moreover, TSPAN8+ chromatin remodeling fibroblasts were detectable in spatial transcriptome data and multiplexed immunofluorescence assays. Compared with other fibroblast subtypes, TSPAN8+ chromatin remodeling fibroblasts exhibited higher scores in cell differentiation and resident fibroblast, mainly interacting with endothelial cells and T cells through ligand VEGFA and receptor F2R, and their presence was associated with poor prognosis. Our analyses comprehensively defined the shared and specific characteristics of fibroblast subtypes across tissues and provided a user-friendly data portal, Fibroblast Atlas.
Collapse
Affiliation(s)
- Kaidong Liu
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yanrui Cui
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Huiming Han
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Erliang Guo
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Xingyang Shi
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Kai Xiong
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Nan Zhang
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Songmei Zhai
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shaocong Sang
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Mingyue Liu
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Bo Chen
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yunyan Gu
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
13
|
Liu S, Liu C, He Y, Li J. Benign non-immune cells in tumor microenvironment. Front Immunol 2025; 16:1561577. [PMID: 40248695 PMCID: PMC12003390 DOI: 10.3389/fimmu.2025.1561577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/24/2025] [Indexed: 04/19/2025] Open
Abstract
The tumor microenvironment (TME) is a highly complex and continuous evolving ecosystem, consisting of a diverse array of cellular and non-cellular components. Among these, benign non-immune cells, including cancer-associated fibroblasts (CAFs), adipocytes, endothelial cells (ECs), pericytes (PCs), Schwann cells (SCs) and others, are crucial factors for tumor development. Benign non-immune cells within the TME interact with both tumor cells and immune cells. These interactions contribute to tumor progression through both direct contact and indirect communication. Numerous studies have highlighted the role that benign non-immune cells exert on tumor progression and potential tumor-promoting mechanisms via multiple signaling pathways and factors. However, these benign non-immune cells may play different roles across cancer types. Therefore, it is important to understand the potential roles of benign non-immune cells within the TME based on tumor heterogeneity. A deep understanding allows us to develop novel cancer therapies by targeting these cells. In this review, we will introduce several types of benign non-immune cells that exert on different cancer types according to tumor heterogeneity and their roles in the TME.
Collapse
Affiliation(s)
- Shaowen Liu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chunhui Liu
- The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
| | - Yuan He
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jun Li
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
- Department of Molecular Pathology, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
14
|
Guo RQ, Li YM, Bie ZX, Peng JZ, Li XG. Microwave ablation of non-small cell lung cancer enhances local T-cell abundance and alters monocyte interactions. BMC Cancer 2025; 25:605. [PMID: 40181307 PMCID: PMC11966799 DOI: 10.1186/s12885-025-14002-5] [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: 04/23/2024] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Minimally invasive thermal therapies show great prospect in non-small cell lung cancer (NSCLC) treatment. However, changes in immune cell populations following microwave ablation (MWA) in NSCLC microenvironment are not fully revealed. OBJECTIVE The present study was conducted to identify changes in immune cell populations and analyse dysregulated genes in immune cells after MWA in NSCLC microenvironment. METHODS The patients received fractionated MWA in two treatments separated by 3 weeks. Tumor biopsy samples were obtained through core-needle biopsy before each fractionated MWA procedure at the same site and used for single-cell RNA sequencing with the 10x Genomics pipeline. RESULTS A total of 9 major cell types were identified after MWA, which include neutrophils, T cells, B cells, monocytes, epithelial cells, chondrocytes, macrophages, tissue stem cells, and endothelial cells. After MWA, the tumor tissue exhibited an increased proportion of T cells. MWA altered gene expression in each cell cluster at the single-cell level. Cell trajectory analysis revealed that the cells at the starting point were most like T helper cells, naïve T cells, and regulatory T cells; they then developed into anergic T cells, T follicular cells, natural killer T cells, T memory cells, and exhausted T cells, and finally ended as γδ T cells and cytotoxic T cells. Moreover, after MWA, more interaction between monocytes and T cells (or B cells) were identified. CONCLUSIONS MWA increases local T-cell abundance and alters monocyte interactions, thereby reshaping the tumor microenvironment. This study lays a foundation for investigating dysregulated genes that may contribute to the MWA-induced immune response in NSCLC. WHAT IS ALREADY KNOWN ON THIS TOPIC Thermal ablation may change the immune profiles of patients by activating various steps in the cancer immunity cycle. However, changes in immune cell populations following MWA of NSCLC have not been fully reported. WHAT THIS STUDY ADDS After MWA, an increase in interactions between monocytes and T cells intratumorally was observed, which promoted antitumor immunity. HOW THIS STUDY MIGHT AFFECT RESEARCH, PRACTICE OR POLICY The current study illuminates the MWA-caused systemic immune response in NSCLC, which may help to identify the dysregulated genes involved in the MWA-caused immune response.
Collapse
Affiliation(s)
- Run-Qi Guo
- Minimally Invasive Tumor Therapies Centre Beijing Hospital, Institute of Geriatric Medicine, National Centre of Gerontology, Chinese Academy of Medical Sciences, No.1 Dongdan Dahua Street, Beijing, 100370, P.R. China.
| | - Yuan-Ming Li
- Minimally Invasive Tumor Therapies Centre Beijing Hospital, Institute of Geriatric Medicine, National Centre of Gerontology, Chinese Academy of Medical Sciences, No.1 Dongdan Dahua Street, Beijing, 100370, P.R. China
| | - Zhi-Xin Bie
- Minimally Invasive Tumor Therapies Centre Beijing Hospital, Institute of Geriatric Medicine, National Centre of Gerontology, Chinese Academy of Medical Sciences, No.1 Dongdan Dahua Street, Beijing, 100370, P.R. China
| | - Jin-Zhao Peng
- Minimally Invasive Tumor Therapies Centre Beijing Hospital, Institute of Geriatric Medicine, National Centre of Gerontology, Chinese Academy of Medical Sciences, No.1 Dongdan Dahua Street, Beijing, 100370, P.R. China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R. China
| | - Xiao-Guang Li
- Minimally Invasive Tumor Therapies Centre Beijing Hospital, Institute of Geriatric Medicine, National Centre of Gerontology, Chinese Academy of Medical Sciences, No.1 Dongdan Dahua Street, Beijing, 100370, P.R. China.
| |
Collapse
|
15
|
Leung RWT, Zhang X, Chen Z, Liang Y, Huang S, Yang Z, Zong X, Jiang X, Lin R, Deng W, Hu Y, Qin J. CORN 2.0 - Condition Orientated Regulatory Networks 2.0. Comput Struct Biotechnol J 2025; 27:1518-1528. [PMID: 40270708 PMCID: PMC12017979 DOI: 10.1016/j.csbj.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/26/2025] [Accepted: 04/02/2025] [Indexed: 04/25/2025] Open
Abstract
Gene regulation is a fundamental process that allows organisms to adapt to their environment and increase complexity through the action of nucleic acid-binding proteins (NBPs), such as transcription factors (TFs), which regulate specific sets of genes under distinct conditions. These regulatory interactions form transcriptional regulatory networks (TRNs), which can be further broken down into transcriptional regulatory sub-networks (TRSNs) centered around individual TFs. TRSNs are more stable and practical for analysis, making them ideal for studying gene regulation under specific conditions. Condition-Oriented Regulatory Networks (CORN, https://qinlab.sysu.edu.cn/corn/home) is a comprehensive library of condition-based TRSNs, including those induced by natural compounds, small molecules, drug treatments, and gene perturbations. CORN 2.0 represents a significant update, associating 7540 specific conditions with 71934 TRSNs across 52 human cell lines, involving 542 transcription factors (TFs). Notably, CORN 2.0 includes 1550 natural compound-triggered TRSNs, providing a valuable resource for studying the pharmacological effects of natural products. This study demonstrates the utility of CORN in three key areas: personalized medicine, induced pluripotency transitions, and natural compound-associated pharmacology. By linking specific conditions to their corresponding TRSNs, CORN enables researchers to explore how gene regulatory networks are altered under various conditions, offering insights into disease mechanisms and potential therapeutic interventions.
Collapse
Affiliation(s)
- Ricky Wai Tak Leung
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
- Division of Science, Engineering and Health Studies, College of Professional and Continuing Education, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xinying Zhang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Zhuobin Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Yuyun Liang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Simei Huang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Zixin Yang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Xueqing Zong
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Xiaosen Jiang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Runming Lin
- BGI-Shenzhen, Shenzhen, Guangdong 518103, China
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Yaohua Hu
- School of Mathematical Sciences, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Jing Qin
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| |
Collapse
|
16
|
Xie Y, Wang X, Wang W, Pu N, Liu L. Epithelial-mesenchymal transition orchestrates tumor microenvironment: current perceptions and challenges. J Transl Med 2025; 23:386. [PMID: 40176117 PMCID: PMC11963649 DOI: 10.1186/s12967-025-06422-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/25/2025] [Indexed: 04/04/2025] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a critical process in cancer progression, facilitating tumor cells to develop invasive traits and augmenting their migratory capabilities. EMT is primed by tumor microenvironment (TME)-derived signals, whereupon cancer cells undergoing EMT in turn remodel the TME, thereby modulating tumor progression and therapeutic response. This review discusses the mechanisms by which EMT coordinates TME dynamics, including secretion of soluble factors, direct cell contact, release of exosomes and enzymes, as well as metabolic reprogramming. Recent evidence also indicates that cells undergoing EMT may differentiate into cancer-associated fibroblasts, thereby establishing themselves as functional constituents of the TME. Elucidating the relationship between EMT and the TME offers novel perspectives for therapeutic strategies to enhance cancer treatment efficacy. Although EMT-directed therapies present significant therapeutic potential, the current lack of effective targeting approaches-attributable to EMT complexity and its microenvironmental context dependency-underscores the necessity for mechanistic investigations and translational clinical validation.
Collapse
Affiliation(s)
- Yuqi Xie
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Xuan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenquan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ning Pu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
17
|
Solomon BJ, Tan DS, de Castro G, Cobo M, Garassino MC, Zhang J, Johnson BE, Lee JM, Garrido P, Butler AA, Pelletier MR, Savchenko A, Ji L, Brase JC, Caparica R, Demanse D, Wu J, Bossen C, Mok T. The Impact of the Tumor Microenvironment on the Effect of IL-1β Blockade in NSCLC: Biomarker Analyses from CANOPY-1 and CANOPY-N Trials. CANCER RESEARCH COMMUNICATIONS 2025; 5:632-646. [PMID: 40116353 PMCID: PMC12006968 DOI: 10.1158/2767-9764.crc-24-0490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 03/23/2025]
Abstract
SIGNIFICANCE Patients with NSCLC with immunosuppressive tumor features and low T-cell infiltration derive less benefit from ICI-based treatment. Biomarker analyses presented here suggest that these patients may benefit from the addition of anti-IL-1β therapy to their treatment.
Collapse
Affiliation(s)
| | - Daniel S.W. Tan
- National Cancer Centre Singapore, Duke-NUS Medical School, Singapore, Singapore
| | | | - Manuel Cobo
- Medical Oncology Intercenter Unit, Regional University Hospital and Virgen de la Victoria University Hospital, IBIMA, Málaga, Spain
| | - Marina Chiara Garassino
- Department of Medicine, Section Hematology Oncology, Thoracic Oncology, University of Chicago, Chicago, Illinois
| | - Jun Zhang
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | | | | | | | | | | | - Alexander Savchenko
- Precision Medicine, Novartis Pharmaceuticals Corporation, Cambridge, Massachusetts
| | - Lexiang Ji
- Novartis BioMedical Research, Cambridge, Massachusetts
| | | | | | | | - Jincheng Wu
- Novartis BioMedical Research, Cambridge, Massachusetts
| | | | - Tony Mok
- The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
18
|
Zeng J, Wu Z, Luo M, Chen Z, Xu X, Xie G, Chen Q, Bai W, Xiao G, Xie J. Identification of a long non-coding RNA signature associated with cuproptosis for prognosis and immunotherapy response prediction in patients with lung adenocarcinoma. Discov Oncol 2025; 16:432. [PMID: 40163162 PMCID: PMC11958909 DOI: 10.1007/s12672-025-02092-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 03/07/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD), the most common histotype of lung cancer, exhibits high heterogeneity due to molecular variations. Cuproptosis is a newly discovered type of cell death that is linked to copper metabolism and long non-coding RNAs (lncRNAs) may play a significant role in this process. We conducted a comprehensive analysis of lncRNA related to cuproptosis and identified a CRLscore to predict the prognosis and immune landscape for LUAD patients. METHODS The LUAD patient cohort obtained from TCGA database was divided into training and validation sets. A range of statistical methods were employed to identify lncRNAs associated with cuproptosis. Multivariate Cox regression was then utilized to develop the CRLscore, which was further used to construct and evaluate a nomogram. Additionally, we investigated the biological functions, gene mutations, and immune landscape. RESULTS A CRLscore, comprising six cuproptosis-related lncRNAs, was developed to stratify patients into high- and low-risk groups. The CRLscore demonstrated its ability to independently predict prognosis in both the training set and the validation set. Utilizing the CRLscore, we constructed a nomogram that exhibited favorable predictive efficiency. Furthermore, the cuproptosis-related lncRNAs exhibited associations with important signaling pathways such as p53 signaling, MYC Targets V1, and G2M Checkpoint. Notably, the CRLscore displayed substantial differences in somatic mutations and immune landscape. Finally, qRT-PCR results showed the significant differential expression of five cuproptosis-related lncRNAs between LUAD and normal cells. CONCLUSION The CRLscore could serve as a potential prognostic indicator and may predict the response to immunotherapy in LUAD patients.
Collapse
Affiliation(s)
- Jie Zeng
- Department of Thoracic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Zhenyu Wu
- Department of Urology, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Meijuan Luo
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Zhibo Chen
- Department of Thoracic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Xie Xu
- Department of Thoracic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Guijing Xie
- Department of Thoracic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Quhai Chen
- Department of Thoracic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Wenjie Bai
- Department of Thoracic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Gang Xiao
- Department of Thoracic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.
| | - Jianjiang Xie
- Department of Thoracic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.
| |
Collapse
|
19
|
Jia H, Chen X, Zhang L, Chen M. Cancer associated fibroblasts in cancer development and therapy. J Hematol Oncol 2025; 18:36. [PMID: 40156055 PMCID: PMC11954198 DOI: 10.1186/s13045-025-01688-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/12/2025] [Indexed: 04/01/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) are key players in cancer development and therapy, and they exhibit multifaceted roles in the tumor microenvironment (TME). From their diverse cellular origins, CAFs undergo phenotypic and functional transformation upon interacting with tumor cells and their presence can adversely influence treatment outcomes and the severity of the cancer. Emerging evidence from single-cell RNA sequencing (scRNA-seq) studies have highlighted the heterogeneity and plasticity of CAFs, with subtypes identifiable through distinct gene expression profiles and functional properties. CAFs influence cancer development through multiple mechanisms, including regulation of extracellular matrix (ECM) remodeling, direct promotion of tumor growth through provision of metabolic support, promoting epithelial-mesenchymal transition (EMT) to enhance cancer invasiveness and growth, as well as stimulating cancer stem cell properties within the tumor. Moreover, CAFs can induce an immunosuppressive TME and contribute to therapeutic resistance. In this review, we summarize the fundamental knowledge and recent advances regarding CAFs, focusing on their sophisticated roles in cancer development and potential as therapeutic targets. We discuss various strategies to target CAFs, including ECM modulation, direct elimination, interruption of CAF-TME crosstalk, and CAF normalization, as approaches to developing more effective treatments. An improved understanding of the complex interplay between CAFs and TME is crucial for developing new and effective targeted therapies for cancer.
Collapse
Affiliation(s)
- Hongyuan Jia
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Xingmin Chen
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Linling Zhang
- Department of Respiratory and Critical Care, Chengdu Third People's Hospital, Chengdu, China
| | - Meihua Chen
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
20
|
Fan J, Chen Y, Gong Y, Sun H, Hou R, Dou X, Zhang Y, Huo C. Single-cell RNA sequencing reveals potential therapeutic targets in the tumor microenvironment of lung squamous cell carcinoma. Sci Rep 2025; 15:10374. [PMID: 40140461 PMCID: PMC11947091 DOI: 10.1038/s41598-025-93916-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Lung squamous cell carcinoma (LUSC), accounting for 30% of lung cancer cases, lacks adequate research due to limited understanding of its molecular abnormalities. Our study analyzed public LUSC datasets to explore the tumor microenvironment (TME) composition using scRNA-seq from two cohorts. Applying non-negative matrix factorization, we identified unique malignant cell phenotypes, or meta-programs (MPs), based on gene expression patterns. Survival analysis revealed the clinical relevance of these MPs. Findings illuminated a TME landscape enriched with immune cells-CD8 + T, exhausted T, CD4 + T, and naive T cells-and suggested roles for myeloid cells, like cDC1 and pDCs, in LUSC progression. Different MPs highlighted the heterogeneity of malignant cells and their clinical implications. Targeting MP-specific genes may enable personalized therapy, especially for early-stage LUSC. This study offers insights into immune cell function in tumor dynamics, identifies MPs, and paves the way for novel LUSC strategies, enhancing early intervention, personalized treatment, and prognosis, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Junda Fan
- Department of Oncology, 242 Hospital Affiliated to Shenyang Medical College, Shenyang, 110034, China
| | - Yu Chen
- Jiamusi Central Hospital, Jiamusi, 154000, China
| | - Yue Gong
- Geneis Beijing Co., Ltd, Beijing, 100102, China
| | - Hongmei Sun
- Department of Medical Oncology, The Cancer Hospital of Jia Mu Si, Jiamusi, 154000, China
| | - Rui Hou
- Geneis Beijing Co., Ltd, Beijing, 100102, China
| | - Xiaoya Dou
- Geneis Beijing Co., Ltd, Beijing, 100102, China
| | - Yanping Zhang
- School of Mathematics and Physics Science and Engineering, Hebei University of Engineering, Handan, 056038, China
| | - Cheng Huo
- Departmen of Pathology, Sinopharm Tongmei General Hospital, Datong, 037003, China.
| |
Collapse
|
21
|
Liu YT, Liu HM, Ren JG, Zhang W, Wang XX, Yu ZL, Fu QY, Xiong XP, Jia J, Liu B, Chen G. Immune-featured stromal niches associate with response to neoadjuvant immunotherapy in oral squamous cell carcinoma. Cell Rep Med 2025; 6:102024. [PMID: 40107247 PMCID: PMC11970382 DOI: 10.1016/j.xcrm.2025.102024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 11/19/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025]
Abstract
Tumor stromal cells (TSCs) play a crucial yet underexplored role in the tumor microenvironment (TME). This study uses single-cell sequencing and spatial transcriptomics on paired tumor specimens from 22 patients with oral squamous cell carcinoma (OSCC) enrolled in a randomized two-arm phase 2 trial, receiving neoadjuvant anti-PD-1 mono-immunotherapy or anti-PD-1 plus docetaxel-cisplatin-5-fluorouracil (TPF) immunochemotherapy. Single-cell analysis reveals increased TSCs within the TME of responders in immunochemotherapy. Notably, significant post-treatment upregulation of SELP+ high endothelial venules (HEVs) and APOD+ myofibroblastic cancer-associated fibroblasts (myCAFs), alongside a decline in STMN1+ capillary endothelial cells (cECs), is specific to the immunochemotherapy cohort. In contrast, MYF5+ muscle satellite cells (MSCs) are upregulated in non-responders to mono-immunotherapy. SELP+ HEVs and APOD+ myCAFs foster favorable immunomodulatory stromal niches for improved outcomes, while STMN1+ cECs and MYF5+ MSCs form immunosuppressive niches in tumor invasion regions, highlighting therapeutic targets. The trial was registered at ClinicalTrials.gov, and the registration number is NCT04649476.
Collapse
Affiliation(s)
- Yu-Tong Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hai-Ming Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jian-Gang Ren
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wei Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xin-Xin Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zi-Li Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Qiu-Yun Fu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xue-Peng Xiong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jun Jia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Gang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China.
| |
Collapse
|
22
|
Xiao Z, Puré E. The fibroinflammatory response in cancer. Nat Rev Cancer 2025:10.1038/s41568-025-00798-8. [PMID: 40097577 DOI: 10.1038/s41568-025-00798-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/06/2025] [Indexed: 03/19/2025]
Abstract
Fibroinflammation refers to the highly integrated fibrogenic and inflammatory responses mediated by the concerted function of fibroblasts and innate immune cells in response to tissue perturbation. This process underlies the desmoplastic remodelling of the tumour microenvironment and thus plays an important role in tumour initiation, growth and metastasis. More specifically, fibroinflammation alters the biochemical and biomechanical signalling in malignant cells to promote their proliferation and survival and further supports an immunosuppressive microenvironment by polarizing the immune status of tumours. Additionally, the presence of fibroinflammation is often associated with therapeutic resistance. As such, there is increasing interest in targeting this process to normalize the tumour microenvironment and thus enhance the treatment of solid tumours. Herein, we review advances made in unravelling the complexity of cancer-associated fibroinflammation that can inform the rational design of therapies targeting this.
Collapse
Affiliation(s)
- Zebin Xiao
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Ellen Puré
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
23
|
Sun Y, Qiao Y, Niu Y, Madhavan BK, Fang C, Hu J, Schuck K, Traub B, Friess H, Herr I, Michalski CW, Kong B. ARP2/3 complex affects myofibroblast differentiation and migration in pancreatic ductal adenocarcinoma. Int J Cancer 2025; 156:1272-1281. [PMID: 39472297 PMCID: PMC11737003 DOI: 10.1002/ijc.35246] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/18/2024] [Accepted: 10/07/2024] [Indexed: 01/18/2025]
Abstract
The ARP2/3 complex, which orchestrates actin cytoskeleton organization and lamellipodia formation, has been implicated in the initiation of pancreatic ductal adenocarcinoma (PDAC). This study aims to clarify its impact on the activity of cancer-associated fibroblasts (CAFs), key players in PDAC progression, and patient outcomes. Early pancreatic carcinogenesis was modeled in p48Cre; LSL-KrasG12D mice with caerulein-induced pancreatitis, complemented by in vitro studies on human immortalized pancreatic stellate cells (PSCs) and primary PDAC-derived CAFs. Data were gained from microarray analysis, RNA sequencing (RNA-seq), and single-cell RNA sequencing (sc-RNA-seq), with subsequent bioinformatics analysis. We uncovered a specific transcriptional signature associated with fibroblast migration in early pancreatic carcinogenesis and linked it to poor survival in patients with PDAC. A pivotal role of the ARP2/3 complex in CAF migration was identified. Inhibition of the ARP2/3 complex markedly decreased CAF motility and induced significant morphological changes in vitro. Furthermore, its inhibition also hindered TGFβ1-mediated myofibroblastic CAF differentiation but had no effect on IL-1-mediated inflammatory CAF differentiation. Our findings position the ARP2/3 complex as central to the migration and differentiation of myofibroblastic CAF. Targeting this complex presents a promising new therapeutic avenue for PDAC treatment.
Collapse
Affiliation(s)
- Yifeng Sun
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
- Beijing Tsinghua Changgung Hospital, School of Clinical MedicineTsinghua UniversityBeijingChina
- Department of General and Visceral SurgeryUlm University HospitalUlmGermany
| | - Yina Qiao
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
| | - Yiqi Niu
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
- Department of General and Visceral SurgeryUlm University HospitalUlmGermany
| | | | - Chao Fang
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
- Department of General and Visceral SurgeryUlm University HospitalUlmGermany
| | - Jingxiong Hu
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
- Department of General and Visceral SurgeryUlm University HospitalUlmGermany
| | - Kathleen Schuck
- Department of General and Visceral SurgeryUlm University HospitalUlmGermany
| | - Benno Traub
- Department of General and Visceral SurgeryUlm University HospitalUlmGermany
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der Isar, School of Medicine and HealthyTechnical University of Munich (TUM)MunichGermany
| | - Ingrid Herr
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
| | - Christoph W. Michalski
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
| | - Bo Kong
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
| |
Collapse
|
24
|
Jing SY, Wang HQ, Lin P, Yuan J, Tang ZX, Li H. Quantifying and interpreting biologically meaningful spatial signatures within tumor microenvironments. NPJ Precis Oncol 2025; 9:68. [PMID: 40069556 PMCID: PMC11897387 DOI: 10.1038/s41698-025-00857-1] [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: 08/22/2024] [Accepted: 02/25/2025] [Indexed: 03/15/2025] Open
Abstract
The tumor microenvironment (TME) plays a crucial role in orchestrating tumor cell behavior and cancer progression. Recent advances in spatial profiling technologies have uncovered novel spatial signatures, including univariate distribution patterns, bivariate spatial relationships, and higher-order structures. These signatures have the potential to revolutionize tumor mechanism and treatment. In this review, we summarize the current state of spatial signature research, highlighting computational methods to uncover spatially relevant biological significance. We discuss the impact of these advances on fundamental cancer biology and translational research, address current challenges and future research directions.
Collapse
Affiliation(s)
- Si-Yu Jing
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - He-Qi Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Ping Lin
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Jiao Yuan
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Zhi-Xuan Tang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Hong Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China.
| |
Collapse
|
25
|
Turlej E, Domaradzka A, Radzka J, Drulis-Fajdasz D, Kulbacka J, Gizak A. Cross-Talk Between Cancer and Its Cellular Environment-A Role in Cancer Progression. Cells 2025; 14:403. [PMID: 40136652 PMCID: PMC11940884 DOI: 10.3390/cells14060403] [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: 01/30/2025] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
The tumor microenvironment is a dynamic and complex three-dimensional network comprising the extracellular matrix and diverse non-cancerous cells, including fibroblasts, adipocytes, endothelial cells and various immune cells (lymphocytes T and B, NK cells, dendritic cells, monocytes/macrophages, myeloid-derived suppressor cells, and innate lymphoid cells). A constantly and rapidly growing number of studies highlight the critical role of these cells in shaping cancer survival, metastatic potential and therapy resistance. This review provides a synthesis of current knowledge on the modulating role of the cellular microenvironment in cancer progression and response to treatment.
Collapse
Affiliation(s)
- Eliza Turlej
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Aleksandra Domaradzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Justyna Radzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Dominika Drulis-Fajdasz
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Julita Kulbacka
- Departament of Molecular and Cellular Biology, Faculty of Pharmacy, Wrocław Medical University, Borowska 211A, 50-556 Wrocław, Poland;
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Agnieszka Gizak
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| |
Collapse
|
26
|
Xiong Y, Lei J, Wen M, Ma Y, Zhao J, Tian Y, Wan Z, Li X, Zhu J, Wang W, Ji X, Sun Y, Yang J, Zhang J, Xin S, Liu Y, Jia L, Han Y, Jiang T. CENPF (+) cancer cells promote malignant progression of early-stage TP53 mutant lung adenocarcinoma. Oncogenesis 2025; 14:5. [PMID: 40044674 PMCID: PMC11882812 DOI: 10.1038/s41389-025-00546-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/18/2024] [Accepted: 01/23/2025] [Indexed: 03/09/2025] Open
Abstract
The prevention and precise treatment of early-stage lung adenocarcinoma (LUAD) characterized by small nodules (stage IA) remains a significant challenge for clinicians, which is due largely to the limited understanding of the oncogenic mechanisms spanning from preneoplasia to invasive adenocarcinoma. Our study highlights the pivotal role of cancer cells exhibiting high expression of centromere protein F (CENPF), driven by TP53 mutations, which become increasingly prevalent during the transition from preneoplasia to invasive LUAD. Biologically, cancer cells (CENPF+) exhibited robust proliferative and stem-like capabilities, thereby propelling the malignant progression of early-stage LUAD. Clinically, autoantibodies against CENPF in the serum and elevated cancer cells (CENPF+) in tissue correlated positively with the progression of early-stage LUAD, especially those in stage IA. Our findings suggest that cancer cells (CENPF+) play a central role in orchestrating the malignant evolution of LUAD and hold potential as a novel biomarker for early-stage detection and management of the disease.
Collapse
Affiliation(s)
- Yanlu Xiong
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
- Innovation Center for Advanced Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
- Department of Thoracic Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jie Lei
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Miaomiao Wen
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yongfu Ma
- Department of Thoracic Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jinbo Zhao
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yahui Tian
- Department of Thoracic Surgery, Air Force Medical Center, PLA, Beijing, China
| | - Zitong Wan
- College of Life Sciences, Northwestern University, Xi'an, China
| | - Xiaoyan Li
- Department of Blood Transfusion, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Jianfei Zhu
- Department of Thoracic Surgery, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Wenchen Wang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaohong Ji
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Ying Sun
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jie Yang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jiao Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Shaowei Xin
- Department of Thoracic Surgery, Air Force Medical Center, PLA, Beijing, China
| | - Yang Liu
- Department of Thoracic Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Lintao Jia
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, China
| | - Yong Han
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
- Department of Thoracic Surgery, Air Force Medical Center, PLA, Beijing, China.
| | - Tao Jiang
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
27
|
Lee D, Oh S, Lawler S, Kim Y. Bistable dynamics of TAN-NK cells in tumor growth and control of radiotherapy-induced neutropenia in lung cancer treatment. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2025; 22:744-809. [PMID: 40296792 DOI: 10.3934/mbe.2025028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Neutrophils play a crucial role in the innate immune response as a first line of defense in many diseases, including cancer. Tumor-associated neutrophils (TANs) can either promote or inhibit tumor growth in various steps of cancer progression via mutual interactions with cancer cells in a complex tumor microenvironment (TME). In this study, we developed and analyzed mathematical models to investigate the role of natural killer cells (NK cells) and the dynamic transition between N1 and N2 TAN phenotypes in killing cancer cells through key signaling networks and how adjuvant therapy with radiation can be used in combination to increase anti-tumor efficacy. We examined the complex immune-tumor dynamics among N1/N2 TANs, NK cells, and tumor cells, communicating through key extracellular mediators (Transforming growth factor (TGF-$ \beta $), Interferon gamma (IFN-$ \gamma $)) and intracellular regulation in the apoptosis signaling network. We developed several tumor prevention strategies to eradicate tumors, including combination (IFN-$ \gamma $, exogenous NK, TGF-$ \beta $ inhibitor) therapy and optimally-controlled ionizing radiation in a complex TME. Using this model, we investigated the fundamental mechanism of radiation-induced changes in the TME and the impact of internal and external immune composition on the tumor cell fate and their response to different treatment schedules.
Collapse
Affiliation(s)
- Donggu Lee
- Department of Mathematics, Konkuk University, Seoul 05029, Republic of Korea
| | - Sunju Oh
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Sean Lawler
- Department of Pathology and Laboratory Medicine, Legorreta Brown Cancer Center, Brown University, Providence, RI 02912, USA
| | - Yangjin Kim
- Department of Mathematics, Konkuk University, Seoul 05029, Republic of Korea
- Department of Pathology and Laboratory Medicine, Legorreta Brown Cancer Center, Brown University, Providence, RI 02912, USA
| |
Collapse
|
28
|
Xia P, Wu W, Liu Q, Huang B, Wu M, Lin Z, Zhu M, Yu M, Qu Y, Li K, Wu L, Zhang R, Wang Q. SCANER: robust and sensitive identification of malignant cells from the scRNA-seq profiled tumor ecosystem. Brief Bioinform 2025; 26:bbaf175. [PMID: 40253692 PMCID: PMC12009548 DOI: 10.1093/bib/bbaf175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/25/2024] [Accepted: 03/26/2025] [Indexed: 04/22/2025] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) has enabled the dissection of complex tumor ecosystems. Recognition of malignant cells as an essential step has a profound impact on downstream interpretation. However, most existing computational strategies are based on prior knowledge of canonical cell-type markers. We have developed a marker-free approach, the Seed-Cluster based Approach for NEoplastic cells Recognition (SCANER), to identify malignant cells based on significant gene expression variations caused by genomic instability. Upon analyzing different cancer types, SCANER achieved superior accuracy and robustness in identifying malignant cells, effectively addressing dropout events and tumor purity variations. Besides, SCANER can significantly detect copy number variations (CNVs) in malignant cells compared to nonmalignant cells, which is further confirmed through the paired whole exome sequencing data. In conclusion, SCANER has the potential to facilitate the biological exploration of the tumor ecosystem by accurately identifying malignant cells and it is applicable across various solid cancer types regardless of prior knowledge. SCANER is available at https://github.com/woolingxiang/SCANER.
Collapse
Affiliation(s)
- Peng Xia
- School of Biological Science & Medical Engineering, Southeast University, 8 Dongnandaxue Road, Jiangning District, Nanjing 211189, Jiangsu, China
- Department of Bioinformatics, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
| | - Wei Wu
- School of Biological Science & Medical Engineering, Southeast University, 8 Dongnandaxue Road, Jiangning District, Nanjing 211189, Jiangsu, China
- Department of Bioinformatics, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
| | - Quanzhong Liu
- Department of Bioinformatics, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
| | - Bin Huang
- School of Biological Science & Medical Engineering, Southeast University, 8 Dongnandaxue Road, Jiangning District, Nanjing 211189, Jiangsu, China
- Department of Bioinformatics, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
| | - Min Wu
- Department of Bioinformatics, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
| | - Zihan Lin
- Department of Bioinformatics, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
| | - Mengyan Zhu
- Department of Bioinformatics, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
| | - Miao Yu
- Department of Bioinformatics, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
| | - Ying Qu
- Department of Bioinformatics, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
| | - Kening Li
- Department of Bioinformatics, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
| | - Lingxiang Wu
- Department of Bioinformatics, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 119 South 4th Ring West Road, Fengtai District, Beijing 100070, China
| | - Ruohan Zhang
- Department of Bioinformatics, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
| | - Qianghu Wang
- School of Biological Science & Medical Engineering, Southeast University, 8 Dongnandaxue Road, Jiangning District, Nanjing 211189, Jiangsu, China
- Department of Bioinformatics, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing 211166, Jiangsu, China
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing 210009, Jiangsu, China
- Department of Pathology, Jiangsu Province Hospital and the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Gulou District, Nanjing 210029, Jiangsu, China
| |
Collapse
|
29
|
Bi L, Wang X, Li J, Li W, Wang Z. Epigenetic modifications in early stage lung cancer: pathogenesis, biomarkers, and early diagnosis. MedComm (Beijing) 2025; 6:e70080. [PMID: 39991629 PMCID: PMC11843169 DOI: 10.1002/mco2.70080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 01/03/2025] [Accepted: 01/09/2025] [Indexed: 02/25/2025] Open
Abstract
The integration of liquid biopsy with epigenetic markers offers significant potential for early lung cancer detection and personalized treatment. Epigenetic alterations, including DNA methylation, histone modifications, and noncoding RNA changes, often precede genetic mutations and are critical in cancer progression. In this study, we explore how liquid biopsy, combined with epigenetic markers, can provide early detection of lung cancer, potentially predicting onset up to 4 years before clinical diagnosis. We discuss the challenges of targeting epigenetic regulators, which could disrupt cellular balance if overexploited, and the need for maintaining key gene expressions in therapeutic applications. This review highlights the promise and challenges of using liquid biopsy and epigenetic markers for early-stage lung cancer diagnosis, with a focus on optimizing treatment strategies for personalized and precision medicine.
Collapse
Affiliation(s)
- Lingfeng Bi
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Respiratory Health and MultimorbidityWest China Hospital, Sichuan UniversityChengduSichuanChina
- Institute of Respiratory Health, Frontiers Science Center for Disease‐Related Molecular NetworkWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Xin Wang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Respiratory Health and MultimorbidityWest China Hospital, Sichuan UniversityChengduSichuanChina
- Institute of Respiratory Health, Frontiers Science Center for Disease‐Related Molecular NetworkWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Jiayi Li
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Respiratory Health and MultimorbidityWest China Hospital, Sichuan UniversityChengduSichuanChina
- Institute of Respiratory Health, Frontiers Science Center for Disease‐Related Molecular NetworkWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Respiratory Health and MultimorbidityWest China Hospital, Sichuan UniversityChengduSichuanChina
- Institute of Respiratory Health, Frontiers Science Center for Disease‐Related Molecular NetworkWest China Hospital, Sichuan UniversityChengduSichuanChina
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan UniversityChengduSichuanChina
- The Research Units of West China, Chinese Academy of Medical SciencesWest China HospitalChengduSichuanChina
| | - Zhoufeng Wang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Respiratory Health and MultimorbidityWest China Hospital, Sichuan UniversityChengduSichuanChina
- Institute of Respiratory Health, Frontiers Science Center for Disease‐Related Molecular NetworkWest China Hospital, Sichuan UniversityChengduSichuanChina
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan UniversityChengduSichuanChina
- The Research Units of West China, Chinese Academy of Medical SciencesWest China HospitalChengduSichuanChina
| |
Collapse
|
30
|
Hua H, Long W, Pan Y, Li S, Zhou J, Wang H, Chen S. scCrab: A Reference-Guided Cancer Cell Identification Method based on Bayesian Neural Networks. Interdiscip Sci 2025; 17:12-26. [PMID: 39348073 DOI: 10.1007/s12539-024-00655-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 10/01/2024]
Abstract
Cancer is a significant global public health concern, where early detection can greatly enhance curative outcomes. Therefore, the identification of cancer cells holds significant importance as the primary method for cancer diagnosis. The advancement of single-cell RNA sequencing (scRNA-seq) technology has made it possible to address the problem of cancer cell identification at the single-cell level more efficiently with computational methods, as opposed to the time-consuming and less reproducible manual identification methods. However, existing computational methods have shown suboptimal identification performance and a lack of capability to incorporate external reference data as prior information. Here, we propose scCrab, a reference-guided automatic cancer cell identification method, which performs ensemble learning based on a Bayesian neural network (BNN) with multi-head self-attention mechanisms and a linear regression model. Through a series of experiments on various datasets, we systematically validated the superior performance of scCrab in both intra- and inter-dataset predictions. Besides, we demonstrated the robustness of scCrab to dropout rate and sample size, and conducted ablation experiments to investigate the contributions of each component in scCrab. Furthermore, as a dedicated model for cancer cell identification, scCrab effectively captures cancer-related biological significance during the identification process.
Collapse
Affiliation(s)
- Heyang Hua
- School of Mathematical Sciences and LPMC, Nankai University, Tianjin, 300071, China
| | - Wenxin Long
- School of Mathematical Sciences and LPMC, Nankai University, Tianjin, 300071, China
| | - Yan Pan
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Siyu Li
- School of Mathematical Sciences and LPMC, Nankai University, Tianjin, 300071, China
| | - Jianyu Zhou
- College of Software, Nankai University, Tianjin, 300071, China.
| | - Haixin Wang
- Cadre Medical Department, The 1St Clinical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Shengquan Chen
- School of Mathematical Sciences and LPMC, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
31
|
Zhang J, Hu D, Fang P, Qi M, Sun G. Deciphering key roles of B cells in prognostication and tailored therapeutic strategies for lung adenocarcinoma: a multi-omics and machine learning approach towards predictive, preventive, and personalized treatment strategies. EPMA J 2025; 16:127-163. [PMID: 39991096 PMCID: PMC11842682 DOI: 10.1007/s13167-024-00390-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/24/2024] [Indexed: 02/25/2025]
Abstract
Background Lung adenocarcinoma (LUAD) remains a significant global health challenge, with an urgent need for innovative predictive, preventive, and personalized medicine (PPPM) strategies to improve patient outcomes. This study leveraged multi-omics and machine learning approaches to uncover the prognostic roles of B cells in LUAD, thereby reinforcing the PPPM approach. Methods We integrated multi-omics data, including bulk RNA, ATAC-seq, single-cell RNA, and spatial transcriptomics sequencing, to characterize the B cell landscape in LUAD within the PPPM framework. Subsequently, we developed an integrative machine learning program that generated the Scissor+ related B cell score (SRBS). This score was validated in the training and validation sets, and its prognostic value was assessed along with clinical features to develop predictive nomograms. This study further assessed the role of SRBS and SRBS genes in response to immunotherapy and identified personalized drug targets for distinct risk subgroups, with gene expression verified experimentally to ensure tailored medical interventions. Results Our analysis identified 79 Scissor+ B cell genes linked to LUAD prognosis, supporting the predictive aspect of PPPM. The SRBS model, which utilizes multiple machine learning algorithms, performed excellently in predicting prognosis and clinical transformation, embodying the preventive and personalized aspects of PPPM. Multifactorial analysis confirmed that SRBS was an independent prognostic factor. We observed varying biological functions and immune cell infiltration in the tumor immune microenvironment (TIME) between the high- and low-SRBS groups, underscoring personalized treatment approaches. Notably, patients with elevated SRBS may exhibit resistance to immunotherapy but show increased sensitivity to chemotherapy and targeted therapies. Additionally, we found that LDHA, as an SRBS gene with significant clinical implications, may regulate the sensitivity of LUAD cells to cisplatin. Conclusion This study presents a B cell-associated gene signature that serves as a prognostic marker to facilitate personalized treatment for patients with LUAD, adhering to the principles of PPPM. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00390-4.
Collapse
Affiliation(s)
- Jinjin Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui Province China
| | - Dingtao Hu
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, China
| | - Pu Fang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui Province China
| | - Min Qi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui Province China
| | - Gengyun Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui Province China
| |
Collapse
|
32
|
Zhi-Xiong C. Single-cell RNA sequencing in ovarian cancer: Current progress and future prospects. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 195:100-129. [PMID: 39778630 DOI: 10.1016/j.pbiomolbio.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 12/25/2024] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
Ovarian cancer is one of the most prevalent gynaecological malignancies. The rapid development of single-cell RNA sequencing (scRNA-seq) has allowed scientists to use this technique to study ovarian cancer development, heterogeneity, and tumour environment. Although multiple original research articles have reported the use of scRNA-seq in understanding ovarian cancer and how therapy resistance occurs, there is a lack of a comprehensive review that could summarize the findings from multiple studies. Therefore, this review aimed to fill this gap by comparing and summarizing the results from different studies that have used scRNA-seq in understanding ovarian cancer development, heterogeneity, tumour microenvironment, and treatment resistance. This review will begin with an overview of scRNA-seq workflow, followed by a discussion of various applications of scRNA-seq in studying ovarian cancer. Next, the limitations and future directions of scRNA-seq in ovarian cancer research will be presented.
Collapse
Affiliation(s)
- Chong Zhi-Xiong
- Faculty of Science and Engineering, University of Nottingham Malaysia, Jalan Broga, Semenyih, 43500 Selangor, Malaysia; Victor Biotech, 81200 Johor Bahru, Johor, Malaysia.
| |
Collapse
|
33
|
Ma M, Jin C, Dong Q. Intratumoral Heterogeneity and Immune Microenvironment in Hepatoblastoma Revealed by Single-Cell RNA Sequencing. J Cell Mol Med 2025; 29:e70482. [PMID: 40099956 PMCID: PMC11915626 DOI: 10.1111/jcmm.70482] [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: 11/09/2024] [Revised: 02/21/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025] Open
Abstract
Hepatoblastoma (HB) is a common paediatric liver malignancy characterised by significant intratumoral heterogeneity and a complex tumour microenvironment (TME). Using single-cell RNA sequencing (scRNA-seq), we analysed 43,592 cells from three tumour regions and adjacent normal tissue of an HB patient. Our study revealed distinct cellular compositions and varying degrees of malignancy across different tumour regions, with the T1 region showing the highest malignancy and overexpression of HMGB2 and TOP2A. Survival analysis demonstrated that high HMGB2 expression is associated with poor prognosis and increased recurrence, suggesting its potential as a prognostic marker. Additionally, we identified a diverse immune microenvironment enriched with regulatory T cells (Tregs) and CD8+ effector memory T cells (Tem), indicating potential immune evasion mechanisms. Notably, CTLA-4 and PD-1 were highly expressed in Tregs and Tem cells, highlighting their potential as immunotherapy targets. Myeloid cells, including Kupffer cells and dendritic cells, also exhibited distinct functional roles in different tumour regions. This study provides the first comprehensive single-cell atlas of HB, revealing critical insights into its intratumoral heterogeneity and immune microenvironment. Our findings not only advance the understanding of HB biology but also offer new directions for precision medicine, including the development of targeted therapies and immunotherapeutic strategies to improve patient outcomes.
Collapse
Affiliation(s)
- Mingdi Ma
- Department of Pediatric SurgeryThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Chen Jin
- Department of Pediatric SurgeryThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Qian Dong
- Department of Pediatric SurgeryThe Affiliated Hospital of Qingdao UniversityQingdaoChina
- Shandong Key Laboratory of Digital Medicine and Computer Assisted SurgeryThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| |
Collapse
|
34
|
Johri S, Bi K, Titchen BM, Fu J, Conway J, Crowdis JP, Vokes NI, Fan Z, Fong L, Park J, Liu D, He MX, Van Allen EM. Dissecting tumor cell programs through group biology estimation in clinical single-cell transcriptomics. Nat Commun 2025; 16:2090. [PMID: 40025015 PMCID: PMC11873288 DOI: 10.1038/s41467-025-57377-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/20/2025] [Indexed: 03/04/2025] Open
Abstract
With the growth of clinical cancer single-cell RNA sequencing studies, robust differential expression methods for case/control analyses (e.g., treatment responders vs. non-responders) using gene signatures are pivotal to nominate hypotheses for further investigation. However, many commonly used methods produce a large number of false positives, do not adequately represent the patient-specific hierarchical structure of clinical single-cell RNA sequencing data, or account for sample-driven confounders. Here, we present a nonparametric statistical method, BEANIE, for differential expression of gene signatures between clinically relevant groups that addresses these issues. We demonstrate its use in simulated and real-world clinical datasets in breast cancer, lung cancer and melanoma. BEANIE outperforms existing methods in specificity while maintaining sensitivity, as demonstrated in simulations. Overall, BEANIE provides a methodological strategy to inform biological insights into unique and shared differentially expressed gene signatures across different tumor states, with utility in single-study, meta-analysis, and cross-validation across cell types.
Collapse
Affiliation(s)
- Shreya Johri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Kevin Bi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Breanna M Titchen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Harvard Graduate Program in Biological and Biomedical Sciences, Boston, MA, USA
| | - Jingxin Fu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jake Conway
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Harvard Graduate Program in Bioinformatics and Integrative Genomics, Boston, MA, USA
| | - Jett P Crowdis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Natalie I Vokes
- Department of Thoracic and Head and Neck Oncology, MD Anderson Cancer Center, Houston, TX, USA
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Zenghua Fan
- Division of Hematology/Oncology, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, CA, USA
| | - Lawrence Fong
- Division of Hematology/Oncology, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, CA, USA
| | - Jihye Park
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - David Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Meng Xiao He
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
35
|
Teng F, Wei H, Che D, Miao K, Dong X. Identifying macrophage-associated subtypes in patients with serous ovarian cancer and exploring potential personalized therapeutic drugs using combined single-cell and bulk RNA sequencing omics. Heliyon 2025; 11:e42429. [PMID: 40028569 PMCID: PMC11870195 DOI: 10.1016/j.heliyon.2025.e42429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 01/14/2025] [Accepted: 01/31/2025] [Indexed: 03/05/2025] Open
Abstract
Purpose We aimed to analyze the sensitivity of patients to chemotherapy drugs and actively explore potential new intervention targets, providing an essential reference for personalized treatment. Methods Candidate markers with significant differential expression in macrophages were identified by analyzing gene expression at the single-cell level. A weighted gene co-expression network (WGCN) was constructed on the GSE26712 dataset to explore the modules most relevant to macrophages. Differentially expressed genes for specific markers were identified. A multi-factor regulatory network was constructed based on single-cell dataset markers screening, differentially expressed genes, and genes commonly present in WGCNA modules. Different macrophage subtypes were identified using this network. Machine learning was used to filter and predict the markers' drug sensitivity, and the potential therapeutic compounds for specific markers were screened. Results We identified 14 and 17 of M1 and M2 macrophage candidate markers, respectively. In the multi-factor regulatory network of M1 macrophages, 6 out of 14 markers recognized 159 transcription factors (TFs) and 48 micro RNAs (miRNAs), whereas 13 of 17 markers recognized 191 TFs and 182 miRNAs in the multi-factor regulatory network of M2 macrophages. Filtering of the identified differentially expressed genes using random forests yielded 15 M1 and M2 macrophage-specific markers. Drug sensitivity prediction analysis and in vitro experiments revealed the close association of these markers with common chemotherapy drug sensitivity. Conclusion We identified specific M1 and M2 macrophage markers and found potential therapeutic compounds (dasatinib and afatinib) in these specific markers. These potential therapeutic compounds provide insight into the underlying mechanisms of serous ovarian cancer (OC) and inspire more effective treatment methods.
Collapse
Affiliation(s)
- Fei Teng
- In-Patient Ultrasound Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Ultrasound Department, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hong Wei
- In-Patient Ultrasound Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dehong Che
- Ultrasound Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kuo Miao
- Ultrasound Department, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoqiu Dong
- Ultrasound Department, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
36
|
Wang L, Wang J, Wang N, Wang X, Song M, Zhou Y, Wang Z, Meng H, Guo E, Miao S. ANLN promotes head and neck squamous cell carcinoma progression by upregulating PD-L1 via the ERK-MAPK pathway. iScience 2025; 28:111633. [PMID: 39967877 PMCID: PMC11834070 DOI: 10.1016/j.isci.2024.111633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 09/27/2024] [Accepted: 12/16/2024] [Indexed: 02/20/2025] Open
Abstract
Anillin (ANLN) is a highly conserved protein involved in cytokinesis and cytoskeletal remodeling. This study investigates the role of ANLN in head and neck squamous cell carcinoma (HNSCC) progression and its impact on the tumor immune microenvironment, with a focus on the combination of ANLN silencing and anti-programmed cell death protein 1 (PD-1) therapy. Through in vitro and in vivo experiments, along with clinical specimen analysis, we discovered that silencing ANLN not only inhibits the malignant progression of HNSCC but also reduces the activation of the extracellular signal-regulated kinase-mitogen-activated protein kinase (ERK-MAPK) signaling pathway and decreases programmed death ligand-1 (PD-L1) expression. Integrating ANLN silencing with anti-PD-1 monoclonal antibody treatment significantly enhances the activation of infiltrating CD8+ T cells, leading to marked tumor growth suppression. Our findings highlight the potential of ANLN as a therapeutic target in HNSCC, providing a foundation for developing innovative and effective combined treatment strategies.
Collapse
Affiliation(s)
- Lei Wang
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, P.R. China
| | - Junrong Wang
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, P.R. China
| | - Nana Wang
- School of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, P.R. China
| | - Xueying Wang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha 410008, Hunan, P.R. China
| | - Ming Song
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, P.R. China
| | - Yang Zhou
- Department of Respiratory Medicine, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, P.R. China
| | - Ziyuan Wang
- Department of Pathology, Harbin Medical University, Harbin 150081, Heilongjiang, P.R. China
| | - Hongxue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Erliang Guo
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, P.R. China
| | - Susheng Miao
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, P.R. China
| |
Collapse
|
37
|
Zhu XR, Zhu JQ, Gu QH, Liu N, Lu JJ, Li XH, Liu YY, Zheng X, Chen MB, Ji Y. A novel identified epithelial ligand-receptor-associated gene signature highlights POPDC3 as a potential therapy target for non-small cell lung cancer. Cell Death Dis 2025; 16:114. [PMID: 39971925 PMCID: PMC11840029 DOI: 10.1038/s41419-025-07410-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 01/13/2025] [Accepted: 01/30/2025] [Indexed: 02/21/2025]
Abstract
The tumor microenvironment (TME) is pivotal in non-small cell lung cancer (NSCLC) progression, influencing drug resistance and immune cell behavior through complex ligand-receptor (LR) interactions. This study developed an epithelial LR-related prognostic risk score (LRrisk) to identify biomarkers and targets in NSCLC. We identified twenty epithelial LRs with significant prognostic implications and delineated three molecular NSCLC subtypes with distinct outcomes, pathological characteristics, biological pathways, and immune profiles. The LRrisk model was constructed using twelve differentially expressed ligand-receptor interaction-related genes (LRGs), with a focus on POPDC3 (popeye domain-containing protein 3), which was overexpressed in NSCLC cells. Functional assays revealed that POPDC3 knockdown reduced cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT), while its overexpression promoted cancerous activities. In vivo, POPDC3 silencing hindered, and its overexpression accelerated the growth of NSCLC xenografts in nude mice. Additionally, high expression levels of POPDC3 in NSCLC tissues were associated with enhanced CD4+ T cell infiltration and increased PD-1 expression within the TME. Moreover, ectopic POPDC3 overexpression in C57BL/6 J mouse Lewis lung carcinoma (LLC) xenografts enhanced CD4+ T cell infiltration and PD-1 expression in the TME. This research establishes a robust epithelial LR-related signature, highlighting POPDC3 as a critical facilitator of NSCLC progression and a potential therapeutic target.
Collapse
Affiliation(s)
- Xiao-Ren Zhu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Medical School of Jiangsu University, Zhenjiang, China
| | - Jia-Qi Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Qian-Hui Gu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Na Liu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Medical School of Jiangsu University, Zhenjiang, China
| | - Jing-Jing Lu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Medical School of Jiangsu University, Zhenjiang, China
| | - Xiao-Hong Li
- Department of Clinical Laboratory, The First People's Hospital of Taicang, Taicang, China
| | - Yuan-Yuan Liu
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Medical School of Jiangsu University, Zhenjiang, China
| | - Xian Zheng
- Medical School of Jiangsu University, Zhenjiang, China.
- Department of Pharmacy, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| | - Min-Bin Chen
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
- Medical School of Jiangsu University, Zhenjiang, China.
| | - Yong Ji
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China.
| |
Collapse
|
38
|
Yuan H, Liang X, Zhang X, Cao Y. Single-cell transcriptomes reveal cell-type-specific and sample-specific gene function in human cancer. Heliyon 2025; 11:e42218. [PMID: 39959484 PMCID: PMC11830296 DOI: 10.1016/j.heliyon.2025.e42218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 02/18/2025] Open
Abstract
Accurate annotation of gene function in individual samples and even in each cell type is essential for understanding the pathogenesis of cancers. Single-cell RNA-sequencing (scRNA-seq) provides unprecedented resolution to decipher gene function. In order to explore how scRNA-seq contributes to the understanding of gene function in cancers, we constructed an assessment framework based on co-expression network and neighbor-voting method using 116,814 cells. Compared with bulk transcriptome, scRNA-seq recalled more experimentally verified gene functions. Surprisingly, scRNA-seq revealed cell-type-specific functions, especially in immune cells, whose expression profile recalled immune-related functions that were not discovered in cancer cells. Furthermore, scRNA-seq discovered sample-specific functions, highlighting that it provided sample-specific information. We also explored factors affecting the performance of gene function prediction. We found that 500 or more cells should be considered in the prediction with scRNA-seq, and that scRNA-seq datasets generated from 10x Genomics platform had a better performance than those from Smart-seq2. Collectively, we compared the prediction performance of bulk data and scRNA-seq data from multiple perspectives, revealing the irreplaceable role of single-cell sequencing in decoding the biological progresses in which the gene involved.
Collapse
Affiliation(s)
- Huating Yuan
- College of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Xin Liang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xinxin Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yu Cao
- Institute of Big Health, Guizhou Medical University, Guiyang, China
| |
Collapse
|
39
|
Regner MJ, Garcia-Recio S, Thennavan A, Wisniewska K, Mendez-Giraldez R, Felsheim B, Spanheimer PM, Parker JS, Perou CM, Franco HL. Defining the regulatory logic of breast cancer using single-cell epigenetic and transcriptome profiling. CELL GENOMICS 2025; 5:100765. [PMID: 39914387 PMCID: PMC11872555 DOI: 10.1016/j.xgen.2025.100765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/04/2024] [Accepted: 01/08/2025] [Indexed: 02/12/2025]
Abstract
Annotation of cis-regulatory elements that drive transcriptional dysregulation in cancer cells is critical to understanding tumor biology. Herein, we present matched chromatin accessibility (single-cell assay for transposase-accessible chromatin by sequencing [scATAC-seq]) and transcriptome (single-cell RNA sequencing [scRNA-seq]) profiles at single-cell resolution from human breast tumors and healthy mammary tissues processed immediately following surgical resection. We identify the most likely cell of origin for subtype-specific breast tumors and implement linear mixed-effects modeling to quantify associations between regulatory elements and gene expression in malignant versus normal cells. These data unveil cancer-specific regulatory elements and putative silencer-to-enhancer switching events in cells that lead to the upregulation of clinically relevant oncogenes. In addition, we generate matched scATAC-seq and scRNA-seq profiles for breast cancer cell lines, revealing a conserved oncogenic gene expression program between in vitro and in vivo cells. This work highlights the importance of non-coding regulatory mechanisms that underlie oncogenic processes and the ability of single-cell multi-omics to define the regulatory logic of cancer cells.
Collapse
Affiliation(s)
- Matthew J Regner
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Susana Garcia-Recio
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Aatish Thennavan
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kamila Wisniewska
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Raul Mendez-Giraldez
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Brooke Felsheim
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Philip M Spanheimer
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hector L Franco
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Division of Clinical and Translational Cancer Research, University of Puerto Rico Comprehensive Cancer Center, San Juan, PR 00935, USA.
| |
Collapse
|
40
|
Peng L, Deng S, Li J, Zhang Y, Zhang L. Single-Cell RNA Sequencing in Unraveling Acquired Resistance to EGFR-TKIs in Non-Small Cell Lung Cancer: New Perspectives. Int J Mol Sci 2025; 26:1483. [PMID: 40003951 PMCID: PMC11855476 DOI: 10.3390/ijms26041483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 02/07/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) have demonstrated remarkable efficacy in treating non-small cell lung cancer (NSCLC), but acquired resistance greatly reduces efficacy and poses a significant challenge to patients. While numerous studies have investigated the mechanisms underlying EGFR-TKI resistance, its complexity and diversity make the existing understanding still incomplete. Traditional approaches frequently struggle to adequately reveal the process of drug resistance development through mean value analysis at the overall cellular level. In recent years, the rapid development of single-cell RNA sequencing technology has introduced a transformative method for analyzing gene expression changes within tumor cells at a single-cell resolution. It not only deepens our understanding of the tumor microenvironment and cellular heterogeneity associated with EGFR-TKI resistance but also identifies potential biomarkers of resistance. In this review, we highlight the critical role of single-cell RNA sequencing in lung cancer research, with a particular focus on its application to exploring the mechanisms of EGFR-TKI-acquired resistance in NSCLC. We emphasize its potential for elucidating the complexity of drug resistance mechanism and its promise in informing more precise and personalized treatment strategies. Ultimately, this approach aims to advance NSCLC treatment toward a new era of precision medicine.
Collapse
Affiliation(s)
| | | | | | | | - Li Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (L.P.); (S.D.); (J.L.); (Y.Z.)
| |
Collapse
|
41
|
Wu S, Hu Y, Sui B. Promotion Mechanisms of Stromal Cell-Mediated Lung Cancer Development Within Tumor Microenvironment. Cancer Manag Res 2025; 17:249-266. [PMID: 39957904 PMCID: PMC11829646 DOI: 10.2147/cmar.s505549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/19/2025] [Indexed: 02/18/2025] Open
Abstract
Lung cancer, with its high incidence and mortality rates, has garnered significant attention in the medical community. The tumor microenvironment (TME), composed of tumor cells, stromal cells, extracellular matrix, surrounding blood vessels, and other signaling molecules, plays a pivotal role in the development of lung cancer. Stromal cells within the TME hold potential as therapeutic targets for lung cancer treatment. However, the precise and comprehensive mechanisms by which stromal cells contribute to lung cancer progression have not been fully elucidated. This review aims to explore the mechanisms through which stromal cells in the tumor microenvironment promote lung cancer development, with a particular focus on how immune cells, tumor-associated fibroblasts, and endothelial cells contribute to immune suppression, inflammation, and angiogenesis. The goal is to provide new insights and potential strategies for the diagnosis and treatment of lung cancer.
Collapse
Affiliation(s)
- Siyu Wu
- Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| | - Yumeng Hu
- Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| | - Bowen Sui
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| |
Collapse
|
42
|
Liu X, Zhang L, Li L, Hou J, Qian M, Zheng N, Liu Y, Song Y. Transcriptomic profiles of single-cell autophagy-related genes (ATGs) in lung diseases. Cell Biol Toxicol 2025; 41:40. [PMID: 39920481 PMCID: PMC11805875 DOI: 10.1007/s10565-025-09990-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/03/2025] [Indexed: 02/09/2025]
Abstract
Autophagy related genes (ATGs) play essential roles in maintaining cellular functions, although biological and pathological alterations of ATG phenotypes remain poorly understood. To address this knowledge gap, we utilized the single-cell sequencing technology to elucidate the transcriptomic atlas of ATGs in lung diseases, with a focus on lung epithelium and lymphocytes. This study conducted a comprehensive investigation into RNA profiles of ATGs in the lung tissues obtained from healthy subjects and patients with different lung diseases through single-cell RNA sequencing (scRNA-seq), including COVID-19 related acute lung damage, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), systemic sclerosis (SSC), and lung adenocarcinoma (LUAD). Our findings revealed significant variations of ATGs expression across lung epithelial cell subsets, e.g., over-expression of MAPK8 in basal cells, ATG10 in club cells, and BCL2 in a goblet cell subset. The changes of autophagy-related pathways varied between lung epithelial and lymphocyte subsets. We identified the disease-associated changes in ATG expression, including significant alterations in BCL2, BCL2L1, PRKCD, and PRKCQ in inflammatory lung diseases (COPD and IPF), and MAP2K7, MAPK3, and RHEB in lung cancer (LUAD), as compared to normal lung tissues. Key ligand-receptor pairs (e.g., CD6-ALCAM, CD99-CD99) and signaling pathways (e.g., APP, CD74) might serve as biomarkers for lung diseases. To evaluate ATGs responses to external challenges, we examined ATGs expression in different epithelial cell lines exposed to cigarette smoking extract (CSE), lysophosphatidylcholine (lysoPC), lipopolysaccharide (LPS), and cholesterol at various doses and durations. Notable changes were observed in CFLAR, EIF2S1, PPP2CA, and PPP2CB in A549 and H1299 against CSE and LPS. The heterogeneity of ATGs expression was dependent on cell subsets, pathologic conditions, and challenges, as well as varied among cellular phenotypes, functions, and behaviors, and the severity of lung diseases. In conclusion, our data might provide new insights into the roles of ATGs in epithelial biology and pulmonary disease pathogenesis, with implications for disease progression and prognosis.
Collapse
Affiliation(s)
- Xuanqi Liu
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China.
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, China.
- Shanghai Institute of Clinical Bioinformatics, Shanghai, China.
| | - Linlin Zhang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, China
| | - Liyang Li
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, China
| | - Jiayun Hou
- Shanghai Institute of Clinical Bioinformatics, Shanghai, China
| | - Mengjia Qian
- Shanghai Institute of Clinical Bioinformatics, Shanghai, China
| | - Nannan Zheng
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, China
| | - Yifei Liu
- Center of Molecular Diagnosis and Therapy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yuanlin Song
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China.
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, China.
- Shanghai Institute of Clinical Bioinformatics, Shanghai, China.
| |
Collapse
|
43
|
Samiea A, Celis G, Yadav R, Rodda LB, Moreau JM. B cells in non-lymphoid tissues. Nat Rev Immunol 2025:10.1038/s41577-025-01137-6. [PMID: 39910240 DOI: 10.1038/s41577-025-01137-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2025] [Indexed: 02/07/2025]
Abstract
B cells have long been understood to be drivers of both humoral and cellular immunity. Recent advances underscore this importance but also indicate that in infection, inflammatory disease and cancer, B cells function directly at sites of inflammation and form tissue-resident memory populations. The spatial organization and cellular niches of tissue B cells have profound effects on their function and on disease outcome, as well as on patient response to therapy. Here we review the role of B cells in peripheral tissues in homeostasis and disease, and discuss the newly identified cellular and molecular signals that are involved in regulating their activity. We integrate emerging data from multi-omic human studies with experimental models to propose a framework for B cell function in tissue inflammation and homeostasis.
Collapse
Affiliation(s)
- Abrar Samiea
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA
| | - George Celis
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Rashi Yadav
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Lauren B Rodda
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA.
| | - Joshua M Moreau
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA.
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA.
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA.
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
44
|
Molla Desta G, Birhanu AG. Advancements in single-cell RNA sequencing and spatial transcriptomics: transforming biomedical research. Acta Biochim Pol 2025; 72:13922. [PMID: 39980637 PMCID: PMC11835515 DOI: 10.3389/abp.2025.13922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/20/2025] [Indexed: 02/22/2025]
Abstract
In recent years, significant advancements in biochemistry, materials science, engineering, and computer-aided testing have driven the development of high-throughput tools for profiling genetic information. Single-cell RNA sequencing (scRNA-seq) technologies have established themselves as key tools for dissecting genetic sequences at the level of single cells. These technologies reveal cellular diversity and allow for the exploration of cell states and transformations with exceptional resolution. Unlike bulk sequencing, which provides population-averaged data, scRNA-seq can detect cell subtypes or gene expression variations that would otherwise be overlooked. However, a key limitation of scRNA-seq is its inability to preserve spatial information about the RNA transcriptome, as the process requires tissue dissociation and cell isolation. Spatial transcriptomics is a pivotal advancement in medical biotechnology, facilitating the identification of molecules such as RNA in their original spatial context within tissue sections at the single-cell level. This capability offers a substantial advantage over traditional single-cell sequencing techniques. Spatial transcriptomics offers valuable insights into a wide range of biomedical fields, including neurology, embryology, cancer research, immunology, and histology. This review highlights single-cell sequencing approaches, recent technological developments, associated challenges, various techniques for expression data analysis, and their applications in disciplines such as cancer research, microbiology, neuroscience, reproductive biology, and immunology. It highlights the critical role of single-cell sequencing tools in characterizing the dynamic nature of individual cells.
Collapse
Affiliation(s)
- Getnet Molla Desta
- College of Veterinary Medicine, Jigjiga University, Jigjiga, Ethiopia
- Institute of Biotechnology, Addis Ababa University, Addis Ababa, Ethiopia
| | | |
Collapse
|
45
|
Huang Q, Yang W, Wang F, Huang R, Wang Q, Li X, Lei T, Yue S, Zou W, An Q, Yue J, Hu Q, Liu C. SELP + TEC:CD8 + T cell crosstalk associates with improved radiotherapy efficacy in cervical cancer. Mol Cancer 2025; 24:41. [PMID: 39901166 PMCID: PMC11789365 DOI: 10.1186/s12943-025-02244-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 01/20/2025] [Indexed: 02/05/2025] Open
Abstract
P-selectin (SELP) expression in tumor cells has been implicated in promoting tumor progression and treatment resistance across various cancers. However, our prior study identified SELP expression in a specific subpopulation of endothelial cells within cervical cancer (CC) and potentially linked to anti-cancer immune response. The precise mechanisms by which SELP influences anti-cancer immunity and its involvement in radiotherapy response in CC, however, remain elusive. To address these gaps, this study analyzed tumor tissue samples from 205 CC patients undergoing radiotherapy, scRNA-seq data from 42,159 cells of eight patients, and bulk RNA-sequencing data from 187 radiotherapy-treated patients. The results revealed that elevated SELP expression in tumor endothelial cells (TECs) was significantly correlated with improved survival outcomes in patients treated with radiotherapy. The SELPhigh group exhibited a prominent enrichment of immune-related pathways, coupled with a diminished enrichment in epithelial cell proliferation and angiogenesis pathways. Notably, this group demonstrated increased infiltration of CD8+ T cells and enhanced expression of chemokine receptors, including ACKR1. Furthermore, our data suggest that SELP+ TECs engage in crosstalk with CD8+ T cells via the ACKR1-CCL5 axis, which is associated with improved radiotherapy efficacy. In conclusion, these findings underscore the pivotal role of SELP+ TEC:CD8+ T cell interactions through the ACKR1-CCL5 pathway in enhancing radiotherapy response in CC. Targeting this crosstalk may offer novel therapeutic strategies to mitigate treatment resistance and improve patient survival.
Collapse
Affiliation(s)
- Qingyu Huang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Wenhui Yang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Fuhao Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Rui Huang
- Department of Oncology, Chongqing University Fuling Hospital, Chongqing, China
| | - Qian Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaohui Li
- Department of Medical Oncology, Peking University First Hospital, Beijing, China
| | - Tianyu Lei
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Shengqin Yue
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wenxue Zou
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
- Department of Oncology, Linyi Center Hospital, Linyi, Shandong, China
| | - Qi An
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jinbo Yue
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China.
| | - Qinyong Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China.
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Chao Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China.
| |
Collapse
|
46
|
Gorría T, Sierra-Boada M, Rojas M, Figueras C, Marin S, Madurga S, Cascante M, Maurel J. Metabolic Singularities in Microsatellite-Stable Colorectal Cancer: Identifying Key Players in Immunosuppression to Improve the Immunotherapy Response. Cancers (Basel) 2025; 17:498. [PMID: 39941865 PMCID: PMC11815897 DOI: 10.3390/cancers17030498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Although immune checkpoint inhibitor (ICI) therapy is currently the standard of care in microsatellite-unstable (MSI) metastatic colorectal cancer (CRC), ICI therapy, alone or in combination with other therapies, is not a treatment approach in microsatellite-stable (MSS) CRC, which is present in 95% of patients. In this review, we focus on metabolic singularities-at the transcriptomic (either bulk or single cell), proteomic, and post-translational modification levels-that induce immunosuppression in cancer and specifically in MSS CRC. First, we evaluate the current efficacy of ICIs in limited and metastatic disease in MSS CRC. Second, we discuss the latest findings on the potential biomarkers for evaluating ICI efficacy in MSS CRC using strict REMARK criteria. Third, we review the current evidence on metabolic patterns in CRC tumors and immune cell metabolism to advance our understanding of metabolic crosstalk and to pave the way for the development of combination strategies to enhance ICI efficacy.
Collapse
Affiliation(s)
- Teresa Gorría
- Medical Oncology Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (T.G.); (M.R.); (C.F.)
- Translational Genomics and Targeted Therapies in Solid Tumors, Agustí Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Medicine Department, University of Barcelona, 08036 Barcelona, Spain
| | - Marina Sierra-Boada
- Medical Oncology Department, Parc Taulí Hospital Universitari, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, 08208 Sabadell, Spain;
| | - Mariam Rojas
- Medical Oncology Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (T.G.); (M.R.); (C.F.)
| | - Carolina Figueras
- Medical Oncology Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (T.G.); (M.R.); (C.F.)
| | - Silvia Marin
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08036 Barcelona, Spain;
- Institute of Biomedicine of University of Barcelona (IBUB), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Sergio Madurga
- Department of Material Science and Physical Chemistry, Research Institute of Theoretical and Computational Chemistry (IQTCUB), University of Barcelona, 08028 Barcelona, Spain;
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08036 Barcelona, Spain;
- Institute of Biomedicine of University of Barcelona (IBUB), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Joan Maurel
- Medical Oncology Department, Hospital Clínic de Barcelona, 08036 Barcelona, Spain; (T.G.); (M.R.); (C.F.)
- Translational Genomics and Targeted Therapies in Solid Tumors, Agustí Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Medicine Department, University of Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| |
Collapse
|
47
|
Cheng PSW, Zaccaria M, Biffi G. Functional heterogeneity of fibroblasts in primary tumors and metastases. Trends Cancer 2025; 11:135-153. [PMID: 39674792 DOI: 10.1016/j.trecan.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/29/2024] [Accepted: 11/18/2024] [Indexed: 12/16/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are abundant components of the tumor microenvironment (TME) of most solid malignancies and have emerged as key regulators of cancer progression and therapy response. Although recent technological advances have uncovered substantial CAF molecular heterogeneity at the single-cell level, defining functional roles for most described CAF populations remains challenging. With the aim of bridging CAF molecular and functional heterogeneity, this review focuses on recently identified functional interactions of CAF subtypes with malignant cells, immune cells, and other stromal cells in primary tumors and metastases. Dissecting the heterogeneous functional crosstalk of specific CAF populations with other components is starting to uncover candidate combinatorial strategies for therapeutically targeting the TME and cancer progression.
Collapse
Affiliation(s)
- Priscilla S W Cheng
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Marta Zaccaria
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Giulia Biffi
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK.
| |
Collapse
|
48
|
Wang P, Wang G, Li H, Yuan Y, Chen H, Wang S, Sun Z, Meng F, Li Y, Yang F, Wang J, Chen K, Qiu M. Nicotinamide N-methyltransferase negatively regulates metastasis-promoting property of cancer-associated fibroblasts in lung adenocarcinoma. Cancer Commun (Lond) 2025; 45:110-137. [PMID: 39623600 PMCID: PMC11833673 DOI: 10.1002/cac2.12633] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 02/19/2025] Open
Abstract
BACKGROUND Recurrence and metastasis remain significant challenges in lung adenocarcinoma (LUAD) after radical resection. The mechanisms behind the recurrence and metastasis of LUAD remain elusive, and deregulated cellular metabolism is suspected to play a significant role. This study explores the metabolic and epigenetic regulation mediated by nicotinamide N-methyl transferase (NNMT) in LUAD. METHODS Untargeted metabolomic analyses were performed to detect metabolism irregularities. Single-cell RNA sequencing (RNA-seq) databases and multiplex immunofluorescence analysis were used to identify the location of NNMT within the tumor microenvironment. The biological functions of NNMT were investigated both in vitro and in vivo, with RNA-seq and chromatin immunoprecipitation-PCR providing insights into underlying mechanisms. Finally, single-cell RNA-seq data and immunohistochemistry of primary tumors were analyzed to validate the main findings. RESULTS Untargeted metabolomic analyses revealed metabolic aberrations in amino acids, organic acids, lipids, and nicotinamide pathways, which are linked to metastasis of non-small cell lung cancer. NNMT is a key enzyme in nicotinamide metabolism, and we found the bulk tissue mRNA level of NNMT gene was inversely associated with LUAD metastasis. NNMT was proved to be predominantly expressed in cancer-associated fibroblasts (CAFs) within the stromal regions of LUAD, and a low stromal NNMT expression was identified as a predictor of poor disease-free survival following radical resection of LUAD. The isolation and primary culture of CAFs from LUAD enabled in vitro and in vivo experiments, which confirmed that NNMT negatively regulated the metastasis-promoting properties of CAFs in LUAD. Mechanistically, the downregulation of NNMT led to an increase in intracellular methyl groups by reducing the activity of the methionine cycle, resulting in heightened methylation at H3K4me3. This alteration triggered the upregulation of genes involved in extracellular matrix remodeling in CAFs, including those encoding collagens, integrins, laminins, and matrix metalloproteinases, thereby facilitating cancer cell invasion and metastasis. Reanalysis of single-cell RNA-seq data and immunohistochemistry assays of primary LUAD tissues substantiated NNMT's negative regulation of these genes in CAFs. CONCLUSIONS This study provides novel insights into the metabolic and epigenetic regulatory functions of NNMT in CAFs, expanding the current understanding of LUAD metastasis regulation and suggesting potential avenues for future research and therapeutic development.
Collapse
Affiliation(s)
- Peiyu Wang
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
- Department of Thoracic SurgeryThe First Affiliated Hospital of Zhengzhou University, ZhengzhouHenanP. R. China
| | - Guangxi Wang
- Department of Pathology, School of Basic Medical SciencesInstitute of Systems Biomedicine, Peking‐Tsinghua Center for Life Sciences, Peking University Health Science CenterBeijingP. R. China
| | - Haoran Li
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Yuyao Yuan
- Department of Pathology, School of Basic Medical SciencesInstitute of Systems Biomedicine, Peking‐Tsinghua Center for Life Sciences, Peking University Health Science CenterBeijingP. R. China
| | - Haiming Chen
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Shaodong Wang
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Zewen Sun
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Fanjie Meng
- Department of Thoracic Surgery, Beijing Institute of Respiratory Medicine and Beijing Chao Yang HospitalCapital Medical UniversityBeijingP. R. China
| | - Yun Li
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Fan Yang
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Jun Wang
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Kezhong Chen
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| | - Mantang Qiu
- Department of Thoracic SurgeryPeking University People's HospitalBeijingP. R. China
- Thoracic Oncology InstitutePeking University People's HospitalBeijingP. R. China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non‐small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002Peking University People's HospitalBeijingP. R. China
| |
Collapse
|
49
|
Du F, Wu X, He Y, Zhao S, Xia M, Zhang B, Tong J, Xia T. Identification of an Amino Acid Metabolism Reprogramming Signature for Predicting Prognosis, Immunotherapy Efficacy, and Drug Candidates in Colon Cancer. Appl Biochem Biotechnol 2025; 197:714-734. [PMID: 39222169 DOI: 10.1007/s12010-024-05049-4] [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] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Colon cancer ranked third among the most frequently diagnosed cancers worldwide. Amino acid metabolic reprogramming was related to the occurrence and development of colon cancer. We looked for the amino acid metabolism genes (AMGs) associated with amino acid metabolism from molecular signatures database as prognostic markers and constructed amino acid metabolism scoring model (AMS). According to AMS, the patients were divided into high AMS and low AMS groups, and the prognostic characteristics, molecular phenotypes, somatic cell mutation characteristics, immune cell infiltration characteristics, and immunotherapy effect of the two groups were systematically analyzed. Finally, the compounds targeting AMGs were also screened. We screen out 6 prognostic AMGs (P < 0.05) and construct an AMS model based on them. K-M curve indicated that OS in low AMS group was significantly higher than that in high group (P < 0.05), which were validated in multiple datasets. And different AMS groups had different molecular phenotypes, somatic cell mutation characteristics and immune cell infiltration characteristics. Low AMS group had a better effect for immunotherapy. In addition, we predicted potential therapeutic compounds that could bind to AMGs target proteins. AMS model can be used as a hierarchical tool to evaluate the prognosis, immune infiltration characteristics and immunotherapy response ability of colon cancer. And the compounds screened based on AMGs may become new anti-tumor drugs.
Collapse
Affiliation(s)
- Fenqi Du
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medial University, Harbin, Heilongjiang Province, People's Republic of China
| | - Xiangxin Wu
- Ganzhou Cancer Hospital, Ganzhou, Jiangxi Province, People's Republic of China
| | - Yibo He
- Department of Acupuncture Massage & Rehabilitation, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, Shandong Province, People's Republic of China
| | - Shihui Zhao
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medial University, Harbin, Heilongjiang Province, People's Republic of China
| | - Mingyu Xia
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medial University, Harbin, Heilongjiang Province, People's Republic of China
| | - Bomiao Zhang
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medial University, Harbin, Heilongjiang Province, People's Republic of China
| | - Jinxue Tong
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medial University, Harbin, Heilongjiang Province, People's Republic of China.
| | - Tianyi Xia
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medial University, Harbin, Heilongjiang Province, People's Republic of China.
| |
Collapse
|
50
|
Onder L, Papadopoulou C, Lütge A, Cheng HW, Lütge M, Perez-Shibayama C, Gil-Cruz C, De Martin A, Kurz L, Cadosch N, Pikor NB, Rodriguez R, Born D, Jochum W, Leskow P, Dutly A, Robinson MD, Ludewig B. Fibroblastic reticular cells generate protective intratumoral T cell environments in lung cancer. Cell 2025; 188:430-446.e20. [PMID: 39566495 DOI: 10.1016/j.cell.2024.10.042] [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: 03/18/2024] [Revised: 08/28/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024]
Abstract
Stringent control of T cell activity in the tumor microenvironment is essential for the generation of protective antitumor immunity. However, the identity, differentiation, and functions of the cells that create critical fibroblastic niches promoting tumor-infiltrating T cells remain elusive. Here, we show that CCL19-expressing fibroblastic reticular cells (FRCs) generate interconnected T cell environments (TEs) in human non-small cell lung cancer, including tertiary lymphoid structures and T cell tracks. Analysis of the FRC-T cell interactome in TEs indicated molecular networks regulating niche-specific differentiation of CCL19-expressing fibroblasts and T cell activation pathways. Single-cell transcriptomics and cell fate-mapping analyses in mice confirmed that FRCs in TEs originate from mural and adventitial progenitors. Ablation of intratumoral FRC precursors decreased antitumor T cell activity, resulting in reduced tumor control during coronavirus vector-based immunotherapy. In summary, specialized FRC niches in the tumor microenvironment govern the quality and extent of antitumor T cell immunity.
Collapse
Affiliation(s)
- Lucas Onder
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland.
| | - Chrysa Papadopoulou
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Almut Lütge
- Department of Molecular Life Sciences and SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich 8057, Switzerland
| | - Hung-Wei Cheng
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Mechthild Lütge
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | | | - Cristina Gil-Cruz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Angelina De Martin
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Lisa Kurz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Nadine Cadosch
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Natalia B Pikor
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland; Institute of Microbiology and Immunology, ETH Zurich, Zurich 8093, Switzerland
| | - Regulo Rodriguez
- Institute of Pathology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Diana Born
- Institute of Pathology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Wolfram Jochum
- Institute of Pathology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Pawel Leskow
- Department of Thoracic Surgery, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Andre Dutly
- Department of Thoracic Surgery, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Mark D Robinson
- Department of Molecular Life Sciences and SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich 8057, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland; University Heart Center, University Hospital Zurich and University of Zurich, Zurich 8091, Switzerland; Center for Translational and Experimental Cardiology, University Hospital Zurich and University of Zurich, Zurich 8091, Switzerland.
| |
Collapse
|