1
|
Liu Q, Song M, Wang Y, Zhang P, Zhang H. CCL20-CCR6 signaling in tumor microenvironment: Functional roles, mechanisms, and immunotherapy targeting. Biochim Biophys Acta Rev Cancer 2025; 1880:189341. [PMID: 40348067 DOI: 10.1016/j.bbcan.2025.189341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 05/01/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Chemokine CC motif ligand 20 (CCL20) is a molecule with immunomodulatory properties that is involved in the regulation of diseases such as chronic inflammation, autoimmune diseases, and cancer. It operates by binding to its specific receptor, CC chemokine receptor type 6 (CCR6), and activating a complex intracellular signaling network. Building on its established role in inflammatory diseases, recent research has expanded our understanding of CCL20 to encompass its critical contributions to the tumor microenvironment (TME), highlighting its significance in cancer progression. Numerous studies have emphasized its prominent role in regulating immune responses. Consequently, Monoclonal antibodies against CCL20 and inhibitors of CCR6 have been successfully developed to block downstream signaling, making the CCL20-CCR6 axis a promising and critical target in the TME. This offers potential immunotherapeutic strategies for cancers. In this review, we summarize the biological consequences of CCL20-CCR6 mediated signaling, its role and mechanisms in the TME, and its potential applications. We suggest that the CCL20-CCR6 axis may be a novel biomarker for tumor diagnosis and prognosis, as well as a therapeutic target in various cancers.
Collapse
Affiliation(s)
- Qi Liu
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Mingyuan Song
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Yan Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Ping Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Hao Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
2
|
Zi X, Ma J, Li X, Wang H, Bao Y, Deng T, Yuan X. BUB1-deficiency suppresses kidney renal clear cell carcinoma progression via the PI3K/Akt pathway: A bioinformatics-oriented validating study. Mol Cell Probes 2025; 81:102024. [PMID: 40081509 DOI: 10.1016/j.mcp.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: 01/05/2025] [Revised: 02/28/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Although great advances have been reached in the diagnosis, treatment and prognosis of kidney renal clear cell carcinoma (KIRC), the advancement of therapeutic strategies for KIRC in clinical practices have been seriously limited due to its unknown molecular mechanisms. To resolve this issue, through analyzing the datasets from the online UCSC database, a novel BUB1 gene was found to be elevated in the cancerous tissues compared to their normal tissues of KIRC, and and KIRC patients with high-expressed BUB1 tended to have a worse prognosis. The subsequent experiments validated that BUB1 protein was located in both nucleus and cytoplasm of KIRC cells, and the expression levels of BUB1 gene were significantly elevated in KIRC tissues and cells, in contrast to their normal counterparts. Loss-of-function experiments verified that knockdown of BUB1 suppressed cell proliferation, mobility, epithelial-mesenchymal transition (EMT) and tumor growth, whereas induced apoptotic cell death in the KIRC cells in vitro and in vivo. In addition, bioinformatics analysis predicted that the differentially-expressed genes (DEGs) in the BUB1-deficient cohorts were enriched in the cell division-related PI3K/Akt signal pathway, and we evidenced that silencing of BUB1 was capable of inactivating the downstream PI3K/Akt signal pathway. Of note, deficiency of BUB1-induced suppressing effects on the malignant phenotypes in KIRC cells were all reversed by co-treating cells with PI3K/Akt pathway activator 740Y-P. Furthermore, it was found that the expression status of BUB1 gene were related with epigenetic modifications, immune infiltration and immunotherapy responses in KIRC. Collectively, silencing of BUB1 inhibited the progression of KIRC through inactivating the downstream PI3K/Akt signal pathway, and BUB1 gene could be potentially used as biomarkers for the diagnosis and treatment of KIRC in clinic.
Collapse
Affiliation(s)
- Xiaolin Zi
- Department of Medical Oncology, Fourth Hospital of Harbin Medical University, Yiyuan Street No. 37, Nangang District, Harbin, 150001, China.
| | - Jinpeng Ma
- Institute of Urology, Lanzhou University Second Hospital, Key Laboratory of Gansu Province for Urological Diseases, Gansu Urological Clinical Center, Lanzhou, China.
| | - Xiaoxia Li
- Department of Medical Laboratory, Fourth Affiliated Hospital of Harbin Medical University, Yiyuan Street No. 37, Nangang District, Harbin, 150001, China.
| | - Honglei Wang
- Urology Surgery Department, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China.
| | - Yuchen Bao
- Urology Surgery Department, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China.
| | - Tao Deng
- Department of Gastroenterology, Fourth Hospital of Harbin Medical University, Yiyuan Street No. 37, Nangang District, Harbin, 150001, China.
| | - Xueli Yuan
- Department of Medical Oncology, Fourth Hospital of Harbin Medical University, Yiyuan Street No. 37, Nangang District, Harbin, 150001, China.
| |
Collapse
|
3
|
Huang Y, Tian H, Yue Z, liang L, Huang C, Zhu H, Yang J. Copper-coordination driven nano-frameworks for efficient colorectal cancer chemo-immunotherapy by suppression of cancer cell stemness. Mater Today Bio 2025; 32:101707. [PMID: 40236810 PMCID: PMC11999347 DOI: 10.1016/j.mtbio.2025.101707] [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: 12/21/2024] [Revised: 03/19/2025] [Accepted: 03/23/2025] [Indexed: 04/17/2025] Open
Abstract
Cancer stemness, characterized by the self-renewal and differentiation capabilities of cancer stem cells (CSCs), is a critical determinant of colorectal cancer (CRC) chemo-immunotherapy. Herein, we repurposed copper-coordination driven metal-organic nano-frameworks (Cu-MOFs) to address the chemo-immunotherapy resistance posed by cancer stemness. These repurposed Cu-MOFs were loaded with the chemotherapeutic agent cisplatin (CDDP), resulting in the formation of Cu-MOF@CDDP. The Cu-MOF@CDDP are efficiently internalized by CRC cells via nanoparticle mediated endocytosis, where they release free copper ions (Cu2+) and CDDP in a high-glutathione (GSH) environment. After that, CDDP forms DNA-CDDP adducts that inhibit DNA synthesis and repair, while Cu2+ induces cuproptosis by disrupting mitochondrial metabolism. Moreover, DNA fragments originating from both the nucleus and mitochondria activate the cGAS-STING pathway, thereby initiating anti-tumor immune responses. Meanwhile, Cu2+ depletes intracellular GSH and induces cuproptosis, leading to the downregulation of stemness-related proteins such as ZEB1 and c-MYC, which enhances the efficacy of chemo-immunotherapy by targeting the critical pathways involved in maintaining stemness. Consequently, our results underscore the substantial promise of Cu-MOFs in overcoming stemness-driven therapeutic resistance, offering a transformative approach to sensitize chemo-immunotherapy.
Collapse
Affiliation(s)
- Yichun Huang
- Department of Surgical Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Hailong Tian
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610041, China
| | - Zhimin Yue
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei liang
- Department of Surgical Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Canhua Huang
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610041, China
| | - Huili Zhu
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610041, China
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, 610041, China
| | - Jun Yang
- Department of Surgical Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| |
Collapse
|
4
|
Hou M, Ji L, Li D, Xiao Q, Hu X. Serum starvation-induced cholesterol reduction increases melanoma cell susceptibility to cytotoxic T lymphocyte killing. Sci Rep 2025; 15:18364. [PMID: 40419522 DOI: 10.1038/s41598-025-00586-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/29/2025] [Indexed: 05/28/2025] Open
Abstract
While calorie restriction has been suggested to reduce tumor incidence and slow tumor progression through promoting anti-tumor immune response, evidence disclosing how absence of specific nutrient component and alterations of its related metabolic pathways contribute to the process of anti-tumor immune response is still vague. Using human HLA-A*02:01 restricted New York esophageal squamous cell carcinoma-1 (NY-ESO-1) T cell receptor-engineered T (TCR-T) cells as a tool to investigate the impact from nutrient factors on tumor cells for targeted cytotoxicity, we serum-starved both human and murine melanoma cells and monitored their responses to TCR-T cell killing. Serum starvation sensitizes melanoma cells predominantly by reducing cholesterol availability without causing unwanted off-target effect, as supplementation of cholesterol compromises the sensitization toward TCR-T cell killing. In response to serum starvation, tumor cells upregulate cholesterol biogenesis pathways as a compensatory mechanism. Our study reveals the critical role of cholesterol reduction in mediating serum starvation-induced enhancement of anti-tumor immune response, highlighting the importance of plasma membrane composition in determining tumor cell response to TCR-T cell killing.
Collapse
MESH Headings
- Cholesterol/metabolism
- Humans
- Animals
- Melanoma/immunology
- Melanoma/metabolism
- Melanoma/pathology
- Cell Line, Tumor
- Mice
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Cytotoxicity, Immunologic
Collapse
Affiliation(s)
- Miaomiao Hou
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Longtao Ji
- Institute of Modern Biology, Nanjing University, Nanjing, China
| | - Dimin Li
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Qian Xiao
- Institute of Modern Biology, Nanjing University, Nanjing, China
| | - Xiao Hu
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China.
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
| |
Collapse
|
5
|
Wang T, Song W, Tang Y, Yi J, Pan H. Breaking the immune desert: Strategies for overcoming the immunological challenges of pancreatic cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189353. [PMID: 40412630 DOI: 10.1016/j.bbcan.2025.189353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 05/18/2025] [Accepted: 05/18/2025] [Indexed: 05/27/2025]
Abstract
Pancreatic cancer is characterised by its highly aggressive nature and extremely poor prognosis, with a uniquely complex tumour immune microenvironment that manifests as a prototypical "immune desert." This immune-desert phenotype primarily arises from the inherently low immunogenicity of the tumour, the formation of a dense fibrotic stroma, severe deficiency in immune cell infiltration, and profound immunosuppressive effects of the metabolic landscape. Specifically, dysregulated tryptophan metabolism, such as indoleamine 2,3-dioxygenase (IDO)-mediated catabolism, and excessive lactate accumulation contribute to impaired T-cell functionality. Collectively, these factors severely limit the efficacy of current immunotherapy strategies, particularly those based on immune checkpoint inhibitors, which have demonstrated significantly lower clinical response rates in pancreatic cancer than in other malignancies. In response to these therapeutic challenges, this review explores integrated treatment strategies that combine metabolic reprogramming, tumour microenvironment remodelling, and next-generation immune checkpoint blockades, such as LAG-3, TIM-3, and VISTA. These emerging approaches hold substantial promise for clinical application. For example, targeting key metabolic pathways, including glycolysis (Warburg effect) and glutamine metabolism, may help restore T-cell activity by alleviating metabolic stress within the tumour milieu. Additionally, localised administration of immune stimulators such as interleukin-12 (IL-12) and CD40 agonists may enhance immune cell infiltration and promote tumour-specific immune activation. Future research should prioritise large-scale, multicentre clinical trials to validate the therapeutic efficacy of these innovative strategies, aiming to achieve meaningful breakthroughs in pancreatic cancer immunotherapy and significantly improve long-term survival and clinical outcomes in affected patients.
Collapse
Affiliation(s)
- Tianming Wang
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China; Zhengning County Community Health Service Center, Qingyang 745300, Gansu Province, China
| | - Wenjing Song
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Yuan Tang
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Jianfeng Yi
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China.
| | - Haibang Pan
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China.
| |
Collapse
|
6
|
Wu Y, Liu J, Yin T, Li X, Liu X, Peng X, Zhan X. SELP can affect the immune microenvironment of gastric cancer and is associated with poor prognosis. Discov Oncol 2025; 16:846. [PMID: 40397261 PMCID: PMC12095770 DOI: 10.1007/s12672-025-02629-6] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 05/08/2025] [Indexed: 05/22/2025] Open
Abstract
The tumor microenvironment (TME) plays a crucial role in the occurrence and progression of gastric cancer. Yet, we still don't understand how immune and stromal components of TMEs are modulated. In this study, we applied the ESTIMATE algorithm to calculate the number of immune and stromal components in 410 STAD cases in the Cancer Genome Atlas (TCGA) database. COX regression analysis and protein-protein interaction (PPI) network construction were used to analyze differentially expressed genes (DEGs). Then, P-selectin (SELP) was identified as a predictor by cross-analysis of univariate COX and PPI. After verifying the clinical significance of SELP for study, we performed an immune infiltration analysis and identified 54 immunomodulators associated with SELP through public data. Immunomodulation associated with gastric cancer prognosis was then confirmed by LASSO regression, and the previous results were further validated with single-cell data. Finally, we verified that SELP can promote EMT on gastric cancer cells. In conclusion, we validated that SELP may affect the biological phenotype of gastric cancer with the immune microenvironment alteration of gastric cancer.
Collapse
Affiliation(s)
- Yue Wu
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Jingyu Liu
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Tong Yin
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Xiaoxiao Li
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Xian Liu
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Xiaobo Peng
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China.
| | - Xianbao Zhan
- Department of Oncology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
7
|
Wang P, Guo W, Liu S, Li S, Li J, Ding B, Yin F, Yang Y, Li X, Cao P, Ma C, Zhang W, Song Y, Geng Y, Liu L, Hu J, Hao J, Feng Y. Novel Pt@PCN-Cu-induced cuproptosis amplifies αPD-L1 immunotherapy in pancreatic ductal adenocarcinoma through mitochondrial HK2-mediated PD-L1 upregulation. J Exp Clin Cancer Res 2025; 44:149. [PMID: 40382627 DOI: 10.1186/s13046-025-03409-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 05/06/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Copper accumulation triggers mitochondrial-driven cell death, known as cuproptosis, offering a promising mechanism for targeted cancer therapy. Recent studies have highlighted the critical role of intratumoral copper levels in regulating the expression of programmed cell death ligand-1 (PD-L1), suggesting that copper-induced cuproptosis not only enhances cancer cell death but may also amplify the effects of anti-PD-L1 antibodies (αPD-L1). However, in tumors where monotherapy with αPD-L1 shows limited efficacy, particularly in pancreatic ductal adenocarcinoma (PDAC), the role of copper-induced cuproptosis in enhancing αPD-L1 treatment efficacy and its underlying mechanisms remain unclear. Meanwhile, inadequate tumor drug accumulation and glycolysis significantly restrict the efficacy of cuproptosis. To address these challenges, we have synthesized a novel nanozyme, Pt@PCN-Cu, designed to stabilize intracellular copper accumulation and effectively induce cuproptosis. Additionally, we aim to determine whether this strong induction of cuproptosis can synergize with αPD-L1 to enhance cancer therapy, ultimately paving the way for novel strategies to improve PDAC treatment. METHODS Pt@PCN-Cu was synthesized via a one-pot method, and its therapeutic potential was assessed in combination with αPD-L1 for the treatment of PDAC. Initially, the material's properties were characterized, and its efficient cellular uptake was confirmed. Anti-tumor efficacy was evaluated by inducing cuproptosis in PDAC cell lines and xenograft models. RNA sequencing (RNA-seq) was utilized to identify key regulators involved in the modulation of PD-L1 expression by cuproptosis. Lastly, the therapeutic efficacy of Pt@PCN-Cu combined with αPD-L1 was evaluated in vivo, focusing on tumor growth inhibition and immune modulation within the tumor microenvironment (TME). RESULTS Pt@PCN-Cu demonstrates excellent physicochemical properties and remarkable cascade catalytic activity, providing a solid foundation for further in vitro and in vivo studies. In vitro, Pt@PCN-Cu efficiently transports copper and induces cuproptosis primarily through mitochondrial dysfunction. Mechanistic studies show that Pt@PCN-Cu triggers the dissociation of hexokinase 2 (HK2) from mitochondria, leading to a reduction in HK2 activity. This decline in HK2 activity impairs glycolysis, a metabolic pathway essential for tumor energy metabolism, which in turn results in elevated PD-L1 levels. In vivo, Pt@PCN-Cu demonstrates excellent safety and accumulates at the tumor site in a subcutaneous PDAC mouse model, inducing cuproptosis. Moreover, the combination of Pt@PCN-Cu with αPD-L1 further enhanced its therapeutic efficacy and effectively reprogrammed the immunosuppressive TME. CONCLUSION This study presents strong evidence confirming the safety and therapeutic potential of Pt@PCN-Cu in PDAC treatment. Importantly, Pt@PCN-Cu not only induces cuproptosis but also significantly enhances antitumor efficacy in combination with αPD-L1 by regulating PD-L1 expression through HK2 modulation. These findings underscore a more effective and innovative approach for treating PDAC.
Collapse
Affiliation(s)
- Pengyu Wang
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Weihua Guo
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Shuyue Liu
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Shouyi Li
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Jiaqi Li
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Bowen Ding
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Fengyi Yin
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yang Yang
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Xingjiang Li
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Pei Cao
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Chaozhe Ma
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Wanying Zhang
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Yidan Song
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Yating Geng
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Lantao Liu
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, 157011, China.
| | - Jing Hu
- School of Basic Medicine, Tianjin Medical University, Tianjin, 300070, China.
| | - Jihui Hao
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Yukuan Feng
- Pancreas Center, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
| |
Collapse
|
8
|
Liu J, Le Y, Wang J, Zheng J, Yuan A, Guo J, Chen H, Wang C, Wang CY, Lu JJ, Lu D. Fruit of Physalis angulata L. and anti-inflammatory potential: An in silico, in vitro, and in vivo study focusing on PFKFB3. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156813. [PMID: 40382942 DOI: 10.1016/j.phymed.2025.156813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 04/14/2025] [Accepted: 04/25/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND Sepsis-associated lung injury (SALI) is a disease characterised by inflammation. The fruit of Physalis angulata L. has been employed as a premium, novel, nutritious, healthcare "herbal fruit", which can be processed into juice, preserved fruit, canned food, and so forth. PURPOSE The objective of this study is to examine the impact of the fruit of Physalis angulata L. on the inhibition of inflammation in sepsis-associated lung injury and to elucidate the underlying mechanisms. METHODS The active components of fruit of Physalis angulata L. were analysed using HPLC-MS/MS. A comprehensive investigation was conducted to elucidate the effects and regulatory mechanisms of fruit of Physalis angulata L. on sepsis-associated lung injury and M1 polarisation of macrophage in mice subjected to acute LPS treatment. The renoprotective effect of fruit of Physalis angulata L. on LPS-treated mice was evaluated by measuring tissue damage and inflammation. In addition, we employed RNA-seq methodologies to analyse the principal regulatory targets of fruit of Physalis angulata L.. Furthermore, the expression of key proteins and markers of inflammation and glucose metabolism, as well as the levels of key indicators related to M1 polarisation of macrophage, were examined by immunoblotting, immunohistochemistry, immunoprecipitation, quantitative real-time PCR (qPCR) and specific probes. RESULTS In murine models, the ethanol extract of the fruit of Physalis angulata L. (EPAF) has been demonstrated to effectively inhibit structural damage and inflammation in the lung tissue of a murine model of LPS-induced acute lung injury. In terms of its mechanism of action, EPAF may inhibit M1 polarisation of macrophage and excessive inflammation by modulating the acetylation and phosphorylation of PFKFB3. This in turn affects glycolysis and the subsequent activation of NF-κB, HIF-1α and STAT3 in macrophages. Furthermore, the capacity of EPAF to markedly diminish LPS-induced lung injury in a murine model indicates that it may serve as a promising adjunctive therapy for acute lung Injury. CONCLUSION These suggest that fruit of Physalis angulata L. alleviates sepsis-associated lung injury through suppressing M1 polarization of macrophage via regulation of PFKFB3.
Collapse
Affiliation(s)
- Jing Liu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China; Zhejiang-Hong Kong Joint Laboratory of Liver and Spleen Simultaneous Treatment in Traditional Chinese Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Key Laboratory for Genetic Improvement and Quality Control of Medicinal Plants, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Yifei Le
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China; Zhejiang-Hong Kong Joint Laboratory of Liver and Spleen Simultaneous Treatment in Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Jingwei Wang
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Jiayu Zheng
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China; Zhejiang-Hong Kong Joint Laboratory of Liver and Spleen Simultaneous Treatment in Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Aini Yuan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China; Zhejiang-Hong Kong Joint Laboratory of Liver and Spleen Simultaneous Treatment in Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Jianan Guo
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China; Zhejiang-Hong Kong Joint Laboratory of Liver and Spleen Simultaneous Treatment in Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Hang Chen
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China; Zhejiang-Hong Kong Joint Laboratory of Liver and Spleen Simultaneous Treatment in Traditional Chinese Medicine, Hangzhou, Zhejiang, China; Department of Medical Research Center, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang, China
| | - Cui Wang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China; Zhejiang-Hong Kong Joint Laboratory of Liver and Spleen Simultaneous Treatment in Traditional Chinese Medicine, Hangzhou, Zhejiang, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Cai-Yi Wang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 311399, Zhejiang, China..
| | - Jiang-Jie Lu
- Zhejiang Provincial Key Laboratory for Genetic Improvement and Quality Control of Medicinal Plants, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China.
| | - Dezhao Lu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China; Zhejiang-Hong Kong Joint Laboratory of Liver and Spleen Simultaneous Treatment in Traditional Chinese Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
9
|
Izadifard M, Ahmadvand M, Chahardouli B, Vaezi M, Janbabai G, Seghatoleslami G, Bahrami M, Yaghmaie M, Barkhordar M. Plasma-circulating miR-638, miR-6511b-5p, miR-3613-5p, miR-455-3p, miR-5787, and miR-548a-3p as noninvasive biomarkers of immune reconstitution post-allogeneic hematopoietic stem cell transplantation in acute myeloid leukemia patients. Transpl Immunol 2025; 91:102240. [PMID: 40347984 DOI: 10.1016/j.trim.2025.102240] [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: 07/01/2024] [Revised: 05/07/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
INTRODUCTION Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a viable treatment option for acute myeloid leukemia (AML), though it carries risks including delayed immune reconstitution and hematopoietic reconstitution failure. This study aimed to explore the potential of circulating miRNA levels as biomarkers for post-transplant immune reconstitution. METHODS This observational study was carried out on de novo non-M3 AML patients receiving allo-HSCT from HLA-matched sibling donors at Shariati Hospital, Iran in 2020-2023. Accordingly, the immunophenotype of NK cells, T cells, and B cells was determined by ten-color multiparameter flow cytometry on blood samples collected pre-transplantation and at day +30 post-transplantation. Concurrently, plasma levels of miR-638, miR-6511b-5p, miR-3613-5p, miR-455-3p, miR-5787, and miR-548a-3p were quantified using quantitative reverse transcription-polymerase chain reaction (RT qPCR). RESULTS The expression of miR-638, miR-3613-5p, miR-455-3p, and miR-548a-3p positively correlated with CD4+ T cells, CD4+/CD8+ T cell ratio, CD3-/16+/56- cells, and platelet count. Elevated miR-455-3p and miR-3613-5p expressions were associated with higher CD3-/16+/56- cells (P = 0.0475 and P = 0.0325, respectively). Similarly, miR-638 upregulation correlated with increases in CD4+ T cells (P = 0.0112) and the CD4+/CD8+ T cell ratio (P = 0.006), while miR-548a-3p upregulation was associated with increases in the CD4+/CD8+ T cell ratio (P = 0.0353) and platelet count (P = 0.0191). Conversely, miR-3613-5p and miR-6511b-5p had considerable negative correlations with CD8+ T cells (P = 0.03 and P = 0.0246, respectively), whereas miR-5787 negatively correlated with CD3+/16-/56+ cells (P = 0.025). CONCLUSION Our findings suggest that differentiation of cell subpopulations is regulated by specific miRNAs. Furthermore, miRNA-based strategies may be developed for immunotherapeutic treatments of AML.
Collapse
Affiliation(s)
- Marzieh Izadifard
- Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ahmadvand
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Bahram Chahardouli
- Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Vaezi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | - Ghasem Janbabai
- Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Marjan Yaghmaie
- Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | - Maryam Barkhordar
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Sun MX, Zhu HC, Yu Y, Yao Y, Li HY, Feng FB, Wang QY, Liu RJ, Sun CG. Role of the Wnt signaling pathway in the complex microenvironment of breast cancer and prospects for therapeutic potential (Review). Int J Oncol 2025; 66:36. [PMID: 40145557 PMCID: PMC12068849 DOI: 10.3892/ijo.2025.5742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
The focus on breast cancer treatment has shifted from the cytotoxic effects of single drugs on tumor cells to multidimensional multi‑pathway synergistic intervention strategies targeting the tumor microenvironment (TME). The activation of the Wnt signaling pathway in the TME of breast cancer cells serves a key regulatory role in tissue homeostasis and is a key driver of the carcinogenic process. Modulating the crosstalk between the Wnt pathway and TME of breast cancer is key for understanding the biological behavior of breast cancer and advancing the development of novel antitumor drugs. The present review aimed to summarize the complex mechanisms of the Wnt signaling pathway in the breast cancer TME, interactions between the Wnt signaling pathway and components of the breast cancer TME and breast cancer‑associated genes, as well as the interactions between the Wnt signaling pathway and other signaling cascades at the molecular level. Furthermore, the present review aimed to highlight the unique advantages of the Wnt signaling pathway in the macro‑regulation of the TME and the current therapeutic strategies targeting the Wnt signaling pathway, their potential clinical value and future research directions in breast cancer treatment.
Collapse
Affiliation(s)
- Meng Xuan Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Han Ci Zhu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Yang Yu
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, P.R. China
| | - Yan Yao
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Hua Yao Li
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Fu Bin Feng
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Qing Yang Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Rui Juan Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Chang Gang Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
11
|
Tian RF, Feng LL, Liang X, Shi Y, Wang H, Fan J, Fan XY, Zhang JJ, Ke Y, Yang T, Huo F, Fu X, Cui HY, Chen ZN, Li L. Carnitine palmitoyltransferase 2 as a novel prognostic biomarker and immunoregulator in colorectal cancer. Int J Biol Macromol 2025; 309:142945. [PMID: 40210071 DOI: 10.1016/j.ijbiomac.2025.142945] [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: 10/31/2024] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Metabolic interventions are critical for enhancing immunotherapy efficacy, but reliable metabolic targets remain absent for colorectal cancer (CRC). This study aims to investigate the interplay between metabolic and immunological factors in CRC, identify metabolic immunoregulatory molecules, and propose targets for prognostic and therapeutic applications. METHODS Immune infiltration and metabolic pathways in CRC were analyzed using CIBERSORT and gene set variation analyses. Cox regression identified survival-related metabolic genes, forming a metabolic-related gene prognostic index (MRGPI), which was validated through survival analysis, timeROC, GSEA, CIBERSORT, and TIDE. Hub genes in the MRGPI were assessed using enrichment and co-expression network analyses. The expression of carnitine palmitoyltransferase 2 (CPT2) was validated through multiplex immunofluorescence of tissue microarrays. While its role was examined by western blot, CCK-8 assay, flow cytometry, qRT-PCR, Elisa, chemotaxis assays, etc. RESULTS: Fatty acid oxidation (FAO) pathways were significantly altered in CRC and correlated with immune cell infiltration and patient survival. The MRGPI, constructed from five survival-related metabolic genes, demonstrated strong prognostic and immunotherapeutic predictive value. Moreover, CPT2, a key hub gene in the MRGPI, whose lower expression in plasma cells predicts unfavorable patients' survival and could be an independent prognostic indicator, while its knockout in tumor cells significantly increases the infiltrating levels of CD8+ T cells via promoting the release of CCL25. CONCLUSION The FAO-dominated MRGPI is a promising biomarker for predicting patient outcomes and immunotherapy response. CPT2 holds potential as a prognostic marker and therapeutic target for CRC metabolic immunotherapy.
Collapse
Affiliation(s)
- Ruo-Fei Tian
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Le-Le Feng
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Xue Liang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Ying Shi
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Hao Wang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Jing Fan
- Air Force Hospital of the Northern Theater Command of the People's Liberation Army of China, Shenyang 110000, China
| | - Xin-Yu Fan
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Jia-Jia Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Yuan Ke
- Department of Radiation and Medical Oncology, Hubei Province Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Ting Yang
- Bayi Orthopedic Hospital, Chengdu 610031, China
| | - Fei Huo
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Xin Fu
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China
| | - Hong-Yong Cui
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China.
| | - Zhi-Nan Chen
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China.
| | - Ling Li
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancer, State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
12
|
Zhang X, Jiang S, Yin H, Zhang H, Yang L, Sun P, Kuai X, Chen C, Huang J. Enhanced IL36RN Expression and Its Association With Immune Microenvironment Predicts Poor Prognosis in Gastric Cancer. Cancer Med 2025; 14:e70954. [PMID: 40347070 PMCID: PMC12065078 DOI: 10.1002/cam4.70954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 03/18/2025] [Accepted: 04/29/2025] [Indexed: 05/12/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) remains a prevalent and lethal malignancy worldwide, underscoring the urgent need to identify novel therapeutic targets and elucidate the tumor microenvironment (TME) to enhance clinical outcomes. METHODS IL36RN mRNA expression in GC tissues was analyzed using The Cancer Genome Atlas (TCGA) dataset. Bioinformatics approaches, cellular models, and clinical tissue microarrays were employed to investigate the functional role of IL36RN, its interactions within the TME, and its prognostic significance. RESULTS IL36RN expression was markedly upregulated in GC tissues and associated with unfavorable survival outcomes. Functional assays demonstrated that IL36RN silencing suppressed GC cell proliferation and invasion. Elevated IL36RN expression correlated with enhanced CD8+ T cell infiltration in the TME and served as an independent prognostic indicator in GC. CONCLUSIONS IL36RN represents a potential prognostic biomarker and therapeutic target in GC, offering novel avenues for precision oncology and immunotherapeutic intervention.
Collapse
Affiliation(s)
- Xiaojing Zhang
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University and Medical School of Nantong UniversityNantongJiangsuChina
- Department of Clinical Biobank and Institute of OncologyAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Sutian Jiang
- Department of PathologyLishui People's HospitalLishuiZhejiangChina
| | - Hang Yin
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University and Medical School of Nantong UniversityNantongJiangsuChina
| | - Hui Zhang
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University and Medical School of Nantong UniversityNantongJiangsuChina
| | - Lei Yang
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University and Medical School of Nantong UniversityNantongJiangsuChina
- Department of Clinical Biobank and Institute of OncologyAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Pingping Sun
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University and Medical School of Nantong UniversityNantongJiangsuChina
- Department of Clinical Biobank and Institute of OncologyAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Xiaoling Kuai
- Department of GastroenterologyNantong University Affiliated HospitalNantongJiangsuChina
| | - Chen Chen
- Department of OncologyJiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and the Affiliated Cancer Hospital of Nanjing Medical UniversityNanjingJiangsuChina
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
| | - Jianfei Huang
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University and Medical School of Nantong UniversityNantongJiangsuChina
- Department of Clinical Biobank and Institute of OncologyAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| |
Collapse
|
13
|
Li M, Wu X, Jiang L, Liu M, Yanju G, Li X, Tian F, Ye F, Wang J, Wang S, Qin C, Zhang L. Exploring the co-morbid relationship between Alzheimer's disease and lung cancer in the 5xFAD transgenic mouse model. Animal Model Exp Med 2025; 8:784-797. [PMID: 39930922 DOI: 10.1002/ame2.12527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/30/2024] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) and lung cancer are leading causes of mortality among the older population. Epidemiological evidence suggests an antagonistic relationship between them, whereby patients with AD exhibit a reduced risk of developing cancer and vice versa. However, the precise mechanism by which AD antagonizes lung cancer progression warrants further elucidation. METHODS To this end, we established a co-morbidity model using 5xFAD transgenic mice induced with the carcinogen urethane. We visualized and quantified surface lung tumor colonies, assessed pathological parameters associated with lung cancer and AD using histopathological analysis, and employed single-cell sequencing and molecular pathological analyses to explore the mechanisms by which AD confers resistance to lung cancer. RESULTS Our findings revealed a significant reduction in lung tumor incidence in the AD group compared with that in the wild-type (WT) group. The results indicated a close association between AD-induced inhibition of lung tumor progression and iron homeostasis imbalance and increased oxidative stress. Moreover, greater CD8+ T cytotoxic lymphocyte and effector natural killer cell infiltration in the lung tumor tissues of AD mice and enhanced CD8+ T cytotoxic lymphocyte-mediated killing of target cells may be the primary factors contributing to the inhibition of lung tumor growth in the presence of AD. CONCLUSION This study identified essential mechanisms through which AD suppresses lung tumorigenesis, thereby providing targets for potential therapeutic interventions in these diseases.
Collapse
Affiliation(s)
- Mingfeng Li
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and Innovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Xinghan Wu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and Innovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Lin Jiang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and Innovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Min Liu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and Innovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Gong Yanju
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and Innovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Xiaomeng Li
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and Innovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Fan Tian
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and Innovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Fan Ye
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and Innovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Jinlong Wang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and Innovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Siyuan Wang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and Innovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Chuan Qin
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and Innovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China
- Changping National Laboratory (CPNL), Beijing, China
| | - Ling Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, International Center for Technology and Innovation of Animal Model, Comparative Medicine Center, Institute of Laboratory Animal Sciences, Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences (CAMS), Beijing, China
| |
Collapse
|
14
|
Liao P, Zhou Y, Qiu Y, Hu R, Li H, Sun H, Li Y. Metal-modulated T cell antitumor immunity and emerging metalloimmunotherapy. Cancer Metastasis Rev 2025; 44:49. [PMID: 40301229 DOI: 10.1007/s10555-025-10266-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/19/2025] [Indexed: 05/01/2025]
Abstract
In recent years, increasing evidence has shown that metals play important roles in both innate and adaptive immunity. An emerging concept of metalloimmunotherapy has been proposed, which may accelerate the development of immunotherapy for cancers. Here, we discuss how metals affect T cell function through different signaling pathways. Metals impact the fate of T cells, including their activation, proliferation, cytotoxicity, and differentiation. Most importantly, metals also participate in mitochondrial operation by regulating energy production and reactive oxygen species homeostasis in T cells. We also identified the metal-based mutual effects between tumor cells and T cells in the tumor microenvironment. Overall, the antitumor effect of T cells can be improved by targeting metal metabolism and metalloimmunotherapy, which will be a step forward in the treatment of cancers.
Collapse
Affiliation(s)
- Peiyun Liao
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying Zhou
- Department of Chemistry, CAS-HKU Joint Laboratory of Metallomics On Health and Environment, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yingqi Qiu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Rong Hu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongyan Li
- Department of Chemistry, CAS-HKU Joint Laboratory of Metallomics On Health and Environment, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Hongzhe Sun
- Department of Chemistry, CAS-HKU Joint Laboratory of Metallomics On Health and Environment, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.
- Guangdong Engineering Research Center of Precision Immune Cell Therapy Technology, Zhujiang Hospital, No. 253, Gongye Road, Guangzhou, China.
| |
Collapse
|
15
|
Han D, Wu C, Jin H. Anti-tumor immune modulation and favorable survival outcomes in uterine corpus endometrial carcinoma: insights from PIK3CA/ARID1A co-mutation analysis. Discov Oncol 2025; 16:641. [PMID: 40301230 PMCID: PMC12040806 DOI: 10.1007/s12672-025-02422-5] [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: 09/30/2024] [Accepted: 04/17/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Uterine corpus endometrial carcinoma (UCEC) is the most prevalent cancer of the female reproductive system, posing significant risks to women's reproductive health and imposing considerable economic burdens on families and society due to high treatment costs. METHODS The study population comprised 529 UCEC patients who were selected and retrieved from the cBioPortal public database for a comprehensive integrated analysis. This study aims to explore the prognostic significance of co-mutation in PIK3CA/ARID1A genes in UCEC, utilizing various bioinformatics approaches, including differential expression genes (DEGs) analysis, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, Gene Set Enrichment Analysis (GSEA), immune infiltration analysis, and the establishment of nomogram model. RESULTS PIK3CA/ARID1A co-mutation group had a better prognosis than the other three groups. The co-mutation of PIK3CA/ARID1A was associated with a significantly improved overall survival (OS) in patients with UCEC and immunotherapy markers. This result was further corroborated in the MSK cohort, reinforcing the robustness of our observations. Our findings revealed that 222 genes were upregulated and 1,464 genes downregulated in the co-mutation group compared to the non-co-mutation (NCM) group, providing a molecular basis for understanding the biological roles of these gene mutations in UCEC. Additionally, pathway analysis identified significant enrichment in immune-related pathways, emphasizing the potential for co-mutation to influence tumor progression via immune modulation. Notably, patients with co-mutations exhibited improved overall survival (P < 0.05), suggesting their role as vital prognostic markers. The developed Cox proportional hazards model demonstrated high predictive accuracy (C-index = 0.835), supporting personalized management for UCEC patients. CONCLUSION In conclusion, this study underscores the importance of PIK3CA and ARID1A co-mutations in UCEC, advocating for their further exploration in clinical applications and therapeutic strategies.
Collapse
Affiliation(s)
- Dongmei Han
- Center for Precision Cancer Medicine and Translation Research, Tianjin Cancer Hospital Airport Hospital, Tianjin, 300181, China
- Center for Precision Cancer Medicine and Translation Research, Tianjin Medical University Cancer Institute & Hospital, No. 99, East 5 th Road, Tianjin Airport Economic Zone, Tianjin, 300060, China
| | - Caihong Wu
- Center for Precision Cancer Medicine and Translation Research, Tianjin Cancer Hospital Airport Hospital, Tianjin, 300181, China
- Center for Precision Cancer Medicine and Translation Research, Tianjin Medical University Cancer Institute & Hospital, No. 99, East 5 th Road, Tianjin Airport Economic Zone, Tianjin, 300060, China
| | - Hao Jin
- Center for Precision Cancer Medicine and Translation Research, Tianjin Cancer Hospital Airport Hospital, Tianjin, 300181, China.
- Center for Precision Cancer Medicine and Translation Research, Tianjin Medical University Cancer Institute & Hospital, No. 99, East 5 th Road, Tianjin Airport Economic Zone, Tianjin, 300060, China.
- Clinical Research Management Department, Tianjin Cancer Hospital Airport Hospital, Tianjin, 300181, China.
| |
Collapse
|
16
|
Ma J, Tang D, Cui G, Zhang X, Wang X, Li Y, Hu E, Zhou X, Liu H, Peng Q, Cai C, Deng X, Zeng S, Chen Y, Xiao Z. The molecular characteristics of DNA damage and repair related to P53 mutation for predicting the recurrence and immunotherapy response in hepatocellular carcinoma. Sci Rep 2025; 15:14939. [PMID: 40301641 PMCID: PMC12041276 DOI: 10.1038/s41598-025-99853-5] [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: 01/31/2025] [Accepted: 04/23/2025] [Indexed: 05/01/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths globally, owing to its high recurrence rate of 50 to 70% within five years. Despite known associations of certain DNA damage and repair (DDR) genes with tumor recurrence and drug resistance, a comprehensive understanding of DDR pathways' role in predicting HCC recurrence and therapeutic responses remains elusive. Addressing this gap could offer significant advancements in prognostic and therapeutic strategies for HCC. This study used 769 RNA sequencing samples from public datasets and 53 samples from Xiangya Hospital for DDR model training and validation. It came out that DDR pathways were significantly enriched in samples with P53 mutations. Next, among the 173 combinations of algorithms and parameters, CoxBoost + RSF, Lasso [fold = 10] + RSF, and Lasso [fold = 50] + RSF demonstrated the best performance. The average AUC values of 1 to 5 years and the average concordance index (C-index) value were around 0.7. The risk scores were increased in tumors with recurrence, P53 mutation, and higher TNM stages. High-risk groups, characterized by enriched DDR pathways, exhibited lower CD8 + T cell infiltration and poorer responses to immunotherapy using atezolizumab and bevacizumab, emphasizing the potential of DDR signatures as valuable prognostic and therapeutic biomarkers. In conclusion, the DDR signatures associated with P53 mutations can predict recurrence and therapeutic response in HCC, highlighting their potential as prognostic and therapeutic biomarkers.
Collapse
Affiliation(s)
- Jiayao Ma
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Diya Tang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Guangzu Cui
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiangyang Zhang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xinwen Wang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yin Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Erya Hu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xin Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Haicong Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Qingping Peng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiangying Deng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yihong Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Zemin Xiao
- Department of Oncology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde, 415000, Hunan, China.
| |
Collapse
|
17
|
Wang N, Yu H, Yin J, Yu X. pH-responsive nano-vaccine combined with anti-PD-1 antibodies for enhanced immunotherapy of breast cancer. Theranostics 2025; 15:6022-6043. [PMID: 40365283 PMCID: PMC12068292 DOI: 10.7150/thno.107200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
Objective: This study aimed to investigate the therapeutic potential and underlying mechanisms of a novel pH-responsive nano-vaccine in combination with anti-Programmed Cell Death Protein 1 (PD-1) antibodies for the treatment of breast cancer (BC), with a focus on tumor growth inhibition, metastasis prevention, and immune microenvironment modulation. Methods: A pH-responsive amphiphilic diblock copolymer was synthesized using reversible addition-fragmentation chain transfer (RAFT) polymerization and conjugated with STING agonist ADU-S100 and mannose to specifically target dendritic cells (DCs). The nano-vaccine was further formulated with antigen peptides and polyethyleneimine (PEI) to enhance antigen delivery. Its particle size, stability, and surface charge were characterized using dynamic light scattering (DLS) and zeta potential analysis. In vitro, the immunostimulatory capacity of the nano-vaccine was evaluated via flow cytometry (FCM) analysis of DC activation markers. In vivo, mouse immune and tumor recurrence models were used to assess the its effects on T-cell activation, tumor suppression, and immune memory induction. The therapeutic efficacy of nano-vaccine/anti-PD-1 combination therapy was further assessed. Results: The nano-vaccine efficiently activated DCs and promoted antigen presentation, as indicated by increased CD80, CD86, and MHC-II expression in vitro. In mouse models, it effectively inhibited tumor growth, induced antigen-specific T-cell responses, and suppressed recurrent and metastatic tumor progression. The combination with anti-PD-1 antibodies further enhanced tumor control, immune cell infiltration, and survival rates compared to monotherapy. Conclusion: The pH-responsive nano-vaccine combined with anti-PD-1 antibodies showed remarkable synergistic effects in BC treatment, highlighting its potential to enhance immune checkpoint blockade therapy and offer a promising strategy for clinical applications in solid tumors.
Collapse
Affiliation(s)
- Ning Wang
- Department of Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Hong Yu
- Department of Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jianqiao Yin
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xiaopeng Yu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
18
|
Wang W, Huang C, Bi S, Liang H, Li S, Lu T, Liu B, Tang Y, Wang Q. A predictive model for the transformation from cervical inflammation to cancer based on tumor immune-related factors. Front Immunol 2025; 16:1532048. [PMID: 40352921 PMCID: PMC12062085 DOI: 10.3389/fimmu.2025.1532048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 04/04/2025] [Indexed: 05/14/2025] Open
Abstract
Introduction Persistent high-risk human papillomavirus (HR-HPV) infection is crucial in transforming cervical intraepithelial neoplasia (CIN) into cervical cancer (CC) by evading immune responses. Additionally, changes in the tumor immune microenvironment (TIME) are increasingly linked to CIN progression to CC. Methods In this study, we used public databases to collect transcriptome data for CIN, CC, and normal cervix, employing LASSO regression to find TIP genes with differential expression. We also used the CIBERSORT algorithm to analyze immune cells in the cervix. ROC curves were plotted to assess tumor-infiltrating immune cells (TICs) and the expression of tumor-infiltrating cell-related genes (TICRGs) for predicting CC efficacy and identifying immune-related genes and cells associated with cervical disease progression for future modeling. We developed a cervical "inflammation-cancer transition" prediction model using the random forest algorithm and assessed its accuracy with internal and external data. Clinical samples from two hospitals were analyzed using multiplexed immunohistochemistry (mIHC) to detect risk factors in various cervical diseases, serving as an independent validation cohort for the model's reliability. Results Four genes, ARG2, HSP90AA1, EZH2, ICAM1, and two immune cells, M1 macrophages and activated CD4 memory T cells, were selected as variables, and a predictive model was constructed. The model achieved an AUC of 1 for internal training sets and 0.912 for testing sets. For validation cohort, the AUC was 0.864 for GSE7803 and 0.918 for TCGA/GTEx. For external validation (GSE39001, GSE149763, and GSE138080), the AUC was 0.703, 0.889 and 0.696. At the same time, the mIHC experimental results also effectively validated the stability of the model. Discussion In conclusion, the developed model enhances the predictive accuracy for the progression of CIN to CC and offers novel insights for the early diagnosis and screening of CC.
Collapse
Affiliation(s)
- Wenjie Wang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment of Regional High-incidence Tumors, Ministry of Education Key Laboratory, Guangxi Medical University, Nanning, Guangxi, China
- University Engineering Research Center of Oncolytic & Nanosystem Development, Nanning, Guangxi, China
- Institute of Life Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Chuntao Huang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment of Regional High-incidence Tumors, Ministry of Education Key Laboratory, Guangxi Medical University, Nanning, Guangxi, China
- University Engineering Research Center of Oncolytic & Nanosystem Development, Nanning, Guangxi, China
| | - Shiwen Bi
- Key Laboratory of Early Prevention and Treatment of Regional High-incidence Tumors, Ministry of Education Key Laboratory, Guangxi Medical University, Nanning, Guangxi, China
- University Engineering Research Center of Oncolytic & Nanosystem Development, Nanning, Guangxi, China
- Department of Pathology, Wuming Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Huiting Liang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment of Regional High-incidence Tumors, Ministry of Education Key Laboratory, Guangxi Medical University, Nanning, Guangxi, China
- University Engineering Research Center of Oncolytic & Nanosystem Development, Nanning, Guangxi, China
| | - Songlin Li
- Key Laboratory of Early Prevention and Treatment of Regional High-incidence Tumors, Ministry of Education Key Laboratory, Guangxi Medical University, Nanning, Guangxi, China
- University Engineering Research Center of Oncolytic & Nanosystem Development, Nanning, Guangxi, China
- Department of Pathology, Wuming Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Tingting Lu
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment of Regional High-incidence Tumors, Ministry of Education Key Laboratory, Guangxi Medical University, Nanning, Guangxi, China
- University Engineering Research Center of Oncolytic & Nanosystem Development, Nanning, Guangxi, China
| | - Ben Liu
- Key Laboratory of Early Prevention and Treatment of Regional High-incidence Tumors, Ministry of Education Key Laboratory, Guangxi Medical University, Nanning, Guangxi, China
- University Engineering Research Center of Oncolytic & Nanosystem Development, Nanning, Guangxi, China
- Institute of Life Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Yong Tang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
- University Engineering Research Center of Oncolytic & Nanosystem Development, Nanning, Guangxi, China
- Department of Pathology, Wuming Hospital of Guangxi Medical University, Nanning, Guangxi, China
- State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning, Guangxi, China
| | - Qi Wang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment of Regional High-incidence Tumors, Ministry of Education Key Laboratory, Guangxi Medical University, Nanning, Guangxi, China
- University Engineering Research Center of Oncolytic & Nanosystem Development, Nanning, Guangxi, China
| |
Collapse
|
19
|
Song B, Shuang L, Zhang S, Tong C, Chen Q, Li Y, Hao M, Niu W, Jin CH. Research progress of nano drug delivery systems in the anti-tumor treatment of traditional Chinese medicine monomers. PeerJ 2025; 13:e19332. [PMID: 40292112 PMCID: PMC12034246 DOI: 10.7717/peerj.19332] [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: 12/04/2024] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Tumors pose a serious threat to global public health and are usually treated from two aspects: tumor cells and tumor microenvironment. Compared with traditional chemotherapy drugs, traditional Chinese medicine (TCM) monomers have advantages in tumor treatment, such as multiple targets, multiple levels and synergistic intervention. However, most TCM active ingredients have disadvantages such as poor water solubility and stability, which restrict their clinical application. Nano drug delivery systems have the functions of improving the bioavailability of TCM anti-tumor active ingredients, enhancing tissue targeting, achieving controlled drug release, and inhibiting tumor multidrug resistance. Compared with free monomers, they have higher therapeutic effects and fewer side effects. This article summarizes five commonly used anti-tumor TCM monomer nanocarriers, including lipid nanomaterials, exosomes, polymer micelles, carbon nanotubes, and dendrimers, and explains their anti-tumor mechanisms after combining with TCM, such as inhibiting tumor cell proliferation and metastasis, regulating tumor microenvironment, etc. At the same time, the potential of nano drug delivery systems combined with radiotherapy and immunotherapy is discussed, as well as the current problems of potential toxicity, long-term stability, and complex amplification process, as well as future development directions, aiming to provide a reference for promoting the clinical application of nano drug delivery systems for TCM anti-tumor active ingredients.
Collapse
Affiliation(s)
- Bocui Song
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Li Shuang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Shuang Zhang
- Comprehensive Service Center, Yongji Economic Development Zone, Jilin, Jilin, China
| | - Chunyu Tong
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Qian Chen
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Yuqi Li
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Meihan Hao
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Wenqi Niu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Cheng-Hao Jin
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
- College of Life Science and Technology, College of Life Science and Technology, Daqing, Heilongjiang, China
| |
Collapse
|
20
|
Zhang N, Qin X, Liu J, Han K, Kang M, Zhu Z, Zhang D, Zhong F. Pan-cancer analysis and validation show GTF2E2's diagnostic, prognostic, and immunological roles in regulating ferroptosis in endometrial cancer. PLoS One 2025; 20:e0321983. [PMID: 40267151 PMCID: PMC12017540 DOI: 10.1371/journal.pone.0321983] [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: 12/18/2024] [Accepted: 03/11/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Transcription initiation factor IIE subunit beta (GTF2E2) is a crucial component of the RNA polymerase II transcription initiation complex. There is a lack of more detailed research on the biological function of GTF2E2 in pan-cancer. METHODS We conducted a comprehensive pan-cancer analysis using data from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) project. Employing a multi-pronged approach with tools including R, Cytoscape, TISIDB, cBioPortal, STRING, GSCALite, and CancerSEA, we investigated GTF2E2's expression patterns, prognostic value, mutational landscape, functional enrichment, and immunological associations across 33 cancer types. Besides, we further validated the bioinformatic results through in vitro experiments in Uterine corpus endometrial carcinoma (UCEC), including western blotting (WB), cell proliferation assays and transwell. DCFH-DA, C11-BODIPY 581/591 and FeRhoNox-1 probes were performed to identify ferroptosis levels in vitro. RESULTS GTF2E2 expression was significantly elevated in most cancers compared to normal tissues, with notable diagnostic potential (AUC > 0.7) in 20 cancer types. GTF2E2 expression varied across molecular and immune subtypes and correlated with tumor stage and patient age in several cancers. Functional enrichment analyses highlighted GTF2E2's involvement in key cancer-related and immunological pathways. Notably, GTF2E2 promoted UCEC progression in vitro, and knockdown of GTF2E2 significantly inhibited the proliferation, migration and invasion of UCEC cells. Compared with the control group, GPX4 expression was down-regulated and ACSL4 expression was up-regulated in the GTF2E2-knockdown group. Knockdown of GTF2E2 also increased the intracellular levels of Fe2+, lipid peroxides (LPOs) and reactive oxygen species (ROS). CONCLUSIONS Our findings underscore GTF2E2's multifaceted roles in cancer biology, highlighting its potential as a diagnostic biomarker, prognostic indicator, and immunotherapeutic target across various malignancies. This investigation has the potential to contribute significantly to a deeper understanding of the substantial involvement of GTF2E2 in human malignancies, particularly UCEC.
Collapse
Affiliation(s)
- Nie Zhang
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, China
- Graduate School of Anhui Medical University, Anhui, China
- Key Laboratory of Gametes and Abnormal Reproductive Tract of National Health Commission, Anhui Medical University, Anhui, China
| | - Xuejin Qin
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, China
| | - Jingjing Liu
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, China
| | - Ke Han
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, China
| | - Manman Kang
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, China
| | - Zhengchun Zhu
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, China
| | - Di Zhang
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, China
| | - Fei Zhong
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, China
- Graduate School of Anhui Medical University, Anhui, China
- Key Laboratory of Gametes and Abnormal Reproductive Tract of National Health Commission, Anhui Medical University, Anhui, China
| |
Collapse
|
21
|
Liu N, Wang X, Wang Z, Kan Y, Fang Y, Gao J, Kong X, Wang J. Nanomaterials-driven in situ vaccination: a novel frontier in tumor immunotherapy. J Hematol Oncol 2025; 18:45. [PMID: 40247328 PMCID: PMC12007348 DOI: 10.1186/s13045-025-01692-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/19/2025] [Indexed: 04/19/2025] Open
Abstract
In situ vaccination (ISV) has emerged as a promising strategy in cancer immunotherapy, offering a targeted approach that uses the tumor microenvironment (TME) to stimulate an immune response directly at the tumor site. This method minimizes systemic exposure while maintaining therapeutic efficacy and enhancing safety. Recent advances in nanotechnology have enabled new approaches to ISV by utilizing nanomaterials with unique properties, including enhanced permeability, retention, and controlled drug release. ISV employing nanomaterials can induce immunogenic cell death and reverse the immunosuppressive and hypoxic TME, thereby converting a "cold" tumor into a "hot" tumor and facilitating a more robust immune response. This review examines the mechanisms through which nanomaterials-based ISV enhances anti-tumor immunity, summarizes clinical applications of these strategies, and evaluates its capacity to serve as a neoadjuvant therapy for eliminating micrometastases in early-stage cancer patients. Challenges associated with the clinical translation of nanomaterials-based ISV, including nanomaterial metabolism, optimization of treatment protocols, and integration with other therapies such as radiotherapy, chemotherapy, and photothermal therapy, are also discussed. Advances in nanotechnology and immunotherapy continue to expand the possible applications of ISV, potentially leading to improved outcomes across a broad range of cancer types.
Collapse
Affiliation(s)
- Naimeng Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiangyu Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhongzhao Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yonemori Kan
- Department of Medical Oncology, National Cancer Center Hospital (NCCH), 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yi Fang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jidong Gao
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518127, China.
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
22
|
Zhang D, Wang W, Tang M, Qu C, Jiang Z, Li X, Luan Y. In Situ Gene Engineering Approach to Overcome Tumor Resistance and Enhance T Cell-Mediated Cancer Immunotherapy. NANO LETTERS 2025; 25:6200-6208. [PMID: 40180619 DOI: 10.1021/acs.nanolett.5c00488] [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: 04/05/2025]
Abstract
T cell-mediated cancer immunotherapy harnesses the power of cytotoxic T lymphocytes (CTLs) to target and eradicate tumor cells. However, tumor cells often evade immune attack through membrane repair mechanisms involving endosomal sorting complexes required for transport (ESCRT) and immune suppression within the tumor microenvironment. Here, we developed a robust TMV@PpCHIL nanomedicine to address these issues by reprogramming tumor cells via in situ gene editing. Using CRISPR/Cas9, we disrupted the Chmp4b gene, a key component of the ESCRT machinery, preventing tumor cells from repairing CTL-induced membrane damage. Simultaneously, we genetically engineered tumor cells to produce interleukin-12 (IL-12), a cytokine that enhances CTL activation. The TMV@PpCHIL nanomedicine, designed by coating tumor membrane vesicles (TMVs) onto polyamidoamine (PAMAM) dendrimer-condensed plasmid complexes, ensured efficient CRISPR/Cas9-based gene editing and sustained IL-12 production. This approach significantly enhanced CTL-mediated tumor cell cytotoxicity, suppressed tumor growth, reduced metastasis, and prolonged survival, providing a promising strategy for durable cancer treatment.
Collapse
Affiliation(s)
- Di Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Wenjuan Wang
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Mingtan Tang
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Chenghao Qu
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, 107 West Wenhua Road, Jinan, 250012, China
| | - Zeyu Jiang
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Xuerui Li
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Yuxia Luan
- Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| |
Collapse
|
23
|
Zhou ZY, Bai N, Zheng WJ, Ni SJ. MultiOmics analysis of metabolic dysregulation and immune features in breast cancer. Int Immunopharmacol 2025; 152:114376. [PMID: 40054322 DOI: 10.1016/j.intimp.2025.114376] [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: 03/15/2024] [Revised: 02/09/2025] [Accepted: 02/24/2025] [Indexed: 03/24/2025]
Abstract
Metabolic disorders and diminished immune response are hallmark characteristics of tumors. However, limited studies have comprehensively integrated metabolic and immunological factors to evaluate or predict the prognosis of cancer patients. In this study, we utilized 72 metabolic pathway gene sets from the MsigDB database to conduct GSVA, univariate regression, and prognostic analyses on 247 breast cancer samples sourced from the TCGA and GEO databases. Consequently, five metabolic pathways with significant research value were identified. Based on these findings, unsupervised clustering was performed on the breast cancer samples to compare differences in gene expression, clinicopathological features, immune infiltration levels, and prognosis across different clusters. This process led to the identification of nine metabolism-related characteristic genes. Additionally, single-cell sequencing analysis was employed to assess the spatial expression patterns of these characteristic genes, revealing significantly higher expression indices in tumor cells compared to non-tumor cells. Subsequently, machine learning algorithms were applied to reconstruct metabolic risk models for evaluating the prognosis of breast cancer patients. The results indicated that the high metabolic risk group exhibited higher gene mutation scores, a greater proportion of unfavorable clinicopathological parameters, and lower chemokine and immune scores compared to the low-risk group. In conclusion, the metabolic risk model constructed using metabolism-related characteristic genes can accurately distinguish and predict the survival prognosis and immunotherapy outcomes of breast cancer patients, offering novel targets and insights for personalized treatment strategies.
Collapse
Affiliation(s)
- Zuo-Yuan Zhou
- Department of Oncology, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China
| | - Nan Bai
- Medical school of Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China
| | - Wen-Jie Zheng
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China.
| | - Su-Jie Ni
- Department of Oncology, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
24
|
Wei W, Li H, Tian S, Zhang C, Liu J, Tao W, Cai T, Dong Y, Wang C, Lu D, Ai Y, Zhang W, Wang H, Liu K, Fan Y, Gao Y, Huang Q, Ma X, Wang B, Zhang X, Huang Y. Asparagine drives immune evasion in bladder cancer via RIG-I stability and type I IFN signaling. J Clin Invest 2025; 135:e186648. [PMID: 39964752 PMCID: PMC11996873 DOI: 10.1172/jci186648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
Tumor cells often employ many ways to restrain type I IFN signaling to evade immune surveillance. However, whether cellular amino acid metabolism regulates this process remains unclear, and its effects on antitumor immunity are relatively unexplored. Here, we found that asparagine inhibited IFN-I signaling and promoted immune escape in bladder cancer. Depletion of asparagine synthetase (ASNS) strongly limited in vivo tumor growth in a CD8+ T cell-dependent manner and boosted immunotherapy efficacy. Moreover, clinically approved L-asparaginase (ASNase),synergized with anti-PD-1 therapy in suppressing tumor growth. Mechanistically, asparagine can directly bind to RIG-I and facilitate CBL-mediated RIG-I degradation, thereby suppressing IFN signaling and antitumor immune responses. Clinically, tumors with higher ASNS expression show decreased responsiveness to immune checkpoint inhibitor therapy. Together, our findings uncover asparagine as a natural metabolite to modulate RIG-I-mediated IFN-I signaling, providing the basis for developing the combinatorial use of ASNase and anti-PD-1 for bladder cancer.
Collapse
Affiliation(s)
- Wenjie Wei
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
- Medical School of PLA, Beijing, China
| | - Hongzhao Li
- Department of Urology, The Third Medical Center and
| | - Shuo Tian
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
- Medical School of PLA, Beijing, China
| | - Chi Zhang
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
- Medical School of PLA, Beijing, China
| | - Junxiao Liu
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
- Medical School of PLA, Beijing, China
| | - Wen Tao
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
- Medical School of PLA, Beijing, China
| | - Tianwei Cai
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
- Medical School of PLA, Beijing, China
| | - Yuhao Dong
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
- Medical School of PLA, Beijing, China
| | - Chuang Wang
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
- Medical School of PLA, Beijing, China
| | - Dingyi Lu
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Yakun Ai
- Department of Pathology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wanlin Zhang
- Department of Pathology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hanfeng Wang
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
- Medical School of PLA, Beijing, China
| | - Kan Liu
- Department of Urology, The Third Medical Center and
| | - Yang Fan
- Department of Urology, The Third Medical Center and
| | - Yu Gao
- Department of Urology, The Third Medical Center and
| | - Qingbo Huang
- Department of Urology, The Third Medical Center and
| | - Xin Ma
- Department of Urology, The Third Medical Center and
| | - Baojun Wang
- Department of Urology, The Third Medical Center and
| | - Xu Zhang
- Department of Urology, The Third Medical Center and
| | - Yan Huang
- Department of Urology, The Third Medical Center and
- Department of Urology Laboratory, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
25
|
Potievskiy MB, Nekrasova LA, Korobov IV, Bykova EA, Moshurov RI, Sokolov PV, Shatalov PA, Falaleeva NA, Petrov LO, Trifanov VS, Ivanov SA, Shegai PV, Kaprin AD. Prognostic Impact of Phenotypic and Genetic Features of Pancreatic Malignancies. Life (Basel) 2025; 15:635. [PMID: 40283189 PMCID: PMC12028913 DOI: 10.3390/life15040635] [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: 01/16/2025] [Revised: 03/28/2025] [Accepted: 04/06/2025] [Indexed: 04/29/2025] Open
Abstract
Pancreatic cancer is a tumor with a poor prognosis, and improving its survival outcomes remains a formidable challenge, requiring a multidisciplinary approach that integrates innovative surgical and pharmacological strategies, guided by molecular and genetic insights. The pathomorphological and genetic characteristics of pancreatic cancer, reflected in morphological, immunohistochemical, and serological marker expression, reveal key patterns of tumor genotypic changes during carcinogenesis, aiding in prognostic evaluation and clinical strategy development. The mutational profile of pancreatic tumors is quite heterogeneous and diverse in terms of mutated genes, including in relation to morphological subtypes, but certain patterns have been identified as a result of studies. Pancreatic adenocarcinoma, for instance, is frequently driven by mutations regulating cell division (KRAS). The disease prognosis often depends on the morphological subtype and tumor microenvironment. Neuroendocrine tumors of the pancreas are characterized by a number of pathogenetic features that distinguish them from adenocarcinomas. Thus, neuroendocrine tumors are characterized by mutations of the MENIN protein, which prevents cells from entering the mitosis phase by stimulating the expression of cell cycle regulators. Thus, epithelial and neuroendocrine malignancies of the pancreas differ in immunohistochemical and genetic features, but there are similar mechanisms of pathogenesis, such as BRCA1 and BRCA2 gene mutations, impaired expression of p53 antioncogene, and HIF-2α and mTOR receptor mutations. The predictive impact of serological markers, such as CA 19-9 and CEA, offers insights into tumor metastasis and long-term outcomes, emphasizing the need for personalized therapeutic strategies. Tailoring treatments based on individual molecular profiles holds promise for improving prognosis, as the genetic landscape of pancreatic tumors varies significantly between patients. This underscores the importance of a systematic, patient-specific approach that addresses tumor heterogeneity, resistance mechanisms, and the molecular underpinnings of carcinogenesis.
Collapse
Affiliation(s)
- Mikhail B. Potievskiy
- FSBI National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (L.A.N.); (I.V.K.); (E.A.B.); (R.I.M.); (P.V.S.); (P.A.S.); (N.A.F.); (L.O.P.); (V.S.T.); (S.A.I.); (P.V.S.); (A.D.K.)
| | - Lidia A. Nekrasova
- FSBI National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (L.A.N.); (I.V.K.); (E.A.B.); (R.I.M.); (P.V.S.); (P.A.S.); (N.A.F.); (L.O.P.); (V.S.T.); (S.A.I.); (P.V.S.); (A.D.K.)
| | - Ivan V. Korobov
- FSBI National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (L.A.N.); (I.V.K.); (E.A.B.); (R.I.M.); (P.V.S.); (P.A.S.); (N.A.F.); (L.O.P.); (V.S.T.); (S.A.I.); (P.V.S.); (A.D.K.)
| | - Ekaterina A. Bykova
- FSBI National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (L.A.N.); (I.V.K.); (E.A.B.); (R.I.M.); (P.V.S.); (P.A.S.); (N.A.F.); (L.O.P.); (V.S.T.); (S.A.I.); (P.V.S.); (A.D.K.)
| | - Ruslan I. Moshurov
- FSBI National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (L.A.N.); (I.V.K.); (E.A.B.); (R.I.M.); (P.V.S.); (P.A.S.); (N.A.F.); (L.O.P.); (V.S.T.); (S.A.I.); (P.V.S.); (A.D.K.)
| | - Pavel V. Sokolov
- FSBI National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (L.A.N.); (I.V.K.); (E.A.B.); (R.I.M.); (P.V.S.); (P.A.S.); (N.A.F.); (L.O.P.); (V.S.T.); (S.A.I.); (P.V.S.); (A.D.K.)
| | - Peter A. Shatalov
- FSBI National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (L.A.N.); (I.V.K.); (E.A.B.); (R.I.M.); (P.V.S.); (P.A.S.); (N.A.F.); (L.O.P.); (V.S.T.); (S.A.I.); (P.V.S.); (A.D.K.)
| | - Natalia A. Falaleeva
- FSBI National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (L.A.N.); (I.V.K.); (E.A.B.); (R.I.M.); (P.V.S.); (P.A.S.); (N.A.F.); (L.O.P.); (V.S.T.); (S.A.I.); (P.V.S.); (A.D.K.)
| | - Leonid O. Petrov
- FSBI National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (L.A.N.); (I.V.K.); (E.A.B.); (R.I.M.); (P.V.S.); (P.A.S.); (N.A.F.); (L.O.P.); (V.S.T.); (S.A.I.); (P.V.S.); (A.D.K.)
| | - Vladimir S. Trifanov
- FSBI National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (L.A.N.); (I.V.K.); (E.A.B.); (R.I.M.); (P.V.S.); (P.A.S.); (N.A.F.); (L.O.P.); (V.S.T.); (S.A.I.); (P.V.S.); (A.D.K.)
| | - Sergey A. Ivanov
- FSBI National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (L.A.N.); (I.V.K.); (E.A.B.); (R.I.M.); (P.V.S.); (P.A.S.); (N.A.F.); (L.O.P.); (V.S.T.); (S.A.I.); (P.V.S.); (A.D.K.)
| | - Peter V. Shegai
- FSBI National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (L.A.N.); (I.V.K.); (E.A.B.); (R.I.M.); (P.V.S.); (P.A.S.); (N.A.F.); (L.O.P.); (V.S.T.); (S.A.I.); (P.V.S.); (A.D.K.)
| | - Andrei D. Kaprin
- FSBI National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 249036 Obninsk, Russia; (L.A.N.); (I.V.K.); (E.A.B.); (R.I.M.); (P.V.S.); (P.A.S.); (N.A.F.); (L.O.P.); (V.S.T.); (S.A.I.); (P.V.S.); (A.D.K.)
- Department of Oncology and Radiology, Institute of Medicine, Peoples’ Friendship University of Russia—RUDN University, 6 Miklukho-Maklaya Street, 117198 Moscow, Russia
| |
Collapse
|
26
|
Shi K, Zhao Y, Ye H, Zhu X, Chen Z. Targeting DKK3 to remodel tumor immune microenvironment and enhance cancer immunotherapy. BMC Cancer 2025; 25:645. [PMID: 40205566 PMCID: PMC11984186 DOI: 10.1186/s12885-025-14075-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 04/02/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer immunotherapy such as immune checkpoint blockade (ICB) therapy has made important breakthroughs in cancer treatment, however, currently only parts of cancer patients benefit from ICB therapy. The suppressive tumor immune microenvironment (TIME) impedes the treatment response of immunotherapy, indicating the necessity to explore new treatment targets. Here, we reported a new potential immunotherapeutic target, Dickkopf-3 (DKK3), for cancer treatment. DKK3 expression is up-regulated in the tumors from multiple cancer types, and high DKK3 expression is associated with worse survival outcome across different cancers. We observed that DKK3 directly inhibits the activation of CD8+ T cells and the Th1 differentiation of CD4+ T cells ex vivo. Also, by establishing four different mouse cancer models, we found that DKK3 blockade triggers effective anti-tumor effects and improve the survival of tumor-bearing mice in vivo. DKK3 blockade also remodels the suppressive TIME of different cancer types, including the increased infiltration of CD8+ T cells, IFN-γ+CD8+ T cells, Th1 cells, and decreased infiltration of M2 macrophages and MDSCs in the TIME. Moreover, we found that combined blockade of DKK3 and PD-1 induces synergistic tumor-control effect in our mouse cancer model. Therefore, our study reveals the impact of DKK3 in the TIME and cancer progression, which suggests that DKK3 is a novel and promising immunotherapeutic target for enhanced cancer immunotherapy.
Collapse
Affiliation(s)
- Kai Shi
- Department of Thoracic Surgery, Huai'an Hospital of Huai'an City & Huai'an Cancer Hospital & The Affiliated Huai'an Hospital of Jiangsu College of Nursing, Huai'an, Jiangsu Province, China
| | - Yan Zhao
- Department of Thoracic Surgery, Huai'an Hospital of Huai'an City & Huai'an Cancer Hospital & The Affiliated Huai'an Hospital of Jiangsu College of Nursing, Huai'an, Jiangsu Province, China
| | - Hao Ye
- Department of Thoracic Surgery, Huai'an Hospital of Huai'an City & Huai'an Cancer Hospital & The Affiliated Huai'an Hospital of Jiangsu College of Nursing, Huai'an, Jiangsu Province, China
| | - Xiaoming Zhu
- Department of Thoracic Surgery, Huai'an Hospital of Huai'an City & Huai'an Cancer Hospital & The Affiliated Huai'an Hospital of Jiangsu College of Nursing, Huai'an, Jiangsu Province, China
| | - Zhenghai Chen
- Department of Thoracic Surgery, Huai'an Hospital of Huai'an City & Huai'an Cancer Hospital & The Affiliated Huai'an Hospital of Jiangsu College of Nursing, Huai'an, Jiangsu Province, China.
| |
Collapse
|
27
|
Zhou X, Zhou Z, Qin X, Cheng J, Fu Y, Wang Y, Wang J, Qin P, Zhang D. Amino Acid Metabolism Subtypes in Neuroblastoma Identifying Distinct Prognosis and Therapeutic Vulnerabilities. J Proteome Res 2025; 24:1560-1578. [PMID: 39442086 DOI: 10.1021/acs.jproteome.4c00554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Although amino acid (AA) metabolism is linked to tumor progression and could serve as an attractive intervention target, its association with neuroblastoma (NB) is unknown. Based on AA metabolism-related genes, we established three NB subtypes associated with distinct prognoses and specific functions, with C1 and C2 having better outcomes. The C1 displayed enhanced metabolic activity and recruited metabolism-associated cells. The C2 exhibited an activated immune microenvironment and was more vulnerable to immunotherapy. The C3, characterized by cell cycle peculiarity, possessed a dismal prognosis and high frequency of gene mutations and was susceptible to chemotherapy. Furthermore, single-cell RNA sequencing analysis revealed that the C3-associated Scissor+ cell subpopulation was characterized by notorious functional states and orchestrated an immunosuppressive microenvironment. Additionally, we identified that ALK and BIRC5 contributed to the shorter lifespan of C3 and their corresponding inhibitors were potential interventions. In conclusion, we identified three distinct subtypes of NB, which help us foster individualized therapeutic strategies to improve the prognosis of NB.
Collapse
Affiliation(s)
- Xing Zhou
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xiaohan Qin
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jian Cheng
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yongcheng Fu
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yuanyuan Wang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jingyue Wang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Pan Qin
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Da Zhang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| |
Collapse
|
28
|
Su B, Chen Q, Li X, Fang M, Wang Y, Song H, You H, Zhou Z, Wu Y, Zhao Z, Chen Y, Fan H, Li C, Jiang C, Sun T. A Methionine Allocation Nanoregulator for the Suppression of Cancer Stem Cells and Support to the Immune Cells by Epigenetic Regulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415207. [PMID: 39985256 PMCID: PMC12005795 DOI: 10.1002/advs.202415207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/13/2025] [Indexed: 02/24/2025]
Abstract
Epigenetic dysregulation is prevalent in human cancers, affecting gene expression and metabolic patterns to meet the demands of malignant evolution and abnormal epigenetic processes, and resulting in a protumor immune microenvironment. Tumors require a steady supply of methionine for maintaining epigenetic flexibility, which is the only exogenous precursor of methyl donor S-adenosylmethionine for methylation, crucial for their resistance to therapies and survival in a nutrient-deficient microenvironment. Thus, tumor cells upregulate the Lat4 transporter to compete and deprive methionine in the microenvironment, sustaining their malignant phenotypes and also impairing immune cell functions. Addressing this methionine addiction is the key to overcoming drug resistance and improving immune response. Despite the challenge of lacking specific Lat4 inhibitors, an oxaliplatin prodrug crosslinked fluorinated polycation/anti-Lat4 small interfering RNA complex nanoregulator (AS-F-NP) has been designed and developed here. This nanoregulator restricted the greedy methionine uptake of tumor cells by knocking down Lat4, which in turn inhibited the malignant evolution of the tumor while restoring the viability and function of tumor-infiltrating immune cells.
Collapse
Affiliation(s)
- Boyu Su
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Qinjun Chen
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Xuwen Li
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Mingzhu Fang
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Yu Wang
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Haolin Song
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Haoyu You
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Zheng Zhou
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Yuxing Wu
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Zhenhao Zhao
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Yun Chen
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Hongrui Fan
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Chufeng Li
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
| | - Chen Jiang
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
- Department of Digestive DiseasesNational Regional Medical CenterBinhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhou350212China
| | - Tao Sun
- Department of PharmaceuticsSchool of PharmacyFudan UniversityKey Laboratory of Smart Drug DeliveryMinistry of EducationState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceShanghai201203China
- Quzhou Fudan InstituteQuzhou324003China
| |
Collapse
|
29
|
Cao C, Lai H, Shi Y. MYCN/MNX1 Axis Drives NSCLC Progression by Inducing Macrophage M2 Polarization and CD8 + T Cell Apoptosis via the Wnt/β-Catenin Pathway. J Biochem Mol Toxicol 2025; 39:e70251. [PMID: 40226978 DOI: 10.1002/jbt.70251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/28/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025]
Abstract
Enhanced macrophage M2 polarization and CD8+ T cell dysfunction contribute to the pathophysiology of non-small cell lung cancer (NSCLC). Motor neuron and pancreatic homeobox 1 (MNX1) has emerged as a potential tumor-promoting player. Here, we clarified the activity of MNX1 in NSCLC. PMA-induced THP-1 M0-like macrophages or CD8+ T cells were co-cultured with NSCLC cells. Cell colony formation, migration, proliferation, apoptosis, and invasiveness were assessed by colony formation, wound healing, CCK-8, flow cytometry, and transwell assays, respectively. The ratio of CD206+ macrophages was analyzed by flow cytometry. Ki-67 expression was tested by immunofluorescence. ChIP and luciferase assays were used to evaluate the relationship between MYCN and MNX1. MNX1 was highly expressed in NSCLC, and its loss-of-function suppressed cell growth, motility, and invasiveness in NSCLC cells. MNX1 depletion also diminished macrophage M2 polarization and CD8+ T cell apoptosis. Mechanistically, MYCN increased MNX1 expression at the transcriptional level. MNX1 increase reversed the impact of MYCN depletion on NSCLC cell malignant behaviors, macrophage M2 polarization, and CD8+ T cell viability. MYCN depletion diminished the in vivo growth of A549 subcutaneous xenografts. Additionally, MNX1 increase counteracted the impact of MYCN depletion on the Wnt/β-catenin pathway. Our findings elucidate the oncogenic role of the MYCN/MNX1/Wnt/β-catenin pathway in NSCLC by driving macrophage M2 polarization and diminishing CD8+ T cell viability. Our study thus uncovers a novel mechanism underlying NSCLC development and highlights potential targets for combating NSCLC.
Collapse
Affiliation(s)
- Chengzhang Cao
- Department of Thoracic Surgery, Longyan First Affiliated Hospital of Fujian Medical Univensity, Longyan, China
| | - Haiyin Lai
- Department of Thoracic Surgery, Longyan First Affiliated Hospital of Fujian Medical Univensity, Longyan, China
| | - Yuzhen Shi
- Department of Critical Care Medicine, Longyan First Affiliated Hospital of Fujian Medical Univensity, Longyan, China
| |
Collapse
|
30
|
Yuan S, Sun R, Shi H, Chapman NM, Hu H, Guy C, Rankin S, Kc A, Palacios G, Meng X, Sun X, Zhou P, Yang X, Gottschalk S, Chi H. VDAC2 loss elicits tumour destruction and inflammation for cancer therapy. Nature 2025; 640:1062-1071. [PMID: 40108474 PMCID: PMC12018455 DOI: 10.1038/s41586-025-08732-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 02/03/2025] [Indexed: 03/22/2025]
Abstract
Tumour cells often evade immune pressure exerted by CD8+ T cells or immunotherapies through mechanisms that are largely unclear1,2. Here, using complementary in vivo and in vitro CRISPR-Cas9 genetic screens to target metabolic factors, we established voltage-dependent anion channel 2 (VDAC2) as an immune signal-dependent checkpoint that curtails interferon-γ (IFNγ)-mediated tumour destruction and inflammatory reprogramming of the tumour microenvironment. Targeting VDAC2 in tumour cells enabled IFNγ-induced cell death and cGAS-STING activation, and markedly improved anti-tumour effects and immunotherapeutic responses. Using a genome-scale genetic interaction screen, we identified BAK as the mediator of VDAC2-deficiency-induced effects. Mechanistically, IFNγ stimulation increased BIM, BID and BAK expression, with VDAC2 deficiency eliciting uncontrolled IFNγ-induced BAK activation and mitochondrial damage. Consequently, mitochondrial DNA was aberrantly released into the cytosol and triggered robust activation of cGAS-STING signalling and type I IFN response. Importantly, co-deletion of STING signalling components dampened the therapeutic effects of VDAC2 depletion in tumour cells, suggesting that targeting VDAC2 integrates CD8+ T cell- and IFNγ-mediated adaptive immunity with a tumour-intrinsic innate immune-like response. Together, our findings reveal VDAC2 as a dual-action target to overcome tumour immune evasion and establish the importance of coordinately destructing and inflaming tumours to enable efficacious cancer immunotherapy.
Collapse
Affiliation(s)
- Sujing Yuan
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Renqiang Sun
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hao Shi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Nicole M Chapman
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Haoran Hu
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Cliff Guy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sherri Rankin
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Anil Kc
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gustavo Palacios
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiaoxi Meng
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiang Sun
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Peipei Zhou
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiaoyang Yang
- Experimental Cellular Therapeutics Laboratory, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
31
|
Yang C, Du J, Qiu X, Jia C, Ding C, Wu Y, Gao C, Wang W, Wang X, Liu SB. ERCC3 serves as a prognostic biomarker for hepatocellular carcinoma and positively regulates cell proliferation and migration. Discov Oncol 2025; 16:419. [PMID: 40155569 PMCID: PMC11953519 DOI: 10.1007/s12672-025-02194-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/20/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND ERCC3, a crucial component of the nucleotide excision repair pathway, is implicated in the development and progression of various cancers and is a potential indicator of poor prognosis. However, the expression and function of ERCC3 in hepatocellular carcinoma (HCC) remain unclear. This study aimed to investigate the expression of ERCC3 in HCC tissues and its clinical significance, focusing on elucidating its potential mechanisms and therapeutic value in immunotherapy. METHODS The differential expression and genetic variation characteristics of ERCC3 across various cancers were evaluated using the TCGA database. The expression and prognostic value of ERCC3 in HCC were analyzed by integrating TCGA, GEO, and ICGC datasets. Independent prognostic value of ERCC3 expression levels in HCC was assessed using Cox regression analysis, Kaplan-Meier survival analysis, receiver operating characteristic curves, and nomograms. Pathway association scores were determined using ssGSEA to reveal the biological functions of ERCC3 in HCC and its potential clinical efficacy in immunotherapy. Stable transient cell lines were established by infecting HepG2 cells with lentivirus overexpressing ERCC3. The effects of ERCC3 on HCC cell biological phenotypes were evaluated using RTCA, wound healing, and Transwell assays. Cell cycle distribution and apoptosis were detected by flow cytometry. Transcriptome sequencing was performed to explore the impact of ERCC3 overexpression on the expression of signaling pathway-related genes in HCC. RESULTS The study revealed that ERCC3 is aberrantly expressed in various tumors, with significantly higher mRNA and protein levels in HCC tissues compared to normal tissues. High ERCC3 expression was significantly correlated with poor survival outcomes in HCC patients. Multivariate Cox regression analysis revealed that ERCC3 expression level is an independent prognostic factor for overall survival (P = 0.014). Gene sets associated with the high ERCC3 group were significantly involved in multiple immune pathways and tumor progression-related pathways, and ERCC3 expression was significantly correlated with immune checkpoints in HCC. Overexpression of ERCC3 promoted the proliferation and migration of HCC cells and influenced cell cycle progression. Transcriptome sequencing analysis indicated that ERCC3 overexpression regulated the proliferation of HCC cells, participated in multiple pro-inflammatory pathways, induced the formation of an inflammatory tumor microenvironment, and promoted HCC progression. CONCLUSION This study is the first to reveal the association between high ERCC3 expression and poor prognosis in HCC and to elucidate its immunomodulatory role in HCC. Unlike previous studies, we found that ERCC3 promotes HCC progression by regulating the inflammatory microenvironment and immune checkpoints. These findings establish a novel theoretical foundation for the development of targeted immunotherapies for HCC and provide new insights into the molecular mechanisms underlying ERCC3's role in HCC.
Collapse
Affiliation(s)
- Chen Yang
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan, 063210, China
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Kehua Road 28, Suzhou, 215009, China
| | - Jiahui Du
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Kehua Road 28, Suzhou, 215009, China
| | - Xiuqin Qiu
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Kehua Road 28, Suzhou, 215009, China
| | - Changhong Jia
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan, 063210, China
| | - Cunbao Ding
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan, 063210, China
| | - Yijie Wu
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan, 063210, China
| | - Chaoxu Gao
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan, 063210, China
| | - Weijie Wang
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan, 063210, China.
| | - Xiaojun Wang
- Department of Laboratory Medicine, Suzhou Wuzhong People's Hospital, Suzhou, 215128, Jiangsu, China.
| | - Song-Bai Liu
- College of Life Science, North China University of Science and Technology, Bohai Avenue 21, Tangshan, 063210, China.
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, Kehua Road 28, Suzhou, 215009, China.
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
32
|
Lin H, Zhu S, Chen Y, Lu J, Xie C, Liao C, Huang X, Li G, Wu Y, Li Z, Hu J, Lin X, Tian Y, Li Q, Wang Z, Chen S. Targeting cTRIP12 counteracts ferroptosis resistance and augments sensitivity to immunotherapy in pancreatic cancer. Drug Resist Updat 2025; 81:101240. [PMID: 40154160 DOI: 10.1016/j.drup.2025.101240] [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: 01/15/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 04/01/2025]
Abstract
AIMS Current therapeutic strategies for pancreatic ductal adenocarcinoma (PDAC) have limited efficacy in increasing patient survival rates, largely due to ferroptosis resistance and immunosuppression. The aim of this study is to identify molecular mechanisms associated with ferroptosis resistance and immunosuppression in PDAC tumour cells. METHODS Circular RNA sequencing (circRNA-seq) was performed on clinical samples to identify potential circRNAs that mediate ferroptosis resistance. C11-BODIPY staining, FerroOrange staining, the glutathione ratio, malondialdehyde quantification, and transmission electron microscopy were employed to assess ferroptosis. RNA pulldown, mass spectrometry, RNA immunoprecipitation, and coimmunoprecipitation assays were conducted to investigate the molecular mechanisms involved. A HuNSG mouse xenograft tumour model was utilized to validate therapeutic agents. RESULTS A circRNA derived from TRIP12 (cTRIP12) was identified in PDAC samples resistant to ferroptosis. cTRIP12 knockdown increased the sensitivity of PDAC cells to ferroptosis and immunotherapy. Subsequent mechanistic studies revealed that cTRIP12 specifically binds to the O-linked N-acetylglucosamine transferase (OGT) protein and increases intracellular O-GlcNAcylation levels, leading to increased protein levels of ferritin heavy chain (FTH) and PD-L1 in tumour cells. Notably, high cTRIP12 expression suppressed ferroptosis sensitivity and increased immune resistance in PDAC cells by functioning as a protein scaffold through its interaction with OGT and protein kinase R-like endoplasmic reticulum kinase (PERK). cTRIP12 inhibition induced ferroptosis in PDAC cells by reducing FTH and PD-L1 expression and synergistically increased the immunotherapy efficacy. In vivo animal experiments confirmed that the triple therapy consisting of GSK2656157, erastin, and anti-CTLA-4 effectively suppressed the progression of PDAC in tumours with high cTRIP12 expression. CONCLUSION We elucidated the molecular mechanisms underlying the simultaneous occurrence of ferroptosis resistance and immune suppression in PDAC patients. Our study provides a novel therapeutic strategy that could promote ferroptosis in tumour cells and increase immunotherapy efficacy.
Collapse
Affiliation(s)
- Hongyi Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Shuncang Zhu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Yinhao Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Jinpeng Lu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Chengke Xie
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Chengyu Liao
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China; Fuzhou University, Fuzhou 350001, China
| | - Xiaoxiao Huang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China; Fuzhou University, Fuzhou 350001, China
| | - Ge Li
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Yongding Wu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Zhiyuan Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Jianfei Hu
- Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China
| | | | - Yifeng Tian
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China; Fuzhou University, Fuzhou 350001, China
| | - Qiaowei Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Fuzhou University, Fuzhou 350001, China; Fujian Provincial Institute of Clinical Geriatrics, Fuzhou 350001, China; Fujian Key Laboratory of Geriatrics, Fuzhou 350001, China; Fujian Provincial Center for Geriatrics, Fuzhou 350001, China.
| | - Zuwei Wang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China; Fuzhou University, Fuzhou 350001, China.
| | - Shi Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China; Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou 350001, China; Fuzhou University, Fuzhou 350001, China; Fujian Provincial Institute of Clinical Geriatrics, Fuzhou 350001, China; Fujian Key Laboratory of Geriatrics, Fuzhou 350001, China; Fujian Provincial Center for Geriatrics, Fuzhou 350001, China.
| |
Collapse
|
33
|
Chi X, Wang SH, Gao J, Su J, Du YZ, Xu XL. Carrier-Free Nanocombo-Sensitized Photoimmunotherapy via Activation of α2-Adrenergic Receptors. ACS APPLIED MATERIALS & INTERFACES 2025; 17:16437-16452. [PMID: 40040324 PMCID: PMC11931486 DOI: 10.1021/acsami.4c18052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 12/10/2024] [Accepted: 12/10/2024] [Indexed: 03/06/2025]
Abstract
Photodynamic therapy (PDT)-based photoimmunotherapy has attracted increasing attention in the field of cancer immunotherapy. Nonetheless, monotherapy alone proves insufficient in eliciting robust and enduring tumor immunogenicity within the "cold" microenvironment of triple-negative breast cancer. Therefore, it is imperative to integrate phototherapy and immunostimulation strategies to achieve synergistic effects. Here, we developed a carrier-free nanocombo comprising a photosensitizer (chlorin e6, Ce6) and an α2-adrenergic receptor (α2-AR) agonist (guanfacine, GFC) to enhance photoimmunotherapy through α2-AR activation. Ce6 and GFC possessed the ability to self-assemble into spherical nanoparticles, with the resulting Ce6-GFC (CeG) exhibiting exceptional drug loading efficiency (approaching 100%) and long-lasting colloidal stability, along with effective in vivo tumor-targeting capabilities. Following near-infrared laser irradiation, CeG-mediated phototherapy instigated a rapid generation of reactive oxygen species, leading to membrane disruption and the release of tumor-associated antigens, thereby facilitating dendritic cell maturation. Furthermore, α2-AR agonists served to repolarize M2 tumor-associated macrophages toward the M1 phenotype via adenylyl cyclase-mediated activation of α2-AR, thereby promoting the recruitment and activation of cytotoxic T lymphocytes. As a result, the carrier-free nanocombo significantly enhanced the efficacy of photoimmunotherapy in combatting poorly immunogenic breast tumors in female mice. Our findings showcase a "killing two birds with one stone" approach that boosts tumor immunogenicity, mitigates tumor immunosuppression, and advances the field of photoimmunotherapy.
Collapse
Affiliation(s)
- XiaoKai Chi
- Shulan International
Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
- College of
Pharmacy, Jiamusi University, Jiamusi 154007, PR China
- Institute
of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Si-Hui Wang
- Shulan International
Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - JingJing Gao
- Shulan International
Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - Jin Su
- College of
Pharmacy, Jiamusi University, Jiamusi 154007, PR China
| | - Yong-Zhong Du
- Institute
of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Xiao-Ling Xu
- Shulan International
Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| |
Collapse
|
34
|
Zhang X, Chen Y, Liu X, Li G, Zhang S, Zhang Q, Cui Z, Qin M, Simon HU, Terzić J, Kocic G, Polić B, Yin C, Li X, Zheng T, Liu B, Zhu Y. STING in cancer immunoediting: Modeling tumor-immune dynamics throughout cancer development. Cancer Lett 2025; 612:217410. [PMID: 39826670 DOI: 10.1016/j.canlet.2024.217410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/16/2024] [Accepted: 12/21/2024] [Indexed: 01/22/2025]
Abstract
Cancer immunoediting is a dynamic process of tumor-immune system interaction that plays a critical role in cancer development and progression. Recent studies have highlighted the importance of innate signaling pathways possessed by both cancer cells and immune cells in this process. The STING molecule, a pivotal innate immune signaling molecule, mediates DNA-triggered immune responses in both cancer cells and immune cells, modulating the anti-tumor immune response and shaping the efficacy of immunotherapy. Emerging evidence has shown that the activation of STING signaling has dual opposing effects in cancer progression, simultaneously provoking and restricting anti-tumor immunity, and participating in every phase of cancer immunoediting, including immune elimination, equilibrium, and escape. In this review, we elucidate the roles of STING in the process of cancer immunoediting and discuss the dichotomous effects of STING agonists in the cancer immunotherapy response or resistance. A profound understanding of the sophisticated roles of STING signaling pathway in cancer immunoediting would potentially inspire the development of novel cancer therapeutic approaches and overcome the undesirable protumor effects of STING activation.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China; Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Yan Chen
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Xi Liu
- Department of Cardiology, ordos central hospital, Ordos, People's Republic of China
| | - Guoli Li
- Department of Colorectal and Anal Surgery, Chifeng Municipal Hospital, Chifeng Clinical Medical School of Inner Mongolia Medical University, Chifeng, People's Republic of China
| | - Shuo Zhang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China
| | - Qi Zhang
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Zihan Cui
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Minglu Qin
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland; Institute of Biochemistry, Brandenburg Medical School, Neuruppin, 16816, Germany
| | - Janoš Terzić
- Laboratory for Cancer Research, University of Split School of Medicine, Split, Croatia
| | - Gordana Kocic
- Department of Biochemistry, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
| | - Bojan Polić
- University of Rijeka Faculty of Medicine, Croatia
| | - Chengliang Yin
- Faculty of Medicine, Macau University of Science and Technology, 999078, Macao.
| | - Xiaobo Li
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China.
| | - Tongsen Zheng
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No.150 Haping Road, Nangang District, Harbin, Heilongjiang, People's Republic of China.
| | - Bing Liu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China; School of Stomatology, Harbin Medical University, Harbin, 150001, People's Republic of China.
| | - Yuanyuan Zhu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China; Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China.
| |
Collapse
|
35
|
Guo D, Cai S, Deng L, Xu W, Fu S, Lin Y, Jiang T, Li Q, Shen Z, Zhang J, Luo P, Tang B, Wang L. Ferroptosis in Pulmonary Disease and Lung Cancer: Molecular Mechanisms, Crosstalk Regulation, and Therapeutic Strategies. MedComm (Beijing) 2025; 6:e70116. [PMID: 39991627 PMCID: PMC11847630 DOI: 10.1002/mco2.70116] [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: 08/23/2024] [Revised: 01/12/2025] [Accepted: 01/15/2025] [Indexed: 02/25/2025] Open
Abstract
Ferroptosis is a distinct form of iron-dependent programmed cell death characterized primarily by intracellular iron accumulation and lipid peroxidation. Multiple cellular processes, including amino acid metabolism, iron metabolism, lipid metabolism, various signaling pathways, and autophagy, have been demonstrated to influence the induction and progression of ferroptosis. Recent investigations have elucidated that ferroptosis plays a crucial role in the pathogenesis of various pulmonary disorders, including lung injury, chronic obstructive pulmonary disease, pulmonary fibrosis, and asthma. Ferroptosis is increasingly recognized as a promising novel strategy for cancer treatment. Various immune cells within the tumor microenvironment, including CD8+ T cells, macrophages, regulatory T cells, natural killer cells, and dendritic cells, have been shown to induce ferroptosis in tumor cells and modulate the process through the regulation of iron and lipid metabolism pathways. Conversely, ferroptosis can reciprocally alter the metabolic environment, leading to the activation or inhibition of immune cell functions, thereby modulating immune responses. This paper reviews the molecular mechanism of ferroptosis and describes the tumor immune microenvironment, discusses the connection between ferroptosis and the tumor microenvironment in lung cancer and pulmonary diseases, and discusses the development prospect of their interaction in the treatment of lung cancer and pulmonary diseases.
Collapse
Affiliation(s)
- Dandan Guo
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Songhua Cai
- Department of Thoracic SurgeryNational Cancer CenterNational Clinical Research Center for CancerCancer Hospital & Shenzhen HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenGuangdongChina
| | - Lvdan Deng
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Wangting Xu
- Department of RespiratoryFirst Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Sentao Fu
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Yaling Lin
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Tong Jiang
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Qing Li
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Zhijun Shen
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Jian Zhang
- The Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Peng Luo
- The Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Bufu Tang
- Department of Radiation OncologyZhongshan HospitalFudan UniversityShanghaiShanghaiChina
| | - Ling Wang
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| |
Collapse
|
36
|
Lu J, Ding F, Sun Y, Zhao Y, Ma W, Zhang H, Shi B. Unveiling the role of MDH1 in breast cancer drug resistance through single-cell sequencing and schottenol intervention. Cell Signal 2025; 127:111608. [PMID: 39818404 DOI: 10.1016/j.cellsig.2025.111608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 12/29/2024] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
This study utilizes single-cell RNA sequencing data to reveal the transcriptomic characteristics of breast cancer and normal epithelial cells. Nine significant cell populations were identified through stringent quality control and batch effect correction. Further classification of breast cancer epithelial cells based on the PAM50 method and clinical subtypes highlighted significant heterogeneity between triple-negative breast cancer (TNBC) and non-triple-negative breast cancer (NTNBC). The study also analyzed myeloid cells and tumor-infiltrating lymphocytes (TILs) within the breast cancer immune microenvironment, identifying 14 TIL subpopulations and assessing their proportion variations across different patients. The CellChat tool revealed a complex cellular communication network within the tumor microenvironment, showing notable differences in communication intensity and patterns between TNBC and NTNBC patients. Additionally, the key regulatory role of the senescence-associated gene MDH1 in breast cancer was confirmed, and its impact on drug sensitivity was explored. Finally, it was discovered that the phytosterol Schottenol inhibits breast cancer cell proliferation by downregulating MDH1 expression and enhances sensitivity to paclitaxel. These findings provide new insights into MDH1 as a therapeutic target and suggest Schottenol as a potential strategy to overcome breast cancer drug resistance.
Collapse
Affiliation(s)
- Jian Lu
- Cheeloo College of Medicine, Shandong University, Jinan 250000, Shandong, China.; Department of Breast Diseases (II), Shandong Second Provincial General Hospital, Jinan 250000, Shandong, China
| | - Feng Ding
- Department of Breast Diseases (II), Shandong Second Provincial General Hospital, Jinan 250000, Shandong, China
| | - Yongjie Sun
- Department of Breast Diseases (II), Shandong Second Provincial General Hospital, Jinan 250000, Shandong, China
| | - Yu Zhao
- Department of Stomatology, Shandong Second Provincial General Hospital, Jinan 250000, Shandong, China
| | - Wenbiao Ma
- Department of Breast and Thyroid Surgery, The Qinghai Provincial People's Hospital, Xining 810007, China
| | - Huan Zhang
- Department of Anesthesiology, The Qinghai Provincial People's Hospital, Xining 810007, China
| | - Bo Shi
- Department of Breast and Thyroid Surgery, The Qinghai Provincial People's Hospital, Xining 810007, China.
| |
Collapse
|
37
|
Alam MR, Akinyemi AO, Wang J, Howlader M, Farahani ME, Nur M, Zhang M, Gu L, Li Z. CD4 +CD8 + double-positive T cells in immune disorders and cancer: Prospects and hurdles in immunotherapy. Autoimmun Rev 2025; 24:103757. [PMID: 39855286 DOI: 10.1016/j.autrev.2025.103757] [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: 08/21/2024] [Revised: 01/10/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
CD4+ and CD8+ T cells play critical roles in both innate and adaptive immune responses, managing and modulating cellular immunity during immune diseases and cancer. Their well-established functions have led to significant clinical benefits. CD4+CD8+ double-positive (DP) T cells, a subset of the T cell population, have been identified in the blood and peripheral lymphoid tissues across various species. They have gained interest due to their involvement in immune disorders, inflammation, and cancer. Although mature DP T cells are present in healthy individuals and contribute to disease contexts, their molecular characteristics and pathophysiological roles remain debated. Notably, the number of DP T cells in the blood is higher in older adults compared to younger individuals, and these cells can stimulate inflammation and viral infections through increased secretion of interleukin (IL)-10, interferon gamma (IFN-γ), and transforming growth factor beta (TGF-β). In cancer, DP T cells have been observed to infiltrate cutaneous T cell lymphomas and are found in greater numbers in nodular lymphocyte predominant Hodgkin lymphoma, melanoma, hepatocellular carcinoma, and breast cancer. The higher prevalence of DP T cells in advanced cancers, coupled with their strong lytic activity and distinct cytokine profile, suggests that these cells may play a crucial role in modulating immune responses to cancer. This insight offers a potential new approach for enhancing the identification and selection of antigen-reactive T cells in immune-based treatments. This review provides a comprehensive overview of the origin, distribution, transcriptional regulation during developmental stages, and functions of DP T cells. A deeper understanding of the diversity and roles of DP T cells may pave the way for their development as a promising tool for immunotherapy in the management of immune disorders and metastatic cancers.
Collapse
Affiliation(s)
- Md Rakibul Alam
- Department of Toxicology and Cancer Biology, Collage of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Amos Olalekan Akinyemi
- Department of Toxicology and Cancer Biology, Collage of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Jianlin Wang
- Department of Toxicology and Cancer Biology, Collage of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Mithu Howlader
- Department of Toxicology and Cancer Biology, Collage of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Mohammad Esfini Farahani
- Department of Toxicology and Cancer Biology, Collage of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Maria Nur
- Department of Toxicology and Cancer Biology, Collage of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Min Zhang
- Department of Toxicology and Cancer Biology, Collage of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Lixiang Gu
- Department of Toxicology and Cancer Biology, Collage of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Zhiguo Li
- Department of Toxicology and Cancer Biology, Collage of Medicine, University of Kentucky, Lexington, KY 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
38
|
Du F, Wang G, Dai Q, Huang J, Li J, Liu C, Du K, Tian H, Deng Q, Xie L, Zhao X, Zhang Q, Yang L, Li Y, Wu Z, Zhang Z. Targeting novel regulated cell death: disulfidptosis in cancer immunotherapy with immune checkpoint inhibitors. Biomark Res 2025; 13:35. [PMID: 40012016 DOI: 10.1186/s40364-025-00748-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
The battle against cancer has evolved over centuries, from the early stages of surgical resection to contemporary treatments including chemotherapy, radiation, targeted therapies, and immunotherapies. Despite significant advances in cancer treatment over recent decades, these therapies remain limited by various challenges. Immune checkpoint inhibitors (ICIs), a cornerstone of tumor immunotherapy, have emerged as one of the most promising advancements in cancer treatment. Although ICIs, such as CTLA-4 and PD-1/PD-L1 inhibitors, have demonstrated clinical efficacy, their therapeutic impact remains suboptimal due to patient-specific variability and tumor immune resistance. Cell death is a fundamental process for maintaining tissue homeostasis and function. Recent research highlights that the combination of induced regulatory cell death (RCD) and ICIs can substantially enhance anti-tumor responses across multiple cancer types. In cells exhibiting high levels of recombinant solute carrier family 7 member 11 (SLC7A11) protein, glucose deprivation triggers a programmed cell death (PCD) pathway characterized by disulfide bond formation and REDOX (reduction-oxidation) reactions, termed "disulfidptosis." Studies suggest that disulfidptosis plays a critical role in the therapeutic efficacy of SLC7A11high cancers. Therefore, to investigate the potential synergy between disulfidptosis and ICIs, this study will explore the mechanisms of both processes in tumor progression, with the goal of enhancing the anti-tumor immune response of ICIs by targeting the intracellular disulfidptosis pathway.
Collapse
Affiliation(s)
- Fei Du
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Guojun Wang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qian Dai
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Jiang Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Junxin Li
- Department of pharmacy, Zigong Fourth People's Hospital, Zigong, 643000, China
| | - Congxing Liu
- Department of Pharmacy, Chengfei Hospital, Chengdu, 610000, China
| | - Ke Du
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pediatrics, Luzhou Maternal and Child Health Hospital, Luzhou Second People's Hospital, Luzhou, 646000, Sichuan, China
| | - Hua Tian
- School of Nursing, Chongqing College of Humanities, Science & Technology, Chongqing, 401520, China
| | - Qiwei Deng
- Heruida Pharmaceutical Co.,ltd, Haikou, Hainan, 570100, China
| | - Longxiang Xie
- The TCM Hospital of Longquanyi District, Chengdu, 610100, Sichuan, China
| | - Xin Zhao
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qimin Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Lan Yang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Yaling Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhigui Wu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhuo Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
39
|
Liu M, Li L, Cao L, Li W, Gu X, Yang M, Wu D, Li Y, Deng Y, Zhang J, Yang C, Liang Q, Liu H, Rong P, Ma X, Wang W. Targeted delivery of CCL3 reprograms macrophage antigen presentation and enhances the efficacy of immune checkpoint blockade therapy in hepatocellular carcinoma. J Immunother Cancer 2025; 13:e010947. [PMID: 39988347 PMCID: PMC11848677 DOI: 10.1136/jitc-2024-010947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 02/05/2025] [Indexed: 02/25/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) remains a leading cause of cancer-related deaths worldwide, especially in advanced stages where limited treatment options result in poor prognosis. The immunosuppressive tumor immune microenvironment (TIME), characterized by low immune cell infiltration and exhaustion, limits immunotherapy efficacy. To address this, our study investigates the role of C-C motif chemokine ligand 3 (CCL3) in modulating the HCC TIME. METHODS We analyzed CCL3 expression in human HCC samples from The Cancer Genome Atlas database, focusing on its correlation with inflammatory gene signatures and immune cell infiltration. High-dimensional single-cell RNA sequencing (scRNA-seq), flow cytometry, and multiplex immunofluorescence were used to investigate CCL3's effects on macrophage function and T cell activation. The biological impact of CCL3 on macrophages was assessed using co-culture systems, confocal imaging, metabolite detection, and inhibition assays. Preclinical HCC models and ex vivo tumor fragment assays further explored how CCL3 modulates immune responses and enhances immune checkpoint blockade efficacy. RESULTS Our study shows that CCL3 is suppressed in the tumor microenvironment and positively correlates with immune infiltration and inflammatory responses. Targeted liver delivery of rAAV-Ccl3 reprograms the immune microenvironment in HCC, promoting immune cell recruitment and tertiary lymphoid structure formation, thus suppressing tumor growth via immune engagement. Through scRNA-seq, flow cytometry, and multiplex immunofluorescence, we found that CCL3 enhances macrophage antigen uptake and activates cytotoxic T cells. In vivo and in vitro experiments confirmed that CCL3 facilitates T cell infiltration and upregulates MHC II expression on macrophages, enhancing antigen presentation. The CCL3-CCR5 pathway also boosts macrophage metabolism, increasing lysosomal activity and antigen uptake, thereby strengthening adaptive immune responses and increasing sensitivity to immune checkpoint blockade therapies in preclinical models. CONCLUSIONS This study highlights the pivotal role of CCL3 in reshaping the TIME and enhancing antitumor immunity in HCC. By promoting immune cell recruitment and enhancing antigen presentation, CCL3 demonstrates significant potential to improve the efficacy of immunotherapy, particularly in combination with immune checkpoint inhibitors. Targeting CCL3 may help to overcome the immunosuppressive TIME in HCC and improve patient outcomes.
Collapse
Affiliation(s)
- Muqi Liu
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Linzhe Li
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Lu Cao
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Wei Li
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Xingshi Gu
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Min Yang
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Di Wu
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Yanan Li
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Yao Deng
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Juan Zhang
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Cejun Yang
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Qi Liang
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Huaping Liu
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Pengfei Rong
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Xiaoqian Ma
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Wei Wang
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| |
Collapse
|
40
|
Wang S, Wang R, Zhang A, Duan X, Shang X, Jiang H. Causal relationship between circulating immune cells and the risk of esophageal cancer: a Mendelian randomization study. Discov Oncol 2025; 16:220. [PMID: 39982541 PMCID: PMC11845655 DOI: 10.1007/s12672-025-01921-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 02/04/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Although relevant research has unveiled the intricate connections between immune cells and the occurrence and prognosis of esophageal cancer (EC), the specific impact of immune cell phenotypes on EC remains unclear. METHODS We employed bidirectional two-sample Mendelian Randomization (MR) analysis to explore the causal relationship between immune cell phenotypes and EC. The summary data for immune cell phenotypes and EC are both sourced from the GWAS (Genome-Wide Association Study) database. Sensitivity analysis was conducted on the results, utilizing a combination of MR-Egger and MR-Presso to assess horizontal pleiotropy, employing Cochran's Q test to evaluate heterogeneity. RESULTS We identified 24 immunophenotypes with potential causal relationships to EC. Our results are presented based on the panel results from flow cytometry detection, categorized into B-cell panel, TBNK panel, cDC panel, Maturation stages of T-cell panel, Monocyte panel, and Myeloid cell panel. In the reverse MR analysis, we found a potential negative correlation between EC and IgD + CD38dim B cell Absolute Count (OR = 0.94, 95% CI, 0.88-0.99, P = 0.023). CONCLUSION This study has unveiled the causal relationship between immune cell phenotypes and EC, providing new insights for the exploration of immunotherapy targets in subsequent EC research and for the assessment of EC prognosis.
Collapse
Affiliation(s)
- Shangren Wang
- Department of Minimally Invasive Esophageal Surgery, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tiyuanbei, Huanhuxi Rd., Hexi District, Tianjin, 300060, China
| | - Ruizhen Wang
- Department of Minimally Invasive Esophageal Surgery, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tiyuanbei, Huanhuxi Rd., Hexi District, Tianjin, 300060, China
| | - Aiqiao Zhang
- Department of Pediatrics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xiaofeng Duan
- Department of Minimally Invasive Esophageal Surgery, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tiyuanbei, Huanhuxi Rd., Hexi District, Tianjin, 300060, China
| | - Xiaobin Shang
- Department of Minimally Invasive Esophageal Surgery, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tiyuanbei, Huanhuxi Rd., Hexi District, Tianjin, 300060, China
| | - Hongjing Jiang
- Department of Minimally Invasive Esophageal Surgery, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tiyuanbei, Huanhuxi Rd., Hexi District, Tianjin, 300060, China.
| |
Collapse
|
41
|
Liu J, Wei T, Quan L, Dou M, Yue J, Yuan P. Identification and multi-dimensional validation of mitochondrial permeability transition-driven necrosis-related model to assess the prognosis and immunotherapy value in breast cancer. Eur J Med Res 2025; 30:113. [PMID: 39966932 PMCID: PMC11834492 DOI: 10.1186/s40001-025-02370-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 02/08/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Breast cancer is a highly prevalent tumor worldwide. Mitochondrial permeability transition (MPT)-driven necrosis is a novel type of cell death induced by mitochondrial membrane disruption. The roles of MPT-driven necrosis in breast cancer remain unclear. METHODS Gene expression and clinicopathologic features were extracted from The Cancer Genome Atlas and Gene Expression Omnibus. We performed a genome landscape analysis of MPT-driven necrosis (MPTdn)-related genes, and a consensus clustering analysis was conducted to construct MPTdn clusters. Next, a risk model was established based on the differentially expressed genes related to MPTdn. We grouped and used external data sets to verify the stability of the model. Subsequently, immune correlation analysis, clinical correlation assessment and drug sensitivity analysis were conducted. Finally, candidate genes were validated in the protein and mRNA levels. RESULTS A total of 39 MPTdn-related genes were identified in our analysis. Most MPTdn-related genes had different expression levels and somatic mutations in breast cancer, and a close interaction was noted among them. A risk model composed of BCL2A1, SCUBE2, NPY1R and CLIC6 was constructed. The low-risk group had better overall survival and higher immune infiltration levels. All three external data sets achieved excellent predictive efficacy. Finally, the immunohistochemistry results indicated that BCL2A1, SCUBE2, NPY1R and CLIC6 were expressed at significantly lower levels in breast cancer tissues, and the transcriptome sequencing results revealed that BCL2A1 and SCUBE2 mRNA expression levels were greater in the nonrecurrence group. CONCLUSIONS We developed a risk model with excellent predictive efficacy based on MPTdn and revealed that BCL2A1, SCUBE2, NPY1R and CLIC6 could be used as the biomarkers, laying a solid foundation for investigations of therapeutic targets of breast cancer.
Collapse
Affiliation(s)
- Jinsong Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tong Wei
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Liuliu Quan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Min Dou
- Bengbu Medical University, BengBu, 233030, China
| | - Jian Yue
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peng Yuan
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
42
|
Wang H, Liu X, Huang H, Tang M, Li J, Huang T, Wang S. Multi-omics analysis identifies UBA family as potential pan-cancer biomarkers for tumor prognosis and immune microenvironment infiltration. Front Immunol 2025; 16:1510503. [PMID: 40046044 PMCID: PMC11880792 DOI: 10.3389/fimmu.2025.1510503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 01/30/2025] [Indexed: 03/17/2025] Open
Abstract
Background UBA1 and UBA6 are classic ubiquitin-activating E1 enzymes, which participate in the ubiquitination degradation of intracellular proteins and are closely related to the occurrence and development of various diseases and tumors. However, at present, comprehensive analysis has not been used to study the role of UBA family in cancers. Methods We extracted the relevant data of cancer patients from the TCGA database and studied the relationship between the expression patterns of UBA family and the survival rate, and stage of patients in pan-cancer, especially breast cancer (BRCA), colorectal cancer (COAD), renal cancer (KIRC) and lung adenocarcinoma (LUAD). In addition, we also evaluated their impact on immune infiltration using TISIDB database and R packages. Results UBA1 and UBA6 are highly expressed in most cancer types, which may be associated with poor prognosis of patients. This study also investigated their expression had a closely tie with clinical stages in some specific tumors. Furthermore, this study also demonstrated that these genes were closely related to immune score, immune subtypes and tumor infiltrating immune cells. Conclusions Our study demonstrated that the differential expression of the UBA family, along with their associated survival landscape and immune infiltration across various cancer types, holds potential as biomarkers linked to cancer immune infiltration. This finding offers a novel perspective for informing the direction of cancer treatment strategies.
Collapse
Affiliation(s)
- Haibin Wang
- Department of Gastrointestinal Oncology Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - XinLi Liu
- Department of Medical Oncology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Hesen Huang
- Department of Otolaryngology-Head and Neck Surgery, Xiang’an Hospital of Xiamen University, Fujian, Xiamen, China
| | - Meng Tang
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Jiwei Li
- Department of Respiratory, Critical Care and Sleep Medicine Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Tingting Huang
- Department of Medical Oncology, Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen, China
| | - Shengjie Wang
- Department of Thyroid and Breast Surgery, Xiamen Humanity Hospital Fujian Medical University, Xiamen, Fujian, China
| |
Collapse
|
43
|
Wu C, Fang S, Wu L, Mi Z, Yin Y, Liao Y, Zhao Y, Wang T, Na J. Identification of the entosis-related prognostic signature and tumour microenvironment in hepatocellular carcinoma on the basis of bioinformatics analysis and experimental validation. Clin Exp Med 2025; 25:55. [PMID: 39937284 PMCID: PMC11821697 DOI: 10.1007/s10238-025-01580-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 01/28/2025] [Indexed: 02/13/2025]
Abstract
Liver cancer ranks among the deadliest cancers worldwide. Entosis, a recently uncovered method of cell death, has not yet been fully explored for its relevance to HCC. A bioinformatics analysis was performed to determine the expression and mutational landscapes of Entosis-related genes (ERGs). A subset of differentially expressed Entosis-related genes (DEERGs) was generated. A risk model for entosis was subsequently constructed employing LASSO and Cox regression methodologies. The correlations among ERGs, genes associated with risk, the developed risk model, and the immune context of the tumour were explored. Furthermore, the study investigated the varying drug sensitivities between high-risk and slight-risk patient groups. The expression patterns of four pivotal risk genes were delineated via qRT‒PCR and WB. A prognostic model comprising four DEERGs (KIF18A, SPP1, LCAT and TRIB3) was developed. The ability of this model to predict the survival outcomes of patients with HCC was confirmed through receiver operating characteristic curve analysis. Patients were grouped according to their risk assessments, revealing that the low-risk population demonstrated a more favourable survival outcome than did the high-risk population. The high-risk population presented reduced tumour stroma, immune and ESTIMATE scores, along with an increased proportion of cancer stem cells and tumour mutation burden. Additionally, a connection between the risk model and the responsiveness of various chemotherapy drugs as well as the efficacy of immunotherapies in patients was noted. These findings provide significant guidance for the development of targeted clinical treatment strategies. qRT‒PCR and WB analysis revealed that the gene expression of KIF18A and SPP1 were elevated in HCCLM3 cells compared with that in THLE2 cells; whereas, the expression level of LCAT and TIRB3 was decreased. The four genes KIF18A, SPP1, LCAT and TRIB3 could effectively predict the survival prognosis of patients with liver cancer. KIF18A and SPP1 were elevated in HCC tissues compared with that in THLE2 cells.
Collapse
Affiliation(s)
- Chen Wu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Shixu Fang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Liangliang Wu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Zhengcheng Mi
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yao Yin
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yuan Liao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yongxiang Zhao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Tinghua Wang
- Laboratory Animal Department, Kunming Medical University, Kunming, 650031, Yunnan, China.
| | - Jintong Na
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| |
Collapse
|
44
|
Li C, Cheng D, Li P. Androgen receptor dynamics in prostate cancer: from disease progression to treatment resistance. Front Oncol 2025; 15:1542811. [PMID: 40008000 PMCID: PMC11850250 DOI: 10.3389/fonc.2025.1542811] [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: 12/10/2024] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
Prostate cancer is the most common cancer among men worldwide, especially in those over 65, and is a leading cause of cancer-related mortality. The disease typically advances from an androgen-dependent state to castration-resistant prostate cancer (CRPC), which poses significant treatment challenges. The androgen receptor (AR) on the X chromosome is a central driver in this process, activating genes that govern proliferation and survival. Mutations and amplifications of the AR are closely associated with disease progression and treatment resistance. While traditional therapies such as androgen deprivation therapy (ADT) and AR antagonists like enzalutamide have been effective, resistance persists due to reactivation of AR signaling through mechanisms like ligand-independent activation. Recent research highlights the role of epigenetic modifications in enhancing AR activity and drug resistance. The tumor microenvironment, particularly interactions with cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs), further complicates treatment by promoting aggressive tumor behavior and immune evasion. Future directions include developing next-generation AR antagonists, identifying AR-related biomarkers for personalized therapy, and exploring combinations with immune checkpoint inhibitors. Additionally, basal cell-lumen-derived organoids provide innovative models that can enhance understanding and treatment strategies in prostate cancer.
Collapse
Affiliation(s)
| | | | - Peng Li
- Center for Reproductive Medicine, Shenyang Jinghua Hospital, Shenyang, China
| |
Collapse
|
45
|
Cao X, Luo N, Liu X, Guo K, Deng M, Lv C. Crosstalk of SPINK4 Expression With Patient Mortality, Immunotherapy and Metastasis in Pan-Cancer Based on Integrated Multi-Omics Analyses. Onco Targets Ther 2025; 18:161-177. [PMID: 39926372 PMCID: PMC11806753 DOI: 10.2147/ott.s487126] [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: 08/27/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025] Open
Abstract
Background Cancer remains a major global health challenge, with early detection and prompt treatment being crucial for reducing mortality rates. The SPINK4 has been linked to the development of several tumors, and there is growing evidence of its involvement. However, its specific functions and effects in different cancer types remain unclear. Methods The association between SPINK4 expression levels and tumor progression was investigated and confirmed using the TCGA dataset. Kaplan-Meier curves were utilized to examine the correlation between SPINK4 expression with survival outcomes in pan-cancer patients. The Pearson method was employed to investigate the association of SPINK4 expression with the tumor microenvironment, stemness score, immunoinfiltrating subtype, and chemotherapy sensitivity in human different cancer types. Wound healing and Transwell assays were performed to confirm the roles of the model gene in colon adenocarcinoma cells. Results The expression of SPINK4 shows heterogeneity across pan-cancer tissues, and is closely associated with poor prognosis, immune cell invasion, tumor cell resistance, and tumor metastasis in a various human cancer. Mutation of SPINK4 hold significant predictive value for poor prognosis of pan-cancer patients. In addition, SPINK4 expression was significantly correlated with the tumor microenvironment (stromal cells and immune cells) and stemness score (DNAss and RNAss) in human pan-cancer tissues, particularly in BLCA and COAD. Single-cell sequencing analysis showed that SPINK4 is mainly expressed in endothelial cells in BLCA and in malignant cells in COAD. Drug resistance analysis showed a significant association between SPINK4 expression and sensitivity to several cancer chemotherapy drugs. Importantly, overexpression of SPINK4 promoted the metastasis of colon cancer cell lines (HCT116 and RKO), whereas SPINK4 knockout markedly inhibited their metastasis. Conclusion These findings reveal the crucial role of SPINK4 in the pan-cancer process and may have significant implications for the diagnosis and treatment of cancer in the future.
Collapse
Affiliation(s)
- Xiuhua Cao
- Center for Basic Medical Research, Southwest Medical University, Luzhou, People’s Republic of China
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Na Luo
- Center for Basic Medical Research, Southwest Medical University, Luzhou, People’s Republic of China
| | - Xiaoyan Liu
- Center for Basic Medical Research, Southwest Medical University, Luzhou, People’s Republic of China
| | - Kan Guo
- Center for Basic Medical Research, Southwest Medical University, Luzhou, People’s Republic of China
| | - Mingming Deng
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Chaoxiang Lv
- Center for Basic Medical Research, Southwest Medical University, Luzhou, People’s Republic of China
| |
Collapse
|
46
|
Qi L, Wang J, Hou S, Liu S, Zhang Q, Zhu S, Liu S, Zhang S. Unraveling the tumor microenvironment of esophageal squamous cell carcinoma through single-cell sequencing: A comprehensive review. Biochim Biophys Acta Rev Cancer 2025; 1880:189264. [PMID: 39805342 DOI: 10.1016/j.bbcan.2025.189264] [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: 08/27/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/16/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a highly heterogeneous and aggressive malignancy. The progression, invasiveness, and metastatic potential of ESCC are shaped by a multitude of cells within the tumor microenvironment (TME), including tumor cells, immune cells, endothelial cells, as well as fibroblasts and other cell types. Recent advancements in single-cell sequencing technologies have significantly enhanced our comprehension of the diverse landscape of ESCC. Single-cell multi-omics technology, particularly single-cell transcriptome sequencing, have shed light on the expression profiles of individual cells and the molecular characteristics of distinct tumor cell populations. This review summarizes the latest literature on single-cell research in the field of ESCC, aiming to elucidate the heterogeneity of tumor cells, immune cells, and stromal cells at the single-cell level. Furthermore, it explores the impact of cellular interactions within the TME on the progression of ESCC. By compiling a comprehensive overview of single-cell omics research on ESCC, this article aims to enhance our understanding of ESCC diagnosis and treatment by elucidating the intricate interplay within the TME. It explores the cellular composition, spatial arrangement, and functional attributes of the ESCC TME, offering potential therapeutic targets and biomarkers for personalized treatment strategies.
Collapse
Affiliation(s)
- Lingyu Qi
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Jiaxin Wang
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Songyuan Hou
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Siying Liu
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Qian Zhang
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Shengtao Zhu
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China
| | - Si Liu
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China.
| | - Shutian Zhang
- State Key Laboratory of Digestive healthy, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing 100050, PR China.
| |
Collapse
|
47
|
Chang TH, Ho PC. Interferon-driven Metabolic Reprogramming and Tumor Microenvironment Remodeling. Immune Netw 2025; 25:e8. [PMID: 40078784 PMCID: PMC11896656 DOI: 10.4110/in.2025.25.e8] [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: 12/24/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 03/14/2025] Open
Abstract
IFNs play a critical role in cancer biology, including impacting tumor cell behavior and instructing the tumor microenvironment (TME). IFNs recently have been shown to reprogram tumor metabolism through distinct mechanisms. Furthermore, IFNs shape the TME by modulating immune cell infiltration and function, contributing to the intricate interaction between the tumor and stromal cells. This review summarizes the effects of IFNs on metabolic reprogramming and their impacts on the function of immune cells within the TME, with a particular focus on the dual roles of IFNs in mediating both anti-tumor and pro-tumor immune responses. Understanding the significance of IFNs-mediated processes aids to advise future therapeutic strategies in cancer treatment.
Collapse
Affiliation(s)
- Tzu-Hsuan Chang
- Department of Fundamental Oncology, University of Lausanne, 1015 Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, 1015 Lausanne, Switzerland
| | - Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, 1015 Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, 1015 Lausanne, Switzerland
| |
Collapse
|
48
|
Leven AS, Wagner N, Nienaber S, Messiha D, Tasdogan A, Ugurel S. Changes in tumor and cardiac metabolism upon immune checkpoint. Basic Res Cardiol 2025; 120:133-152. [PMID: 39658699 PMCID: PMC11790718 DOI: 10.1007/s00395-024-01092-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/06/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024]
Abstract
Cardiovascular disease and cancer are the leading causes of death in the Western world. The associated risk factors are increased by smoking, hypertension, diabetes, sedentary lifestyle, aging, unbalanced diet, and alcohol consumption. Therefore, the study of cellular metabolism has become of increasing importance, with current research focusing on the alterations and adjustments of the metabolism of cancer patients. This may also affect the efficacy and tolerability of anti-cancer therapies such as immune-checkpoint inhibition (ICI). This review will focus on metabolic adaptations and their consequences for various cell types, including cancer cells, cardiac myocytes, and immune cells. Focusing on ICI, we illustrate how anti-cancer therapies interact with metabolism. In addition to the desired tumor response, we highlight that ICI can also lead to a variety of side effects that may impact metabolism or vice versa. With regard to the cardiovascular system, ICI-induced cardiotoxicity is increasingly recognized as one of the most life-threatening adverse events with a mortality of up to 50%. As such, significant efforts are being made to assess the specific interactions and associated metabolic changes associated with ICIs to improve both efficacy and management of side effects.
Collapse
Affiliation(s)
- Anna-Sophia Leven
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| | - Natalie Wagner
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Stephan Nienaber
- Clinic III for Internal Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Daniel Messiha
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Centre, University of Duisburg-Essen, Essen, Germany
| | - Alpaslan Tasdogan
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
- National Center for Tumor Diseases (NCT)-West, Campus Essen, and Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
- National Center for Tumor Diseases (NCT)-West, Campus Essen, and Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
49
|
Ni G, Sun Y, Jia H, Xiahou Z, Li Y, Zhao F, Zang H. MAZ-mediated tumor progression and immune evasion in hormone receptor-positive breast cancer: Targeting tumor microenvironment and PCLAF+ subtype-specific therapy. Transl Oncol 2025; 52:102280. [PMID: 39805182 PMCID: PMC11780959 DOI: 10.1016/j.tranon.2025.102280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/19/2024] [Accepted: 01/05/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Breast cancer had been the most frequently diagnosed cancer among women, making up nearly one-third of all female cancers. Hormone receptor-positive breast cancer (HR+BC) was the most prevalent subtype of breast cancer and exhibited significant heterogeneity. Despite advancements in endocrine therapies, patients with advanced HR+BC often faced poor outcomes due to the development of resistance to treatment. Understanding the molecular mechanisms behind this resistance, including tumor heterogeneity and changes in the tumor microenvironment, was crucial for overcoming resistance, identifying new therapeutic targets, and developing more effective personalized treatments. METHODS The study utilized single-cell RNA sequencing (scRNA-seq) data sourced from the Gene Expression Omnibus database and The Cancer Genome Atlas to analyze HR+BC and identify key cellular characteristics. Cell type identification was achieved through Seurat's analytical tools, and subtype differentiation trajectories were inferred using Slingshot. Cellular communication dynamics between tumor cell subtypes and other cells were analyzed with the CellChat. The pySCENIC package was utilized to analyze transcription factors regulatory networks in the identified tumor cell subtypes. The results were verified by in vitro experiments. A risk scoring model was developed to assess patient outcomes. RESULTS This study employed scRNA-seq to conduct a comprehensive analysis of HR+BC tumor subtypes, identifying the C3 PCLAF+ tumor cells subtype, which demonstrated high proliferation and differentiation potential. C3 PCLAF+ tumor cells subtype was found to be closely associated with cancer-associated fibroblasts through the MK signaling pathway, facilitating tumor progression. Additionally, we discovered that MAZ was significantly expressed in C3 PCLAF+ tumor cells subtype, and in vitro experiments confirmed that MAZ knockdown inhibited tumor growth, accentuating its underlying ability as a therapeutic target. Furthermore, we developed a novel prognostic model based on the expression profile of key prognostic genes within the PCLAF+/MAZ regulatory network. This model linked high PCLAF+ tumor risk scores with poor survival outcomes and specific immune microenvironment characteristics. CONCLUSION This study utilized scRNA-seq to reveal the role of the C3 PCLAF+ tumor cells subtype in HR+BC, emphasizing its association with poor prognosis and resistance to endocrine therapies. MAZ, identified as a key regulator, contributed to tumor progression, while the tumor microenvironment had a pivotal identity in immune evasion. The findings underscored the importance of overcoming drug resistance, recognizing novel treatment targets, and crafting tailored diagnosis regimens.
Collapse
Affiliation(s)
- Gaofeng Ni
- Department of Breast Surgery, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai 264003, China
| | - Yuwei Sun
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Hongling Jia
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Zhikai Xiahou
- China Institute of Sport and Health Science, Beijing Sport University, Beijing 100084, China
| | - Yumeng Li
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Fu Zhao
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Hongyan Zang
- Department of Breast Surgery, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai 264003, China.
| |
Collapse
|
50
|
Guo Z, Liu Y, Chen D, Sun Y, Li D, Meng Y, Zhou Q, Zeng F, Deng G, Chen X. Targeting regulated cell death: Apoptosis, necroptosis, pyroptosis, ferroptosis, and cuproptosis in anticancer immunity. J Transl Int Med 2025; 13:10-32. [PMID: 40115032 PMCID: PMC11921819 DOI: 10.1515/jtim-2025-0004] [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] [Indexed: 03/22/2025] Open
Abstract
In the evolving landscape of cancer treatment, the strategic manipulation of regulated cell death (RCD) pathways has emerged as a crucial component of effective anti-tumor immunity. Evidence suggests that tumor cells undergoing RCD can modify the immunogenicity of the tumor microenvironment (TME), potentially enhancing its ability to suppress cancer progression and metastasis. In this review, we first explore the mechanisms of apoptosis, necroptosis, pyroptosis, ferroptosis, and cuproptosis, along with the crosstalk between these cell death modalities. We then discuss how these processes activate antigen-presenting cells, facilitate the cross-priming of CD8+ T cells, and trigger anti-tumor immune responses, highlighting the complex effects of novel forms of tumor cell death on TME and tumor biology. Furthermore, we summarize potential drugs and nanoparticles that can induce or inhibit these emerging RCD pathways and their therapeutic roles in cancer treatment. Finally, we put forward existing challenges and future prospects for targeting RCD in anti-cancer immunity. Overall, this review enhances our understanding of the molecular mechanisms and biological impacts of RCD-based therapies, providing new perspectives and strategies for cancer treatment.
Collapse
Affiliation(s)
- Ziyu Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Yihuang Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Danyao Chen
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Yuming Sun
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Daishi Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Yu Meng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Qian Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Furong Zeng
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| |
Collapse
|