51
|
Yang Y, Zhao L, Gao F, Wu G, Luo Y, An M. Modulation of renal fibrosis-related signaling pathways by traditional Chinese medicine: molecular mechanisms and experimental evidence. Int Urol Nephrol 2025:10.1007/s11255-025-04532-z. [PMID: 40293615 DOI: 10.1007/s11255-025-04532-z] [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: 02/10/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025]
Abstract
Renal fibrosis (RF), characterized by excessive deposition of extracellular matrix leading to tissue damage and scar formation, represents a refractory disease and a pivotal pathological basis for the progression to end-stage renal disease. The pathogenesis of RF is intricate, prominently implicating multiple key signaling pathways, including adenosine monophosphate-activated protein kinase/mammalian target of rapamycin (AMPK/mTOR), phosphoinositide 3-kinase/protein kinase B (PI3K/Akt), transforming growth factor-β1/small mother against decapentaplegic (TGF-β1/Smad), toll-like receptor 4/nuclear factor kappa B (TLR4/NF-κB), wingless integrated/β-catenin (Wnt/β-catenin), hypoxia-inducible factor-1α (HIF-1α), Hedgehog, and mitogen-activated protein kinase (MAPK). The current Western medical practices primarily rely on supportive and replacement therapies, which are often costly and suboptimal in efficacy. In contrast, traditional Chinese medicine (TCM), with its inherent advantages of multi-target, multi-pathway, and multi-effect modulation, emerges as a promising new strategy for RF treatment. However, a systematic, comprehensive, and detailed summary of these advancements remains absent. Therefore, this review consolidates the recent research progress on TCM modulation of RF-related signaling pathways, aiming to provide a theoretical foundation for further investigations into RF and the development of TCM interventions.
Collapse
Affiliation(s)
- Yufei Yang
- Department of Pharmacy, Baotou Medical College, 31 Jianshe Road, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China
| | - Longshan Zhao
- Department of Pharmacy, Baotou Medical College, 31 Jianshe Road, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China
- Department of Pharmacy, Shenyang Pharmaceutical University, Benxi, 117004, China
| | - Fengli Gao
- Department of Pharmacy, Second Affiliated Hospital of Baotou Medical College, Baotou, 014030, China
| | - Guodong Wu
- Department of Pharmacy, Baotou Medical College, 31 Jianshe Road, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China
| | - Yiduo Luo
- Department of Pharmacy, Baotou Medical College, 31 Jianshe Road, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China
| | - Ming An
- Department of Pharmacy, Baotou Medical College, 31 Jianshe Road, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China.
| |
Collapse
|
52
|
Zhang Z, Lu M, Shen P, Xu T, Tan S, Tang H, Yu Z, Zhou J. TGFBI promotes EMT and perineural invasion of pancreatic cancer via PI3K/AKT pathway. Med Oncol 2025; 42:181. [PMID: 40286004 DOI: 10.1007/s12032-025-02736-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Pancreatic cancer is a highly lethal malignancy, and perineural invasion (PNI) is a common pathological feature that significantly contributes to poor prognosis. Our research identified TGFBI as a key player in PNI development. The expression of TGFBI in tissue and cancer cells were detected by RT-qPCR, Western blot, Immunohistochemistry, and ELISA. The localization of TGFBI in cells was analyzed by Immunofluorescence staining (IF). The neural invasion ability of cancer cells were assessed by in vitro neural invasion model. Moreover, Western blot was used to investigate epithelial-mesenchymal transition (EMT) markers and PI3K/AKT pathway markers to elucidate the underlying mechanisms. Finally, an in vivo neural invasion model was used to verify the tumorigenic ability of the cancer cells in the sciatic nerve. Our findings highlight that TGFBI is up-regulated in PNI tissue and significantly correlates with poor prognosis in pancreatic cancer patients. Based on in vitro experiments, knockdown of TGFBI reduced neural invasion, as well as EMT, whereas rTGFBI exhibited the reverse effect. Knockdown of TGFBI reduced PI3K/AKT phosphorylation in Capan-2 and CFPAC-1. Moreover, PI3K inhibitor LY294002 was observed to counteract the effects of TGFBI on neural invasion, and EMT in Capan-2 and CFPAC-1. In vivo, knockdown of TGFBI inhibited tumor formation in the sciatic nerve of mice. Finally, we confirmed TGFBI as potential biomarker for PNI and prognosis of pancreatic cancer. Collectively, we concluded that TGFBI activates the PI3K-AKT pathway in pancreatic cancer cells, ultimately promoting EMT and leading to PNI.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Surgery, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu Province, China
| | - Miao Lu
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, 210009, Jiangsu Province, China
| | - Peiyuan Shen
- Department of Surgery, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu Province, China
| | - Tonglei Xu
- Department of Surgery, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu Province, China
| | - Siyuan Tan
- Department of Surgery, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu Province, China
| | - Haodong Tang
- Department of Surgery, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu Province, China
| | - Zeqian Yu
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, 210009, Jiangsu Province, China
| | - Jiahua Zhou
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, 210009, Jiangsu Province, China.
| |
Collapse
|
53
|
Jing H, Gao Y, Jing L, Yang H, Liu S. Recent advances in therapeutic use of transforming growth factor-beta inhibitors in cancer and fibrosis. Front Oncol 2025; 15:1489701. [PMID: 40352593 PMCID: PMC12061708 DOI: 10.3389/fonc.2025.1489701] [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: 09/01/2024] [Accepted: 04/03/2025] [Indexed: 05/14/2025] Open
Abstract
Transforming growth factor-beta (TGF-β) has long been known to be associated with early embryonic development and organogenesis, immune supervision, and tissue repair and homeostasis in adults. TGF-β has complex roles in fibrosis and cancer that may be opposing at different stages of these diseases. Under pathological conditions, overexpression of TGF-β causes epithelial-mesenchymal transition, deposition of extracellular matrix, and formation of cancer-associated fibroblasts, leading to fibrotic disease or cancer. Fibroblasts, epithelial cells, and immune cells are the most common targets of TGF-β, while fibrosis and cancer are the most common TGF-β-associated diseases. Given the critical role of TGF-β and its downstream molecules in fibrosis and progression of cancer, therapies targeting TGF-β signaling appear to be a promising strategy. Preclinical and clinical studies have investigated therapies targeting TGF-β, including antisense oligonucleotides, monoclonal antibodies, and ligand traps. However, development of targeted TGF-β therapy has been hindered by systemic cytotoxicity. This review discusses the molecular mechanisms of TGF-β signaling and highlights targeted TGF-β therapy for cancer and fibrosis as a therapeutic strategy for related diseases.
Collapse
Affiliation(s)
- Hanhui Jing
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yan Gao
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Linyuan Jing
- Department of Integrated Chinese and Western Medicine, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Hanyu Yang
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shanglong Liu
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
54
|
Guo Y, Deng F, Jiang Y, Cao G, Zhang Y, Liu G, Alimujiang M, Ayati M, Chen Y, Chen L, Lv S, Dou X. IL-37 Alleviates Sepsis-Induced Lung Injury by Inhibiting Inflammatory Response Through the TGF-β/Smad3 Pathway. Immunol Invest 2025:1-15. [PMID: 40270428 DOI: 10.1080/08820139.2025.2495958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Introduction: Sepsis is caused by an uncontrolled inflammatory response and immune dysfunction, with lung injury being the most common complication and one of the leading causes of death in clinically ill patients. Interleukin 37 (IL-37) is a multifunctional cytokine that plays a vital role in various pathophysiological processes, including inflammation, infection, and immunity.Methods: The study involved both clinical and animal experiments (establishing an animal model of sepsis-induced lung injury). Firstly, 50 patients with sepsis-induced lung injury and 50 healthy controls were included. In addition, a more in-depth study was conducted using animal models.Results: IL-37, IL-6, PCT, and CRP levels were significantly higher in the sepsis-induced lung injury group. Correlation analysis revealed that IL-37 significantly correlated with IL-6, PCT, and CRP levels. In animal experiments, IL-37 significantly attenuated CLP-induced pulmonary edema and cellular injury while reducing the levels of inflammatory factors IL-6 and TNF-α, as well as sepsis-related inflammatory markers PCT and CRP. Moreover, IL-37 significantly downregulated the expression levels of genes and proteins of apoptosis-related molecules Caspase-3 and Bax and pathway molecules TGF-β and Smad3. Discussion: The TGF-β/Smad3 pathway is involved in the process of IL-37 inhibiting inflammatory response and ameliorating sepsis-induced lung injury.
Collapse
Affiliation(s)
- Yufang Guo
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, Xinjiang, P.R. China
| | - Feifei Deng
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Yali Jiang
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
- Ili & Jiangsu Joint Institute of Health, Yili, Xinjiang, P.R. China
| | - Guodong Cao
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, Xinjiang, P.R. China
- Ili & Jiangsu Joint Institute of Health, Yili, Xinjiang, P.R. China
| | - Yixin Zhang
- Department of Critical Care Medicine, Medical School of Shihezi University, Shihezi, Xinjiang, P.R. China
| | - Gaowu Liu
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Mayinur Alimujiang
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Mairhaba Ayati
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Yufeng Chen
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Lili Chen
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Su Lv
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Xueqin Dou
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| |
Collapse
|
55
|
Balhara N, Yadav R, Chauhan MB. Role of signaling pathways in endometrial cancer. Mol Biol Rep 2025; 52:408. [PMID: 40257522 DOI: 10.1007/s11033-025-10523-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
Endometrial cancer (EC) is a prevalent gynecological malignancy with a complex molecular landscape, contributing to significant global morbidity and mortality. Dysregulated signaling pathways such as PI3K/AKT/mTOR and RAS/RAF/MEK drive EC progression by promoting uncontrolled cell proliferation, survival, angiogenesis, and metastasis. Mutations in genes like PTEN and PIK3CA further underpin tumor aggressiveness. Molecular alterations in these pathways not only serve as biomarkers for prognosis but also guide the formulation of targeted therapies, such as mTOR inhibitors and anti-angiogenic agents. While such therapies show promise, optimizing their efficacy and minimizing adverse effects requires further research. A comprehensive approach integrating early detection (e.g., addressing postmenopausal bleeding), preventive strategies (e.g., managing obesity), increasing diagnostic sensitivity (e.g., transvaginal ultrasound) and advanced molecularly tailored treatments (e.g., AI & ML) is critical to reducing the burden of this disease. By targeting key signaling pathways, leveraging AI-driven methodologies, and addressing treatment resistance, we can enhance patient outcomes, also mitigate the rising global impact of EC.
Collapse
Affiliation(s)
- Nikita Balhara
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Ritu Yadav
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India.
| | - Meenakshi B Chauhan
- Department of Obstetrics and Gynecology, PGIMS, Rohtak, Haryana, 124001, India
| |
Collapse
|
56
|
Zhang H, Zhuang Z, Hong L, Wang R, Xu J, Tang Y. The malignant signature gene of cancer-associated fibroblasts serves as a potential prognostic biomarker for colon adenocarcinoma patients. Front Immunol 2025; 16:1589678. [PMID: 40313961 PMCID: PMC12043632 DOI: 10.3389/fimmu.2025.1589678] [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: 03/07/2025] [Accepted: 04/01/2025] [Indexed: 05/03/2025] Open
Abstract
Background Colon adenocarcinoma (COAD) is the most frequently occurring type of colon cancer. Cancer-associated fibroblasts (CAFs) are pivotal in facilitating tumor growth and metastasis; however, their specific role in COAD is not yet fully understood. This research utilizes single-cell RNA sequencing (scRNA-seq) to identify and validate gene markers linked to the malignancy of CAFs. Methods ScRNA-seq data was downloaded from a database and subjected to quality control, dimensionality reduction, clustering, cell annotation, cell communication analysis, and enrichment analysis, specifically focusing on fibroblasts in tumor tissues compared to normal tissues. Fibroblast subsets were isolated, dimensionally reduced, and clustered, then combined with copy number variation (CNV) inference and pseudotime trajectory analysis to identify genes related to malignancy. A Cox regression model was constructed based on these genes, incorporating LASSO analysis, nomogram construction, and validation.Subsequently, we established two FNDC5-knockdown cell lines and utilized colony formation and transwell assays to investigate the impact of FNDC5 on cellular biological behaviors. Results Using scRNA-seq data, we analyzed 8,911 cells from normal and tumor samples, identifying six distinct cell types. Cell communication analysis highlighted interactions between these cell types mediated by ligands and receptors. CNV analysis classified CAFs into three groups based on malignancy levels. Pseudo-time analysis identified 622 pseudotime-related genes and generated a forest plot using univariate Cox regression. Lasso regression identified the independent prognostic gene FNDC5, which was visualized in a nomogram. Kaplan-Meier survival analysis confirmed the prognostic value of FNDC5, showing associations with T stage and distant metastasis. In vitro experiment results demonstrated a strong association between FNDC5 expression levels and the proliferative, migratory, and invasive abilities of colon cancer cells. Conclusion We developed a risk model for genes related to the malignancy of CAFs and identified FNDC5 as a potential therapeutic target for COAD.
Collapse
Affiliation(s)
| | | | | | | | | | - Youyuan Tang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
57
|
Zhao Q, Jin M, Zhao Q, Wang Z, Zhao C, Xue X, Qiao X, Qu P, Han D, Tao R. Natural products in traditional Chinese medicine for renal fibrosis: a comprehensive review. Front Pharmacol 2025; 16:1560567. [PMID: 40308781 PMCID: PMC12041090 DOI: 10.3389/fphar.2025.1560567] [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: 01/14/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Renal fibrosis represents the terminal pathological manifestation of most chronic kidney diseases, driving progressive loss of renal function. Natural products have emerged as promising therapeutic agents for preventing and ameliorating renal fibrosis due to their multi-target efficacy and favorable safety profiles. In this review, we conducted a comprehensive literature search on PubMed using the keywords "natural product" and "renal fibrosis" from 2004 to 2025, identifying 704 relevant articles. We systematically categorize and discuss the biological effects of key natural products and formulations with antifibrotic potential, focusing on five major classes: glycosides, flavonoids, phenolic compounds, anthraquinones, and terpenoids. Representative compounds from each category are highlighted for their mechanisms of action, including modulation of oxidative stress, inflammation, autophagy, and fibrosis signaling pathways. This review aims to provide a theoretical foundation for the development of natural product-based therapies to combat renal fibrosis, offering insights into their therapeutic potential and future research directions.
Collapse
Affiliation(s)
- Qianqian Zhao
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| | - Meihua Jin
- Department of Immunology, Medical College, Dalian University, Dalian, Liaoning, China
| | - Qiang Zhao
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| | - Zhimei Wang
- Department of Immunology, Medical College, Dalian University, Dalian, Liaoning, China
| | - Chun Zhao
- Department of Immunology, Medical College, Dalian University, Dalian, Liaoning, China
| | - Xiaocong Xue
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| | - Xikai Qiao
- Department of Immunology, Medical College, Dalian University, Dalian, Liaoning, China
| | - Peng Qu
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| | - Donghe Han
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| | - Ran Tao
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| |
Collapse
|
58
|
Huang C, Wang L, Zhuo C, Chen W, Fan H, Hong Y, Zhang Y, Zhou D, Lin W, Zhang L, Zhao J, Chen S, Yu C, Ye Y. ID3 enhances PD-L1 expression by restructuring MYC to promote colorectal cancer immune evasion. Proc Natl Acad Sci U S A 2025; 122:e2423490122. [PMID: 40208940 PMCID: PMC12012548 DOI: 10.1073/pnas.2423490122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/12/2025] [Indexed: 04/12/2025] Open
Abstract
The inhibitor of DNA binding protein ID3 has been associated with the progression of colorectal cancer (CRC). Despite its significance, its specific role in the immune evasion strategies utilized by CRC remains unclear. RNA-seq analysis revealed that ID3 was positively associated with the PD-L1 immune checkpoint. We further demonstrated that tumor cell-expressed ID3 enhanced PD-L1 expression, suppressed the infiltration and activation of CD8+ T cells, and facilitated the immune evasion of CRC cells. Additionally, we found that knockdown of ID3 significantly enhanced the effectiveness of PD-L1 antibody blockade treatment in combating CRC, reduced the upregulation of PD-L1 induced by the antibody, and altered the immune microenvironment within CRC. Mechanistically, ID3 interacted with the transcription factor MYC and reconstructed the four-dimensional structure of MYC, thereby enhancing its binding affinity to the PD-L1 promoter and augmenting PD-L1 transcriptional activity. By integrating analysis of ChIP-seq, RNA-seq, and ImmPort gene sets, we found that ID3's DNA-assisted binding function was widespread and could either enhance or suppress gene transcription, not only affecting tumor immune escape through immune checkpoints but also regulating various cytokines and immune cells involved in tumor immunity. In conclusion, our study uncovers a mechanism by which ID3 promotes immune evasion in CRC and implicates that targeting ID3 may improve the efficacy of anti-PD-1/PD-L1 immunotherapy.
Collapse
Affiliation(s)
- Chuanzhong Huang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou350122, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou350014, People’s Republic of China
| | - Ling Wang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou350122, China
| | - Changhua Zhuo
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou350122, China
| | - Wenxin Chen
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou350122, China
| | - Hongmei Fan
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou350122, China
| | - Yilin Hong
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen361102, People’s Republic of China
| | - Yu Zhang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
| | - Dongmei Zhou
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou350122, China
| | - Wansong Lin
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou350122, China
| | - Lingyu Zhang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou350122, China
| | - Jingjing Zhao
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
| | - Shuping Chen
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou350122, China
| | - Chundong Yu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen361102, People’s Republic of China
| | - Yunbin Ye
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
| |
Collapse
|
59
|
Chen X, Chen B, Zhao H. Role of Neutrophils in Anti-Tumor Activity: Characteristics and Mechanisms of Action. Cancers (Basel) 2025; 17:1298. [PMID: 40282474 PMCID: PMC12025517 DOI: 10.3390/cancers17081298] [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: 02/25/2025] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
As one of the leading components in the immune system, neutrophils in the tumor microenvironment (TME) have received considerable attention in recent years. The tumor-killing effects of neutrophils in a variety of tumors have been reported. However, the functions of neutrophils in tumors remain to be completely elucidated, and both anti-tumor and tumor-promotion activities have been reported. This review focuses on the characteristics of neutrophils and their mechanisms of action in the TME, with an emphasis on their anti-tumor activity, including reactive oxygen species (ROS)-induced tumor killing, cytotoxic T lymphocytes (CTLs)-induced tumor killing, trogocytosis, cytotoxic enzymes, and trained immunity. Furthermore, the possible targets and methods of tumor treatment regimens for neutrophils are explored, with the aim of exploring the use of neutrophils in the future as a potential anti-tumor treatment strategy.
Collapse
Affiliation(s)
- Xin Chen
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi’an 710032, China;
| | - Bingdi Chen
- The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai 200092, China
| | - Huadong Zhao
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi’an 710032, China;
| |
Collapse
|
60
|
Sun Z, Zhao W, Fei X, He B, Shi L, Zhang Z, Cai S. Static magnetic field inhibits epithelial mesenchymal transition and metastasis of glioma. Sci Rep 2025; 15:12430. [PMID: 40216876 PMCID: PMC11992211 DOI: 10.1038/s41598-025-96047-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Gliomas exhibit suboptimal responses to conventional treatments, with tumor cell migration remaining a significant challenge in therapy. Epithelial-mesenchymal transition (EMT) is crucial for glioma cell invasion, and transforming growth factor β1(TGF-β1) is a key factor promoting proliferation, migration, and EMT in glioblastoma (GBM). Although magnetic fields are widely used in the diagnosis and treatment of various diseases, their effects on EMT in glioma cells remain unclear. In this study, we investigated whether a static magnetic field (SMF) could inhibit EMT and metastasis in glioma cells. Cellular functional assays using the U251 and U87 glioma cell lines were performed to investigate their functional and phenotypic changes. Results showed that TGF-β1 treatment increased the invasion and migration capabilities of glioma cells, while simultaneously reducing apoptosis. However, when SMF was combined with TGF-β1 treatment, a significant reduction in cell migration and invasion was observed, along with an increase in apoptosis. Additionally, this combination treatment significantly decreased the protein expression of mesenchymal markers N-cadherin and β-catenin, as well as reduced the levels of the matrix metalloproteinase (MMP)-2. Collectively, these findings suggest that SMFs may attenuate glioma cell metastasis by inhibiting EMT. Therefore, SMFs could represent a promising therapeutic strategy for diminishing glioma metastasis.
Collapse
Affiliation(s)
- Ziyu Sun
- Department of Neurosurgery, Gusu School, Nanjing Medical University, The First People's Hospital of Kunshan, Suzhou, People's Republic of China
| | - Wenxuan Zhao
- Department of Neurosurgery, Gusu School, Nanjing Medical University, The First People's Hospital of Kunshan, Suzhou, People's Republic of China
| | - Xifeng Fei
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, People's Republic of China
| | - Bao He
- Department of Neurosurgery, Gusu School, Nanjing Medical University, The First People's Hospital of Kunshan, Suzhou, People's Republic of China
| | - Lei Shi
- Department of Neurosurgery, Gusu School, Nanjing Medical University, The First People's Hospital of Kunshan, Suzhou, People's Republic of China.
| | - Zhen Zhang
- Department of Radiology, Affiliated Kunshan Hospital of Jiangsu University, China Medical University, Gusu School Nanjing Medical University, Suzhou, People's Republic of China.
| | - Shizhong Cai
- Department of Child and Adolescent Healthcare, Children's Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China.
| |
Collapse
|
61
|
Zhang M, Zhang B. Extracellular matrix stiffness: mechanisms in tumor progression and therapeutic potential in cancer. Exp Hematol Oncol 2025; 14:54. [PMID: 40211368 PMCID: PMC11984264 DOI: 10.1186/s40164-025-00647-2] [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: 02/06/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025] Open
Abstract
Tumor microenvironment (TME) is a complex ecosystem composed of both cellular and non-cellular components that surround tumor tissue. The extracellular matrix (ECM) is a key component of the TME, performing multiple essential functions by providing mechanical support, shaping the TME, regulating metabolism and signaling, and modulating immune responses, all of which profoundly influence cell behavior. The quantity and cross-linking status of stromal components are primary determinants of tissue stiffness. During tumor development, ECM stiffness not only serves as a barrier to hinder drug delivery but also promotes cancer progression by inducing mechanical stimulation that activates cell membrane receptors and mechanical sensors. Thus, a comprehensive understanding of how ECM stiffness regulates tumor progression is crucial for identifying potential therapeutic targets for cancer. This review examines the effects of ECM stiffness on tumor progression, encompassing proliferation, migration, metastasis, drug resistance, angiogenesis, epithelial-mesenchymal transition (EMT), immune evasion, stemness, metabolic reprogramming, and genomic stability. Finally, we explore therapeutic strategies that target ECM stiffness and their implications for tumor progression.
Collapse
Affiliation(s)
- Meiling Zhang
- School of Basic Medicine, China Three Gorges University, 8 Daxue Road, Yichang, 443002, Hubei, China
- Central Laboratory, The First Affiliated Hospital of Jinan University, No. 613 Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China
| | - Bin Zhang
- School of Basic Medicine, China Three Gorges University, 8 Daxue Road, Yichang, 443002, Hubei, China.
- Central Laboratory, The First Affiliated Hospital of Jinan University, No. 613 Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.
| |
Collapse
|
62
|
Bagheri L, Javanbakht M, Malekian S, Ghahderijani BH, Taghipour S, Tanha FD, Ranjkesh M, Cegolon L, Zhao S. Antifibrotic therapeutic strategies in systemic sclerosis: Critical role of the Wnt/β-catenin and TGF-β signal transduction pathways as potential targets. Eur J Pharmacol 2025; 999:177607. [PMID: 40209848 DOI: 10.1016/j.ejphar.2025.177607] [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: 11/14/2024] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Systemic sclerosis (SSc) is a prototypic fibrosing disorder characterized by widespread fibrosis and immune dysregulation. Current evidence highlights the intricate cross-talk between the canonical Wnt/β-catenin signaling pathway and transforming growth factor-beta (TGF-β) signaling, both of which play fundamental roles in the pathogenesis of fibrosis. This review aims to elucidate the central role of the Wnt/β-catenin-TGF-β pathway and TGF-β signal transduction pathway in fibrotic diseases, focusing on SSc. We summarized evidence from cellular biology studies, animal model investigations and clinical observations to provide a comprehensive view of the mechanisms causing pathological fibrosis. In addition, we explore the possibilities of antifibrotic therapeutic strategies against Wnt/β-catenin-TGF-β signaling to counteract fibrosis, delineating approaches for treatment of SSc patients by targeting these interconnected signaling pathways.
Collapse
Affiliation(s)
- Leyla Bagheri
- Department of Internal Medicine, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Javanbakht
- Nephrology and Urology Research Center, Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Sheida Malekian
- Department of Internal Medicine, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Sadra Taghipour
- Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Fatemeh Davari Tanha
- Department of Infertility, Yas Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Luca Cegolon
- Department of Medical, Surgical & Health Sciences, University of Trieste, 34128, Trieste, Italy; Public Health Unit, University Health Agency Giuliano-Isontina (ASUGI), 34148, Trieste, Italy
| | - Shi Zhao
- School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
63
|
Wen J, Cui W, Yin X, Chen Y, Liu A, Wang Q, Meng X. Application and future prospects of bispecific antibodies in the treatment of non-small cell lung cancer. Cancer Biol Med 2025; 22:j.issn.2095-3941.2024.0470. [PMID: 40192238 PMCID: PMC12032835 DOI: 10.20892/j.issn.2095-3941.2024.0470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/05/2025] [Indexed: 04/29/2025] Open
Abstract
As the leading cause of cancer-related deaths, lung cancer remains a noteworthy threat to human health. Although immunotherapies, such as immune checkpoint inhibitors (ICIs), have significantly increased the efficacy of lung cancer treatment, a significant percentage of patients are not sensitive to immunotherapies and patients who initially respond to treatment can quickly develop acquired drug resistance. Bispecific antibodies (bsAbs) bind two different antigens or epitopes simultaneously and have been shown to enhance antitumor efficacy with suitable safety profiles, thus attracting increasing attention as novel antitumor therapies. At present, in addition to the approved bsAb, amivantamab, three novel bsAbs (KN046, AK112, and SHR-1701) are being evaluated in phase 3 clinical trials and many bsAbs are being evaluated in phase 1/2 clinical trials for patients with non-small cell lung cancer (NSCLC). Herein we present the structure, classification, and mechanism of action underlying bsAbs in NSCLC and introduce related clinical trials. Finally, we discuss challenges, potential solutions, and future prospects in the context of cancer treatment with bsAbs.
Collapse
Affiliation(s)
- Junxu Wen
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Wenxing Cui
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xiaoyan Yin
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Yu Chen
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Ailing Liu
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Qian Wang
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| |
Collapse
|
64
|
Khongpiroon C, Buakaew W, Brindley PJ, Potikanond S, Daowtak K, Thongsri Y, Potup P, Usuwanthim K. Anti-Fibrotic Effect of Oleamide Identified from the Moringa oleifera Lam. Leaves via Inhibition of TGF-β1-Induced SMAD2/3 Signaling Pathway. Int J Mol Sci 2025; 26:3388. [PMID: 40244247 PMCID: PMC11989637 DOI: 10.3390/ijms26073388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
Moringa oleifera (MO) is a prominent plant in traditional medicine, widely recognized for its phytochemicals with anti-inflammatory properties. Liver fibrosis characterized by chronic inflammation and excessive extracellular matrix deposition may benefit from the therapeutic properties of MO. This report focuses on the potential of oleamide (OLA), a bioactive compound identified from MO, in mitigating liver fibrosis. The anti-fibrotic effects of OLA were evaluated by assessing the production of pro-inflammatory cytokines, gelatinase activity and the expression of genes and proteins associated with the TGF-β/SMAD2/3 pathway. The LX-2 human hepatic stellate cell line, in conjunction with TGF-β1, was employed to model fibrotic conditions. OLA treatment significantly reduced the production of pro-fibrotic effectors in the activated LX-2 cells. Molecular docking analysis demonstrated a high binding affinity of OLA to key proteins in the TGF-β/SMAD2/3 pathway, while qRT-PCR and Western blotting revealed that OLA suppressed the expression of COL1A1, COL4A1, SMAD2, SMAD3, SMAD4, MMP2, MMP9, ACTA2 and TIMP1. These findings indicate that OLA effectively attenuates the pro-inflammatory responses induced by TGF-β1 and inhibits the activation of LX-2 cells. Collectively, OLA holds significant potential as a therapeutic agent for the prevention and treatment of liver fibrosis via the modulation of the TGF-β/SMAD2/3 signaling pathway.
Collapse
Affiliation(s)
- Chavisa Khongpiroon
- Cellular and Molecular Immunology Research Unit, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand (K.D.); (Y.T.); (P.P.)
| | - Watunyoo Buakaew
- Department of Microbiology, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand;
| | - Paul J. Brindley
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA;
| | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Krai Daowtak
- Cellular and Molecular Immunology Research Unit, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand (K.D.); (Y.T.); (P.P.)
| | - Yordhathai Thongsri
- Cellular and Molecular Immunology Research Unit, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand (K.D.); (Y.T.); (P.P.)
| | - Pachuen Potup
- Cellular and Molecular Immunology Research Unit, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand (K.D.); (Y.T.); (P.P.)
| | - Kanchana Usuwanthim
- Cellular and Molecular Immunology Research Unit, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand (K.D.); (Y.T.); (P.P.)
| |
Collapse
|
65
|
Wei Y, Jia Z, Ma J, Zhang W, Li H, Wu J, Wang X, Yu X, Shi Y, Kong X, Pang M. Proteomics and Metabolomics Analyses Reveal a Dynamic Landscape of Coal Workers' Pneumoconiosis: An Insight into Disease Progression. J Proteome Res 2025; 24:1715-1731. [PMID: 40036136 PMCID: PMC11976863 DOI: 10.1021/acs.jproteome.4c00715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 03/06/2025]
Abstract
Coal worker's pneumoconiosis (CWP) is characterized by chronic inflammation and pulmonary fibrosis. The key factor contributing to the incurability of CWP is the unclear pathogenesis. This study explored the characteristic changes in proteomics and metabolomics of early and advanced CWP patients through proteomics and metabolomics techniques. Proteomics identified proteins that change with the progression of CWP, with significant enrichment in the TGF-β signaling pathway and autoimmune disease pathways. Metabolomics revealed the metabolic characteristics of CWP at different stages. These metabolites mainly include changes in amino acid metabolism, unsaturated fatty acid synthesis, and related metabolites. Integrated analysis found that ABC transporters are a shared pathway among the three groups, and ABCD2 is involved in the ABC transporter pathway. In the subsequent independent sample verification analysis, consistent with proteomics experiments, compared to the CM group, FMOD expression level was upregulated in the NIC group. TFR expression level was consistently downregulated in both the IC and NIC groups. Additionally, ABCD2 increased in the IC group but decreased in the NIC group. In summary, this study revealed the metabolic characteristics of CWP at different stages. These findings may provide valuable insights for the early prediction, diagnosis, and treatment of CWP.
Collapse
Affiliation(s)
- Yangyang Wei
- Department
of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
- NHC
Key Laboratory of Pneumoconiosis, Taiyuan 030001, China
| | - Zhenzhen Jia
- Academy
of Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi 030001, China
- NHC
Key Laboratory of Pneumoconiosis, Taiyuan 030001, China
| | - Jing Ma
- Shanxi
Cardiovascular Hospital, Taiyuan, Shanxi 030001, China
| | - Wei Zhang
- Department
of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
- NHC
Key Laboratory of Pneumoconiosis, Taiyuan 030001, China
| | - Hui Li
- Department
of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
- NHC
Key Laboratory of Pneumoconiosis, Taiyuan 030001, China
| | - Juan Wu
- Department
of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
- NHC
Key Laboratory of Pneumoconiosis, Taiyuan 030001, China
| | - Xiaojing Wang
- Department
of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
- NHC
Key Laboratory of Pneumoconiosis, Taiyuan 030001, China
| | - Xiao Yu
- Department
of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
- NHC
Key Laboratory of Pneumoconiosis, Taiyuan 030001, China
| | - Yiwei Shi
- Department
of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
- NHC
Key Laboratory of Pneumoconiosis, Taiyuan 030001, China
| | - Xiaomei Kong
- Department
of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
- NHC
Key Laboratory of Pneumoconiosis, Taiyuan 030001, China
| | - Min Pang
- Department
of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
- NHC
Key Laboratory of Pneumoconiosis, Taiyuan 030001, China
| |
Collapse
|
66
|
Xie Y, Wang X, Wang W, Pu N, Liu L. Epithelial-mesenchymal transition orchestrates tumor microenvironment: current perceptions and challenges. J Transl Med 2025; 23:386. [PMID: 40176117 PMCID: PMC11963649 DOI: 10.1186/s12967-025-06422-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/25/2025] [Indexed: 04/04/2025] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a critical process in cancer progression, facilitating tumor cells to develop invasive traits and augmenting their migratory capabilities. EMT is primed by tumor microenvironment (TME)-derived signals, whereupon cancer cells undergoing EMT in turn remodel the TME, thereby modulating tumor progression and therapeutic response. This review discusses the mechanisms by which EMT coordinates TME dynamics, including secretion of soluble factors, direct cell contact, release of exosomes and enzymes, as well as metabolic reprogramming. Recent evidence also indicates that cells undergoing EMT may differentiate into cancer-associated fibroblasts, thereby establishing themselves as functional constituents of the TME. Elucidating the relationship between EMT and the TME offers novel perspectives for therapeutic strategies to enhance cancer treatment efficacy. Although EMT-directed therapies present significant therapeutic potential, the current lack of effective targeting approaches-attributable to EMT complexity and its microenvironmental context dependency-underscores the necessity for mechanistic investigations and translational clinical validation.
Collapse
Affiliation(s)
- Yuqi Xie
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Xuan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenquan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ning Pu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
67
|
Shan B, Guo C, Zhou H, Chen J. Tanshinone IIA alleviates pulmonary fibrosis by modulating glutamine metabolic reprogramming based on [U- 13C 5]-glutamine metabolic flux analysis. J Adv Res 2025; 70:531-544. [PMID: 38697470 PMCID: PMC11976427 DOI: 10.1016/j.jare.2024.04.029] [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: 01/21/2024] [Revised: 03/28/2024] [Accepted: 04/28/2024] [Indexed: 05/05/2024] Open
Abstract
INTRODUCTION Glutamine metabolic reprogramming, mediated by glutaminase (GLS), is an important signal during pulmonary fibrosis (PF) progression. Tanshinone IIA (Tan IIA) is a naturally lipophilic diterpene with antioxidant and antifibrotic properties. However, the potential mechanisms of Tan IIA for regulating glutamine metabolic reprogramming are not yet clear. OBJECTIVES This study aimed was to evaluate the role of Tan IIA in intervening in glutamine metabolic reprogramming to exert anti-PF and to explore the potential new mechanisms of metabolic regulation. METHODS Fibrotic characteristics was detected via immunofluorescence and western blotting analysis. Cell proliferation was examined with EdU Assay. Cell metabolites were labeled by using stable isotope [U-13C5]-glutamine. By utilizing 100% 13C glutamine tracers and employing network analysis to investigate the activation of metabolic pathways in fibroblasts, as well as evaluating the impact of Tan IIA on these pathways, we accurately quantified the absolute flux of glutaminolysis, proline synthesis, and the TCA cycle pathway using isotopomer network compartmental analysis (INCA), a user-friendly software tool for 13C metabolic flux analysis (13C-MFA). Molecular docking was used for identifying the binding of Tan IIA with target protein. RESULTS Tan IIA ameliorate TGF-β1-induced myofibroblast proliferation, reduce collagen I and III and α-SMA protein expression in MRC-5 and NIH-3T3 cells. Furthermore, Tan IIA regulate mitochondrial energy metabolism by modulating TGF-β1-stimulated glutamine metabolic reprogramming in NIH-3T3 cells and inhibiting GLS1 expression, which reduced the metabolic flux of glutamine into mitochondria in myofibroblasts, and also targeted inhibited the expression of Δ1-pyrroline-5-carboxylate synthase (P5CS), P5C reductase 1 (PYCR1), and phosphoserine aminotransferase 1 (PSAT1), and reduced proline hydroxylation and blocked the collagen synthesis pathway. CONCLUSION Tan IIA reverses glutamine metabolic reprogramming, reduces mitochondrial energy expenditure, and inhibits collagen matrix synthesis by modulating potential targets in glutamine metabolism. This novel perspective sheds light on the essential role of glutamine metabolic reprogramming in PF.
Collapse
Affiliation(s)
- Baixi Shan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Congying Guo
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Haoyan Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jun Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
68
|
Selvam P, Tseng CH, Wang CT, Sun YY, Chen YL, Kao YT, Dahms HU, Cheng CM. 4-Anilinoquinolinylchalcone derivatives mediate antifibrotic effects through ERK/MRTF-a signaling pathway crosstalk. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2025; 32:11685-11696. [PMID: 40234319 DOI: 10.1007/s11356-025-36382-8] [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: 08/07/2024] [Accepted: 04/03/2025] [Indexed: 04/17/2025]
Abstract
Quinolones and their analogues are a remarkable group of drugs that have multiple impacts on the human immune system. They are suspected to mediate anti-cancer and anti-inflammatory responses. However, due to their effectiveness in treating a number of significant diseases, such as genitourinary cancer and breast cancer, as well as their antiangiogenic and immunomodulatory qualities, interest in this group of traditional medicines has recently increased. Unfortunately, numerous side effects were observed, such as diarrhea, skin rashes, nausea, vomiting, bleeding, and abnormal liver functions. To overcome these restrictions and to enhance the pharmacological profile, research efforts are focusing on the synthesis and optimization of novel quinolone analogues that lack severe side effects. The present study focuses on the mechanism of action and the signaling pathway involving the 4-anilinoquinolinylchalcone derivative. The objective of the present work was to better understand the mechanism by which anti-fibrosis is mediated by screening 6 synthesized 4-anilinoquinolinylchalcone derivatives for their potential as novel anti-fibrosis therapeutics.
Collapse
Affiliation(s)
- Padhmavathi Selvam
- Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung City, 807, Taiwan
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No. 100, Shih-Chuan 1st Road, Kaohsiung City, 807, Taiwan
| | - Chih Hua Tseng
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Department of Fragrance & Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Department of Pharmacy, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, 801, Taiwan
| | - Ching Tung Wang
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No. 100, Shih-Chuan 1st Road, Kaohsiung City, 807, Taiwan
| | - Yu-Yo Sun
- Institute of BioPharmaceutical Sciences, National Sun Yat-Sen University, Kaohsiung City, 804, Taiwan
| | - Yeh-Long Chen
- Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung City, 807, Taiwan
| | - Yu-Tse Kao
- Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung City, 807, Taiwan
| | - Hans-Uwe Dahms
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No. 100, Shih-Chuan 1st Road, Kaohsiung City, 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung City, 807, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung City, 804, Taiwan
| | - Chih Mei Cheng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No. 100, Shih-Chuan 1st Road, Kaohsiung City, 807, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan.
| |
Collapse
|
69
|
Liu S, Chen Y, Zhou G, Sun C, Ma M, Huang R, Li X, Liang X, Shi C, Wu W, Yan X, Wang L, Han J. Uniform and controllable surface nano-structure on polyetheretherketone implants can regulate mechanical property to enhance soft tissue integration through Piezo1/TGF-β1 signaling axis. Mater Today Bio 2025; 31:101645. [PMID: 40151615 PMCID: PMC11946874 DOI: 10.1016/j.mtbio.2025.101645] [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: 12/17/2024] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Polyetheretherketone (PEEK) has potential to repair the orbital floor bone defects following craniofacial trauma and orbital surgery. However, the inertness of the material impedes the soft tissue integration of implants, leading to complications such as implant migration and infection. Surface patterning modification on PEEK can promote the surface hydrophily to enhance better soft tissue integration, but it is difficult to obtain the uniform and controllable nano-structure. In this study, hot-pressing technology on PEEK implant was used to produce surface nanopores with a uniform diameter of 200, 500, 800 nm. Depending on the controllable craft, the surficial mechanical properties of PEEK can be regulated and assessed by finite element analysis. Furthermore, 500 nm interface has better mechanical properties to promote the proliferation, migration, and fibrosis of fibroblasts and achieved optimal integration effects in animal implantation experiments. To explore the mechanism of biological responses, transcriptomics and molecular biology experiments revealed that Piezo1/TGF-β1 axis played a critical role in the response of soft tissue cells to the mechanical stimulation of PEEK. Our study has established a novel modification technology for constructing uniform and controllable nanostructures on the surface of PEEK, thereby promoting the soft tissues integration with implants and improving the anchoring effect.
Collapse
Affiliation(s)
- Sida Liu
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
- The 940 Hospital of the Joint Logistic Support Force, 730050, Lanzhou, Gansu Province, China
| | - Yixuan Chen
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| | - Gandong Zhou
- School of Advanced Materials and Nanotechnology, Xidian University, 710126, Xi'an, Shaanxi Province, China
| | - Changning Sun
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, 710054, Xi'an, Shaanxi Province, China
| | - Minghai Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| | - Rou Huang
- School of Advanced Materials and Nanotechnology, Xidian University, 710126, Xi'an, Shaanxi Province, China
| | - Xing Li
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| | - Xiao Liang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| | - Changquan Shi
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, 710054, Xi'an, Shaanxi Province, China
| | - Weiwei Wu
- School of Advanced Materials and Nanotechnology, Xidian University, 710126, Xi'an, Shaanxi Province, China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| | - Lei Wang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| | - Jing Han
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| |
Collapse
|
70
|
Qian Y, Zhao X, Wu F, Wang X, Chen T. TGF-β-induced acetylation of KLF5 drives TNFAIP2 transcription and EMT in nasopharyngeal carcinoma: Unveiling a novel regulatory mechanism. Exp Cell Res 2025; 447:114498. [PMID: 40054652 DOI: 10.1016/j.yexcr.2025.114498] [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: 12/18/2024] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 03/12/2025]
Abstract
Epithelial-mesenchymal transition (EMT) is one of the critical mechanisms underlying migration, invasion, and metastasis of nasopharyngeal carcinoma (NPC) cells. The transcription factor KLF5 plays a pivotal role in various cancers, however, its precise functions in NPC remain incompletely understood. This study aims to explore the detailed mechanisms by which TGF-β enhances TNFAIP2 transcription by acetylating KLF5, thereby inducing EMT in NPC. KLF5 was significantly overexpressed in NPC tissues and closely associated with adverse clinicopathological features of the patients. Further studies revealed that TGF-β markedly increased the expression of KLF5 and its acetylated form, Ac-KLF5, in NPC cells, with the acetylation status of KLF5 being crucial for its function. KLF5 induced EMT in NPC cells by directly binding to the TNFAIP2 promoter and promoting its transcription. The pro-migratory and pro-invasive effects of acetylated KLF5 on NPC cells depended on TNFAIP2. Additionally, in vivo experiments confirmed that TGF-β treatment induced tumors in NPC mouse models to exhibit apparent EMT characteristics. These results collectively support the central role of the TGF-β-KLF5-TNFAIP2 axis in EMT of NPC. This study elucidates the specific mechanisms by which TGF-β promotes TNFAIP2 transcription by acetylating KLF5, thereby inducing EMT in NPC. This discovery not only provides new insights into the pathogenesis of NPC but also identifies potential therapeutic targets for NPC treatment.
Collapse
Affiliation(s)
- Yi Qian
- Department of Health Management Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing City, China
| | - Xuxu Zhao
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Feiyang Wu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoqiang Wang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Chen
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
71
|
Plichta J, Karbownik M, Kuna P, Panek M. In Silico-Designed TGFβRI/TGFβRII Receptor Complex Peptide Inhibitors Exhibit Biological Activity In Vitro. J Cell Mol Med 2025; 29:e70548. [PMID: 40245195 PMCID: PMC12005349 DOI: 10.1111/jcmm.70548] [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: 01/18/2025] [Revised: 03/22/2025] [Accepted: 03/28/2025] [Indexed: 04/19/2025] Open
Abstract
TGF-β (transforming growth factor β) is a pleiotropic cytokine found in three isoforms in humans. It regulates cell proliferation, wound healing, immune cell recruitment, contributes to epithelial-to-mesenchymal transition (EMT) and to the conversion of fibroblasts to myofibroblasts. TGF-β signalling pathway hyperactivity underlies many human disorders. The aim of this study was to evaluate a series of novel, in silico-designed peptide inhibitors (PIs) of the TGFβ/TGFβRI/TGFβRII complex. Luciferase-based luminescence assays on HEK293T cells were used to comparatively assess PI biological activity and calculate IC50 values. Flow cytometry was used to assess PI cytotoxicity on HEK293T cells. The PIs caused significant luminescence level reductions compared to controls. Additionally, three of the PIs caused luminescence reductions that did not differ significantly from the effects of SD-208, a small molecule TGFβ inhibitor. None of the PIs exhibited cytotoxicity. Our TGFBR PIs have demonstrated activity in vitro, with no observed cytotoxicity. Our results suggest the PIs may be of interest in the treatment of fibrotic disorders, chronic inflammatory diseases, or certain neoplastic cancers. The PIs will be further refined in silico and tested via assays carried out on cancer cell lines and CD4+/CD8+ T cells.
Collapse
Affiliation(s)
- Jacek Plichta
- Department of Internal Medicine, Asthma and AllergyMedical University of LodzLodzPoland
| | - Michał Karbownik
- Department of Pharmacology and ToxicologyMedical University of LodzLodzPoland
| | - Piotr Kuna
- Department of Internal Medicine, Asthma and AllergyMedical University of LodzLodzPoland
| | - Michał Panek
- Department of Internal Medicine, Asthma and AllergyMedical University of LodzLodzPoland
| |
Collapse
|
72
|
Tian M, Wang X, Zhang M, Li C, Xu Y, Chen X, Chen C, Wei Z, Li X, Ding G, Zhang L, Wang H, Gan H. DEAD-box protein 21 promotes renal fibrosis via p21-dependent cell cycle arrest in proximal tubular epithelial cells. Cell Signal 2025; 128:111654. [PMID: 39938704 DOI: 10.1016/j.cellsig.2025.111654] [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: 01/13/2025] [Accepted: 02/08/2025] [Indexed: 02/14/2025]
Abstract
Renal interstitial fibrosis is the final common outcome of various chronic kidney diseases (CKD). Renal tubular epithelial cells (TECs) G2/M cell cycle arrest play a pivotal role in renal fibrosis. Although RNA-binding proteins (RBPs) are implicated in organ fibrosis, the underlying mechanisms remain poorly understood. Here, we identify DEAD-box protein 21 (DDX21), a representative RBP, as highly expressed in fibrotic renal tissues, especially in TECs. Moreover, DDX21 expression is positively correlated with renal function decline in CKD patients, underscoring its role in disease progression. TECs-specific deletion of Ddx21 alleviates cell cycle arrest in G2/M, and attenuates fibrotic responses. Mechanistically, silencing DDX21 reduces p21 expression at both the mRNA and protein levels and decreases cell apoptosis, indicating that DDX21 promotes G2/M cell cycle arrest by regulating the p21 signaling pathway. This study suggests that DDX21 may serve as a promising therapeutic target for kidney fibrosis.
Collapse
Affiliation(s)
- Maoqing Tian
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaofei Wang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Meng Zhang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Chen Li
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yuhan Xu
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xinghua Chen
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Hubei Provincial Clinical Research Center for Kidney Disease, Wuhan 430060, China
| | - Cheng Chen
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Hubei Provincial Clinical Research Center for Kidney Disease, Wuhan 430060, China
| | - Zhongping Wei
- Department of Nephrology, Hubei Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan 430060, China
| | - Xiaoyan Li
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Guohua Ding
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Hubei Provincial Clinical Research Center for Kidney Disease, Wuhan 430060, China
| | - Lu Zhang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Hubei Provincial Clinical Research Center for Kidney Disease, Wuhan 430060, China.
| | - Huiming Wang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Hubei Provincial Clinical Research Center for Kidney Disease, Wuhan 430060, China.
| | - Hua Gan
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
73
|
Cao JF, Zhang X, Xia Q, Hang K, Men J, Tian J, Liao D, Xia Z, Li K. Insights into curcumin's anticancer activity in pancreatic ductal adenocarcinoma: Experimental and computational evidence targeting HRAS, CCND1, EGFR and AKT1. Bioorg Chem 2025; 157:108264. [PMID: 39954354 DOI: 10.1016/j.bioorg.2025.108264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
PURPOSE Curcumin, as a natural polyphenolic compound, possesses antitumor, antioxidant properties and anti-inflammatory. Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor, and there is a lack of molecular mechanisms and therapeutic options regarding relevant therapeutic agents. Therefore, we investigated the mechanism of curcumin inhibiting pancreatic cancer growth by modulating proliferation of cells and cellular metabolism. METHODS Bioinformatics analysis was involved in analyzing the intersecting targets of curcumin and pancreatic ductal adenocarcinoma. The effect of curcumin on proliferation of PANC-1 cells was tested by CCK-8, and total RNA from PANC-1 was also analysed by transcriptome sequencing. Molecular docking was involved in verifying binding stability of curcumin to protein targets. Molecular dynamics simulated and evaluated binding free energy, hydrogen bonds and root mean square fluctuation of the complex. RESULTS PPI, GO and KEGG were involved in screening and analysing key interacting protein targets. 40 μg/mL curcumin significantly inhibited the growth and proliferation of PANC-1. Transcriptome sequencing results showed gene expression of Cyclin D1 (CCND1), AKT serine/threonine kinase 1 (AKT1), HRas proto-oncogene (HRAS), epidermal growth factor receptor (EGFR) was significantly down-regulated by curcumin treatment. Result of molecular dynamics and molecular docking inhibited the free binding energies of CCND1/Curcumin, HRAS/Curcumin, AKT1/Curcumin and EGFR/Curcumin were -21.13 ± 3.41 kcal/mol, -21.84 ± 4.38 kcal/mol, -20.61 ± 1.82 kcal/mol and -27.37 ± 1.94 kcal/mol, respectively. CONCLUSION We found curcumin may not only regulate cell cycle progression in PDAC and apoptosis by down-regulating HRAS, thereby inhibiting CCND1 and its downstream signaling pathways, but also inhibit energy metabolism reprogramming, Ras-RAF-MEK-ERK and other downstream signalling pathways by down-regulating EGFR and AKT1, thereby affecting tumor cell metastasis, survival and proliferation.
Collapse
Affiliation(s)
- Jun-Feng Cao
- College of Medicine, Southwest Jiaotong University, Chengdu 610031 Sichuan, China
| | - Xiao Zhang
- Chengdu Medical College, Chengdu 610500 Sichuan, China
| | - Qingjie Xia
- Institute of Neurological Diseases, Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041 Sichuan, China
| | - Kuan Hang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041 Sichuan, China
| | - Jie Men
- College of Medicine, Southwest Jiaotong University, Chengdu 610031 Sichuan, China
| | - Jin Tian
- College of Medicine, Southwest Jiaotong University, Chengdu 610031 Sichuan, China
| | - Dunshui Liao
- Institute of Neurological Diseases, Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041 Sichuan, China
| | - Zengliang Xia
- Institute of Neurological Diseases, Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041 Sichuan, China
| | - Kezhou Li
- College of Medicine, Southwest Jiaotong University, Chengdu 610031 Sichuan, China; Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041 Sichuan, China.
| |
Collapse
|
74
|
Zeng C, Chen X, Lin M, Jin Y, Guo Q, Zhou T, Wang X, Li Y, Wang X, Han Y, Du L, Tang Q, Liu P, Zhang J. Overcoming matrix barriers for enhanced immune infiltration using siRNA-coated metal-organic frameworks. Acta Biomater 2025; 196:410-422. [PMID: 40054648 DOI: 10.1016/j.actbio.2025.03.001] [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: 10/21/2024] [Revised: 02/27/2025] [Accepted: 03/02/2025] [Indexed: 03/16/2025]
Abstract
The extracellular matrix (ECM) of solid tumor constitutes a formidable physical barrier that impedes immune cell infiltration, contributing to immunotherapy resistance. Breast cancer, particularly triple-negative breast cancer (TNBC), is characterized by a collagen-rich tumor microenvironment, which is associated with T cell exclusion and poor therapeutic outcomes. Discoidin domain receptor 2 (DDR2) and integrins, key ECM regulatory receptors on cancer cells, play pivotal role in maintaining this barrier. In this study, we developed a dual-receptor-targeted strategy using metal-organic frameworks (MOFs) to deliver DDR2-specific siRNA (siDDR2) and ITGAV-specific siRNA (siITGAV) to disrupt the ECM barrier. siDDR2 modulates immune infiltration by regulating collagen-cell interactions, while siITGAV suppresses TGF-β1 activation. The MOF@siDDR2+siITGAV complex significantly reduced collagen deposition, enhanced CD8+ T cell infiltration, and downregulated programmed cell death ligand 1 (PD-L1) expression in TNBC. Consequently, this approach markedly inhibited tumor growth. Our findings demonstrate that dual-receptor-targeted MOF-based nanocarriers (MOF@siDDR2+siITGAV) can effectively reprogram the tumor ECM to enhance immune cell access, offering a promising prospect for synergistic cancer immunotherapy. STATEMENT OF SIGNIFICANCE: A dual-receptor-targeted MOF nanocarrier is developed to improve immune accessibility in tumors. Concurrent blockade of DDR2 and ITGAV effectively decreases collagen deposition, increases CD8+ T cell infiltration, and suppresses PD-L1 expression. Modulating the mechanical properties of the extracellular matrix (ECM) to enhance immune accessibility offers an innovative strategy for cancer treatment.
Collapse
Affiliation(s)
- Cheng Zeng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaojing Chen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China; Central Laboratory, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Mingxi Lin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yizi Jin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qing Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Teng Zhou
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xingang Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Yiping Li
- Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Xinghui Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Yongming Han
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Ling Du
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Qianyun Tang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China.
| | - Peifeng Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China; Central Laboratory, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
75
|
Lan J, Cai D, Gou S, Bai Y, Lei H, Li Y, Chen Y, Zhao Y, Shen J, Wu X, Li M, Chen M, Li X, Sun Y, Gu L, Li W, Wang F, Cho CH, Zhang Y, Zheng X, Xiao Z, Du F. The dynamic role of ferroptosis in cancer immunoediting: Implications for immunotherapy. Pharmacol Res 2025; 214:107674. [PMID: 40020885 DOI: 10.1016/j.phrs.2025.107674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/14/2025] [Accepted: 02/23/2025] [Indexed: 03/03/2025]
Abstract
Currently, cancer immunotherapy strategies are primarily formulated based on the patient's present condition, representing a "static" treatment approach. However, cancer progression is inherently "dynamic," as the immune environment is not fixed but undergoes continuous changes. This dynamism is characterized by the ongoing interactions between tumor cells and immune cells, which ultimately lead to alterations in the tumor immune microenvironment. This process can be effectively elucidated by the concept of cancer immunoediting, which divides tumor development into three phases: "elimination," "equilibrium," and "escape." Consequently, adjusting immunotherapy regimens based on these distinct phases may enhance patient survival and improve prognosis. Targeting ferroptosis is an emerging area in cancer immunotherapy, and our findings reveal that the antioxidant systems associated with ferroptosis possess dual roles, functioning differently across the three phases of cancer immunoediting. Therefore, this review delve into the dual role of the ferroptosis antioxidant system in tumor development and progression. It also propose immunotherapy strategies targeting ferroptosis at different stages, ultimately aiming to illuminate the significant implications of targeting ferroptosis at various phases for cancer immunotherapy.
Collapse
Affiliation(s)
- Jiarui Lan
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Dan Cai
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Shuang Gou
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China
| | - Yulin Bai
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China
| | - Huaqing Lei
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Yan Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Meijuan Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Li Gu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Fang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yan Zhang
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, China
| | - Xin Zheng
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, China.
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China.
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China.
| |
Collapse
|
76
|
Li L, Liu Y, Wang K, Mo J, Weng Z, Jiang H, Jin C. Stem cell exosomes: new hope and future potential for relieving liver fibrosis. Clin Mol Hepatol 2025; 31:333-349. [PMID: 39510097 PMCID: PMC12016649 DOI: 10.3350/cmh.2024.0854] [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: 09/29/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/15/2024] Open
Abstract
Liver fibrosis is a chronic liver injury resulting from factors like viral hepatitis, autoimmune hepatitis, non-alcoholic steatohepatitis, fatty liver disease, and cholestatic liver disease. Liver transplantation is currently the gold standard for treating severe liver diseases. However, it is limited by a shortage of donor organs and the necessity for lifelong immunosuppressive therapy. Mesenchymal stem cells (MSCs) can differentiate into various liver cells and enhance liver function when transplanted into patients due to their differentiation and proliferation capabilities. Therefore, it can be used as an alternative therapy for treating liver diseases, especially for liver cirrhosis, liver failure, and liver transplant complications. However, due to the potential tumorigenic effects of MSCs, researchers are exploring a new approach to treating liver fibrosis using extracellular vesicles (exosomes) secreted by stem cells. Many studies show that exosomes released by stem cells can promote liver injury repair through various pathways, contributing to the treatment of liver fibrosis. In this review, we focus on the molecular mechanisms by which stem cell exosomes affect liver fibrosis through different pathways and their potential therapeutic targets. Additionally, we discuss the advantages of exosome therapy over stem cell therapy and the possible future directions of exosome research, including the prospects for clinical applications and the challenges to be overcome.
Collapse
Affiliation(s)
- Lihua Li
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Yongjie Liu
- Department of Cell biology, School of Medicine, Taizhou University, Taizhou, Zhejiang Province, P. R. China
- Department of Pathophysiology, School of Basic Medicine, Shenyang Medical College, Shenyang, Liaoning Province, P. R. China
| | - Kunpeng Wang
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Jinggang Mo
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Zhiyong Weng
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Hao Jiang
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Chong Jin
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| |
Collapse
|
77
|
Tang X, Lu L, Li X, Huang P. Bridging Cancer and COVID-19: The Complex Interplay of ACE2 and TMPRSS2. Cancer Med 2025; 14:e70829. [PMID: 40145441 PMCID: PMC11947763 DOI: 10.1002/cam4.70829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/09/2025] [Accepted: 03/16/2025] [Indexed: 03/28/2025] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic presents heightened risks for cancer patients, who are more susceptible to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and severe outcomes due to immunosuppression from both the malignancy and anticancer therapies. This review investigates the dual roles of angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2) in SARS-CoV-2 infection among cancer patients. ACE2, the vital entry receptor for SARS-CoV-2, is overexpressed in certain tumors such as colon adenocarcinoma, renal carcinomas, pancreatic adenocarcinoma, and lung adenocarcinoma, potentially increasing viral susceptibility. Paradoxically, ACE2 also exhibits tumor-suppressive properties by inhibiting angiogenesis and modulating the tumor microenvironment, leading to improved patient prognoses in some cancers like breast cancer. TMPRSS2, essential for viral entry, shows decreased expression in several tumors but acts as a prognostic biomarker in prostate and lung cancers. This review illustrates the complexity of therapeutically targeting ACE2 and TMPRSS2 due to their contrasting roles in cancer progression and viral entry. We analyze the expression levels of ACE2 and TMPRSS2 in relation to immune cell infiltration and patient outcomes, and propose personalized therapeutic strategies. Furthermore, we underscore the necessity for multidisciplinary approaches, integrating antiviral treatments with cancer therapies and tailoring interventions based on individual molecular profiles. This approach to personalized medicine seeks to enhance treatment results and better manage cancer patients who have contracted SARS-CoV-2.
Collapse
Affiliation(s)
- Xuerui Tang
- School of Basic MedicineGannan Medical UniversityGanzhouJiangxiChina
| | - Liuzhi Lu
- School of Basic MedicineGannan Medical UniversityGanzhouJiangxiChina
| | - Xiaoping Li
- Clinical LaboratoryTongxiang First People's HospitalZhejiangChina
| | - Panpan Huang
- School of Basic MedicineGannan Medical UniversityGanzhouJiangxiChina
| |
Collapse
|
78
|
Yang S, Fang Y, Ma Y, Wang F, Wang Y, Jia J, Yang Y, Sun W, Zhou Q, Li Z. Angiogenesis and targeted therapy in the tumour microenvironment: From basic to clinical practice. Clin Transl Med 2025; 15:e70313. [PMID: 40268524 PMCID: PMC12017902 DOI: 10.1002/ctm2.70313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
Angiogenesis, as a core marker of cancer survival and growth, is integral to the processes of tumour growth, invasion and metastasis. In recent years, targeted angiogenesis treatment strategies have gradually become an important direction in cancer treatment. Single-cell sequencing technology can provide new insights into targeted angiogenesis by providing a deeper understanding of the heterogeneity of tumour endothelial cells and exploring the interactions between endothelial cells and surrounding cells in the tumour microenvironment. Here, we systematically review the research progress in endothelial cell pathophysiology and its endothelial‒mesenchymal transition and illustrate the heterogeneity of endothelial cells from a single-cell perspective. Finally, we examine the contributions of different cell types within the tumour microenvironment in relation to tumour angiogenesis, as well as the latest progress and strategies in targeted angiogenesis therapy, hoping to provide useful insights into the clinical application of antiangiogenic treatment. Furthermore, a summary of the present progress in the development of potential angiogenesis inhibitors and the ongoing clinical trials for combination therapies is provided. KEY POINTS: Angiogenesis plays a key role in tumour progression, invasion and metastasis, so strategies targeting angiogenesis are gradually becoming an important direction in cancer therapy. Interactions between endothelial cells and stromal cells and immune cells in the tumour microenvironment are significant in angiogenesis. The application of antiangiogenic immunotherapy and nanotechnology in antiangiogenic therapy provides a vital strategy for prolonging the survival of cancer patients.
Collapse
Affiliation(s)
- Shuaixi Yang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yingshuai Fang
- The First Clinical School of MedicineZhengzhou UniversityZhengzhouChina
| | - Yangcheng Ma
- Department of OrthopedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Fuqi Wang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yuhang Wang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jiachi Jia
- The First Clinical School of MedicineZhengzhou UniversityZhengzhouChina
| | - Yabing Yang
- The First Clinical School of MedicineZhengzhou UniversityZhengzhouChina
| | - Weipeng Sun
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Quanbo Zhou
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhen Li
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
79
|
Fan Y, Gao L, Huang Y, Zhao L, Zhao Y, Wang X, Mo D, Lu H, Wang D. Effects and Significance of Dicliptera chinensis Polysaccharide on the Expression of Transforming Growth Factor β1/Connective Tissue Growth Factor Pathway in the Masseter and Head and Neck Skin of Rats With Radiation-Induced Fibrosis. Int Dent J 2025; 75:784-796. [PMID: 38991877 PMCID: PMC11976479 DOI: 10.1016/j.identj.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/26/2024] [Accepted: 06/13/2024] [Indexed: 07/13/2024] Open
Abstract
PURPOSE To investigate whether Dicliptera chinensis polysaccharide (DCP) can alleviate radiation-induced fibrosis of masseter and head and neck skin. METHODS SD rats were divided into the control, the irradiation (IR), the IR + low dose DCP (200 mg/kg), and the IR + high dose DCP (400 mg/kg) groups. The head and neck of rats in the last 3 groups received a single dose of 18 Gy X-ray. At 1st, 2nd, 4th week (w) after radiation, haematoxylin and eosin staining were performed on masseter and skin to observe the histopathological changes; immunohistochemistry staining was performed to observe the pathological changes of the skin; Masson staining was performed on masseter and skin to observe the collagen deposition; western blot analysis was used on masseter to calculate the relative transforming growth factor β1 (TGF-β1), connective tissue growth factor (CTGF) expressions; ELISA was used to detect the contents of TGF-β1 and CTGF in skin and the contents of type I and type III collagens in masseter and skin. RESULTS In terms of skin, compared to the IR group, the IR + high-dose DCP group exhibited relatively smaller changes in skin structure, lower levels of TGF-β1 and CTGF; thinner skin thickness was observed at the 4th w after radiation; and the positive rates of collagen fibre and the optical densities of type I and type III collagens were lower at the 2nd and 4th w. For the masseter, compared to the IR group, the morphological changes were improved and the expression levels of TGF-β1 and CTGF proteins decreased in the 2 DCP dose groups at 2nd and 4th w. CONCLUSION DCP can reduce the formation and accumulation of type I and type III collagens after IR and ameliorate radiation-induced fibrosis of masseter and skin by down-regulating the expressions of TGF-β1 and CTGF.
Collapse
Affiliation(s)
- Yiyang Fan
- College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China; Guangxi Key Laboratory of the Rehabilitation and Reconstruction for Oral and Maxillofacial Research, Guangxi Clinical Research Center for Craniofacial Deformity, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Deformity, Nanning, China; Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
| | - Linjing Gao
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yude Huang
- College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China; Guangxi Key Laboratory of the Rehabilitation and Reconstruction for Oral and Maxillofacial Research, Guangxi Clinical Research Center for Craniofacial Deformity, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Deformity, Nanning, China
| | - Lixiang Zhao
- College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China; Guangxi Key Laboratory of the Rehabilitation and Reconstruction for Oral and Maxillofacial Research, Guangxi Clinical Research Center for Craniofacial Deformity, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Deformity, Nanning, China
| | - Yanfei Zhao
- College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China; Guangxi Key Laboratory of the Rehabilitation and Reconstruction for Oral and Maxillofacial Research, Guangxi Clinical Research Center for Craniofacial Deformity, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Deformity, Nanning, China
| | - Xian Wang
- College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China; Guangxi Key Laboratory of the Rehabilitation and Reconstruction for Oral and Maxillofacial Research, Guangxi Clinical Research Center for Craniofacial Deformity, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Deformity, Nanning, China
| | - Dongqin Mo
- College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China; Guangxi Key Laboratory of the Rehabilitation and Reconstruction for Oral and Maxillofacial Research, Guangxi Clinical Research Center for Craniofacial Deformity, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Deformity, Nanning, China
| | - Haoyu Lu
- College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China; Guangxi Key Laboratory of the Rehabilitation and Reconstruction for Oral and Maxillofacial Research, Guangxi Clinical Research Center for Craniofacial Deformity, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Deformity, Nanning, China
| | - Daiyou Wang
- College of Stomatology, Hospital of Stomatology, Guangxi Medical University, Nanning, China; Guangxi Key Laboratory of the Rehabilitation and Reconstruction for Oral and Maxillofacial Research, Guangxi Clinical Research Center for Craniofacial Deformity, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Deformity, Nanning, China.
| |
Collapse
|
80
|
Vasuthas K, Kjesbu JS, Brambilla A, Levitan M, Coron AE, Fonseca DM, Strand BL, Slupphaug G, Rokstad AMA. Fucoidan alginate and sulfated alginate microbeads induce distinct coagulation, inflammatory and fibrotic responses. Mater Today Bio 2025; 31:101474. [PMID: 39896282 PMCID: PMC11783016 DOI: 10.1016/j.mtbio.2025.101474] [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: 10/23/2024] [Revised: 12/20/2024] [Accepted: 01/08/2025] [Indexed: 02/04/2025] Open
Abstract
This study investigates the host response to fucoidan alginate microbeads in comparison to sulfated alginate microbeads, which are relevant for immune protection in cell therapy. While sulfated alginate microbeads reduce fibrosis and inflammation, fucoidan, a kelp-derived polysaccharide rich in sulfate groups, has not been evaluated in this context. The study assesses surface reactivity to acute-phase proteins and cytokines using ex vivo human whole blood and plasma models. It also examines pericapsular overgrowth (PFO) in C57BL/6JRj mice, incorporating protein pattern mapping through LC-MS/MS proteomics. Fucoidan alginate microbeads activated complement and coagulation, while both fucoidan and sulfated alginate microbeads induced plasmin activity. Fucoidan alginate microbeads exhibited a distinct cytokine profile, characterized by high levels of MCP-1, IL-8, IFN-γ, and reduced levels of RANTES, Eotaxin, PDGF-BB, TGF-β isoforms, along with higher PFO. The balance between plasmin activity and coagulation emerged as a potential predictor of fibrosis resistance, favouring sulfated alginate microbeads. Explanted materials were enriched with both complement and coagulation activators (Complement C1q and C3, Factor 12, Kallikrein, HMW-kininogen) and inhibitors (C1-inhibitor, Factor H, Factor I). Fucoidan alginate microbeads predominantly enriched extracellular matrix factors (Fibrinogen, Collagen, TGF-β, Bmp), while sulfated alginate microbeads favoured ECM-degrading proteases (Metalloproteases and Cathepsins). This study reveals significant differences in host responses to fucoidan and sulfated alginate in microbeads. The plasmin activity to coagulation ratio is highlighted as a key indicator of fibrosis resistance. Additionally, the preferential enrichment of ECM-degrading proteases on the material surface post-implantation proved to be another crucial factor.
Collapse
Affiliation(s)
- Kalaiyarasi Vasuthas
- Centre of Molecular Inflammation Research (CEMIR), NTNU, Norway
- Department of Clinical and Molecular Medicine, NTNU, Norway
| | | | - Alessandro Brambilla
- Department of Clinical and Molecular Medicine, NTNU, Norway
- Clinic of Laboratory Medicine, St Olavs Hospital, Trondheim, Norway
- Proteomics and Modomics Experimental Core (PROMEC), NTNU and the Central Norway Health Authority, Norway
| | - Maya Levitan
- Centre of Molecular Inflammation Research (CEMIR), NTNU, Norway
- Department of Clinical and Molecular Medicine, NTNU, Norway
| | | | - Davi M. Fonseca
- Department of Clinical and Molecular Medicine, NTNU, Norway
- Clinic of Laboratory Medicine, St Olavs Hospital, Trondheim, Norway
- Proteomics and Modomics Experimental Core (PROMEC), NTNU and the Central Norway Health Authority, Norway
| | | | - Geir Slupphaug
- Department of Clinical and Molecular Medicine, NTNU, Norway
- Clinic of Laboratory Medicine, St Olavs Hospital, Trondheim, Norway
- Proteomics and Modomics Experimental Core (PROMEC), NTNU and the Central Norway Health Authority, Norway
| | - Anne Mari A. Rokstad
- Centre of Molecular Inflammation Research (CEMIR), NTNU, Norway
- Department of Clinical and Molecular Medicine, NTNU, Norway
- Clinic of Laboratory Medicine, St Olavs Hospital, Trondheim, Norway
| |
Collapse
|
81
|
Himani, Kaur C, Kumar R, Mishra R, Singh G. Targeting TGF-β: a promising strategy for cancer therapy. Med Oncol 2025; 42:142. [PMID: 40155496 DOI: 10.1007/s12032-025-02667-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
Transforming growth factor β (TGF-β) has important role in regulating the cellular processes including cell growth, differentiation, and migration. TGF-β exerts its effect by binding with transcellular membranes and kinases. Our findings demonstrate that TGF- β possess dual role as tumor suppressor and tumor promoter in different stages of cancer. TGF-β emerged as a promising anticancer agent that exhibits the apoptosis by acting on the suppressor of mothers against decapentaplegic (SMAD) and non-SMAD pathways. In this review we are focusing on the different types of TGF- β inhibitors active against skin cancer, breast cancer, colorectal cancer, lung cancer and ovarian cancer. TGF-β inhibitors includes ligand traps, monoclonal antibodies and receptor kinase inhibitors. In recent studies, TGF- β inhibitors have also been used in combination therapies in the treatment of cancer. The TGF-β has important role in vaccine therapy, Chemo and Radio Resistance in Cancer. TGF-β inhibitors present the novel therapeutic approach for the cancer therapy, highlighting the mechanism of action involved, clinical trials, challenges and exploring therapeutic opportunities. This will help to develop the novel TGF-β inhibitors as anticancer agents as well as help to resolve the problem of drug resistance by developing new drugs as anticancer agents.
Collapse
Affiliation(s)
- Himani
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Charanjit Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Rajesh Kumar
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Rakhi Mishra
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh, India
| | - Gurvinder Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India.
| |
Collapse
|
82
|
Liu W, Wang B, Wei M, Hai Z. In situ self-assembled peptide nanoparticles improve the anti-hepatic fibrosis effect. J Mater Chem B 2025; 13:4080-4084. [PMID: 40079763 DOI: 10.1039/d4tb02819h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Antagonistic peptide Leu-Ser-Lys-Leu (LSKL) is capable of blocking the transforming growth factor-β1 (TGF-β1) signaling pathway and exhibits anti-fibrotic effects. Herein, we constructed LSKL nanoparticles (NPs) in situ based on an alkaline phosphatase (ALP)-instructed self-assembly strategy for improving its specific therapeutic effect against liver fibrosis.
Collapse
Affiliation(s)
- Wenbin Liu
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China.
| | - Beibei Wang
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China.
| | - Mengxing Wei
- Department of Radiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
- Medical Imagine Research Center, Anhui Medical University, Hefei, Anhui 230601, China
| | - Zijuan Hai
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China.
| |
Collapse
|
83
|
Lai G, Zhao X, Chen Y, Xie T, Su Z, Lin J, Chen Y, Chen K. The origin and polarization of Macrophages and their role in the formation of the Pre-Metastatic niche in osteosarcoma. Int Immunopharmacol 2025; 150:114260. [PMID: 39938167 DOI: 10.1016/j.intimp.2025.114260] [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: 10/26/2024] [Revised: 01/21/2025] [Accepted: 02/06/2025] [Indexed: 02/14/2025]
Abstract
Osteosarcoma, a primary malignant bone tumor commonly found in adolescents, is highly aggressive, with a high rate of disability and mortality. It has a profound negative impact on both the physical and psychological well-being of patients. The standard treatment approach, comprising surgery and chemotherapy, has seen little improvement in patient outcomes over the past several decades. Once relapse or metastasis occurs, prognosis worsens significantly. Therefore, there is an urgent need to explore new therapeutic approaches. In recent years, the successful application of immunotherapy in certain cancers has demonstrated its potential in the field of cancer treatment. Macrophages are the predominant components of the immune microenvironment in osteosarcoma and represent critical targets for immunotherapy. Macrophages exhibit dual characteristics; while they play a key role in maintaining tumor-promoting properties within the microenvironment, such as inflammation, angiogenesis, and immune suppression, they also possess antitumor potential as part of the innate immune system. A deeper understanding of macrophages and their relationship with osteosarcoma is essential for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Guisen Lai
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Xinyi Zhao
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Yuanquan Chen
- Department of Orthopaedic Sun Yat-sen Memorial Hospital Sun Yat-sen University PR China
| | - Tianwei Xie
- The People's Hospital of Hezhou, No.150 Xiyue Street, Hezhou 542800 PR China
| | - Zepeng Su
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Jiajie Lin
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Yuanhai Chen
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Keng Chen
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China.
| |
Collapse
|
84
|
Jimenez-Socha M, Dion GR, Mora-Navarro C, Wang Z, Nolan MW, Freytes DO. Radiation-Induced Fibrosis in Head and Neck Cancer: Challenges and Future Therapeutic Strategies for Vocal Fold Treatments. Cancers (Basel) 2025; 17:1108. [PMID: 40227628 PMCID: PMC11987993 DOI: 10.3390/cancers17071108] [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: 02/07/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 04/15/2025] Open
Abstract
Head and neck cancer encompasses a diverse group of malignant neoplasms originating in regions such as the oral cavity, oropharynx, hypopharynx, larynx, sinonasal cavities, and salivary glands. HNC represents a significant public health challenge, and recent reports indicate an increment in the incidence of HNC in young adults. In 2020, approximately 377,700 new HNC cases and 177,800 HNC-related deaths were reported globally. Major risk factors include tobacco smoking, alcohol consumption, and human papillomavirus (HPV) infections. HNC impacts vital functions such as breathing, swallowing, and speech. Treatments for this type of cancer within this complex anatomy include surgery, radiotherapy, and chemotherapy combinations. Radiotherapy is often an essential component of both curative and palliative HNC treatment, balancing tumor control with the preservation of function and appearance. However, its use can damage adjacent normal tissues, causing acute or chronic toxicity. One complication of HNC irradiation is VF fibrosis, which leads to severe voice impairments, significantly affecting patients' quality of life. Fibrosis involves excessive and aberrant deposition of extracellular matrix, driven by factors such as TGF-β1 and inflammatory cytokines, which ultimately impair the flexibility and function of VF. Current radiation-induced fibrosis treatments primarily focus on symptom management and include systemic therapies like corticosteroids, anti-inflammatory drugs, and antioxidants. However, these treatments have limited efficacy. Experimental approaches targeting molecular pathways involved in fibrosis are being explored. Given the limitations of these treatments, advancing research is crucial to develop more effective therapeutic strategies that can significantly improve the quality of life for HNC patients, especially those vulnerable to VF fibrosis.
Collapse
Affiliation(s)
- Maria Jimenez-Socha
- Lampe Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina-Chapel Hill, Raleigh, NC 27606, USA; (M.J.-S.); (Z.W.)
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27606, USA;
| | - Gregory R. Dion
- Department of Otolaryngology-Head & Neck Surgery, University of Cincinnati, Cincinnati, OH 45267, USA;
| | - Camilo Mora-Navarro
- Department of Chemical Engineering, University of Puerto Rico-Mayaguez, Mayagüez, PR 00680, USA;
| | - Ziyu Wang
- Lampe Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina-Chapel Hill, Raleigh, NC 27606, USA; (M.J.-S.); (Z.W.)
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27606, USA;
| | - Michael W. Nolan
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27606, USA;
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC 27606, USA
| | - Donald O. Freytes
- Lampe Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina-Chapel Hill, Raleigh, NC 27606, USA; (M.J.-S.); (Z.W.)
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27606, USA;
| |
Collapse
|
85
|
Jiang N, Jiang M, Chen J, Mohsin A, Mu Y, Yi X, Zhuang Y, Qian J, Huang J. Living hybrid material based on probiotic with photothermal properties inhibits PD-L1 expression after tumouricidal photothermal therapy. BIOMATERIALS TRANSLATIONAL 2025; 6:73-84. [PMID: 40313568 PMCID: PMC12041808 DOI: 10.12336/biomatertransl.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/22/2024] [Accepted: 02/25/2025] [Indexed: 05/03/2025]
Abstract
Photothermal therapy is a safe and effective tumour treatment strategy due to its excellent spatiotemporal controllability. However, interferon gamma in the tumour microenvironment is upregulated after photothermal therapy, which enhances the expression of programmed cell death ligand 1 (PD-L1) in tumour cells. This further promotes immunosuppression and tumour metastasis, resulting in a poor prognosis in cancer therapy. Traditional nanodrugs often face challenges in penetrating the dense extracellular matrix of solid tumours, whereas certain probiotics possess the ability to specifically colonise the core regions of tumours. In this research, we used Escherichia coli Nissle 1917 (ECN) as a chassis cell and self-assembly polydopamine (PDA) on the ECN surface. The black PDA@ECN (notes as PE) actively colonises at the tumour site and produces a photothermal effect under 808 nm laser irradiation to kill tumour cells. To overcome the high expression of PD-L1 induced after photothermal therapy, metformin (MET) was also encapsulated in PE to form PDA@MET@ECN (notes as PME). In vivo experiments demonstrated that PME effectively inhibited the PD-L1 expression and growth of CT26 tumour cells. Overall, PME reverses the immunosuppressive tumour microenvironment and enhances the effect of photothermal/immune therapy in tumour treatment.
Collapse
Affiliation(s)
- Ning Jiang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Mingyan Jiang
- College of Life Sciences, Jiangxi Normal University, Nanchang, Jiangxi Province, China
| | - Jianshu Chen
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Ali Mohsin
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yuqing Mu
- Institute for biomedicine and glycomics, Griffith University School of Medicine and Dentistry, Griffith University, Queensland, Australia
| | - Xiaoping Yi
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yingping Zhuang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Jiangchao Qian
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Jiaofang Huang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- College of Life Sciences, Jiangxi Normal University, Nanchang, Jiangxi Province, China
| |
Collapse
|
86
|
Marques-Magalhães Â, Monteiro-Ferreira S, Canão PA, Rios E, Costa ÂM, Castro F, Velho S, Paredes J, Carneiro F, Oliveira MJ, Cardoso AP. Patient-Derived Colorectal Cancer Extracellular Matrices Modulate Cancer Cell Stemness Markers. Int J Mol Sci 2025; 26:2890. [PMID: 40243470 PMCID: PMC11988371 DOI: 10.3390/ijms26072890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/14/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Although it has been shown that the tumor extracellular matrix (ECM) may sustain the cancer stem cell (CSC) niche, its role in the modulation of CSC properties remains poorly characterized. To elucidate this, paired tumor and adjacent normal mucosa, derived from colon cancer patients' surgical resections, were decellularized and recellularized with two distinct colon cancer cells, HT-29 or HCT-15. Methods: The matrix impact on cancer stem cell marker expression was evaluated by flow cytometry and qRT-PCR, while transforming growth factor-β (TGF-β) secretion and matrix metalloprotease (MMP) activity were quantified by ELISA and zymography. Results: In contrast to their paired normal counterparts, the tumor decellularized matrices enhanced HT-29 expression of the pluripotency and stemness genes NANOG (p = 0.0117), SOX2 (p = 0.0156), and OCT4 (p = 0.0312) and of the epithelial-to-mesenchymal transition (EMT)-associated transcription factor SNAI1 (p = 0.0156). Notably, no significant differences were found in the expression of SLUG or TGFB on HT-29 or of the six transcripts on HCT-15 cells. HT-29 mRNA alterations were followed by enhanced expression of the stemness-associated receptors cluster of differentiation 44 (CD44), CD133, and CD166 (p = 0.0078), the secretion of TGF-β (p = 0.0286), and MMP-2 (p = 0.0081) and MMP-9 (p = 0.0402) proteolysis. To infer the clinical relevance of these findings, we assessed cohort databases and evidenced that patients expressing higher levels of the four stemness-associated genes (NANOG/SOX2/OCT4/SNAI1) had worse overall survival. This study demonstrates that normal and tumor matrices harbor different stemness potential and suggest patient-derived decellularized matrices as an excellent three-dimensional (3D) model to unveil stemness signatures, appointing candidates for future therapeutic strategies.
Collapse
Affiliation(s)
- Ângela Marques-Magalhães
- i3S—Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (Â.M.-M.); (S.M.-F.); (E.R.); (Â.M.C.); (F.C.); (S.V.); (J.P.); (F.C.); (A.P.C.)
- ICBAS—School of Medicine and Biomedical Sciences, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Sara Monteiro-Ferreira
- i3S—Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (Â.M.-M.); (S.M.-F.); (E.R.); (Â.M.C.); (F.C.); (S.V.); (J.P.); (F.C.); (A.P.C.)
- Champalimaud Research, Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Pedro Amoroso Canão
- Centro Hospitalar Universitário São João, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal;
| | - Elisabete Rios
- i3S—Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (Â.M.-M.); (S.M.-F.); (E.R.); (Â.M.C.); (F.C.); (S.V.); (J.P.); (F.C.); (A.P.C.)
- Centro Hospitalar Universitário São João, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal;
- FMUP—Faculty of Medicine of the University of Porto, Pathology Department, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Ângela Margarida Costa
- i3S—Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (Â.M.-M.); (S.M.-F.); (E.R.); (Â.M.C.); (F.C.); (S.V.); (J.P.); (F.C.); (A.P.C.)
| | - Flávia Castro
- i3S—Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (Â.M.-M.); (S.M.-F.); (E.R.); (Â.M.C.); (F.C.); (S.V.); (J.P.); (F.C.); (A.P.C.)
| | - Sérgia Velho
- i3S—Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (Â.M.-M.); (S.M.-F.); (E.R.); (Â.M.C.); (F.C.); (S.V.); (J.P.); (F.C.); (A.P.C.)
| | - Joana Paredes
- i3S—Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (Â.M.-M.); (S.M.-F.); (E.R.); (Â.M.C.); (F.C.); (S.V.); (J.P.); (F.C.); (A.P.C.)
- FMUP—Faculty of Medicine of the University of Porto, Pathology Department, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Fátima Carneiro
- i3S—Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (Â.M.-M.); (S.M.-F.); (E.R.); (Â.M.C.); (F.C.); (S.V.); (J.P.); (F.C.); (A.P.C.)
- Centro Hospitalar Universitário São João, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal;
- FMUP—Faculty of Medicine of the University of Porto, Pathology Department, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Maria José Oliveira
- i3S—Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (Â.M.-M.); (S.M.-F.); (E.R.); (Â.M.C.); (F.C.); (S.V.); (J.P.); (F.C.); (A.P.C.)
- ICBAS—School of Medicine and Biomedical Sciences, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- FMUP—Faculty of Medicine of the University of Porto, Pathology Department, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Ana Patrícia Cardoso
- i3S—Institute for Research and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (Â.M.-M.); (S.M.-F.); (E.R.); (Â.M.C.); (F.C.); (S.V.); (J.P.); (F.C.); (A.P.C.)
| |
Collapse
|
87
|
Zhao Z, Zhu Y, Wan D. Exercise and tissue fibrosis: recent advances in therapeutic potential and molecular mechanisms. Front Endocrinol (Lausanne) 2025; 16:1557797. [PMID: 40182630 PMCID: PMC11965137 DOI: 10.3389/fendo.2025.1557797] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Tissue fibrosis represents an aberrant repair process, occurring because of prolonged injury, sustained inflammatory response, or metabolic disorders. It is characterized by an excessive accumulation of extracellular matrix (ECM), resulting in tissue hardening, structural remodeling, and loss of function. This pathological phenomenon is a common feature in the end stage of numerous chronic diseases. Despite the advent of novel therapeutic modalities, including antifibrotic agents, these have only modest efficacy in reversing established fibrosis and are associated with adverse effects. In recent years, a growing body of research has demonstrated that exercise has significant benefits and potential in the treatment of tissue fibrosis. The anti-fibrotic effects of exercise are mediated by multiple mechanisms, including direct inhibition of fibroblast activation, reduction in the expression of pro-fibrotic factors such as transforming growth factor-β (TGF-β) and slowing of collagen deposition. Furthermore, exercise has been demonstrated to assist in maintaining the dynamic equilibrium of tissue repair, thereby indirectly reducing tissue damage and fibrosis. It can also help maintain the dynamic balance of tissue repair by improving metabolic disorders, exerting anti-inflammatory and antioxidant effects, regulating cellular autophagy, restoring mitochondrial function, activating stem cell activity, and reducing cell apoptosis, thereby indirectly alleviating tissue. This paper presents a review of the therapeutic potential of exercise and its underlying mechanisms for the treatment of a range of tissue fibrosis, including cardiac, pulmonary, renal, hepatic, and skeletal muscle. It offers a valuable reference point for non-pharmacological intervention strategies for the comprehensive treatment of fibrotic diseases.
Collapse
Affiliation(s)
- Zheng Zhao
- School of Physical Education, Anyang Normal University, Anyang, Henan, China
| | - Yongjia Zhu
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Dongfeng Wan
- School of Health, Shanghai Normal University Tianhua College, Shanghai, China
| |
Collapse
|
88
|
Li Q, Xiao N, Zhang H, Liang G, Lin Y, Qian Z, Yang X, Yang J, Fu Y, Zhang C, Liu A. Systemic aging and aging-related diseases. FASEB J 2025; 39:e70430. [PMID: 40022602 DOI: 10.1096/fj.202402479rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/07/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Aging is a biological process along with systemic and multiple organ dysfunction. It is more and more recognized that aging is a systemic disease instead of a single-organ functional disorder. Systemic aging plays a profound role in multiple diseases including neurodegenerative diseases, cardiovascular diseases, and malignant diseases. Aged organs communicate with other organs and accelerate aging. Skeletal muscle, heart, bone marrow, skin, and liver communicate with each other through organ-organ crosstalk. The crosstalk can be mediated by metabolites including lipids, glucose, short-chain fatty acids (SCFA), inflammatory cytokines, and exosomes. Metabolic disorders including hyperglycemia, hyperinsulinemia, and hypercholesterolemia caused by chronic diseases accelerate hallmarks of aging. Systemic aging leads to the destruction of systemic hemostasis, causes the release of inflammatory cytokines, senescence-associated secretory phenotype (SASP), and the imbalance of microbiota composition. Released inflammatory factors further aggregate senescence, which promotes the aging of multiple solid organs. Targeting senescence or delaying aging is emerging as a critical health strategy for solving age-related diseases, especially in the old population. In the current review, we will delineate the mechanisms of organ crosstalk in systemic aging and age-related diseases to provide therapeutic targets for delaying aging.
Collapse
Affiliation(s)
- Qiao Li
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Nanyin Xiao
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Heng Zhang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Guangyu Liang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yan Lin
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Zonghao Qian
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Xiao Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jiankun Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yanguang Fu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Cuntai Zhang
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anding Liu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| |
Collapse
|
89
|
Wang S, Xu D, Xiao L, Liu B, Yuan X. Radiation-induced lung injury: from mechanism to prognosis and drug therapy. Radiat Oncol 2025; 20:39. [PMID: 40082925 PMCID: PMC11907960 DOI: 10.1186/s13014-025-02617-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/28/2025] [Indexed: 03/16/2025] Open
Abstract
Radiation induced lung injury, known as the main complication of thoracic radiation, remains to be a major resistance to tumor treatment. Based on the recent studies on radiation-induced lung injury, this review collated the possible mechanisms at the level of target cells and key pathways, corresponding prognostic models including predictors, patient size, number of centers, radiotherapy technology, construction methods and accuracy, and pharmacotherapy including drugs, targets, therapeutic effects, impact on anti-tumor treatment and research types. The research priorities and limitations are summarized to provide a reference for the research and management of radiation-induced lung injury.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu, 210000, China
| | - Duo Xu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Lingyan Xiao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
90
|
Shi Z, Liu J, Qin J, Liang X, Ou X, Zhang T, Yan X, Hu Q, Huang W, Hu K. Astilbin Alleviates Radiation-Induced Pulmonary Fibrosis via circPRKCE Targeting the TGF-β/Smad7 Pathway to Inhibit Epithelial-Mesenchymal Transition. Biomedicines 2025; 13:689. [PMID: 40149664 PMCID: PMC11939908 DOI: 10.3390/biomedicines13030689] [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: 02/05/2025] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025] Open
Abstract
Purpose: This study aimed to clarify the protective effect of astilbin (AST) on radiation-induced pulmonary fibrosis (RIPF) and explore its underlying molecular mechanism, focusing on non-coding RNAs. Methods: Mouse lung epithelial cells (MLE-12 and TC-1) and C57BL/6J mice were used to establish in vitro radiation injury models and in vivo RIPF models, respectively. Cell viability, apoptosis, the epithelial-to-mesenchymal transition (EMT), and fibrosis-related markers were assessed using cell-counting kit-8 assays, Western blotting, immunohistochemistry, and histological staining. High-throughput sequencing identified differentially expressed circRNAs. The mechanistic studies included RNA-FISH, a dual-luciferase reporter assay, an RNA immunoprecipitation (RIP) assay, and loss-of-function experiments. Results: AST significantly alleviated radiation-induced apoptosis and EMT in vitro, as well as RIPF in vivo. AST treatment reduced collagen deposition, fibrosis-related protein expression, and EMT marker changes. High-throughput sequencing revealed that AST upregulated circPRKCE, a non-coding RNA that functions through a ceRNA mechanism by binding to miR-15b-5p, thereby promoting Smad7 expression and suppressing the TGF-β/Smad7 pathway. Knockdown of circPRKCE abolished AST's protective effects, confirming its pivotal role in mediating AST's anti-fibrotic activity. Conclusions: This study demonstrates that Astilbin alleviates radiation-induced pulmonary fibrosis via circPRKCE targeting the TGF-β/Smad7 pathway to inhibit EMT, suggesting AST as a potential therapeutic agent for managing this severe complication of radiotherapy.
Collapse
Affiliation(s)
- Zhiling Shi
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning 530021, China
- State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Jing Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning 530021, China
- State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Jing Qin
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
| | - Xian Liang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
| | - Xue Ou
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China
| | - Tingting Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
| | - Xueting Yan
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
| | - Qianxin Hu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
| | - Weimei Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning 530021, China
- State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Kai Hu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning 530021, China
- State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
91
|
Du Z, Liu Q, Wang M, Gao Y, Li Q, Yang Y, Lu T, Bao L, Pang Y, Wang H, Niu Y, Zhang R. Reticulophagy promotes EMT-induced fibrosis in offspring's lung tissue after maternal exposure to carbon black nanoparticles during gestation by a m 5C-dependent manner. JOURNAL OF HAZARDOUS MATERIALS 2025; 485:136873. [PMID: 39694008 DOI: 10.1016/j.jhazmat.2024.136873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 12/20/2024]
Abstract
Accumulating evidence indicates that maternal exposure to carbon black nanoparticles (CBNPs) during gestation can induce multiple system abnormalities in offspring, whereas its potential mechanism in respiratory disease is still largely unknown. In order to explore the effect of maternal exposure to CBNPs on offspring's lung and latent pathogenesis, we respectively established in vivo model of pregnant rats exposed to CBNPs and ex vivo model of lung epithelial cells treated with pups' serum of pregnant rats exposed to CBNPs. After maternal exposure to CBNPs, epithelial-mesenchymal transition (EMT) and fibrosis levels increased as a result of DDRGK1-mediated reticulophagy upregulated in offspring's lung. DDRGK1 as FAM134B's cargo bound with FAM134B to mediate reticulophagy. Transcription factor "SP1" positively regulated DDRGK1 gene expression by binding to its promoter. Furthermore, the upregulation of NSUN2 elevated m5C methylation of SP1 mRNA and the protein level of SP1 subsequently increased through Ybx1 recognizing and stabilizing m5C-methylated SP1 mRNA, followed by the increased levels of reticulophagy and fibrosis in lung epithelial cells treated with offspring's serum of matrix exposed to CBNPs during gestation. In conclusion, NSUN2/Ybx1/m5C-SP1 axis promoted DDRGK1-mediated reticulophagy, which played an important role in EMT-induced fibrosis in offspring's lung tissue after maternal exposure to CBNPs during gestation.
Collapse
Affiliation(s)
- Zhe Du
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Qingping Liu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Mengruo Wang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Yifu Gao
- Hebei Province Center for Disease Control and Prevention, Shijiazhuang 050021, PR China
| | - Qi Li
- Hunan Institute for Drug Control, Changsha 410001, PR China
| | - Yizhe Yang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Tianyu Lu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Lei Bao
- Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Yaxian Pang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Haijun Wang
- Department of Maternal and Child Health, Peking University, Beijing 100191, PR China
| | - Yujie Niu
- Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Rong Zhang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China.
| |
Collapse
|
92
|
Li T, Chen H, Guo Y, Huang M, Liu P, Aikemu A, Mohammadtursun N, Pan X, Yang X. Nuciferine Restores Autophagy via the PI3K-AKT-mTOR Pathway to Alleviate Renal Fibrosis in Diabetic Kidney Disease. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:5223-5235. [PMID: 39989251 DOI: 10.1021/acs.jafc.4c08844] [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: 02/25/2025]
Abstract
Diabetic kidney disease (DKD) is one of the complications of diabetes mellitus, which triggers kidney fibrosis and eventually develops into end-stage renal disease. Nuciferine (NF) is one of the most important functional components in lotus leaves (LL), but its role and mechanism for the treatment of DKD are unclear. A high-fat-diet (HFD)-induced DKD model in KK-AY mice was established in this study. NF treatment significantly improved blood glucose and blood biochemical indices in DKD mice. Furthermore, NF reduced the levels of mALB, UCRE, Scr, and BUN in mice urine. Further, the extent of renal lesions in the mice in this study was at stage IV according to the Mogensen staging method. NF treatment was effective in ameliorating renal injury during this period. Concurrently, the protein levels of FN, N-cadherin, TGFβ, p-Smad3, p-PI3K, p-AKT, p-mTOR, and p62 were decreased. In contrast, the level of expression of Beclin-1 was increased. In the high glucose-exposed HK-2 cell model, the expression of p-PI3K, p-AKT, and p-mTOR was all downregulated, and autophagy proteins were increased after NF intervention. In addition, HK-2 cells were treated with high glucose in combination with Wortmannin and 3-MA, respectively. The results demonstrated that NF inhibited the expression of TGFβ and p-Smad3 by regulating autophagy through the PI3K-AKT-mTOR pathway, thereby ameliorating renal fibrosis at stage IV in mice. Therefore, LL can be used as a dietary component for the prevention of renal fibrosis in DKD patients.
Collapse
Affiliation(s)
- Tongqing Li
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, 182 min-Zu Road, Wuhan 430074, China
| | - Huijian Chen
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, 182 min-Zu Road, Wuhan 430074, China
| | - Yan Guo
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, 182 min-Zu Road, Wuhan 430074, China
| | - Mi Huang
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, 182 min-Zu Road, Wuhan 430074, China
| | - Pengxin Liu
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, 182 min-Zu Road, Wuhan 430074, China
| | - Ainiwaer Aikemu
- Xinjiang Key Laboratory of Hotan Characteristic Chinese Traditional Medicine Research, College of Xinjiang Uyghur Medicine, Hotan 848000, China
| | - Nabijan Mohammadtursun
- Xinjiang Key Laboratory of Hotan Characteristic Chinese Traditional Medicine Research, College of Xinjiang Uyghur Medicine, Hotan 848000, China
| | - Xin Pan
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, 182 min-Zu Road, Wuhan 430074, China
| | - Xinzhou Yang
- International Cooperation Base for Active Substances in Traditional Chinese Medicine in Hubei Province, School of Pharmaceutical Sciences, South-Central Minzu University, 182 min-Zu Road, Wuhan 430074, China
| |
Collapse
|
93
|
Liu Z, Zhang X, Ben T, Li M, Jin Y, Wang T, Song Y. Focal adhesion in the tumour metastasis: from molecular mechanisms to therapeutic targets. Biomark Res 2025; 13:38. [PMID: 40045379 PMCID: PMC11884212 DOI: 10.1186/s40364-025-00745-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/11/2025] [Indexed: 03/09/2025] Open
Abstract
The tumour microenvironment is the "hotbed" of tumour cells, providing abundant extracellular support for growth and metastasis. However, the tumour microenvironment is not static and is constantly remodelled by a variety of cellular components, including tumour cells, through mechanical, biological and chemical means to promote metastasis. Focal adhesion plays an important role in cell-extracellular matrix adhesion. An in-depth exploration of the role of focal adhesion in tumour metastasis, especially their contribution at the biomechanical level, is an important direction of current research. In this review, we first summarize the assembly of focal adhesions and explore their kinetics in tumour cells. Then, we describe in detail the role of focal adhesion in various stages of tumour metastasis, especially its key functions in cell migration, invasion, and matrix remodelling. Finally, we describe the anti-tumour strategies targeting focal adhesion and the current progress in the development of some inhibitors against focal adhesion proteins. In this paper, we summarize for the first time that focal adhesion play a positive feedback role in pro-tumour metastatic matrix remodelling by summarizing the five processes of focal adhesion assembly in a multidimensional way. It is beneficial for researchers to have a deeper understanding of the role of focal adhesion in the biological behaviour of tumour metastasis and the potential of focal adhesion as a therapeutic target, providing new ideas for the prevention and treatment of metastases.
Collapse
Affiliation(s)
- Zonghao Liu
- Department of Radiotherapy, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China
- The First Clinical College, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| | - Xiaofang Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Tianru Ben
- The First Clinical College, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| | - Mo Li
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China
| | - Yi Jin
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China
| | - Tianlu Wang
- Department of Radiotherapy, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China.
- Department of Radiotherapy, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning Province, 110042, People's Republic of China.
- Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning Province, 116024, P. R. China.
| | - Yingqiu Song
- Department of Radiotherapy, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China.
- Department of Radiotherapy, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China.
| |
Collapse
|
94
|
Cao X, Wan S, Wu B, Liu Z, Xu L, Ding Y, Huang H. Antitumor Research Based on Drug Delivery Carriers: Reversing the Polarization of Tumor-Associated Macrophages. Mol Pharm 2025; 22:1174-1197. [PMID: 39868820 DOI: 10.1021/acs.molpharmaceut.4c01277] [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: 01/28/2025]
Abstract
The development of malignant tumors is a complex process that involves the tumor microenvironment (TME). An immunosuppressive TME presents significant challenges to current cancer therapies, serving as a key mechanism through which tumor cells evade immune detection and play a crucial role in tumor progression and metastasis. This impedes the optimal effectiveness of immunotherapeutic approaches, including cytokines, immune checkpoint inhibitors, and cancer vaccines. Tumor-associated macrophages (TAMs), a major component of tumor-infiltrating immune cells, exhibit dual functionalities: M1-like TAMs suppress tumorigenesis, while M2-like TAMs promote tumor growth and metastasis. Consequently, the development of various nanocarriers aimed at polarizing M2-like TAMs to M1-like phenotypes through distinct mechanisms has emerged as a promising therapeutic strategy to inhibit tumor immune escape and enhance antitumor responses. This Review covers the origin and types of TAMs, common pathways regulating macrophage polarization, the role of TAMs in tumor progression, and therapeutic strategies targeting TAMs, aiming to provide a comprehensive understanding and guidance for future research and clinical applications.
Collapse
Affiliation(s)
- Xinyu Cao
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Shen Wan
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Bingyu Wu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Zhikuan Liu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Lixing Xu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Yu Ding
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Haiqin Huang
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| |
Collapse
|
95
|
Wang Y, Huang S, Cai Y, Wang T, Zhao H, Lin X, Wang X, Li P. Programmed cell death protein 5 inhibits hepatocellular carcinoma progression by inducing pyroptosis through regulation of TGF-β/Smad2/3/Snail pathway. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167696. [PMID: 39884472 DOI: 10.1016/j.bbadis.2025.167696] [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: 09/17/2024] [Revised: 11/11/2024] [Accepted: 01/22/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Programmed cell death protein 5 (PDCD5) is involved in apoptosis and is regarded as a tumor suppressor in various tumors. However, its role and underlying molecular mechanisms in hepatocellular carcinoma (HCC) remain unclear. METHODS PDCD5-overexpressing cell and xenograft tumor models were developed. Cell Counting Kit-8, 5-Ethynyl-2'-deoxyuridine, wound healing, Transwell, flow cytometry, immunohistochemistry, and hematoxylin-eosin staining were employed to explore the effects of PDCD5 on HCC cell behaviors and tumor growth. The enzyme-linked immunosorbent assay and western blot were used to detect pyroptosis-related marker levels. The molecular mechanisms underlying PDCD5's role in HCC were investigated through transcriptome sequencing and coimmunoprecipitation. SRI-011381, a TGF-β signaling activator, was applied to evaluate the impact of PDCD5 in modulating the TGF-β/Smad2/3/Snail pathway. RESULTS PDCD5 expression was reduced in HCC cells. Overexpression of PDCD5 inhibited HCC cell proliferation, migration, invasion, and xenograft tumor growth. Additionally, PDCD5 overexpression promoted apoptosis and pyroptosis, with corresponding increases in inflammatory factors and Caspase-1, GSDMD, and NLRP3 protein levels. Mechanistically, PDCD5 bound to receptor-regulated Smads (Smad2/3), inhibiting the TGF-β pathway. Treatment with the TGF-β pathway activator SRI-011381 significantly counteracted the inhibitory effects of PDCD5 overexpression on HCC progression. CONCLUSION Our findings suggest that PDCD5 impedes the progression of HCC by promoting pyroptosis via regulation of TGF-β/Smad2/3/Snail pathway, which could be a possible therapeutic target for HCC.
Collapse
Affiliation(s)
- Yiqiao Wang
- Department of Hepatobiliary and Pancreatic Surgery, Yueqing City People's Hospital, No. 338 Qingyuan Road, Chengnan Street, Yueqing City 325699, Zhejiang Province, China
| | - Shihao Huang
- Department of Hepatobiliary and Pancreatic Surgery, Yueqing City People's Hospital, No. 338 Qingyuan Road, Chengnan Street, Yueqing City 325699, Zhejiang Province, China
| | - Yangbai Cai
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Hainan Medical University, No. 48 Baishuitang Road, Haikou City 570100, Hainan Province, China
| | - Taicheng Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Hainan Medical University, No. 48 Baishuitang Road, Haikou City 570100, Hainan Province, China
| | - Hongyan Zhao
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Hainan Medical University, No. 48 Baishuitang Road, Haikou City 570100, Hainan Province, China
| | - Xianke Lin
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Hainan Medical University, No. 48 Baishuitang Road, Haikou City 570100, Hainan Province, China
| | - Xueguo Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Hainan Medical University, No. 48 Baishuitang Road, Haikou City 570100, Hainan Province, China
| | - Peng Li
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Hainan Medical University, No. 48 Baishuitang Road, Haikou City 570100, Hainan Province, China.
| |
Collapse
|
96
|
Sousa CH, Mercier M, Rioux‐Leclercq N, Flecher E, Bendavid C, Val‐Laillet D, Ferrant J, Jaillard S, Loiseau E, Branchereau J, Berkane Y, Nyangoh Timoh K, Carton I, Le Lous M, Lavoue V, Dion L. Hypothermic machine perfusion in uterus transplantation in a porcine model: A proof of concept and the first results in graft preservation. Acta Obstet Gynecol Scand 2025; 104:461-473. [PMID: 39868864 PMCID: PMC11871101 DOI: 10.1111/aogs.15056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/05/2025] [Accepted: 01/10/2025] [Indexed: 01/28/2025]
Abstract
INTRODUCTION Graft optimization is a necessity in order to develop uterus transplantation from brain-dead donors, as a complement to living donors, as these grafts are rare and the last organs retrieved in multiple organ donation. The aim of this study was to assess the feasibility and interest of hypothermic machine perfusion (HMP) in uterus transplantation using a porcine model; secondary outcomes were the evaluation of the graft's tolerance to a prolonged cold ischaemia time and to find new biomarkers of uterus viability. MATERIAL AND METHODS Fifteen uterus allotransplantations were performed in a porcine model, after 18 h of cold ischaemia, divided in three groups: Static cold storage in a HTK solution, HMP (with the VitaSmart (™) machine Bridge to Life Ltd.) with a UW-MP solution, and static cold storage in a UW solution. The main outcome was macroscopic: uterine arteries pulsatility, recoloration, and bleeding at the cut. Secondary outcomes were histological analyses (Zitkute and inflammation scores), caspase3 immunohistochemistry and plasmatic dosage of biomarkers. RESULTS 14/15 allotransplantations were performed according to the protocol and met the criteria of macroscopic vitality. Grafts treated with HMP (MP did not show significantly more tissue) damage than the recipient's uterus, contrary to grafts in static cold storage, independently of the solution used. This difference disappeared one and 3 h after uterus transplantation. Plasma dosages before and after uterus transplantation did not allow to identify a new biomarker of uterus viability. CONCLUSIONS HMP is feasible in a porcine model, without inflicting damage on the grafts during cold ischaemia time. Grafts exposed to HMP seemed to better endure reperfusion phenomena, but this advantage did not last over time.
Collapse
Affiliation(s)
- Carla Héléna Sousa
- Department of GynecologyRennes University Hospital, Hôpital SudRennesFrance
- Institute for Research in HealthEnvironment and Work (Irset)RennesFrance
| | - Marion Mercier
- Department of GynecologyRennes University Hospital, Hôpital SudRennesFrance
- Institute for Research in HealthEnvironment and Work (Irset)RennesFrance
| | - Nathalie Rioux‐Leclercq
- Institute for Research in HealthEnvironment and Work (Irset)RennesFrance
- Department of PathologyRennes University Hospital, Hôpital PontchaillouFrance
| | - Erwan Flecher
- Department of Cardiothoracic and Vascular SurgeryRennes University Hospital, Hôpital PontchaillouRennesFrance
| | - Claude Bendavid
- Laboratory of BiochemistryRennes University Hospital, Hôpital PontchaillouRennesFrance
| | - David Val‐Laillet
- INRAE, INSERMUniversity Of Rennes, NuMeCan Institute, Nutrition Metabolisms And CancerRennesFrance
| | - Juliette Ferrant
- Unité Mixte de Recherche (UMR)1236, Université RennesINSERM, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue Contre le CancerRennesFrance
| | - Sylvie Jaillard
- Institute for Research in HealthEnvironment and Work (Irset)RennesFrance
- Department of Cytogenetics and Cellular BiologyRennes University HospitalRennesFrance
| | - Emma Loiseau
- Department of GynecologyCHU Nantes Hôtel DieuNantesFrance
| | | | - Yanis Berkane
- Unité Mixte de Recherche (UMR)1236, Université RennesINSERM, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue Contre le CancerRennesFrance
- Department of Plastic, Reconstructive and Aesthetic SurgeryRennes University Hospital, Hôpital SudRennesFrance
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | | | - Isis Carton
- Department of GynecologyRennes University Hospital, Hôpital SudRennesFrance
| | - Maëla Le Lous
- Department of GynecologyRennes University Hospital, Hôpital SudRennesFrance
| | - Vincent Lavoue
- Department of GynecologyRennes University Hospital, Hôpital SudRennesFrance
- Institute for Research in HealthEnvironment and Work (Irset)RennesFrance
| | - Ludivine Dion
- Department of GynecologyRennes University Hospital, Hôpital SudRennesFrance
- Institute for Research in HealthEnvironment and Work (Irset)RennesFrance
| |
Collapse
|
97
|
Avolio E, Bassani B, Campanile M, Mohammed KA, Muti P, Bruno A, Spinetti G, Madeddu P. Shared molecular, cellular, and environmental hallmarks in cardiovascular disease and cancer: Any place for drug repurposing? Pharmacol Rev 2025; 77:100033. [PMID: 40148035 DOI: 10.1016/j.pharmr.2024.100033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 03/29/2025] Open
Abstract
Cancer and cardiovascular disease (CVD) are the 2 biggest killers worldwide. Specific treatments have been developed for the 2 diseases. However, mutual therapeutic targets should be considered because of the overlap of cellular and molecular mechanisms. Cancer research has grown at a fast pace, leading to an increasing number of new mechanistic treatments. Some of these drugs could prove useful for treating CVD, which realizes the concept of cancer drug repurposing. This review provides a comprehensive outline of the shared hallmarks of cancer and CVD, primarily ischemic heart disease and heart failure. We focus on chronic inflammation, altered immune response, stromal and vascular cell activation, and underlying signaling pathways causing pathological tissue remodeling. There is an obvious scope for targeting those shared mechanisms, thereby achieving reciprocal preventive and therapeutic benefits. Major attention is devoted to illustrating the logic, advantages, challenges, and viable examples of drug repurposing and discussing the potential influence of sex, gender, age, and ethnicity in realizing this approach. Artificial intelligence will help to refine the personalized application of drug repurposing for patients with CVD. SIGNIFICANCE STATEMENT: Cancer and cardiovascular disease (CVD), the 2 biggest killers worldwide, share several underlying cellular and molecular mechanisms. So far, specific therapies have been developed to tackle the 2 diseases. However, the development of new cardiovascular drugs has been slow compared with cancer drugs. Understanding the intersection between pathological mechanisms of the 2 diseases provides the basis for repurposing cancer therapeutics for CVD treatment. This approach could allow the rapid development of new drugs for patients with CVDs.
Collapse
Affiliation(s)
- Elisa Avolio
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| | - Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy
| | - Marzia Campanile
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy; Department of Biosciences, University of Milan, Milan, Italy
| | - Khaled Ak Mohammed
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom; Department of Cardiothoracic Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Paola Muti
- IRCCS MultiMedica, Milan, Italy; Department of Biomedical, Surgical and Dental Health Sciences, University of Milan, Italy
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy; Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy.
| | - Gaia Spinetti
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy.
| | - Paolo Madeddu
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| |
Collapse
|
98
|
Zhang X, Cong L, Yu R, Yu Q, Hou X, Zhou Y. MicroRNA‑96 promotes the proliferation and migration of breast cancer cells by inhibiting Smad7 expression. Oncol Lett 2025; 29:151. [PMID: 39898288 PMCID: PMC11783994 DOI: 10.3892/ol.2025.14897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 10/26/2023] [Indexed: 02/04/2025] Open
Abstract
The present study aimed to investigate the effects of microRNA (miR)-96 on the proliferation and migration of breast cancer cells, and indicated that miR-96 may have a promoting role in breast cancer by inhibiting Smad7. Reverse transcription-quantitative (RT-q)PCR was used to detect the expression levels of miR-96 and Smad7 in breast cancer tissues and adjacent tissues. Western blotting and immunohistochemistry were conducted to determine the expression levels of SMAD7 in breast cancer and adjacent tissues. A dual luciferase assay was performed to verify the targeted binding between miR-96 and Smad7. Furthermore, the different expression patterns of miR-96 and Smad7 were compared in various breast cancer cell lines using RT-qPCR and western blotting. Among these cell lines, MDA-MB-231, which exhibited the highest expression of miR-96, was chosen for subsequent functional verification. The expression levels of miR-96 were significantly higher in breast cancer tissues compared with those in adjacent tissues. By contrast, the expression levels of Smad7 were significantly lower in breast cancer tissues compared with those in adjacent tissues. The dual luciferase assay revealed a targeted binding effect between miR-96 and Smad7. Notably, transfection with miR-96-5p mimics and short hairpin RNA-Smad7 markedly promoted the proliferation, adhesion, invasion and migration of breast cancer cells. Conversely, transfection with a miR-96-5p inhibitor and Smad7 overexpression plasmid exhibited the opposite trend. In conclusion, the expression levels of miR-96 were significantly elevated in breast cancer tissues compared with those in adjacent tissues. Overexpression of miR-96 was shown to promote the migration of breast cancer cells by downregulating the expression of Smad7. These findings indicated that miR-96 may serve as a prognostic marker for breast cancer.
Collapse
Affiliation(s)
- Xiumei Zhang
- Department of Pathology, The People's Hospital of Xinghua City, Xinghua, Jiangsu 225700, P.R. China
| | - Lin Cong
- Department of Pathology, The People's Hospital of Xinghua City, Xinghua, Jiangsu 225700, P.R. China
| | - Rong Yu
- Department of Gastrointestinal Surgery, The People's Hospital of Xinghua City, Xinghua, Jiangsu 225700, P.R. China
| | - Qianwen Yu
- Department of Pathology, The People's Hospital of Xinghua City, Xinghua, Jiangsu 225700, P.R. China
| | - Xian Hou
- Department of Radiology, The People's Hospital of Xinghua City, Xinghua, Jiangsu 225700, P.R. China
| | - Yonghua Zhou
- Department of Breast Surgery, The People's Hospital of Xinghua City, Xinghua, Jiangsu 225700, P.R. China
| |
Collapse
|
99
|
Hsu JY, Hsu KC, Chou CH, He TY, Lin TE, Sung TY, Yen SC, Hsieh JH, Yang CR, Huang WJ. Structural optimization and biological evaluation of indolin-2-one derivatives as novel CDK8 inhibitors for idiopathic pulmonary fibrosis. Biomed Pharmacother 2025; 184:117891. [PMID: 39955852 DOI: 10.1016/j.biopha.2025.117891] [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: 10/14/2024] [Revised: 01/27/2025] [Accepted: 02/03/2025] [Indexed: 02/18/2025] Open
Abstract
Cyclin-dependent kinase 8 (CDK8) plays a crucial role in the transforming growth factor beta (TGF-β) signaling pathway, which is critical to the pathology of idiopathic pulmonary fibrosis (IPF). CDK8 promotes the epithelial-mesenchymal transition (EMT) and excessive extracellular matrix (ECM) deposition, making it a promising target for IPF treatment. This study focused on optimizing F059-1017, a previously identified CDK8 inhibitor, to enhance its potency. Through integrated structure-based modifications, a series of compounds was synthesized, and their inhibitory effects on CDK8 were tested. Results indicated that substituting with cyclopentanone significantly improved the inhibitory activity, and compound 4j demonstrated the best potency (IC50 = 16 nM). Notably, compared to F059-1017, its potency increased 35-fold, and kinase profiling revealed that the compound was selective for CDK8. Compound 4j inhibited the TGF-β1-induced EMT, cell migration, and morphological changes in A549 cells at a concentration of 0.1 μM and inhibited ECM and EMT protein expressions. In addition, the compound blocked TGF-β1-induced transcriptional changes and inhibited Smad3 and RNA polymerase II phosphorylation. These results highlight the potential of the optimized CDK8 inhibitor as a prospective drug for IPF treatment.
Collapse
Affiliation(s)
- Jui-Yi Hsu
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan; Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Kai-Cheng Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan; Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ching-Hsuan Chou
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tzu-Ying He
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tony Eight Lin
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Ying Sung
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Shih-Chung Yen
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong (Shenzhen), Shenzhen, China
| | - Jui-Hua Hsieh
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, USA
| | - Chia-Ron Yang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Wei-Jan Huang
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; School of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
100
|
Zhang X, Zhang L, Tian J, Li Y, Wu M, Zhang L, Qin X, Gong L. The application and prospects of drug delivery systems in idiopathic pulmonary fibrosis. BIOMATERIALS ADVANCES 2025; 168:214123. [PMID: 39615374 DOI: 10.1016/j.bioadv.2024.214123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/06/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease primarily affecting elderly individuals aged >65 years and has a poor prognosis. No effective treatment is currently available for IPF. The two antipulmonary fibrosis drugs nintedanib and pirfenidone approved by the FDA in the United States have somewhat decelerated IPF progression. However, the side effects of these drugs can lead to poor patient tolerance and compliance with the medications. Researchers have recently developed various methods for IPF treatment, such as gene silencing and pathway inhibitors, which hold great promise in IPF treatment. Nevertheless, the nonselectivity and nonspecificity of drugs often affect their efficacies. Drug delivery systems (DDS) are crucial for delivering drugs to specific target tissues or cells, thereby minimizing potential side effects, enhancing drug bioavailability, and reducing lung deposition. This review comprehensively summarizes the current state of DDS and various delivery strategies for IPF treatment (e.g., nano-delivery, hydrogel delivery, and biological carrier delivery) to completely expound the delivery mechanisms of different drug delivery carriers. Subsequently, the advantages and disadvantages of different DDS are fully discussed. Finally, the challenges and difficulties associated with the use of different DDS are addressed so as to accelerate their rapid clinical translation.
Collapse
Affiliation(s)
- Xi Zhang
- School of Biological Engineering, Zunyi Medical University, Guangdong 519000, China; Department of Clinical Medicine, The Fifth Clinical Institution, Zhuhai Campus of Zunyi Medical University, Guangdong 519000, China
| | - Ling Zhang
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), GuiZhou 563000, China
| | - Jiahua Tian
- Department of Clinical Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Yunfei Li
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), GuiZhou 563000, China
| | - Manli Wu
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), GuiZhou 563000, China
| | - Longju Zhang
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), GuiZhou 563000, China
| | - Xiaofei Qin
- School of Biological Engineering, Zunyi Medical University, Guangdong 519000, China.
| | - Ling Gong
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), GuiZhou 563000, China.
| |
Collapse
|