1
|
Wang Y, Tang C, Wang K, Zhang X, Zhang L, Xiao X, Lin H, Xiong L. The role of ferroptosis in breast cancer: Tumor progression, immune microenvironment interactions and therapeutic interventions. Eur J Pharmacol 2025; 996:177561. [PMID: 40154567 DOI: 10.1016/j.ejphar.2025.177561] [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: 12/29/2024] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Ferroptosis represents a distinctive and distinct form of regulated cellular death, which is driven by the accumulation of lipid peroxidation. It is distinguished by altered redox lipid metabolism and is linked to a spectrum of cellular activities, including cancer. In breast cancer (BC), with triple negative breast cancer (TNBC) being an iron-and lipid-rich tumor, inducing ferroptosis was thought to be a novel approach to killing breast tumor cells. However, in the recent past, a novel conceptual framework has emerged which posits that in addition to the promotion of tumor cell death, ferritin deposition has a potent immunosuppressive effect on the tumor immune microenvironment (TIME) via the influence on both innate and adaptive immune responses. TIME of BC includes various cell populations from both the innate and adaptive immune systems. In this review, the internal association between iron homeostasis and the progression of ferroptosis, along with the common inducers and protectors of ferroptosis in BC, are discussed in detail. Furthermore, a comprehensive analysis is conducted on the dual role of ferroptosis in immune cells and proto-oncogenic functions, along with an evaluation of the potential applications of immunogenic cell death-targeted immunotherapy in TIME of BC. It is anticipated that our review will inform future research endeavors that seek to integrate ferroptosis and immunotherapy in the management of BC.
Collapse
Affiliation(s)
- Yi Wang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Chuanyun Tang
- First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Keqin Wang
- First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Xiaoan Zhang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lifang Zhang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xinghua Xiao
- Department of Pathology, The First Affiliated Hospital, Nanchang University, 17 Yongwaizheng Road, Nanschang, 330066, China
| | - Hui Lin
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lixia Xiong
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
2
|
Yuan X, Yang X. CAPRIN1 Transcriptionally Activated PLPP4 to Inhibit DOX Sensitivity and Promote Breast Cancer Progression. Cell Biochem Biophys 2025; 83:2035-2045. [PMID: 39556159 DOI: 10.1007/s12013-024-01614-0] [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] [Accepted: 11/06/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Phospholipid phosphatase 4 (PLPP4) has been identified as a potential regulator of cancer cell dynamics, however, the role of PLPP4 in breast cancer (BC) progression and the sensitivity of BC cells to doxorubicin (DOX) remain elusive. METHODS The study analyzed the expression of PLPP4 and cell cycle-associated protein 1 (CAPRIN1) expression in BC tissues and cells using quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) and western blotting assays. Functional assays including colony formation, EdU, Transwell, and flow cytometry were employed to assess cellular behaviors. The sensitivity of BC cells to DOX was analyzed by CCK-8 assay and an in vivo xenograft model assay. The association between PLPP4 and CAPRIN1 was investigated using RNA immunoprecipitation assay and dual-luciferase reporter assay. RESULTS Upregulation of PLPP4 expression was observed in BC tissues and cells. Downregulation of PLPP4 expression in BC cells resulted in a suppression of their proliferative capacity, as well as a reduction in migratory and invasive capabilities. Additionally, this manipulation enhanced cell susceptibility to apoptosis and improved the sensitivity of these cells to DOX. When PLPP4 was knocked down in vivo in transplantable tumors, there was a marked enhancement in the responsiveness to DOX treatment. The transcription factor CAPRIN1 was found to regulate the expression of PLPP4 in the HCC1937 and MDA-MB-231 cell lines. Upregulation of CAPRIN1 was observed in both BC tissues and cells, and overexpression of PLPP4 reversed the effects of CAPRIN1 silencing on BC cell proliferation, migration, invasion, apoptosis, and DOX sensitivity. CONCLUSION This study demonstrates that CAPRIN1 transcriptionally activates PLPP4 to inhibit DOX sensitivity and promote BC progression. Targeting PLPP4 may represent a novel therapeutic strategy to enhance the efficacy of DOX in BC patients.
Collapse
Affiliation(s)
- Xiaorong Yuan
- Department of Lymphatic Breast Oncology, Baotou Cancer Hospital, Baotou, 014030, China
| | - Xuejie Yang
- Department of Lymphatic Breast Oncology, Baotou Cancer Hospital, Baotou, 014030, China.
| |
Collapse
|
3
|
Yang Y, Wen J, Lou S, Han Y, Pan Y, Zhong Y, He Q, Zhang Y, Mo X, Ma J, Shen N. DNAJC12 downregulation induces neuroblastoma progression via increased histone H4K5 lactylation. J Mol Cell Biol 2025; 16:mjae056. [PMID: 39716470 PMCID: PMC12096081 DOI: 10.1093/jmcb/mjae056] [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: 06/27/2024] [Revised: 12/09/2024] [Accepted: 12/22/2024] [Indexed: 12/25/2024] Open
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor in children. Despite treatment advances, the survival rates of high-risk NB patients remain low. This highlights the urgent need for a deeper understanding of the molecular mechanisms driving NB progression to support the development of new therapeutic strategies. In this study, we demonstrated that the reduced levels of DNAJC12, a protein involved in metabolic regulation, are associated with poor prognosis in NB patients. Our data indicate that low DNAJC12 expression activates glycolysis in NB cells, leading to increased lactic acid production and histone H4 lysine 5 lactylation (H4K5la). Elevated H4K5la upregulates the transcription of COL1A1, a gene implicated in cell metastasis. Immunohistochemistry staining of NB patient samples confirmed that high H4K5la levels correlate with poor clinical outcomes. Furthermore, we showed that inhibiting glycolysis, reducing H4K5la, or targeting COL1A1 can mitigate the invasive behavior of NB cells. These findings reveal a critical link between metabolic reprogramming and epigenetic modifications in the context of NB progression, suggesting that H4K5la could serve as a novel diagnostic and prognostic marker, and shed light on identifying new therapeutic targets within metabolic pathways for the treatment of this aggressive pediatric cancer.
Collapse
Affiliation(s)
- Yaqi Yang
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Jiejun Wen
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Susu Lou
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Pediatric Department, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310000, China
| | - Yali Han
- Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yi Pan
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Ying Zhong
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Qiao He
- Department of Pathology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yinfeng Zhang
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Xi Mo
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Jing Ma
- Department of Pathology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Nan Shen
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Department of Infectious Diseases, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| |
Collapse
|
4
|
Yang L, Wang R, Zhang L. HSPB1/KDM1 A facilitates ANXA2 expression via hypomethylated DNA promoter to inhibit ferroptosis and enhance gemcitabine resistance in pancreatic cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04228-2. [PMID: 40366396 DOI: 10.1007/s00210-025-04228-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025]
Abstract
Chemotherapy resistance contributes to the unsatisfied prognosis in pancreatic cancer (PC) patients. Heat shock protein beta-1 (HSPB1) plays a tumor promoting role in PC by inhibiting ferroptosis. This study aims to explore whether high expression of HSPB1 was responsible for ferroptosis and gemcitabine (GEM) resistance in PC. Here, we found that HSPB1 was upregulated in GEM-resistant PC cells and tumor tissues, as confirmed by RT-qPCR and Western blotting assays. Knockdown of HSPB1 enhanced GEM sensitivity, decreased the abilities of proliferation and invasion, and promoted apoptosis in GEM-resistant PC cells. Utilizing commercial kits, HSPB1 inhibition triggered ferroptosis, as indicated by increased levels of reactive oxygen species, malondialdehyde, and Fe2+, along with reduced glutathione (GSH) levels. Furthermore, the methylation specific PCR (MSP) results demonstrated a significant decrease in the methylation level of annexin A2 (ANXA2) CpG. The Chromatin immunoprecipitation (ChIP), ChIP-Re-ChIP, and Co-IP experiments revealed that HSPB1 interacts with lysine-specific histone demethylase 1A (KDM1A), recruiting KDM1A-CoREST complex to the ANXA2 promoter to enhance ANXA2 expression through demethylation of H3K9me2. Additionally, ANXA2 depletion further inhibited cell proliferation and invasion and induced ferroptosis in KDM1A-silenced cells, whereas ANXA2 overexpression produced the opposite effects. Finally, HSPB1 overexpression reduced gemcitabine sensitivity by promoting tumor growth in nude mice. Altogether, HSPB1 promoted ANXA2 expression by facilitating H3K9me2 demethylation through the recruitment of KDM1A-CoREST complex to the ANXA2 promoter, thereby inhibiting ferroptosis and enhancing GEM resistance in PC. These data provided a new insight for overcoming GEM-resistant PC.
Collapse
Affiliation(s)
- Liuxu Yang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an JiaoTong University, Xian, Shaanxi, 710061, China
| | - Ruizhe Wang
- Health Science Center, Xi'an JiaoTong University, Xian, Shaanxi, 710061, China
| | - Lun Zhang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an JiaoTong University, Xian, Shaanxi, 710061, China.
| |
Collapse
|
5
|
Chen X, Ma G, Li J, Gao X, Li Y. Supramolecular self-assembly of multifunctional carrageenan-based Nanomicelles for effective tumor therapy via apoptosis and immunogenic cell death. Int J Biol Macromol 2025; 306:141518. [PMID: 40020827 DOI: 10.1016/j.ijbiomac.2025.141518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/27/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Combination therapy that promotes both apoptosis and immunogenic cell death (ICD) holds promise for enhanced tumor treatment, yet its effectiveness is often hindered by challenges including inadequate targeted delivery, intricate nanoformulations processes, and cost inefficiency. Here, we present novel tumor-targeted, stimuli-responsive nanomicelles (D@CGO-G) through a straightforward supramolecular self-assembly method, incorporating targeting peptide GE11, doxorubicin (DOX), and a labile carrageenan (CG) linkage. Mechanistic insights into nanomicelles assembly process are thoroughly investigated using molecular dynamics (MD) simulations. D@CGO-G exhibit superior targeting capability both in vitro and in vivo, while the released DOX induces tumors apoptosis and ICD, stimulating macrophage polarization and enhancing the infiltration of tumor cytotoxic T lymphocytes. Furthermore, D@CGO-G effectively suppress both primary and distant tumors in vivo through combined apoptosis and anti-tumor immune responses. Our results highlight a simple, cost-effective approach to develop responsive CG-based nanomicelles for precise tumor targeting and robust multimodal therapeutic effects, advancing their clinical potential in cancer therapy.
Collapse
Affiliation(s)
- Xiangyan Chen
- State Key Laboratory Base of Eco-Chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Guohua Ma
- State Key Laboratory Base of Eco-Chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Junyao Li
- State Key Laboratory Base of Eco-Chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xintao Gao
- State Key Laboratory Base of Eco-Chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yantao Li
- State Key Laboratory Base of Eco-Chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| |
Collapse
|
6
|
Jiang L, Lu X, Li W. circFADS2 inhibits ferroptosis associated with IGF2BP2-dependent SLC7A11 m6A modification in colorectal cancer cells. J Gastrointest Oncol 2025; 16:503-516. [PMID: 40386612 PMCID: PMC12078837 DOI: 10.21037/jgo-2024-1014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 03/03/2025] [Indexed: 05/20/2025] Open
Abstract
Background Colorectal cancer (CRC) is one of the most common malignancies worldwide, and its pathogenesis is highly complex. The aim of this study was to explore the mechanism of action of circFADS2- and IGF2BP2-mediated SLC7A11 m6A modification in CRC. Methods In vitro experiments were conducted to knock down circFADS2 and overexpress SLC7A11 in CRC cells, and circFADS2 expression was detected by reverse transcription quantitative polymerase chain reaction (RT-qPCR); Cell Counting Kit-8 (CCK-8) and colony formation experiments were used to detect cell proliferation; cell migration was detected by Transwell; terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) was used for detection of cell apoptosis; western blot (WB) was employed for detection of SLC7A11 expression; the levels of malondialdehyde (MDA), glutathione (GSH), reactive oxygen species (ROS), and Fe2+ levels were detected; and messenger RNA (mRNA) stability testing, nuclear cytoplasmic separation experiments, RNA immunoprecipitation, fluorescence in situ hybridization (FISH), and immunofluorescence (IF) were used to verify the binding and mechanism of action of circFADS2, IGF2BP2, and SLC7A11 mRNA. In vivo experiments were conducted by injecting CRC cells from each group subcutaneously into the right side of mice, and the growth of tumor cells was measured in each group in vivo. Results After knocking down circFADS2, the expression of circFADS2 was downregulated in CRC cells. There was a significant reduction in cell proliferation and migration and a significant increase in cell apoptosis. The expression of SLC7A11 was significantly reduced; MDA content significantly decreased; GSH levels decreased; ROS levels increased; and the concentration of Fe2+ significantly increased. After circFADS2 knockdown, the growth of CRC cells in vivo was inhibited. In addition, mRNA stability testing showed that circFADS2 knockdown significantly reduced the stability of SLC7A11 mRNA. The nuclear cytoplasmic separation experiment showed that circFADS2 was mainly expressed in the cytoplasm. RNA immunoprecipitation indicated a binding relationship between IGF2BP2 and circFADS2, as well as between IGF2BP2 and SLC7A11 mRNA. The results of FISH and IF analysis showed that circFADS2, IGF2BP2, and SLC7A11 mRNA were co-localized with IGF2BP2 in the cytoplasm. Conclusions circFADS2 facilitates the formation of the circFADS2/IGF2BP2/SLC7A11 mRNA-protein complex, thereby enhancing the m6A methylation of SLC7A11. This process significantly promotes ferroptosis in CRC cells.
Collapse
Affiliation(s)
- Liangjun Jiang
- Department of Gastroenterology, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Xianzhou Lu
- Department of Hepatobiliary Surgery, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Wei Li
- Department of Hepatobiliary Surgery, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| |
Collapse
|
7
|
Wu KY, Crucho A, Su M, Chen ST, Hung CH, Kou YL, Liu YJ, Hsu TC, Yeh FY, Lien CF, Chen CC, Cai BH. Synergistic Anticancer Activity of HSP70 Inhibitor and Doxorubicin in Gain-of-Function Mutated p53 Breast Cancer Cells. Biomedicines 2025; 13:1034. [PMID: 40426862 PMCID: PMC12109493 DOI: 10.3390/biomedicines13051034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/09/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
Background: The mutation rate of p53 in breast cancer is around 20%. Specific p53 mutations exhibit prion-like abnormal misfolding and aggregation and gain oncogenic function, causing resistance to the chemotherapy drug doxorubicin. In this study, we identified key upstream regulatory molecules that inhibit the aggregation of p53 with the aim of increasing the anticancer effect of doxorubicin. Methods: Thioflavin T was employed as a fluorescent probe to detect prion-like protein aggregates within cells, the response to various inhibitors was evaluated using CCK8 assay, and the coefficient of drug interaction was calculated. The cell apoptosis ratio was evaluated using Caspase-3/7 based flow cytometry assay. Results: MDA-MB-231 cells (with p53 R280K mutation) and T47D cells (with p53 L194F mutation) had a strong Thioflavin T staining signal, but MDA-MB-468 cells (with p53 R273H mutation) had a weak Thioflavin T signal. Compared to MDA-MB-468 cells, which had a good response to doxorubicin, both MDA-MB-231 and T47D showed high doxorubicin drug resistance. Co-treatment with various misfolding p53 aggregation inhibitors and doxorubicin found that only the HSP70 inhibitor and doxorubicin had synergistic anticancer activity in both MDA-MB-231 and T47D cells. Furthermore, this co-treatment induced cell apoptosis in MDA-MB-231, which was reversed by a pan-caspase inhibitor. Conclusions: Doxorubicin resistance caused by specific p53 mutants can be resolved by co-treatment with a HSP70 inhibitor in breast cancer cells.
Collapse
Affiliation(s)
- Kuan-Yo Wu
- Department of Biomedical Engineering, I-Shou University, Kaohsiung City 82445, Taiwan;
| | - Ana Crucho
- Lisbon School of Medicine, Lisbon University, 1649-004 Lisboa, Portugal;
| | - Mia Su
- Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Sih-Tong Chen
- Department of Medical Science and Biotechnology, I-Shou University, Kaohsiung City 82445, Taiwan;
| | - Chen-Hsiu Hung
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung City 82445, Taiwan; (C.-H.H.); (Y.-L.K.); (Y.-J.L.); (T.-C.H.)
| | - Yu-Ling Kou
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung City 82445, Taiwan; (C.-H.H.); (Y.-L.K.); (Y.-J.L.); (T.-C.H.)
| | - Yu-Jie Liu
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung City 82445, Taiwan; (C.-H.H.); (Y.-L.K.); (Y.-J.L.); (T.-C.H.)
| | - Tzu-Chi Hsu
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung City 82445, Taiwan; (C.-H.H.); (Y.-L.K.); (Y.-J.L.); (T.-C.H.)
| | - Fang-Yu Yeh
- School of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan; (F.-Y.Y.); (C.-F.L.)
| | - Ching-Feng Lien
- School of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan; (F.-Y.Y.); (C.-F.L.)
- Department of Otolaryngology-Head and Neck Surgery, E-Da Hospital, Kaohsiung City 82445, Taiwan
| | - Chia-Chi Chen
- School of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan; (F.-Y.Y.); (C.-F.L.)
- Department of Pathology, E-Da Hospital, Kaohsiung City 82445, Taiwan
- Department of Physical Therapy, I-Shou University, Kaohsiung City 82445, Taiwan
- Department of Occupational Therapy, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Bi-He Cai
- School of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan; (F.-Y.Y.); (C.-F.L.)
| |
Collapse
|
8
|
Yin H, Chen Q, Gao S, Shoucair S, Xie Y, Habib JR, He T, Gan W, Wang J, Zhang L, Xu H, Shi C, He J, Wang W, Jin Y, Goggins MG, Liu L, Lou W, Wu W, Yu J, Pu N. The Crosstalk with CXCL10-Rich Tumor-Associated Mast Cells Fuels Pancreatic Cancer Progression and Immune Escape. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417724. [PMID: 39965084 PMCID: PMC11984875 DOI: 10.1002/advs.202417724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/30/2025] [Indexed: 02/20/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease, necessitating approaches to improve prognosis. As the mediator of allergic process, mast cells have been found in various cancers and are associated with survival. However, the biological behaviors of tumor-associated mast cells (TAMCs) remain unclear. Herein, an excessive infiltration of TAMCs in PDAC is demonstrated, which apparently associated with poor survival in PDAC patients. PDAC cells are found to recruit CXCR2+ MCs into TME, and then inhibited MCs ferroptosis, and maintained their proliferation. Concomitantly, the tumor-derived exosome miR-188-5p activated the PTEN/AKT/GSK3β signaling, further stabilized transcriptional factor ERG by inhibiting its ubiquitin degradation, and finally enhanced the transcription of cxcl10 within TAMCs. In reverse, TAMCs-derived CXCL10 reversely promoted tumor epithelial-mesenchymal transition and induced immunosuppressive tumor microenvironment by recruiting CXCR3+ Tregs. Sodium cromoglycate (SCG) is a membrane stabilizer for MCs and confirmed as an effective and widely used agent to block TAMCs-derived CXCL10 and further sensitize the therapeutic efficacy of anti-PD-1 antibody plus gemcitabine for PDAC. These findings illuminate a critical and innovative crosstalk between TAMCs and PDAC cells that promote PDAC progression, and SCG sensitizes PDAC to the current immuno-chemotherapy, which reveals its potential to be a valuable adjuvant for PDAC patients.
Collapse
Affiliation(s)
- Hanlin Yin
- Department of Pancreatic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
| | - Qiangda Chen
- Department of Pancreatic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
| | - Shanshan Gao
- Department of RadiologyZhongshan HospitalFudan UniversityShanghai200032China
- Departments of Medicine, Oncology and SurgeryJohns Hopkins University School of MedicineBaltimoreMD21287USA
| | - Sami Shoucair
- Departments of Medicine, Oncology and SurgeryJohns Hopkins University School of MedicineBaltimoreMD21287USA
| | - Yuqi Xie
- Department of Pancreatic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
| | - Joseph R. Habib
- Department of SurgeryNew York University School of Medicine and NYU‐Langone Medical CenterNew YorkNY10016USA
| | - Taochen He
- Department of Pancreatic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
| | - Wei Gan
- Department of Pancreatic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
| | - Juan Wang
- HANGZHOU CHEXMED TECHNOLOGY CO., LTDHangzhou310000China
| | - Lei Zhang
- Department of Pancreatic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
| | - Huaxiang Xu
- Department of Pancreatic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
| | - Chenye Shi
- Department of Pancreatic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
| | - Junyi He
- Department of Pancreatic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
| | - Wenquan Wang
- Department of Pancreatic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
| | - Yun Jin
- Department of Hepatobiliary and Pancreatic SurgeryThe First People's Hospital of Yunnan ProvinceThe Affiliated Hospital of Kunming University of Science and TechnologyKunming650500China
| | - Michael G Goggins
- Departments of Medicine and PathologyThe Sol Goldman Pancreatic Cancer Research CenterJohns Hopkins University School of MedicineBaltimoreMD21287USA
| | - Liang Liu
- Department of Pancreatic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
| | - Wenhui Lou
- Department of Pancreatic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
| | - Wenchuan Wu
- Department of Pancreatic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
| | - Jun Yu
- Departments of Medicine, Oncology and SurgeryJohns Hopkins University School of MedicineBaltimoreMD21287USA
- Pancreas CenterTianjin Medical University Cancer Institute & HospitalTianjin Medical UniversityTianjin300060China
| | - Ning Pu
- Department of Pancreatic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
| |
Collapse
|
9
|
Wang Y, Pei W, Yang Y, Xia C, Zhang Q, Geng Z, Shi X, Wang F. Inhibition of XIST restrains paclitaxel resistance in breast cancer cells by targeting hsa-let-7d-5p/ATG16L1 through regulation of autophagy. Cell Signal 2025; 127:111534. [PMID: 39638138 DOI: 10.1016/j.cellsig.2024.111534] [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: 06/03/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Breast cancer is a fatal malignant tumor in women worldwide. The development of paclitaxel resistance remains a challenge. Autophagy is considered to have a significant part in the chemotherapeutic stress mechanism. This study aimed to investigate the function of long non-coding RNA (lncRNA) in breast cancer cell chemoresistance and autophagy. The paclitaxel (PTX)-resistant breast cancer cells were established. The function of X-inactive specific transcript (XIST) was demonstrated using in vitro and in vivo experiments. Transmission electron microscope (TEM) was used to observe autophagy vesicles. Protein and mRNA levels were determined using western blotting and quantitative real time polymerase chain reaction (qRT-PCR). We discovered that autophagic activity was correlated with chemoresistance in PTX-resistant breast cancer cells. In vitro and in vivo studies showed that XIST inhibition reduced cell resistance to paclitaxel, caused autophagy to be suppressed by regulating hsa-let-7d-5p and ATG16L1 expression. Mechanically, threonine protein kinase B (PKB; also known as AKT) - mammalian target of rapamycin (mTOR) pathway was activated when knockdown of XIST, while was reversed by inhibition of hsa-let-7d-5p. Our results verified that XIST played a significant role in developing chemoresistance via mediating autophagy in PTX-resistant breast cancer cells. It may be a potential target for breast cancer treatment strategies.
Collapse
Affiliation(s)
- Yueyue Wang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu 233004, Anhui, China; The Fifth Clinical Medical College of Anhui Medical University, Hefei 230000, Anhui, China
| | - Wenhao Pei
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Bengbu 233030, Anhui, China
| | - Yuping Yang
- Department of Clinical Laboratory, Second Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China
| | - Chaoqun Xia
- Department of Clinical Laboratory, Second Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China
| | - Qiang Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China
| | - Zhijun Geng
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu 233004, Anhui, China; Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China
| | - Xiuru Shi
- Department of Blood Transfusion, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China
| | - Fengchao Wang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, Anhui, China.
| |
Collapse
|
10
|
Zhang M, Guan X, Dong Z, Yang C, Xiong C, Cheng W, Shang A, Liu Y, Guo X, Zhang B, Zhang B, Jin S, Qi W, Alexandrovna BT, Jiang Y, Du Z, Xu C. Targeting Zfp36 to combat cardiac hypertrophy: Insights into ferroptosis pathways. Clin Transl Med 2025; 15:e70247. [PMID: 40000392 PMCID: PMC11859123 DOI: 10.1002/ctm2.70247] [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: 10/05/2024] [Revised: 01/11/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Cardiac hypertrophy is a precursor to heart failure and represents a significant global cause of mortality, thereby necessitating timely and effective therapeutic interventions. Zinc finger protein 36 (Zfp36) is recognised as a critical regulator of ferroptosis; however, its role and underlying mechanisms in cardiac hypertrophy remain largely unexplored. This study aims to investigate the regulatory function of Zfp36 in ferroptosis within the context of cardiac hypertrophy. METHODS AND RESULTS Single-cell sequencing analysis demonstrated a reduction in Zfp36 expression associated with cardiac hypertrophy. Zfp36 was observed to mitigate ferroptosis and reduce hypertrophic phenotypes in cardiomyocytes subjected to Angiotensin II (Ang II) and in myocardial tissues induced by transverse aortic constriction. The ferroptosis inhibitor Ferrostatin-1 was shown to alleviate hypertrophy when co-incubated with si-Zfp36 and Ang II. Mechanistically, Zfp36 binds to the 3' untranslated region (3'UTR) of Ythdc2 mRNA, facilitating its degradation. Ythdc2 subsequently binds to SLC7A11 mRNA, enhancing its decay, which leads to a reduction in glutathione (GSH) levels, thereby exacerbating ferroptosis and cardiac hypertrophy. Furthermore, overexpression of Ythdc2 reversed the protective effects conferred by Zfp36, while silencing of Ythdc2 counteracted the effects of Zfp36 knockdown. CONCLUSIONS This study elucidates the role of Zfp36 in cardiac hypertrophy, specifically detailing its modulatory mechanism via the Ythdc2/SLC7A11/GSH ferroptosis pathway. These insights lay the groundwork for innovative approaches to understanding the pathological mechanisms underlying cardiac hypertrophy and enhancing clinical interventions. KEY POINTS Zfp36 was initially demonstrated to attenuate cardiac hypertrophy through the inhibition of ferroptosis in cardiomyocytes, providing a new target for therapeutic strategies targeting ferroptosis. Zfp36 facilitated the degradation of Ythdc2 mRNA by binding to it, subsequently inhibiting Ythdc2-mediated degradation of SLC7A11 mRNA, and maintaining GSH levels. This elucidates a previously unrecognized regulatory pathway in the context of cardiac hypertrophy.
Collapse
Affiliation(s)
- Mingyu Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (State Key Labratoray ‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinChina
| | - Xiaoxiang Guan
- Department of Clinical Pharmacythe First Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Zheng Dong
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (State Key Labratoray ‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinChina
| | - Chenxu Yang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (State Key Labratoray ‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinChina
| | - Chao Xiong
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (State Key Labratoray ‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinChina
| | - Wenzheng Cheng
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (State Key Labratoray ‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinChina
| | - Aijing Shang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (State Key Labratoray ‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinChina
| | - Yaru Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (State Key Labratoray ‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinChina
| | - Xiaofei Guo
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (State Key Labratoray ‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinChina
| | - Bowen Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (State Key Labratoray ‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinChina
| | - Bo Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (State Key Labratoray ‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinChina
| | - Saidi Jin
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (State Key Labratoray ‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinChina
| | - Wenyi Qi
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (State Key Labratoray ‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinChina
| | | | - Yuan Jiang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (State Key Labratoray ‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinChina
- Department of Biopharmaceutical Sciences, College of PharmacyHarbin Medical UniversityHarbinChina
| | - Zhimin Du
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (State Key Labratoray ‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinChina
- State Key Laboratory of Quality Research in Chinese MedicinesMacau University of Science and TechnologyMacauChina
| | - Chaoqian Xu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (State Key Labratoray ‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular Research, Ministry of Education)College of PharmacyHarbin Medical UniversityHarbinChina
- Department of Clinical Pharmacythe First Affiliated Hospital of Harbin Medical UniversityHarbinChina
- Joint International Research Laboratory of Cardiovascular Medicine ResearchMinistry of EducationHarbinChina
| |
Collapse
|
11
|
Du F, Wang G, Dai Q, Huang J, Li J, Liu C, Du K, Tian H, Deng Q, Xie L, Zhao X, Zhang Q, Yang L, Li Y, Wu Z, Zhang Z. Targeting novel regulated cell death: disulfidptosis in cancer immunotherapy with immune checkpoint inhibitors. Biomark Res 2025; 13:35. [PMID: 40012016 DOI: 10.1186/s40364-025-00748-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
The battle against cancer has evolved over centuries, from the early stages of surgical resection to contemporary treatments including chemotherapy, radiation, targeted therapies, and immunotherapies. Despite significant advances in cancer treatment over recent decades, these therapies remain limited by various challenges. Immune checkpoint inhibitors (ICIs), a cornerstone of tumor immunotherapy, have emerged as one of the most promising advancements in cancer treatment. Although ICIs, such as CTLA-4 and PD-1/PD-L1 inhibitors, have demonstrated clinical efficacy, their therapeutic impact remains suboptimal due to patient-specific variability and tumor immune resistance. Cell death is a fundamental process for maintaining tissue homeostasis and function. Recent research highlights that the combination of induced regulatory cell death (RCD) and ICIs can substantially enhance anti-tumor responses across multiple cancer types. In cells exhibiting high levels of recombinant solute carrier family 7 member 11 (SLC7A11) protein, glucose deprivation triggers a programmed cell death (PCD) pathway characterized by disulfide bond formation and REDOX (reduction-oxidation) reactions, termed "disulfidptosis." Studies suggest that disulfidptosis plays a critical role in the therapeutic efficacy of SLC7A11high cancers. Therefore, to investigate the potential synergy between disulfidptosis and ICIs, this study will explore the mechanisms of both processes in tumor progression, with the goal of enhancing the anti-tumor immune response of ICIs by targeting the intracellular disulfidptosis pathway.
Collapse
Affiliation(s)
- Fei Du
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Guojun Wang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qian Dai
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Jiang Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Junxin Li
- Department of pharmacy, Zigong Fourth People's Hospital, Zigong, 643000, China
| | - Congxing Liu
- Department of Pharmacy, Chengfei Hospital, Chengdu, 610000, China
| | - Ke Du
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pediatrics, Luzhou Maternal and Child Health Hospital, Luzhou Second People's Hospital, Luzhou, 646000, Sichuan, China
| | - Hua Tian
- School of Nursing, Chongqing College of Humanities, Science & Technology, Chongqing, 401520, China
| | - Qiwei Deng
- Heruida Pharmaceutical Co.,ltd, Haikou, Hainan, 570100, China
| | - Longxiang Xie
- The TCM Hospital of Longquanyi District, Chengdu, 610100, Sichuan, China
| | - Xin Zhao
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qimin Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Lan Yang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Yaling Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhigui Wu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhuo Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
12
|
Luo JY, Deng YL, Lu SY, Chen SY, He RQ, Qin DY, Chi BT, Chen G, Yang X, Peng W. Current Status and Future Directions of Ferroptosis Research in Breast Cancer: Bibliometric Analysis. Interact J Med Res 2025; 14:e66286. [PMID: 40009842 PMCID: PMC11904379 DOI: 10.2196/66286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/26/2024] [Accepted: 12/05/2024] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Ferroptosis, as a novel modality of cell death, holds significant potential in elucidating the pathogenesis and advancing therapeutic strategies for breast cancer. OBJECTIVE This study aims to comprehensively analyze current ferroptosis research and future trends, guiding breast cancer research advancements and innovative treatment strategies. METHODS This research used the R package Bibliometrix (Department of Economic and Statistical Sciences at the University of Naples Federico II), VOSviewer (Centre for Science and Technology Studies at Leiden University), and CiteSpace (Drexel University's College of Information Science and Technology), to conduct a bibliometric analysis of 387 papers on breast cancer and ferroptosis from the Web of Science Core Collection. The analysis covers authors, institutions, journals, countries or regions, publication volumes, citations, and keywords. RESULTS The number of publications related to this field has surged annually, with China and the United States collaborating closely and leading in output. Sun Yat-sen University stands out among the institutions, while the journal Frontiers in Oncology and the author Efferth T contribute significantly to the field. Highly cited papers within the domain primarily focus on the induction of ferroptosis, protein regulation, and comparisons with other modes of cell death, providing a foundation for breast cancer treatment. Keyword analysis highlights the maturity of glutathione peroxidase 4-related research, with breast cancer subtypes emerging as motor themes and the tumor microenvironment, immunotherapy, and prognostic models identified as basic themes. Furthermore, the application of nanoparticles serves as an additional complement to the basic themes. CONCLUSIONS The current research status in the field of ferroptosis and breast cancer primarily focuses on the exploration of relevant theoretical mechanisms, whereas future trends and mechanisms emphasize the investigation of therapeutic strategies, particularly the clinical application of immunotherapy related to the tumor microenvironment. Nanotherapy has demonstrated significant clinical potential in this domain. Future research directions should deepen the exploration in this field and accelerate the clinical translation of research findings to provide new insights and directions for the innovation and development of breast cancer treatment strategies.
Collapse
Affiliation(s)
- Jia-Yuan Luo
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yu-Long Deng
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shang-Yi Lu
- Department of Hepatological and Gland Surgery, Wuzhou Gongren Hospital/The Seventh Affiliated Hospital of Guangxi Medical University, Wuzhou, China
| | - Si-Yan Chen
- Day Chemotherapy Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Rong-Quan He
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Di-Yuan Qin
- Department of Computer Science and Technology, School of Computer and Electronic Information, Guangxi University, Nanning, China
| | - Bang-Teng Chi
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xia Yang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wei Peng
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
13
|
Qian Y, Wang C, Xu R, Wang J, Chen Q, Zhu Z, Hu Q, Shen Q, Shen JW. Copper-based metal-organic frameworks for antitumor application. J Nanobiotechnology 2025; 23:135. [PMID: 39987136 PMCID: PMC11847370 DOI: 10.1186/s12951-025-03220-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/10/2025] [Indexed: 02/24/2025] Open
Abstract
It is urgent to exploit multifunctional materials and combined approaches for efficient antitumor effects. Copper-based metal-organic frameworks (Cu-MOFs) have excellent performances in catalysis, biocompatibility, photothermal conversion, and regulate metabolism, which make them attract more and more attention in antitumor application. Therefore, in this review, representative ligands, synthetic methods, antitumor mechanism, and antitumor applications of Cu-MOFs were provided. Special emphasis is placed on the recent antitumor applications of Cu-MOFs in drug carriers, antitumor therapy, tumor imaging, and theranostic, which are summarized with examples. Finally, we presented the dilemma faced by Cu-MOFs and offered a new perspective for future antitumor application. Hopefully, this review may serve as a reference for further development and application of Cu-MOFs.
Collapse
Affiliation(s)
- Yangwei Qian
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, People's Republic of China
| | - Chenxi Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, People's Republic of China
| | - Ruru Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, People's Republic of China
| | - Jin Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, People's Republic of China
| | - Qinyue Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, People's Republic of China
| | - Zirui Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, People's Republic of China
| | - Quan Hu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, People's Republic of China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, People's Republic of China.
| | - Qiying Shen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, People's Republic of China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, People's Republic of China.
| | - Jia-Wei Shen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, People's Republic of China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, People's Republic of China.
| |
Collapse
|
14
|
Zhou C, Wu K, Gu M, Yang Y, Tu J, Huang X. Reversal of chemotherapy resistance in gastric cancer with traditional Chinese medicine as sensitizer: potential mechanism of action. Front Oncol 2025; 15:1524182. [PMID: 40052129 PMCID: PMC11882405 DOI: 10.3389/fonc.2025.1524182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
Gastric cancer (GC) remains one of the most common types of cancer, ranking fifth among cancer-related deaths worldwide. Chemotherapy is an effective treatment for advanced GC. However, the development of chemotherapy resistance, which involves the malfunction of several signaling pathways and is the consequence of numerous variables interacting, seriously affects patient treatment and leads to poor clinical outcomes. Therefore, in order to treat GC, it is imperative to find novel medications that will increase chemotherapy sensitivity and reverse chemotherapy resistance. Traditional Chinese medicine (TCM) has been extensively researched as an adjuvant medication in recent years. It has been shown to have anticancer benefits and to be crucial in enhancing chemotherapy sensitivity and reducing chemotherapy resistance. Given this, the mechanism of treatment resistance in GC is summed up in this work. The theoretical foundation for TCM as a sensitizer in adjuvant treatment of GC is established by introducing the primary signal pathways and possible targets implicated in improving chemotherapy sensitivity and reversing chemotherapy resistance of GC by TCM and active ingredients.
Collapse
Affiliation(s)
| | | | | | | | | | - Xuan Huang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese
Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
15
|
Chen JC, Chen MS, Jiang SK, Eaw CY, Han YJ, Tang CH. Transcriptomic data integration and analysis revealing potential mechanisms of doxorubicin resistance in chondrosarcoma cells. Biochem Pharmacol 2025; 232:116733. [PMID: 39732441 DOI: 10.1016/j.bcp.2024.116733] [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: 09/25/2024] [Revised: 12/11/2024] [Accepted: 12/19/2024] [Indexed: 12/30/2024]
Abstract
Chondrosarcoma is a type of bone cancer that originates from cartilage cells. In clinical practice, surgical resection is the primary treatment for chondrosarcoma, but chemotherapy becomes essential for patients with metastasis or tumors in surgically inaccessible sites. However, drug resistance often leads to treatment failure. Tumor microenvironment proteins modulate intercellular communication, contributing to drug resistance. Doxorubicin (Dox) is a common chemotherapeutic agent. The present study aimed to establish Dox-resistant chondrosarcoma cells and compare their secretome with parental cells using antibody arrays. Results showed significantly heightened secretion of hepatocyte growth factor (HGF). Knockdown of both HGF and its receptor MET increased Dox sensitivity in chondrosarcoma cells. Treatment of chondrosarcoma cells with conditioned media (CM) from cells secreting high levels of HGF resulted in MET activation. Additionally, the expression levels of HGF and MET were significantly elevated in chondrosarcoma tissues compared to normal cartilage tissues, as confirmed by analysis of GEO database. RNA sequencing and Gene Set Enrichment Analysis (GSEA) elucidated the mechanism involving HGF. Additionally, genes with log fold change > 1 underwent bioinformatics analysis using the ShinyGO web server. The results from both GSEA and ShinyGO analyses corroborate each other, indicating the significance of HGF in cellular signal transduction, regulation of cell motility, developmental processes, immune-inflammatory responses, and functions related to blood and neural systems. In summary, highly secreted HGF can activate signaling pathways through its receptor MET, particularly Ras and Akt activation, enhancing drug resistance in chondrosarcoma cells. The present study may guide the development of novel therapeutic strategies targeting HGF, ultimately improving treatment outcomes and prognosis for malignant chondrosarcoma patients.
Collapse
Affiliation(s)
- Jui-Chieh Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 600355, Taiwan
| | - Ming-Shan Chen
- Department of Anesthesiology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan; Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 41354, Taiwan
| | - Shin-Kuang Jiang
- Department of Neurology, China Medical University Hospital, Taichung 404332, Taiwan
| | - Chi-Yang Eaw
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 600355, Taiwan
| | - Yu-Jiao Han
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 600355, Taiwan
| | - Chih-Hsin Tang
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 41354, Taiwan; Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan; Department of Pharmacology, School of Medicine, China Medical University, Taichung 40402, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan.
| |
Collapse
|
16
|
Wu P, Chen J, Li H, Lu H, Li Y, Zhang J. Interactions between ferroptosis and tumour development mechanisms: Implications for gynaecological cancer therapy (Review). Oncol Rep 2025; 53:18. [PMID: 39635847 PMCID: PMC11638741 DOI: 10.3892/or.2024.8851] [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: 09/19/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
Ferroptosis is a form of programmed cell death that is distinct from apoptosis. The mechanism involves redox‑active metallic iron and is characterized by an abnormal increase in iron‑dependent lipid reactive oxygen species, which results in high levels of membrane lipid peroxides. The relationship between ferroptosis and gynaecological tumours is complex. Ferroptosis can regulate tumour proliferation, metastasis and chemotherapy resistance, and targeting ferroptosis is a promising antitumour approach. Ferroptosis interacts with mechanisms related to tumorigenesis and development, such as macrophage polarization, the neutrophil trap network, mitochondrial autophagy and cuproptosis. The present review examines recent information on the interaction between the molecular mechanism of ferroptosis and other tumour‑related mechanisms, as well as the involvement of ferroptosis in gynaecological tumours, to identify implications for gynaecological cancer therapy.
Collapse
Affiliation(s)
- Peiting Wu
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan 410013, P.R. China
| | - Jianlin Chen
- Department of Assisted Reproductive Centre, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Hui Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan 410013, P.R. China
| | - Haiyuan Lu
- Department of Clinical Laboratory Department, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
- Department of Hunan Vigorzoe Biotechnology Co., Ltd., Hunan 417700, P.R. China
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan 410013, P.R. China
| | - Juan Zhang
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan 410013, P.R. China
| |
Collapse
|
17
|
Xu Y, Xu X, Chai R, Wu X. Targeting ferroptosis to enhance the efficacy of mesenchymal stem cell-based treatments for intervertebral disc degeneration. Int J Biol Sci 2025; 21:1222-1241. [PMID: 39897051 PMCID: PMC11781166 DOI: 10.7150/ijbs.107021] [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: 11/15/2024] [Accepted: 01/11/2025] [Indexed: 02/04/2025] Open
Abstract
Although mesenchymal stromal cell (MSC) implantation shows promise for repairing intervertebral disc (IVD) degeneration (IVDD), their limited retention within degenerative IVDs compromises therapeutic efficacy. The oxidative stress in the microenvironment of degenerated IVDs induces a surge in reactive oxygen species production within MSCs, disrupting the balance between oxidation and antioxidation, and ultimately inducing ferroptosis. Recent evidence has suggested that targeting ferroptosis in MSCs could enhance MSC retention, extend the survival of transplanted MSCs, and markedly delay the pathological progression of IVDD. By targeting ferroptosis, a novel approach emerges to boost the efficacy of MSC transplantation therapy for IVDD. In this review, current research on targeting ferroptosis in MSCs is discussed from various perspectives, including the targeting of specific genes and pathways, drug preconditioning, and hydrogel encapsulation. A detailed discussion on the effects of targeting ferroptosis in MSCs on the transplantation repair of degenerated IVDs is provided. Insights that could guide improvements in stem cell transplantation therapies are also offered. Significantly, this review presents specific ideas for our future foundational research. These insights outline promising avenues for future clinical translation and will contribute to developing and optimizing treatment strategies for MSC transplantation therapy, maximizing benefits for patients with lumbar IVDD.
Collapse
Affiliation(s)
- Yuzhu Xu
- Department of Spine Center, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Xuanfei Xu
- Department of Nuclear Medicine, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Renjie Chai
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Xiaotao Wu
- Department of Spine Center, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, 210009, China
| |
Collapse
|
18
|
Huang Fu ZM, Xiao M, Xie H, Zhang S, Yi T, Li Q, Li M, Wang Y. Suppressing GDF15 enhances the chemotherapeutic effect of 5 FU on MSI-H CRC by regulating the ferroptosis pathway SLC7A11/GSH/GPX4. J Cancer Res Clin Oncol 2024; 151:6. [PMID: 39636345 PMCID: PMC11621149 DOI: 10.1007/s00432-024-06036-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024]
Abstract
Growth differentiation factor 15 (GDF15) is a member of the transforming growth factor beta (TGF-β) superfamily and is related to metabolism, injury, and aging. GDF15 has both tumor-promoting and tumor-suppressing effects. However, its role in colorectal cancer (CRC) with high microsatellite instability (MSI-H) must be further clarified. In our study, we found that GDF15 is generally elevated in pancarcinoma, particularly in colorectal cancer, and serves as an early indicator of the development of colorectal cancer. IHC and WB confirmed that GDF15 was elevated in MSI-H CRC clinical tissues and MSI-H CRC cell lines (HCT-116 and LoVo). Suppressing GDF15 by siRNA resulted in a substantial decrease in cell viability and proliferation. Furthermore, suppressing GDF15 can increase the sensitivity of MSI-H CRC cells to 5-fluorouracil (5-FU), which decreases cell viability and increases the apoptosis rate. In vivo experiments also demonstrated that mouse xenografts with suppressed GDF15 expression were more susceptible to 5-FU chemotherapy. We examined alterations in mitochondria via electron microscopy and changes in the mitochondrial membrane potential, ferroptosis-related signals (MDA, Fe2+), and SLC7A11/GSH/GPX4 protein pathway. Our research indicates that inhibiting GDF15 affects ferroptosis-related pathways, leading to ferroptosis and improving the MSI-H CRC response to 5-FU therapy. As a result, GDF15 could be a promising target for diagnosing and treating MSI-H CRC, potentially enhancing the overall effectiveness of therapy for patients with MSI-H CRC.
Collapse
Affiliation(s)
- Zhi Min Huang Fu
- Department of Pathology, Molecular Medicine and Cancer Research Center, Department of Clinical Pathololgy, Laboratory of Pathology Diagnostic Center, Chongqing Medical University, Chongqing, China
- Department of Pathology, The Second Hospital Affiliated to Third Military Medical University, XinQiao Hospital, Chongqing, China
| | - Ming Xiao
- Department of Pathology, Molecular Medicine and Cancer Research Center, Department of Clinical Pathololgy, Laboratory of Pathology Diagnostic Center, Chongqing Medical University, Chongqing, China
| | - Hailun Xie
- Department of Pathology, Molecular Medicine and Cancer Research Center, Department of Clinical Pathololgy, Laboratory of Pathology Diagnostic Center, Chongqing Medical University, Chongqing, China
| | - Shuxian Zhang
- Department of Pathology, Molecular Medicine and Cancer Research Center, Department of Clinical Pathololgy, Laboratory of Pathology Diagnostic Center, Chongqing Medical University, Chongqing, China
| | - Tang Yi
- Department of Pathology, Molecular Medicine and Cancer Research Center, Department of Clinical Pathololgy, Laboratory of Pathology Diagnostic Center, Chongqing Medical University, Chongqing, China
| | - Qingshu Li
- Department of Pathology, Molecular Medicine and Cancer Research Center, Department of Clinical Pathololgy, Laboratory of Pathology Diagnostic Center, Chongqing Medical University, Chongqing, China
| | - Ming Li
- Department of Pathology, Molecular Medicine and Cancer Research Center, Department of Clinical Pathololgy, Laboratory of Pathology Diagnostic Center, Chongqing Medical University, Chongqing, China.
| | - Yalan Wang
- Department of Pathology, Molecular Medicine and Cancer Research Center, Department of Clinical Pathololgy, Laboratory of Pathology Diagnostic Center, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
19
|
Zhang Y, Pan R, Li K, Cheang LH, Zhao J, Zhong Z, Li S, Wang J, Zhang X, Cheng Y, Zheng X, He R, Wang H. HSPD1 Supports Osteosarcoma Progression through Stabilizing ATP5A1 and thus Activation of AKT/mTOR Signaling. Int J Biol Sci 2024; 20:5162-5190. [PMID: 39430254 PMCID: PMC11489178 DOI: 10.7150/ijbs.100015] [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: 06/24/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024] Open
Abstract
Malignant transformation is concomitant with excessive activation of stress response pathways. Heat shock proteins (HSPs) are stress-inducible proteins that play a role in folding and processing proteins, contributing to the non-oncogene addiction of stressed tumor cells. However, the detailed role of the HSP family in osteosarcoma has not been investigated. Bulk and single-cell transcriptomic data from the GEO and TARGET databases were used to identify HSPs associated with prognosis in osteosarcoma patients. The expression level of HSPD1 was markedly increased in osteosarcoma, correlating with a negative prognosis. Through in vitro and in vivo experiments, we systematically identified HSPD1 as an important contributor to the regulation of proliferation, metastasis, and apoptosis in osteosarcoma by promoting the epithelial-mesenchymal transition (EMT) and activating AKT/mTOR signaling. Subsequently, ATP5A1 was determined as a potential target of HSPD1 using immunoprecipitation followed by mass spectrometry. Mechanistically, HSPD1 may interact with ATP5A1 to reduce the K48-linked ubiquitination and degradation of ATP5A1, which ultimately activates the AKT/mTOR pathway to ensure osteosarcoma progression and EMT process. These findings expand the potential mechanisms by which HSPD1 exerts biological effects and provide strong evidence for its inclusion as a potential therapeutic target in osteosarcoma.
Collapse
Affiliation(s)
- Yiming Zhang
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Ruilin Pan
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Kun Li
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, China
- State Key Laboratory of Bioactive Molecules and Drug Ability Assessment, Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of the Chinese Ministry of Education, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Lek Hang Cheang
- Department of Orthopedic Surgery, Centro Hospitalar Conde de Sao Januario, Macau, China
| | - Jing Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, China
| | - Zhangfeng Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, China
| | - Shaoping Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, China
| | - Jinghao Wang
- Department of Pharmacy, the First Affiliated Hospital, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Jinan University, Guangzhou, China
- Department of Orthopedics, NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaofang Zhang
- Department of Pharmacy, the First Affiliated Hospital, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Jinan University, Guangzhou, China
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, Heilongjiang, China
| | - Yanmei Cheng
- Department of Cardiothoracic Surgery ICU, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Xiaofei Zheng
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Rongrong He
- State Key Laboratory of Bioactive Molecules and Drug Ability Assessment, Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of the Chinese Ministry of Education, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Huajun Wang
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
20
|
Qian J, Zhao L, Xu L, Zhao J, Tang Y, Yu M, Lin J, Ding L, Cui Q. Cell Death: Mechanisms and Potential Targets in Breast Cancer Therapy. Int J Mol Sci 2024; 25:9703. [PMID: 39273650 PMCID: PMC11395276 DOI: 10.3390/ijms25179703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/31/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Breast cancer (BC) has become the most life-threatening cancer to women worldwide, with multiple subtypes, poor prognosis, and rising mortality. The molecular heterogeneity of BC limits the efficacy and represents challenges for existing therapies, mainly due to the unpredictable clinical response, the reason for which probably lies in the interactions and alterations of diverse cell death pathways. However, most studies and drugs have focused on a single type of cell death, while the therapeutic opportunities related to other cell death pathways are often neglected. Therefore, it is critical to identify the predominant type of cell death, the transition to different cell death patterns during treatment, and the underlying regulatory mechanisms in BC. In this review, we summarize the characteristics of various forms of cell death, including PANoptosis (pyroptosis, apoptosis, necroptosis), autophagy, ferroptosis, and cuproptosis, and discuss their triggers and signaling cascades in BC, which may provide a reference for future pathogenesis research and allow for the development of novel targeted therapeutics in BC.
Collapse
Affiliation(s)
- Jiangying Qian
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Linna Zhao
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Ling Xu
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Jin Zhao
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Yongxu Tang
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Min Yu
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Jie Lin
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Lei Ding
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Qinghua Cui
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China
| |
Collapse
|