1
|
Chen C, Liu X, Zhu S, Wang Y, Ma Y, Hu Z, Wu Y, Jiang L. Circ-0069561 as a novel diagnostic biomarker for progression of diabetic kidney disease. Ren Fail 2025; 47:2490200. [PMID: 40260530 PMCID: PMC12016256 DOI: 10.1080/0886022x.2025.2490200] [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: 12/17/2024] [Revised: 03/21/2025] [Accepted: 04/02/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) are non-coding RNAs that are key regulators of the initiation and progression of various human diseases. However, the role of circRNAs in diabetic kidney disease (DKD) remains unknown. METHODS Whole high-throughput RNA sequencing (RNA-seq) was performed on kidney tissues from clinical DKD patients and controls. Circ-0069561 with significantly up-regulated expression level was selected by real-time PCR (RT-PCR) analysis. RT-PCR and fluorescent in situ hybridization (FISH) further validated the expression and subcellular localization of circ-0069561 in type 2 diabetic mice and DKD patients. The clinical significance of circ-0069561 in DKD was evaluated. The circRNA-miRNA-ferroptosis associated mRNA network was constructed. The biological function of circ-0069561 in mouse podocyte clone 5 (MPC5) was analyzed. RESULTS The top 10 up-regulated circular RNAs were selected by RT-PCR validation, and the results demonstrated a significant elevation in the expression level of circ-0069561. The RT-PCR and FISH results demonstrated that the expression of circ-0069561 was elevated in renal tissues of type 2 diabetic mice and DKD patients, with a predominant localization in glomerulus. The ROC curves showed that circ-0069561 had a good diagnostic value in massive proteinuria (area under the curve = 0.889). Kaplan-Meier analysis showed that high expression of circ-0069561 was associated with an increased risk of primary endpoints. The circRNA-miRNA-mRNA network indicated that ferroptosis might be involved in the pathogenesis of DKD. In vitro experiments demonstrated that circ-0069561 aggravated glucose-induced podocyte damage and ferroptosis. CONCLUSION Circ-0069561 has the potential to be an ideal biomarker and therapeutic target for DKD progression.
Collapse
Affiliation(s)
- Chaoyi Chen
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xinran Liu
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Sai Zhu
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yukai Wang
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yu Ma
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ziyun Hu
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yonggui Wu
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Center for Scientific Research, Anhui Medical University, Hefei, China
| | - Ling Jiang
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
2
|
Liu H, Xue H, Guo Q, Xue X, Yang L, Zhao K, Liu Y. Ferroptosis meets inflammation: A new frontier in cancer therapy. Cancer Lett 2025; 620:217696. [PMID: 40189012 DOI: 10.1016/j.canlet.2025.217696] [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: 02/02/2025] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, has emerged as a critical player in cancer pathogenesis. Concurrently, inflammation, a key biological response to tissue injury or infection, significantly influences cancer development and progression. The interplay between ferroptosis and inflammation represents a promising yet underexplored area of research. This review synthesizes recent advances in understanding the molecular mechanisms governing their interaction, emphasizing how ferroptosis triggers inflammatory responses and how inflammatory mediators, such as TNF-α, regulate ferroptosis through iron metabolism and lipid peroxidation pathways. Key molecular targets within the ferroptosis-inflammation axis, including GPX4, ACSL4, and the NF-κB signaling pathway, offer therapeutic potential for cancer treatment. By modulating these targets, it may be possible to enhance ferroptosis and fine-tune inflammatory responses, thereby improving therapeutic outcomes. Additionally, this review explores the broader implications of targeting the ferroptosis-inflammation interplay in disease treatment, highlighting opportunities for developing innovative strategies to combat cancer. By bridging the gap in current knowledge, this review provides a comprehensive resource for researchers and clinicians, offering insights into the therapeutic potential of this intricate biological relationship.
Collapse
Affiliation(s)
- Hu Liu
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Hui Xue
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Qian Guo
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xutong Xue
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Lixue Yang
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China.
| | - Kaijun Zhao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Yu'e Liu
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA; Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
3
|
Gong L, Wu L, Zhao S, Xiao S, Chu X, Zhang Y, Li F, Li S, Yang H, Jiang P. Epigenetic regulation of ferroptosis in gastrointestinal cancers (Review). Int J Mol Med 2025; 55:93. [PMID: 40242977 PMCID: PMC12045471 DOI: 10.3892/ijmm.2025.5534] [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: 11/13/2024] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
Ferroptosis is a type of iron‑dependent cell death characterized by excessive lipid peroxidation and may serve as a potential therapeutic target in cancer treatment. While the mechanisms governing ferroptosis continue to be explored and elucidated, an increasing body of research highlights the significant impact of epigenetic modifications on the sensitivity of cancer cells to ferroptosis. Epigenetic processes, such as DNA methylation, histone modifications and non‑coding RNAs, have been identified as key regulators that modulate the expression of ferroptosis‑related genes. These alterations can either enhance or inhibit the sensitivity of gastrointestinal cancer (GIC) cells to ferroptosis, thereby affecting the fate of GICs. Drugs that target epigenetic markers for advanced‑stage cancer have shown promising results in enhancing ferroptosis and inhibiting tumor growth. This review explores the intricate relationship between epigenetic regulation and ferroptosis in GICs. Additionally, the potential of leveraging epigenetic modifications to trigger ferroptosis in GICs is investigated. This review highlights the importance of further research to elucidate the specific mechanisms underlying epigenetic control of ferroptosis and to advance the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Linqiang Gong
- Department of Gastroenterology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Linlin Wu
- Oncology Department, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Shiyuan Zhao
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, Shandong 272000, P.R. China
| | - Shuai Xiao
- Department of Intensive Care Medicine, Tengzhou Central People's Hospital, Jining Medical University, Tengzhou, Shandong 277500, P.R. China
| | - Xue Chu
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
| | - Yazhou Zhang
- Department of Foot and Ankle Surgery, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Fengfeng Li
- Neurosurgery Department, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Shuhui Li
- Department of Gastroenterology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Hui Yang
- Department of Gynecology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, Shandong 272000, P.R. China
| |
Collapse
|
4
|
Stachura P, Lu Z, Kronberg RM, Xu HC, Liu W, Tu JW, Schaal K, Kameri E, Picard D, von Karstedt S, Fischer U, Bhatia S, Lang PA, Borkhardt A, Pandyra AA. Deep transfer learning approach for automated cell death classification reveals novel ferroptosis-inducing agents in subsets of B-ALL. Cell Death Dis 2025; 16:396. [PMID: 40382332 DOI: 10.1038/s41419-025-07704-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/20/2025]
Abstract
Ferroptosis is a recently described type of regulated necrotic cell death whose induction has anti-cancer therapeutic potential, especially in hematological malignancies. However, efforts to uncover novel ferroptosis-inducing therapeutics have been largely unsuccessful. In the current investigation, we classified brightfield microscopy images of tumor cells undergoing defined modes of cell death using deep transfer learning (DTL). The trained DTL network was subsequently combined with high-throughput pharmacological screening approaches using automated live cell imaging to identify novel ferroptosis-inducing functions of the polo-like kinase inhibitor volasertib. Secondary validation showed that subsets of B-cell acute lymphoblastic leukemia (B-ALL) cell lines, namely 697, NALM6, HAL01, REH and primary patient B-ALL samples were sensitive to ferroptosis induction by volasertib. This was accompanied by an upregulation of ferroptosis-related genes post-volasertib treatment in cell lines and patient samples. Importantly, using several leukemia models, we determined that volasertib delayed tumor growth and induced ferroptosis in vivo. Taken together, we have applied DTL to automated live-cell imaging in pharmacological screening to identify novel ferroptosis-inducing functions of a clinically relevant anti-cancer therapeutic.
Collapse
Affiliation(s)
- Paweł Stachura
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Zhe Lu
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Raphael M Kronberg
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225, Düsseldorf, Germany
- Mathematical Modelling of Biological Systems, Heinrich Heine University, Düsseldorf, North Rhine-Westphalia, Germany
- Deep-Sea Ecology and Technology, Alfred Wegener Institute Helmholtz Centre for Polar and Marine Research, Bremerhaven, Germany
| | - Haifeng C Xu
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Wei Liu
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Jia-Wey Tu
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Katerina Schaal
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Ersen Kameri
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Cancer Prevention Graduate School (CPGS), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel Picard
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Silvia von Karstedt
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Weyertal 115b, Cologne, 50931, Germany
- CECAD Cluster of Excellence, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Straße 26, Cologne, 50931, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Robert-Koch-Straße 21, Cologne, 50931, Germany
| | - Ute Fischer
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Cancer Prevention Graduate School (CPGS), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sanil Bhatia
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Philipp A Lang
- Department of Molecular Medicine II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Aleksandra A Pandyra
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225, Düsseldorf, Germany.
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany.
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany.
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany.
| |
Collapse
|
5
|
Gu J, Ruan J, Guo C, Li Z, Fu H, Xie Y, Xie H, Gong X, Shi H. Organelles Ca 2+ redistribution contributes to cadmium-induced EMT of renal cancer cells through p-cPLA 2-mediated arachidonic acid release. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 298:118317. [PMID: 40383071 DOI: 10.1016/j.ecoenv.2025.118317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/28/2025] [Accepted: 05/10/2025] [Indexed: 05/20/2025]
Abstract
Cadmium ion (Cd2+) is a non-essential metal that can increase cancer risk, including potentially renal cell carcinoma (RCC), though this link is not definitive. Cd2+ exposure impairs fatty acid metabolism in the kidneys, particularly affecting arachidonic acid (AA) levels, which are crucial for health. Previous studies have suggested that Cd2+-altered the AA metabolism associates with renal dysfunction. However, the role and mechanism of Cd2+-regulated AA source in promoting RCC progression are still unclear. This study aims to investigate how Cd2+ exposure affects AA levels in renal cancer cells and its role in promoting cell migration. Cd2+ exposure increases AA levels through cPLA2-mediated release. It also induces calcium ion (Ca2+) redistribution from the endoplasmic reticulum (ER) to mitochondria, activating the p38 MAPK/cPLA2 signaling pathway, and epithelial-mesenchymal transition (EMT) of Caki-1 cells. Cd2+-induced ER Ca2+ release, p38 MAPK/cPLA2 signaling activation, AA levels, and EMT of Caki-1 cells were effectively reversed by siRNA knockdown of IP3R. Both exogenous AA treatments and Cd2+-induced AA metabolite PGD2 promoted EMT and cell migration of Caki-1 cells. This study highlights Cd2+'s impact on fatty acid metabolism and organelle function in renal cancer cells, identifying potential therapeutic targets for RCC.
Collapse
Affiliation(s)
- Jie Gu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212000, China.
| | - Jiacheng Ruan
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212000, China
| | - Chuanzhi Guo
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212000, China
| | - Zehua Li
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212000, China
| | - Huilin Fu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212000, China
| | - Yimin Xie
- Affiliated Hospital of Jiangsu University-Yixing Hospital, Yixing, Jiangsu 214200, China
| | - Hebing Xie
- Jiangsu Shenhou Pharmaceutical Research Co., Ltd., Nantong, 226133, China
| | - Xun Gong
- Department of Rheumatology & Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang 212013, China
| | - Haifeng Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212000, China.
| |
Collapse
|
6
|
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
|
7
|
Xiao J, Feng N, Li Q, Cao X, Huang Q, Zhou B, Fan Z, Wei L, Liu Y. Mitochondria-specific GPX4 inhibition enhances ferroptosis and antitumor immunity. J Control Release 2025:113841. [PMID: 40373937 DOI: 10.1016/j.jconrel.2025.113841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 05/03/2025] [Accepted: 05/11/2025] [Indexed: 05/17/2025]
Abstract
Ferroptosis is gaining attention as a potential cancer immunotherapy strategy, as it can stimulate antitumor responses by enhancing dendritic cell (DC) activation and the infiltration of cytotoxic T cells (CTLs). However, cancer cells often develop resistance to ferroptosis, reducing the effectiveness of existing treatments. This study demonstrates a novel mitochondria-targeted ferroptosis inducer, designated mitoFePDA@R, which is engineered to achieve a "closed-loop" cancer immunotherapy strategy of ferroptosis induction, antitumor immune activation, and ferroptosis enhancement. In this strategy, mitoFePDA@R is designed to release Fe2+ and the mitoGPX4 inhibitor RSL3 within tumor mitochondria, thereby effectively inducing ferroptosis and activating strong antitumor immune responses. Additionally, interferon γ (IFN-γ) released from CTLs inhibits GSH synthesis, which further enhances the ferroptosis sensitivity of tumor cells to form a "closed-loop" strategy. In vitro studies indicated that mitoFePDA@R induced strong ferroptosis in tumor cells by accumulating lipid peroxides (LPO) in mitochondria (which lacks mitochondria targeting). Animal studies confirmed that mitoFePDA@R effectively triggered ferroptosis and activated subsequent antitumor immune responses, leading to significant tumor growth inhibition. This provides a viable and effective strategy for ferroptosis-associated cancer immunotherapy.
Collapse
Affiliation(s)
- Jian Xiao
- Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China; Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, China
| | - Nana Feng
- Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Qiushi Li
- Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xianghui Cao
- Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Qingqing Huang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Biyu Zhou
- Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Zhenrui Fan
- Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Liping Wei
- Department of Cardiology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin 300121, China.
| | - Yang Liu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, China; Department of Thyroid and Breast Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin 300121, China.
| |
Collapse
|
8
|
Wang Z, Dai Z, Gao Y, Zhao Z, Li Z, Wang L, Gao X, Qiu Q, Qiu X, Liu Z. Development of a machine learning-based predictive risk model combining fatty acid metabolism and ferroptosis for immunotherapy response and prognosis in prostate cancer. Discov Oncol 2025; 16:744. [PMID: 40355680 PMCID: PMC12069205 DOI: 10.1007/s12672-025-02484-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
Prostate cancer (PCa) remains a leading cause of cancer-related mortality, necessitating robust prognostic models and personalized therapeutic strategies. This study integrated bulk RNA sequencing, single-cell RNA sequencing (scRNA-seq), and spatial transcriptomics to construct a prognostic model based on genes shared between ferroptosis and fatty acid metabolism (FAM). Using the TCGA-PRAD dataset, we identified 73 differentially expressed genes (DEGs) at the intersection of ferroptosis and FAM, of which 19 were significantly associated with progression-free survival (PFS). A machine learning-based prognostic model, optimized using the Lasso + Random Survival Forest (RSF) algorithm, achieved a high C-index of 0.876 and demonstrated strong predictive accuracy (1-, 2-, and 3-year AUCs: 0.77, 0.75, and 0.78, respectively). The model, validated in the DFKZ cohort, stratified patients into high- and low-risk groups, with the high-risk group exhibiting worse PFS and higher tumor mutation burden (TMB). Functional enrichment analysis revealed distinct pathway activities, with high-risk patients showing enrichment in immune-related and proliferative pathways, while low-risk patients were enriched in metabolic pathways. Immune microenvironment analysis revealed heightened immune activity in high-risk patients, characterized by increased infiltration of CD8 + T cells, regulatory T cells, and M2 macrophages, alongside elevated TIDE scores, suggesting immune evasion and resistance to immunotherapy. In contrast, low-risk patients exhibited higher infiltration of plasma cells and neutrophils and demonstrated better responses to immune checkpoint inhibitors (ICIs). Spatial transcriptomics and scRNA-seq further elucidated the spatial distribution of model genes, highlighting the central role of macrophages in mediating risk stratification. Additionally, chemotherapy sensitivity analysis identified potential therapeutic agents, such as Erlotinib and Picolinic acid, for low-risk patients. In vitro experiments showed that overexpression of CD38 in the PC-3 cell line led to elevated lipid peroxidation (C11-BODIPY) and reactive oxygen species (ROS), suggesting increased cell ferroptosis. These findings provide a comprehensive framework for risk stratification and personalized treatment in PCa, bridging molecular mechanisms with clinical outcomes.
Collapse
Affiliation(s)
- Zhenwei Wang
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Zhihong Dai
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Yuren Gao
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Zhongxiang Zhao
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Zhen Li
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Liang Wang
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Xiang Gao
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Qiuqiu Qiu
- Department of Urology, Gaohou People's Hospital, Maoming, 525200, China.
| | - Xiaofu Qiu
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510317, China.
| | - Zhiyu Liu
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China.
| |
Collapse
|
9
|
Uchihara D, Shimajiri S, Harada Y, Kumamoto K, Oe S, Miyagawa K, Nakamura K, Katafuchi E, Nuratdinova F, Honma Y, Shibata M, Harada M, Nakayama T. Long-chain fatty acyl CoA synthetase 4 expression in pancreatic cancer: a marker for malignant lesions and prognostic indicator for recurrence. Diagn Pathol 2025; 20:59. [PMID: 40349055 PMCID: PMC12065365 DOI: 10.1186/s13000-025-01659-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Long-chain fatty acyl CoA synthetase 4 (ACSL4) is crucial for lipid metabolism, primarily catalyzing the formation of 12-20 carbon chain fatty acids. ACSL4 is upregulated in various cancers and linked to aggressive behavior and poor survival. A bioinformatics study showing ACSL4 upregulation in pancreatic cancer. However, utility for actual pathological diagnosis and clinical significance in pancreatic ductal adenocarcinoma (PDAC) and intraductal papillary mucinous neoplasm (IPMN) are unexplored. This study aimed to investigate ACSL4 expression in PDAC and IPMN, and evaluate its clinical implications. METHODS We examined ACSL4 expression using immunohistochemistry in 165 patients with PDAC and IPMN. Differences in ACSL4 expression between malignant and benign lesions were evaluated using the Pearson χ2 test. The association between ACSL4 expression, pathological parameters, and survival was assessed through Kaplan-Meier and Cox regression analyses in 96 patients with invasive cancer. RESULTS Compared to normal pancreatic ducts, low-grade pancreatic intraepithelial neoplasm, and intraductal papillary mucinous adenoma (IPMA) (3.3%, 3.4%, and 2.7%, respectively), ACSL4 expression was significantly higher in invasive PDAC, noninvasive intraductal papillary mucinous carcinoma (IPMC), and invasive IPMC (77%, 86.7%, and 93.9%, respectively). In invasive cancers, low ACSL4 expression was associated with a higher frequency of lymphovascular invasion and recurrence and shorter disease-free survival (P = 0.006). Additionally, low ACSL4 expression was an independent prognostic factor for shorter disease-free survival in multivariable Cox regression analysis (HR = 2.409, 95% CI: 1.121-5.180, P = 0.024). CONCLUSION ACSL4 expression helps differentiate cancerous from precancerous lesions in pancreatic cancer, but low expression is linked to a higher frequency of lymphovascular invasion and shorter disease-free survival in invasive cases. Due to the limited sample size and broad confidence intervals, the findings of this study should be interpreted with caution and require validation in larger, independent cohorts.
Collapse
Affiliation(s)
- Daiki Uchihara
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan.
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan.
| | - Shohei Shimajiri
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoshikazu Harada
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Keiichiro Kumamoto
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Shinji Oe
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Koichiro Miyagawa
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Koichi Nakamura
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Eisuke Katafuchi
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Fariza Nuratdinova
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yuichi Honma
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Michihiko Shibata
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Masaru Harada
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Toshiyuki Nakayama
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
10
|
Yu Y, Hu J, Wang W, Lei H, Xi Z, Zhang P, Zhao E, Lu C, Chen H, Liu C, Li L. Targeting PSMD14 combined with arachidonic acid induces synthetic lethality via FADS1 m 6A modification in triple-negative breast cancer. SCIENCE ADVANCES 2025; 11:eadr3173. [PMID: 40344056 PMCID: PMC12063657 DOI: 10.1126/sciadv.adr3173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 04/03/2025] [Indexed: 05/11/2025]
Abstract
Dysregulation of deubiquitination is essential for cancer growth. However, the role of 26S proteasome non-ATPase regulatory subunit 14 (PSMD14) in the progression of triple-negative breast cancer (TNBC) remains to be determined. Gain- and loss-of-function experiments showed that silencing PSMD14 notably attenuated the growth, invasion, and metastasis of TNBC cells in vitro and in vivo. Overexpression of PSMD14 produced the opposite results. Mechanistically, PSMD14 decreased K63-linked ubiquitination on SF3B4 protein to de-ubiquitin and stabilize SF3B4 protein. Then, SF3B4/HNRNPC complex bound to FADS1 mRNA and promoted exon inclusion in the target mRNA through m6A site on FADS1 mRNA recognized by HNRNPC, thereby up-regulating the expression of FADS1 and activating Akt/mTOR signaling. Exogenous arachidonic acid supplementation combined with PSMD14 knockdown induced synthetic lethality, which was further confirmed in TNBC organoid (PDO) and TNBC patient-derived xenograft (PDX) mouse models. Overall, our findings reveal an oncogenic role of PSMD14 in TNBC progression, which indicates a potential biomarker and ferroptosis-mediated therapeutic strategy for TNBC.
Collapse
Affiliation(s)
- Yuanhang Yu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jin Hu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wenwen Wang
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hao Lei
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University and Key Laboratory of Tropical Translational Medicine of Ministry of Education and School of Tropical Medicine, Hainan Medical University, Haikou 570311, China
| | - Zihan Xi
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Peiyi Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Ende Zhao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chong Lu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hengyu Chen
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University and Key Laboratory of Tropical Translational Medicine of Ministry of Education and School of Tropical Medicine, Hainan Medical University, Haikou 570311, China
| | - Chunping Liu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lei Li
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
11
|
Wang Z, Di Y, Ye L, Fang W, Wen X, Zhang X, Qin J, Wang Y, Hu K, Zhu Z, He W, Chen Y. NANS suppresses NF-κB signaling to promote ferroptosis by perturbing iron homeostasis. Cell Rep 2025; 44:115701. [PMID: 40349344 DOI: 10.1016/j.celrep.2025.115701] [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: 09/12/2024] [Revised: 03/20/2025] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
Metastatic colorectal cancer (CRC) cells endure survival challenges, including treatment-induced ferroptosis. While adaptation to ferroptosis stress facilitates metastasis, reciprocal regulatory mechanisms remain unclear. Here, a CRISPR-Cas9 screen identifies N-acetylneuraminate synthase (NANS) as a ferroptosis promoter in CRC, regardless of its metabolic function. NANS expression is downregulated and correlates with poor prognosis in patients with CRC. Under ferroptotic stress, cyclin-dependent kinase 1 (CDK1) phosphorylates NANS at serine 275 (S275), triggering its dissociation from TAK1. Phosphorylated NANS is ubiquitinated by UBE2N at K246, leading to degradation, which activates TAK1-NF-κB signaling and upregulates the ferroptosis inhibitor FTH1, enabling metastasis via ferroptosis resistance. NANS pS275 levels are associated with tumor aggressiveness and clinical outcomes in patients with CRC. These findings indicate that NANS suppresses CRC metastasis by enhancing ferroptosis susceptibility, while CDK1-mediated phosphorylation at S275 drives adaptive resistance. Targeting this phosphorylation axis may improve ferroptosis-inducing therapies to restrict metastatic progression in CRC.
Collapse
Affiliation(s)
- Ziyang Wang
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| | - Yuqin Di
- Molecular Diagnosis and Gene Testing Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Lvlan Ye
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Wenzheng Fang
- Department of Oncology, People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fujian 350000, China
| | - Xiangqiong Wen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Xiang Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Jiale Qin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Youpeng Wang
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Kunhua Hu
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Zhenxin Zhu
- Gastrointestinal Surgery Department, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, P.R. China.
| | - Weiling He
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China.
| | - Ying Chen
- Department of Gastroenterology, Changhai Hospital affiliated to the Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
12
|
Edwards DN, Wang S, Kane K, Song W, Kim LC, Ngwa VM, Hwang Y, Ess K, Boothby MR, Chen J. Increased fatty acid delivery by tumor endothelium promotes metastatic outgrowth. JCI Insight 2025; 10:e187531. [PMID: 40198126 DOI: 10.1172/jci.insight.187531] [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: 09/27/2024] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
Metastatic outgrowth in distant microscopic niches requires sufficient nutrients, including fatty acids (FAs), to support tumor growth and to generate an immunosuppressive tumor microenvironment (TME). However, despite the important role of FAs in metastasis, the regulation of FA supply in metastatic niches has not been defined. In this report, we show that tumor endothelium actively promotes outgrowth and restricts antitumor cytolysis by transferring FAs into developing metastatic tumors. We describe a process of transendothelial FA delivery via endosomes that requires mTORC1 activity. Thus, endothelial cell-specific targeted deletion of Raptor (RptorECKO), a unique component of the mTORC1 complex, significantly reduced metastatic tumor burden that was associated with improved markers of T cell cytotoxicity. Low-dose everolimus that selectively inhibited endothelial mTORC1 improves immune checkpoint responses in metastatic disease models. This work reveals the importance of transendothelial nutrient delivery to the TME, highlighting a future target for therapeutic development.
Collapse
Affiliation(s)
- Deanna N Edwards
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, USA
| | - Shan Wang
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kelby Kane
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Wenqiang Song
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Division of Epidemiology, and
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Laura C Kim
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Verra M Ngwa
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yoonha Hwang
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kevin Ess
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Denver, Colorado, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mark R Boothby
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, Tennessee, USA
| | - Jin Chen
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, USA
- Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
13
|
Huang S, Liu K, Liu Q, Tao S, Wang H. Comprehensive analysis of ferroptosis-related long non-coding RNA and its association with tumor progression and ferroptosis in gastric cancer. BMC Gastroenterol 2025; 25:349. [PMID: 40340563 PMCID: PMC12063400 DOI: 10.1186/s12876-025-03951-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 04/29/2025] [Indexed: 05/10/2025] Open
Abstract
Gastric cancer (GC) is one of the most common malignant tumors with a poor prognosis. Ferroptosis is an distinct type of non-apoptotic cell death that is closely associated with tumor prognosis. Thus, we aimed to develop an novel prognosis risk model based on ferroptosis-related lncRNAs and excavate novel diagnostic markers. In this study, eight ferroptosis-related lncRNAs were obtained for constructing the prognosis model in GC based on TCGA database. The patients in the high-risk group had worse survival than those in the low-risk group, and the risk-grouping could be used as an independent prognostic factor for OS. Receiver operating characteristic curve analysis demonstrated this risk model was superior to traditional clinicopathological features in predicting GC prognosis. GSEA revealed that these lncRNAs were mainly involved in cell adhesion, cancer pathways, and immune function regulation. The key gene HAGLR of this risk signature was up-regulated in GC tissues and cells. Function assays showed that knockdown of HAGLR could effectively inhibit the GC cells proliferation and migration, whereas silencing HAGLR accelerated apoptosis and ferroptosis cell death process. In conclusion, we established a novel ferroptosis-related prognostic risk signature including eight lncRNAs, which may improve prognostic predictive accuracy for patients with GC.
Collapse
Affiliation(s)
- Shenglan Huang
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, Jiangxi Province, 330006, P.R. China
| | - Kan Liu
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, Jiangxi Province, 330006, P.R. China
| | - Queling Liu
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, Jiangxi Province, 330006, P.R. China
| | - Si Tao
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, Jiangxi Province, 330006, P.R. China
| | - Hua Wang
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, Jiangxi Province, 330006, P.R. China.
| |
Collapse
|
14
|
Huang J, Yu S, Luo J, Luo X, Yang J, Wang X. IFN-γ could induce ferroptosis in keloid fibroblasts by inhibiting the expression of serpine2. Cell Death Discov 2025; 11:217. [PMID: 40324981 PMCID: PMC12053758 DOI: 10.1038/s41420-025-02401-3] [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: 10/17/2024] [Revised: 02/14/2025] [Accepted: 03/17/2025] [Indexed: 05/07/2025] Open
Abstract
Keloids are common pathological scars resulting from previous trauma or inflammation. Interferon-gamma (IFN-γ) has shown significant therapeutic effects when used alone or in combination with other agents. While IFN-γ has been found to regulate ferroptosis in tumor cells, its ability to regulate ferroptosis in keloid fibroblasts (KFs) is unclear. Here, we have demonstrated a direct causal relationship between IFN-γ levels and ferroptosis in KFs. To explore the intrinsic mechanism, we performed genome-wide RNA and proteomics sequencing and found that serpine2 was the most significantly downregulated gene in KFs after exogenous overexpression of IFN-γ. Serpine2, which belongs to a family of serine protease inhibitors, has been shown to play an important role in fibrotic diseases. Therefore, we hypothesized that serpine2 is a downstream gene in the regulation of ferroptosis in KFs by IFN-γ. Our results showed that serpine2 overexpression promotes collagen synthesis, which in turn promotes the proliferation, migration, and invasive functions of KFs. We further demonstrated that serpine2 overexpression promoted system Xc- transporter expression, cystine uptake, and glutathione synthesis, enhanced GPX4 activity; and inhibited reactive oxygen species generation. This resulted in a reduction in intracellular lipid peroxidation and the levels of its metabolite malondialdehyde, as well as inhibited ferroptosis in KFs. IFN-γ reversed these effects of serpine2 overexpression. These results were largely confirmed in in vivo keloid models too. These findings imply that IFN-γ not only directly induces ferroptosis in KFs but also enhances their sensitivity to ferroptosis by inhibiting the synthesis of SLC7A11 and SLC3A2 through downregulation of serpine2. In summary, we suggest that the serpine2-system Xc- axis is a promising therapeutic target for the treatment of keloids.
Collapse
Affiliation(s)
- Jingyan Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shun Yu
- The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Jing Luo
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xusong Luo
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jun Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Xiuxia Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
15
|
Huang C, Li J, Wu R, Li Y, Zhang C. Targeting pyroptosis for cancer immunotherapy: mechanistic insights and clinical perspectives. Mol Cancer 2025; 24:131. [PMID: 40319304 PMCID: PMC12049004 DOI: 10.1186/s12943-025-02344-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025] Open
Abstract
Pyroptosis is a distinct form of programmed cell death characterized by the rupture of the cell membrane and robust inflammatory responses. Increasing evidence suggests that pyroptosis significantly affects the tumor microenvironment and antitumor immunity by releasing damage-associated molecular patterns (DAMPs) and pro-inflammatory mediators, thereby establishing it as a pivotal target in cancer immunotherapy. This review thoroughly explores the molecular mechanisms underlying pyroptosis, with a particular focus on inflammasome activation and the gasdermin family of proteins (GSDMs). It examines the role of pyroptotic cell death in reshaping the tumor immune microenvironment (TIME) involving both tumor and immune cells, and discusses recent advancements in targeting pyroptotic pathways through therapeutic strategies such as small molecule modulators, engineered nanocarriers, and combinatory treatments with immune checkpoint inhibitors. We also review recent advances and future directions in targeting pyroptosis to enhance tumor immunotherapy with immune checkpoint inhibitors, adoptive cell therapy, and tumor vaccines. This study suggested that targeting pyroptosis offers a promising avenue to amplify antitumor immune responses and surmount resistance to existing immunotherapies, potentially leading to more efficacious cancer treatments.
Collapse
Affiliation(s)
- Chen Huang
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiayi Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ruiyan Wu
- West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yangqian Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Chenliang Zhang
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
16
|
Liao P, Zhou Y, Qiu Y, Hu R, Li H, Sun H, Li Y. Metal-modulated T cell antitumor immunity and emerging metalloimmunotherapy. Cancer Metastasis Rev 2025; 44:49. [PMID: 40301229 DOI: 10.1007/s10555-025-10266-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/19/2025] [Indexed: 05/01/2025]
Abstract
In recent years, increasing evidence has shown that metals play important roles in both innate and adaptive immunity. An emerging concept of metalloimmunotherapy has been proposed, which may accelerate the development of immunotherapy for cancers. Here, we discuss how metals affect T cell function through different signaling pathways. Metals impact the fate of T cells, including their activation, proliferation, cytotoxicity, and differentiation. Most importantly, metals also participate in mitochondrial operation by regulating energy production and reactive oxygen species homeostasis in T cells. We also identified the metal-based mutual effects between tumor cells and T cells in the tumor microenvironment. Overall, the antitumor effect of T cells can be improved by targeting metal metabolism and metalloimmunotherapy, which will be a step forward in the treatment of cancers.
Collapse
Affiliation(s)
- Peiyun Liao
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying Zhou
- Department of Chemistry, CAS-HKU Joint Laboratory of Metallomics On Health and Environment, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yingqi Qiu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Rong Hu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongyan Li
- Department of Chemistry, CAS-HKU Joint Laboratory of Metallomics On Health and Environment, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Hongzhe Sun
- Department of Chemistry, CAS-HKU Joint Laboratory of Metallomics On Health and Environment, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.
- Guangdong Engineering Research Center of Precision Immune Cell Therapy Technology, Zhujiang Hospital, No. 253, Gongye Road, Guangzhou, China.
| |
Collapse
|
17
|
Yang J, Han J. Comprehensive analysis based on IFN-γ and SASP related genes, bulk RNA and single-cell sequencing to evaluate the prognosis and immune landscape of stomach adenocarcinoma. Genes Genomics 2025:10.1007/s13258-025-01646-7. [PMID: 40293675 DOI: 10.1007/s13258-025-01646-7] [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: 12/05/2024] [Accepted: 04/15/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Stomach adenocarcinoma (STAD) represents the predominant subtype of gastric cancer, known for its drug resistance, unfavorable prognosis, and low cure rates. IFN-γ serves as a cytokine generated by immune cells, instrumental in tumor immune clearance and essential to the tumor microenvironment. The aging-associated secretory phenotype (SASP) can modify the local tissue environment, facilitating gastric cancer progression and chemotherapy resistance. OBJECTIVE This study intends to identify STAD subtypes based on IFN-γ and SASP-related genes and to develop a risk prognostic model for predicting patient survival, tumor immune microenvironment, and responses to drug treatment. METHODS The genomic and clinical datasets originate from the Cancer Genome Atlas (TCGA) database, while the genes associated with IFN-γ and SASP come from pertinent scholarly articles. We discovered the prognostic genes linked to IFN-γ and SASP in STAD using Cox regression analysis. Next, we applied non-negative matrix factorization (NMF) to categorize LIHC into distinct molecular subtypes, identifying differentially expressed genes across these subtypes. Following this, we developed a predictive model using Cox and LASSO regression analyses to stratify patients into specific risk categories, validating the model to assess the prognostic significance of the identified signatures. Lastly, we integrated single-cell data to elucidate the immune landscape of STAD and identified potential drugs along with their sensitivity profiles. RESULTS We identified 17 prognostic genes related to IFN-γ and SASP, successfully classifying patients into two distinct molecular subtypes. These subtypes exhibited notable differences in immune profiles and prognostic outcomes. We pinpointed three differentially expressed genes to establish risk characteristics and created a prognostic model capable of accurately predicting patient outcomes. Our findings revealed a strong association between STAD and the extracellular matrix, low-risk group exhibited favorable prognosis, and may derive greater benefits from immunotherapy. CONCLUSION We developed a risk model using IFN-γ and SASP-associated genes to predict the prognosis of STAD patients more accurately. Additionally, we assessed the immune landscape of STAD by integrating bulk RNA and single-cell sequencing analyses. This approach may yield valuable insights for clinical decision-making and immunotherapy strategies in STAD.
Collapse
Affiliation(s)
- Jie Yang
- Department of Gastrointestinal Surgery, Xi'an Daxing Hospital, No.353 North Labor Road, Xi'an, 710016, Shanxi, China
| | - Junwei Han
- Department of Gastrointestinal Surgery, Xi'an Daxing Hospital, No.353 North Labor Road, Xi'an, 710016, Shanxi, China.
| |
Collapse
|
18
|
Ma C, Hu H, Liu H, Zhong C, Wu B, Lv C, Tian Y. Lipotoxicity, lipid peroxidation and ferroptosis: a dilemma in cancer therapy. Cell Biol Toxicol 2025; 41:75. [PMID: 40285867 PMCID: PMC12033115 DOI: 10.1007/s10565-025-10025-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/13/2025] [Indexed: 04/29/2025]
Abstract
The vulnerability of tumor cells to lipid peroxidation, driven by redox imbalance and lipid overabundance within the tumor microenvironment (TME), has become a focal point for novel antitumor strategies. Ferroptosis, a form of regulated cell death predicated on lipid peroxidation, is emerging as a promising approach. Beyond their role in directly eliminating tumor cells, lipid peroxidation and its products, such as 4-hydroxynonenal (HNE), exert an additional influence by damaging DNA and shaping an environment conducive to tumor growth and metastasis. This process polarizes macrophages towards a pro-inflammatory phenotype, dampens the antigen-presenting capacity of dendritic cells (DCs), and undermines the cytotoxic functions of T and NK cells. Furthermore, it transforms neutrophils into pro-tumorigenic polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs). The lipid peroxidation of stroma cells also contributes to tumor progression. Although advanced nanotherapies have shown the ability to target tumor cells precisely, they often overlook the nuanced effects of lipid peroxidation products. In this review, we highlight a synergistic mechanism in which lipid peroxidation products and ferroptosis contribute to an immunosuppressive state that is temporally distinct from cell death. This insight broadens our understanding of ferroptosis-derived immunosuppression, encompassing all types of immune cells within the TME. This review aims to catalyze further research in this underexplored area, emphasizing the potential of lipid peroxidation products to hinder the clinical translation of ferroptosis-based therapies.
Collapse
Affiliation(s)
- Chuhan Ma
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Huixin Hu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Hao Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Chongli Zhong
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Baokang Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Chao Lv
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| |
Collapse
|
19
|
Zheng P, Hu Z, Shen Y, Gu L, Ouyang Y, Duan Y, Ji G, Dong B, Lin Y, Wen T, Tian Q, Hou Y, Zhou Q, Sun X, Chen X, Wang KL, Luo S, Wu S, Sun Y, Li M, Xiao L, Wu Q, Meng Y, Liu G, Wang Z, Bai X, Duan S, Ding Y, Bi Y, Wang Y, Li G, Liu X, Lu Z, Wu X, Tang Z, Xu D. PSAT1 impairs ferroptosis and reduces immunotherapy efficacy via GPX4 hydroxylation. Nat Chem Biol 2025:10.1038/s41589-025-01887-3. [PMID: 40281343 DOI: 10.1038/s41589-025-01887-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 03/14/2025] [Indexed: 04/29/2025]
Abstract
Tumor cells adapt to the inflammatory tumor microenvironment (TME) and develop resistance to immunotherapy, with ferroptosis being a major form of tumor cell death. However, the mechanisms by which tumor cells coordinate TME stimuli and their unique metabolic traits to evade ferroptosis and develop resistance to immunotherapy remain unclear. Here we showed that interferon-γ (IFNγ)-activated calcium/calmodulin-dependent protein kinase II phosphorylates phosphoserine aminotransferase 1 (PSAT1) at serine 337 (S337), allowing it to interact with glutathione peroxidase 4 (GPX4) and stabilize the protein, counteracting ferroptosis. PSAT1 elevates GPX4 stability by promoting α-ketoglutarate-dependent PHD3-mediated GPX4 proline 159 (P159) hydroxylation, disrupting its binding to HSC70 and inhibiting autophagy-mediated degradation. In mice, reconstitution of PSAT1 S337A or GPX4 P159A promotes ferroptosis and suppresses triple-negative breast cancer (TNBC) progression. Blocking PSAT1 pS337 with CPP elevates IFNγ-induced ferroptosis and enhances the efficacy of programmed cell death protein 1 (PD-1) antibodies in TNBC. Additionally, PSAT1-mediated GPX4 hydroxylation correlates with poor immunotherapy outcomes in patients with TNBC, highlighting PSAT1's noncanonical role in suppressing ferroptosis and immunotherapy sensitivity.
Collapse
Affiliation(s)
- Peixiang Zheng
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Zhiqiang Hu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Yuli Shen
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Lina Gu
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuan Ouyang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yuran Duan
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Guimei Ji
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Bofei Dong
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Yanni Lin
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Ting Wen
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Qi Tian
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Yueru Hou
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Qimin Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue Sun
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaohan Chen
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | | | - Shudi Luo
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Shiqi Wu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Yuening Sun
- Department of Pharmacy, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Min Li
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Liwei Xiao
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Qingang Wu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Ying Meng
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Guijun Liu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Zheng Wang
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shengzhong Duan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanli Bi
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Yuhao Wang
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Gaopeng Li
- Department of Colorectal Surgery and Oncology of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoguang Liu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhimin Lu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Xiaohong Wu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, China.
| | - Zhiyuan Tang
- Department of Pharmacy, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
| | - Daqian Xu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China.
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China.
| |
Collapse
|
20
|
Li J, Zhou Y, Wang W. Interfering with GPX4 degradation. Nat Chem Biol 2025:10.1038/s41589-025-01873-9. [PMID: 40281342 DOI: 10.1038/s41589-025-01873-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Affiliation(s)
- Jing Li
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Yuhan Zhou
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Weimin Wang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
21
|
Yapici FI, Seidel E, Dahlhaus A, Weber J, Schmidt C, de Britto Chaves Filho A, Yang M, Nenchova M, Güngör E, Stroh J, Kotouza I, Beck J, Abdallah AT, Lackmann JW, Bebber CM, Androulidaki A, Kreuzaler P, Schulze A, Frezza C, von Karstedt S. An atlas of ferroptosis-induced secretomes. Cell Death Differ 2025:10.1038/s41418-025-01517-4. [PMID: 40281125 DOI: 10.1038/s41418-025-01517-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Cells undergoing regulated necrosis systemically communicate with the immune system via the release of protein and non-protein secretomes. Ferroptosis is a recently described iron-dependent type of regulated necrosis driven by massive lipid peroxidation. While membrane rupture occurs during ferroptosis, a comprehensive appraisal of ferroptotic secretomes and their potential biological activity has been lacking. Here, we apply a multi-omics approach to provide an atlas of ferroptosis-induced secretomes and reveal a novel function in macrophage priming. Proteins with assigned DAMP and innate immune system function, such as MIF, heat shock proteins (HSPs), and chaperones, were released from ferroptotic cells. Non-protein secretomes with assigned inflammatory function contained oxylipins as well as TCA- and methionine-cycle metabolites. Interestingly, incubation of bone marrow-derived macrophages (BMDMs) with ferroptotic supernatants induced transcriptional reprogramming consistent with priming. Indeed, exposure to ferroptotic supernatants enhanced LPS-induced cytokine production. These results define a catalog of ferroptosis-induced secretomes and identify a biological activity in macrophage priming with important implications for the fine-tuning of inflammatory processes.
Collapse
Affiliation(s)
- F Isil Yapici
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Eric Seidel
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Alina Dahlhaus
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Josephine Weber
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Christina Schmidt
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, Institute for Metabolomics in Ageing, University of Cologne, Cologne, Germany
- University of Cologne, Faculty of Mathematics and Natural Sciences, Institute for Genetics, Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
- Institute for Computational Biomedicine, Faculty of Medicine, and Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
| | - Adriano de Britto Chaves Filho
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Ming Yang
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, Institute for Metabolomics in Ageing, University of Cologne, Cologne, Germany
- University of Cologne, Faculty of Mathematics and Natural Sciences, Institute for Genetics, Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Maria Nenchova
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Emre Güngör
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Jenny Stroh
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Ioanna Kotouza
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Julia Beck
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Ali T Abdallah
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
- Institute of Medical Statistics and Computational Biology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Jan-Wilm Lackmann
- University of Cologne, Faculty of Mathematics and Natural Sciences, Institute for Genetics, Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Christina M Bebber
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Ariadne Androulidaki
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Peter Kreuzaler
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, Institute for Metabolomics in Ageing, University of Cologne, Cologne, Germany
| | - Almut Schulze
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Christian Frezza
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, Institute for Metabolomics in Ageing, University of Cologne, Cologne, Germany
- University of Cologne, Faculty of Mathematics and Natural Sciences, Institute for Genetics, Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Silvia von Karstedt
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
- Faculty of Medicine and University Hospital Cologne, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
22
|
Shan G, Bian Y, Ren S, Hu Z, Pan B, Zeng D, Zheng Z, Fan H, Bi G, Yao G, Zhan C. Sarcosine sensitizes lung adenocarcinoma to chemotherapy by dual activation of ferroptosis via PDK4/PDHA1 signaling and NMDAR-mediated iron export. Exp Hematol Oncol 2025; 14:60. [PMID: 40275333 PMCID: PMC12023509 DOI: 10.1186/s40164-025-00657-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Ferroptosis, a regulated cell death driven by iron-dependent lipid peroxidation, is associated with chemoresistance in lung adenocarcinoma (LUAD). This study aims to investigate the role of sarcosine in ferroptosis and its underlying mechanisms. METHODS An RSL3-induced ferroptosis model was used to screen a library of 889 human endogenous metabolites and metabolomic profiling was harnessed to identify metabolites associated with ferroptosis. Cell viability, lipid-reactive oxygen species (ROS), ferrous iron, malondialdehyde (MDA), and mitochondrial integrity were assessed to evaluate sarcosine's effects on ferroptosis. Metabolic fate was studied using 15N-labeled sarcosine. Next, we used untargeted metabolomic profiling and next-generation sequencing to dissect metabolic and transcriptomic changes upon sarcosine supplementation. The effects of sarcosine on ferroptosis and chemotherapy were further validated in patient-derived organoids (PDOs), xenograft models, and LUAD tissues. RESULTS Sarcosine emerged as a potent ferroptosis inducer in the metabolic library screening, which was further confirmed via cell viability, lipid-ROS, ferrous iron, and MDA measurements. Metabolic flux analysis showed limited conversion of sarcosine to other metabolites in LUAD cells, while untargeted metabolomic profiling and seahorse assays indicated a metabolic shift from glycolysis to oxidative phosphorylation. Sarcosine enhanced pyruvate dehydrogenase activity to generate more ROS by interacting with PDK4, reducing PDHA1 phosphorylation. As a co-activator of N-methyl-D-aspartate receptor (NMDAR), sarcosine also exerted its pro-ferroptosis effect via regulating ferrous export through the NMDAR/MXD3/SLC40A1 axis. Given the significance of ferroptosis in chemotherapy, we validated that sarcosine enhanced the sensitization of cisplatin by promoting ferroptosis in LUAD cells, PDOs, and xenograft models. CONCLUSION Sarcosine promotes ferroptosis and enhances chemosensitivity, suggesting its potential as a therapeutic agent in treating LUAD.
Collapse
Affiliation(s)
- Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China
| | - Yunyi Bian
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China
| | - Shencheng Ren
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China
| | - Zhengyang Hu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China
| | - Binyang Pan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China
| | - Dejun Zeng
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China
| | - Zhaolin Zheng
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China
| | - Hong Fan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University (Xiamen Branch), No. 668 Jinhu Road, Huli District, Xiamen, China
| | - Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China.
| | - Guangyu Yao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China.
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, China.
| |
Collapse
|
23
|
Teng D, Swanson KD, Wang R, Zhuang A, Wu H, Niu Z, Cai L, Avritt FR, Gu L, Asara JM, Zhang Y, Zheng B. DHODH modulates immune evasion of cancer cells via CDP-Choline dependent regulation of phospholipid metabolism and ferroptosis. Nat Commun 2025; 16:3867. [PMID: 40274823 PMCID: PMC12022163 DOI: 10.1038/s41467-025-59307-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: 02/29/2024] [Accepted: 04/15/2025] [Indexed: 04/26/2025] Open
Abstract
The ability of cancer cells to evade immune destruction is governed by various intrinsic factors including their metabolic state. Here we demonstrate that inactivation of dihydroorotate dehydrogenase (DHODH), a pyrimidine synthesis enzyme, increases cancer cell sensitivity to T cell cytotoxicity through induction of ferroptosis. Lipidomic and metabolomic analyses reveal that DHODH inhibition reduces CDP-choline level and attenuates the synthesis of phosphatidylcholine (PC) via the CDP-choline-dependent Kennedy pathway. To compensate this loss, there is increased synthesis from phosphatidylethanolamine via the phospholipid methylation pathway resulting in increased generation of very long chain polyunsaturated fatty acid-containing PCs. Importantly, inactivation of Dhodh in cancer cells promotes the infiltration of interferon γ-secreting CD8+ T cells and enhances the anti-tumor activity of PD-1 blockade in female mouse models. Our findings reveal the importance of DHODH in regulating immune evasion through a CDP-choline dependent mechanism and implicate DHODH as a promising target to improve the efficacy of cancer immunotherapies.
Collapse
Affiliation(s)
- Da Teng
- Cedars-Sinai Cancer Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, USA
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Kenneth D Swanson
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Ruiheng Wang
- Cedars-Sinai Cancer Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, USA
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Aojia Zhuang
- Cedars-Sinai Cancer Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Haofeng Wu
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Zhixin Niu
- Epigenetics Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Li Cai
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Faith R Avritt
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, USA
| | - Lei Gu
- Epigenetics Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - John M Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Yaqing Zhang
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Bin Zheng
- Cedars-Sinai Cancer Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, USA.
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA.
| |
Collapse
|
24
|
Lu Y, Chen L, Lin Y, Zhang Y, Wang Y, Yu W, Ren F, Guo H. Fatty acid metabolism: The crossroads in intestinal homeostasis and tumor. Metabolism 2025; 169:156273. [PMID: 40280478 DOI: 10.1016/j.metabol.2025.156273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/09/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
Fatty acids (FAs) have various functions on cell regulation considering their abundant types and metabolic pathways. In addition, the relation between FA and other nutritional metabolism makes their functions more complex. As the first place for diet-derived FA metabolism, intestine is significantly influenced despite lack of clear conclusions due to the inconsistent findings. In this review, we discuss the regulation of fatty acid metabolism on the fate of intestinal stem cells in homeostasis and disorders, and also focus on the intestinal tumor development and treatment from the aspect of gut microbiota-epithelium-immune interaction. We summarize that the balances between FA oxidation and glycolysis, between oxidative phosphorylation and ketogenesis, between catabolism and anabolism, and the specific roles of individual FA types determine the diverse effects of intestinal FA metabolism in different cases. We hope this will inspire further dissection and suggest precise dietary/metabolic intervention for different demands related to intestinal health.
Collapse
Affiliation(s)
- Yao Lu
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Lining Chen
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yingying Lin
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yafei Zhang
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yuqi Wang
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Weiru Yu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Fazheng Ren
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| | - Huiyuan Guo
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
25
|
Jiang P, Jiang Z, Li S, Li YX, Chen Y, Li X. The suppressive role of GLS in radiosensitivity and irradiation-induced immune response in LUAD: integrating bioinformatics and experimental insights. Front Immunol 2025; 16:1582587. [PMID: 40308578 PMCID: PMC12040943 DOI: 10.3389/fimmu.2025.1582587] [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: 02/24/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Background Radiotherapy elicits immune activation, thereby synergistically enhancing systemic tumor control when combined with immunotherapy. Glutaminase (GLS), a key enzyme for glutamine metabolism, has been found to regulate glutamine availability within tumor microenvironment (TME). However, the precise mechanisms through which GLS modulates radiosensitivity and irradiation-induced immune responses in lung adenocarcinoma (LUAD) and its clinical value remain to be fully elucidated. Methods We employed bulk RNA-seq and single-cell transcriptomics to explore the role of GLS expression in radiosensitivity and immune infiltration. The bioinformatic results were validated by in vitro and in vivo experiments. Co-culture assays and flow cytometry were used to validate the impact of GLS expression on CD8+ T cell activation and cytotoxicity. Moreover, a GLS-DSBr (double strand break repair) prognostic model was developed using machine learning with data from 2,066 LUAD patients. Results In vitro and in vivo experiments demonstrated that GLS silence inhibited DSB repair and promoted ferroptosis, therefore enhancing radiosensitivity. Single-cell and spatial transcriptomics revealed the immunomodulatory effects of GLS expression in the TME. Further, Co-culture assays and flow cytometry experiments indicated that silencing GLS in LUAD cells potentiated the activation and cytotoxicity of CD8+ T cells in the context of radiotherapy. The GLS-DSBr model demonstrated robust predictive performance for overall survival, as well as the efficacy of radiotherapy and immunotherapy in LUAD. The applicability of GLS-DSBr model was further validated through pan-cancer analysis. Conclusion In the contexts of radiotherapy, GLS downregulation exerts dual regulatory effects by modulating ferroptosis and remodeling the immune landscapes, particularly enhancing CD8+ T cell cytotoxicity. Our work suggests that strategies preferentially targeting GLS in tumor cells may represent promising and translatable therapeutic approaches to promote antitumor efficacy of radiotherapy plus immune checkpoint blockade in LUAD patients. Furthermore, the established GLS-DSBr model serves as a robust predictive tool for prognosis and effects of radiotherapy and immunotherapy, which assists personalized treatment optimization in LUAD.
Collapse
Affiliation(s)
- Peicheng Jiang
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhifeng Jiang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Su Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- Department of Cardiology, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Ye-Xiong Li
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuqiong Chen
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Xinyan Li
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
26
|
Jinson S, Zhang Z, Lancaster GI, Murphy AJ, Morgan PK. Iron, lipid peroxidation, and ferroptosis play pathogenic roles in atherosclerosis. Cardiovasc Res 2025; 121:44-61. [PMID: 39739567 DOI: 10.1093/cvr/cvae270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/22/2024] [Accepted: 12/05/2024] [Indexed: 01/02/2025] Open
Abstract
Oxidation of lipids, excessive cell death, and iron deposition are prominent features of human atherosclerotic plaques. While extensive research has established the detrimental roles of lipid oxidation and apoptosis in atherosclerosis development, the involvement of iron in atherogenesis is not yet fully understood. With the emergence of an iron-dependent form of cell death termed ferroptosis, new attention has been brought to the complex inter-play among iron, ferroptosis, and atherosclerosis. Mechanistically, ferroptosis is caused by the lethal accumulation of iron-mediated lipid peroxides. Emerging studies have underscored ferroptosis as a contributor to worsened atherosclerosis. Herein, we review the evidence that oxidative damage and iron overload in the context of atherosclerosis may promote ferroptosis within plaques. Furthermore, we summarize recent findings of lipid peroxidation, thereby potentially ferroptosis, in various plaque cell types-such as endothelial cells, macrophages, dendritic cells, T cells, and vascular smooth muscle cells-across different stages of atherosclerosis. Understanding how these processes influence atherosclerotic plaque progression may permit targeting stage-dependent ferroptosis in each cell population and could provide a rationale for developing cell type-specific intervention strategies to mitigate atherogenic ferroptosis effectively.
Collapse
Affiliation(s)
- Swetha Jinson
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Ziyang Zhang
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Graeme I Lancaster
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Pooranee K Morgan
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| |
Collapse
|
27
|
Jiang J, Yan Y, Yang C, Cai H. Immunogenic Cell Death and Metabolic Reprogramming in Cancer: Mechanisms, Synergies, and Innovative Therapeutic Strategies. Biomedicines 2025; 13:950. [PMID: 40299564 PMCID: PMC12024911 DOI: 10.3390/biomedicines13040950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/02/2025] [Accepted: 04/08/2025] [Indexed: 05/01/2025] Open
Abstract
Immunogenic cell death (ICD) is a promising cancer therapy where dying tumor cells release damage-associated molecular patterns (DAMPs) to activate immune responses. Recent research highlights the critical role of metabolic reprogramming in tumor cells, including the Warburg effect, oxidative stress, and lipid metabolism, in modulating ICD and shaping the immune microenvironment. These metabolic changes enhance immune activation, making tumors more susceptible to immune surveillance. This review explores the molecular mechanisms linking ICD and metabolism, including mitochondrial oxidative stress, endoplasmic reticulum (ER) stress, and ferroptosis. It also discusses innovative therapeutic strategies, such as personalized combination therapies, metabolic inhibitors, and targeted delivery systems, to improve ICD efficacy. The future of cancer immunotherapy lies in integrating metabolic reprogramming and immune activation to overcome tumor immune evasion, with multi-omics approaches and microbiome modulation offering new avenues for enhanced treatment outcomes.
Collapse
Affiliation(s)
| | | | - Chunhui Yang
- Department of Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian 116023, China; (J.J.); (Y.Y.)
| | - Hong Cai
- Department of Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian 116023, China; (J.J.); (Y.Y.)
| |
Collapse
|
28
|
Ru Z, Li S, Wang M, Ni Y, Qiao H. Exploring Immune-Related Ferroptosis Genes in Thyroid Cancer: A Comprehensive Analysis. Biomedicines 2025; 13:903. [PMID: 40299520 PMCID: PMC12024864 DOI: 10.3390/biomedicines13040903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Accepted: 04/05/2025] [Indexed: 04/30/2025] Open
Abstract
Background: The increasing incidence and poor outcomes of recurrent thyroid cancer highlight the need for innovative therapies. Ferroptosis, a regulated cell death process linked to the tumour microenvironment (TME), offers a promising antitumour strategy. This study explored immune-related ferroptosis genes (IRFGs) in thyroid cancer to uncover novel therapeutic targets. Methods: CIBERSORTx and WGCNA were applied to data from TCGA-THCA to identify hub genes. A prognostic model composed of IRFGs was constructed using LASSO Cox regression. Pearson correlation was employed to analyse the relationships between IRFGs and immune features. Single-cell RNA sequencing (scRNA-seq) revealed gene expression in cell subsets, and qRT-PCR was used for validation. Results: Twelve IRFGs were identified through WGCNA, leading to the classification of thyroid cancer samples into three distinct subtypes. There were significant differences in patient outcomes among these subtypes. A prognostic risk score model was developed based on six key IRFGs (ACSL5, HSD17B11, CCL5, NCF2, PSME1, and ACTB), which were found to be closely associated with immune cell infiltration and immune responses within the TME. The prognostic risk score was identified as a risk factor for thyroid cancer outcomes (HR = 14.737, 95% CI = 1.95-111.65; p = 0.009). ScRNA-seq revealed the predominant expression of these genes in myeloid cells, with differential expression validated using qRT-PCR in thyroid tumour and normal tissues. Conclusions: This study integrates bulk and single-cell RNA sequencing data to identify IRFGs and construct a robust prognostic model, offering new therapeutic targets and improving prognostic evaluation for thyroid cancer patients.
Collapse
Affiliation(s)
- Zixuan Ru
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; (Z.R.)
| | - Siwei Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China;
| | - Minnan Wang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; (Z.R.)
| | - Yanan Ni
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; (Z.R.)
| | - Hong Qiao
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; (Z.R.)
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
29
|
Xu Y, Ge M, Xu Y, Yin K. Ferroptosis: a novel perspective on tumor immunotherapy. Front Immunol 2025; 16:1524711. [PMID: 40260246 PMCID: PMC12009862 DOI: 10.3389/fimmu.2025.1524711] [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: 11/08/2024] [Accepted: 03/11/2025] [Indexed: 04/23/2025] Open
Abstract
Ferroptosis is a novel form of programmed cell death characterized by iron-dependent accumulation of reactive oxygen species (ROS) and lipid peroxidation. The execution of ferroptosis is intricately linked to both iron and lipid metabolism. Intriguingly, iron and lipid metabolism are also pivotal for maintaining the physiological function of immune cells. Research has revealed that ferroptosis can potentiate the immunogenicity of tumor cells and engage in intricate interactions with immune cells. Certain ferroptosis inducers have the capacity to augment the efficacy of immunotherapy by modulating the tumor immune microenvironment. Ferroptosis holds immense potential in cancer immunotherapy and is anticipated to emerge as a novel therapeutic target in the future landscape of cancer treatment. In this review, we primarily delineate the ferroptosis signaling pathways and metabolic processes pertinent to immune cells, and further summarize the roles of ferroptosis in tumor-infiltrating immune cells. Ultimately, we anticipate further elucidation of the mechanisms of ferroptosis in immunotherapy and envision that strategies targeting ferroptosis and immunotherapy will be expeditiously applied in clinical oncology practice.
Collapse
Affiliation(s)
| | | | | | - Kai Yin
- Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
30
|
Yuan Q, Shi Y, Wang J, Xie Y, Li X, Zhao J, Jiang Y, Qiao Y, Guo Y, Zhang C, Lu J, Zhao T, Dong Z, Li P, Dong Z, Liu K. p38 mediated ACSL4 phosphorylation drives stress-induced esophageal squamous cell carcinoma growth through Src myristoylation. Nat Commun 2025; 16:3319. [PMID: 40195298 PMCID: PMC11976994 DOI: 10.1038/s41467-025-58342-z] [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: 12/30/2023] [Accepted: 03/18/2025] [Indexed: 04/09/2025] Open
Abstract
The comprehension of intricate molecular mechanisms underlying how external stimuli promote malignancy is conducive to cancer early prevention. Esophageal squamous cell carcinoma (ESCC) is considered as an external stimuli (hot foods, tobacco, chemo-compounds) induced cancer, characterized by stepwise progression from hyperplasia, dysplasia, carcinoma in situ and invasive carcinoma. However, the underlying molecular mechanism governing the transition from normal epithelium to neoplastic processes in ESCC under persistent external stimuli has remained elusive. Herein, we show that a positive correlation between p38 and ERK1/2 activation during the progression of ESCC. We identify that phosphorylation of ACSL4 at T679 by p38 enhances its enzymatic activity, resulting in increased production of myristoyl-CoA (C14:0 CoA). This subsequently promotes Src myristoylation and activates downstream ERK signaling. Our results partially elucidate the role of ACSL4 in mediating stress-induced signaling pathways that activate growth cascades and contribute to tumorigenesis.
Collapse
Affiliation(s)
- Qiang Yuan
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Metabolic dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, China
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Yunshu Shi
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Metabolic dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, China
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Junyong Wang
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China
| | - Yifei Xie
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoyu Li
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Metabolic dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, China
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Jimin Zhao
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China
| | - Yanan Jiang
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China
| | - Yan Qiao
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China
| | - Yaping Guo
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China
| | - Chengjuan Zhang
- Center of Bio-Repository, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Lu
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China
| | - Tongjin Zhao
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Zhongshan Hospital, Fudan University, Shanghai Qi Zhi Institute, Shanghai, China
| | - Ziming Dong
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
- Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China
| | - Peng Li
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- State Key Laboratory of Metabolic dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, China.
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China.
| | - Zigang Dong
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China.
| | - Kangdong Liu
- The Pathophysiology Department, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- State Key Laboratory of Metabolic dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, China.
- Tianjian Laboratory for Advanced Biomedical Sciences, Zhengzhou, Henan, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China.
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
31
|
Wang D, Huang W, Li G. miR-145-5p regulates hepatocellular carcinoma malignant advancement and immune escape via down-regulation of AcylCoA synthase ACSL4. BIOMOLECULES & BIOMEDICINE 2025; 25:1184-1196. [PMID: 39652084 PMCID: PMC11984366 DOI: 10.17305/bb.2024.11209] [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: 08/29/2024] [Revised: 12/02/2024] [Accepted: 12/02/2024] [Indexed: 04/04/2025]
Abstract
Hepatocellular carcinoma (HCC) exhibits a subtle onset, high incidence rates, and low survival rates, becoming a substantial threat to human health. Hence, it is crucial to discover fresh biomarkers and treatment targets for the early detection and management of HCC. CCK-8, scratch-wound, and transwell assays were used to evaluate the biological properties of HCC cell lines (Huh-7 and Hep3B). Bioinformatics analysis identified the downstream target mRNA of miR-145-5p as acyl-CoA synthetase long-chain family member 4 (ACSL4). RT-qPCR was used to test miR-145-5p and ACSL4 levels. Transwell chambers were used to co-incubate purified CD8+ T cells and HCC cells for 48 h, and the effect of CD8+ T cells on apoptosis in HCC cells was detected by flow cytometry. A subcutaneous graft tumor model was constructed, and ELISA kits were used to assess cytokine levels and CD8+ T cell activation markers. HCC cells showed a decline in miR-145-5p levels and a rise in ACSL4 levels. Overexpression of miR-145-5p hindered HCC cell proliferation, migration, and invasion, while stimulating CD8+ T cell activation. Conversely, overexpression of ACSL4 enhanced the malignant biological properties of HCC cells and reduced the effect of CD8+ T cells, while silencing ACSL4 had the opposite effect. miR-145-5p targeted and downregulated ACSL4, while overexpression of miR-145-5p weakened the promotion of HCC malignant progression caused by ACSL4 overexpression. Additionally, overexpression of miR-145-5p and silencing ACSL4 were effective in inhibiting tumor growth in vivo. In conclusion, miR-145-5p targets and downregulates ACSL4, leading to the inhibition of HCC malignant progression and preventing immune escape in HCC cells.
Collapse
Affiliation(s)
- Dingxue Wang
- Oncology Department, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Wenqi Huang
- Oncology Department, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Gao Li
- Oncology Department, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
32
|
Jiang Y, Li J, Wang T, Gu X, Li X, Liu Z, Yue W, Li M. VIPAS39 confers ferroptosis resistance in epithelial ovarian cancer through exporting ACSL4. EBioMedicine 2025; 114:105646. [PMID: 40088627 PMCID: PMC11957506 DOI: 10.1016/j.ebiom.2025.105646] [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: 09/24/2024] [Revised: 02/11/2025] [Accepted: 03/01/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND The high mortality rate associated with epithelial ovarian cancer (EOC) is primarily due to recurrence and chemoresistance, underscoring the urgent need for innovative therapeutic approaches that leverage newly identified vulnerabilities in cancer cells. While conventional chemotherapies induce apoptosis by targeting DNA or mitotic machinery, ferroptosis represents a new distinct form of programmed cell death characterised by the accumulation of lipid peroxides. METHODS The sensitivity of different EOC cell lines to ferroptosis inducers was evaluated using cell viability assays and lipid peroxidation measurements. Live-cell imaging with the pH-sensitive CD63-pHuji reporter was performed to track the extracellular export of acyl-CoA synthetase long-chain family member 4 (ACSL4) via exosomes. The upstream regulator of ACSL4 were identified through immunoprecipitation-mass spectrometry (IP-MS) and validated using protein binding assays. Finally, cell-derived xenograft (CDX) and patient-derived xenograft (PDX) models were utilised to evaluate the therapeutic potential overcoming ferroptosis resistance. FINDINGS In this study, we found that interferon (IFN)-γ combined with arachidonic acid (AA), which are endogenous ferroptosis inducers, could initiate ferroptosis in most EOC cells. However, some EOC cells displayed significant resistance. Contrary to the typical increase in ACSL4 protein observed in ferroptosis-sensitive cells, resistant EOC cells exhibited surprisingly low levels of this pro-ferroptotic lipid metabolic protein. Intriguingly, this reduction is attributed to the exosomal expulsion of ACSL4 protein, revealing a distinct cellular mechanism to evade ferroptosis. We further identified VIPAS39 as a pivotal regulator in sorting ACSL4 into late endosomes, thereby facilitating their subsequent release as exosomes. Notably, targeting VIPAS39 not only overcomes the resistance to ferroptotic cell death but also markedly suppresses tumour growth. INTERPRETATION Our findings uncover the crucial role of VIPAS39 in ferroptosis evasion by facilitating the exporting of ACSL4 protein via exosomes, highlighting VIPAS39 as a promising target for ferroptosis-based anti-cancer therapy. FUNDING Funded by Beijing Municipal Natural Science Foundation (Key program Z220011), National Natural Science Foundation of China (NSFC) (T2225006, T2488301, 82272948), Peking University Medicine Youth Science and Technology Innovation 'Sail Plan' Project Type B Medical Interdisciplinary Seed Fund (71006Y3171), GuangDong Basic and Applied Basic Research Foundation (2021A1515110820), and the special fund of the National Clinical Key Speciality Construction Program, P. R. China (2023).
Collapse
Affiliation(s)
- Yuening Jiang
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology, Third Hospital, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, 100191, China
| | - Jie Li
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology, Third Hospital, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, 100191, China
| | - Tianzhen Wang
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology, Third Hospital, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, 100191, China
| | - Xiaoyang Gu
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology, Third Hospital, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, 100191, China
| | - Xinyu Li
- Department of Animal Science, College of Animal Science, Hebei North University, Zhangjiakou, Hebei Province, China; Department of Gynecology and Obstetrics, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhaofei Liu
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Wei Yue
- Interdisciplinary Eye Research Institute (EYE-X Institute) Bengbu Medical University, Bengbu, Anhui, 233030, China.
| | - Mo Li
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology, Third Hospital, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, 100191, China; Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
33
|
Liu Y, Xie Y, Wang Y. Exploring lipid metabolism-associated gene biomarkers and their regulatory mechanisms in nasopharyngeal carcinoma. Cancer Biomark 2025; 42:18758592241301683. [PMID: 40294966 DOI: 10.1177/18758592241301683] [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: 04/30/2025]
Abstract
BackgroundNasopharyngeal carcinoma (NPC) is a neoplasm that arises from the mucosal lining of the nasopharynx. Recent investigations have underscored that reprogramming of lipid metabolism is a salient metabolic alteration in neoplastic cells. Consequently, identifying lipid metabolism-associated biomarkers in NPC is of paramount importance.MethodsUtilizing transcriptomic datasets, differentially expressed genes (DEGs) were identified from GSE12452, contrasting NPC specimens with normal controls. The Weighted Gene Co-expression Network Analysis (WGCNA) was employed to discern key module genes pertinent to NPC. Lipid metabolism-related differentially expressed genes (LMR-DEGs) were ascertained by intersecting DEGs, key module genes linked to NPC, and lipid metabolism-related genes (LMRGs) using a Venn diagram approach. Subsequently, the MCODE algorithm was applied within the protein-protein interaction (PPI) framework to pinpoint lipid metabolism-centric biomarkers for NPC. The diagnostic potential of these biomarkers was assessed through ROC analysis. In the concluding phase, a 'TF-mRNA-miRNA' interaction network was delineated using Cytoscape.ResultsIn our analysis, a total of 5026 DEGs were discerned when contrasting NPC specimens with normal controls. From this pool, 1835 genes were pinpointed as key module genes pertinent to NPC. Through a Venn diagram approach, 64 LMR-DEGs were isolated. Further analysis led to the identification of six lipid metabolism-centric biomarkers for NPC, namely GALC, SPTLC2, SMPD2, DEGS2, DEGS1, and SMPD3. Notably, these biomarkers demonstrated robust diagnostic efficacy. We found that DEGS1 was negatively correlated with SMPD2 and DEGS2. A comparative expression analysis revealed diminished expression levels of GALC, SPTLC2, SMPD2, DEGS2, and SMPD3 in the NPC cohort relative to the control group. In the terminal phase of our study, the 'TF-mRNA-miRNA' regulatory network was delineated, comprising 309 nodes and 360 interaction pairs.ConclusionIn summary, our investigation identified six lipid metabolism-associated biomarkers (GALC, SPTLC2, SMPD2, DEGS2, DEGS1, and SMPD3) linked to NPC, providing a foundational framework for potential therapeutic interventions for NPC.
Collapse
Affiliation(s)
- Yiyi Liu
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yingying Xie
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yong Wang
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
34
|
Lancaster GI, Murphy AJ. Do physiological changes in fatty acid composition alter cellular ferroptosis susceptibility and influence cell function? J Lipid Res 2025; 66:100765. [PMID: 40021010 PMCID: PMC11981815 DOI: 10.1016/j.jlr.2025.100765] [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: 12/19/2024] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
Ferroptosis is an iron-dependent form of cell death driven by the excessive peroxidation of poly-unsaturated fatty acids (PUFAs) within membrane phospholipids. Ferroptosis is a hallmark of many diseases and preventing or inducing ferroptosis has considerable therapeutic potential. Like other forms of cell death, the pathological importance and therapeutic potential of ferroptosis is well appreciated. However, while cell death modalities such as apoptosis and necroptosis have critical physiological roles, such as in development and tissue homeostasis, whether ferroptosis has important physiological roles is largely unknown. In this regard, key questions for field are as follows: Is ferroptosis used for physiological processes? Are certain cell-types purposely adapted to be either resistant or sensitive to ferroptosis to be able to function optimally? Do physiological perturbations such as aging and diet impact ferroptosis susceptibility? Herein, we have reviewed emerging evidence that supports the idea that being able to selectively and controllably induce or resist ferroptosis is essential for development and cell function. While several factors regulate ferroptosis, it appears that the ability of cells and tissues to control their lipid composition, specifically the abundance of phospholipids containing PUFAs, is crucial for cells to be able to either resist or be sensitized to ferroptosis. Finally, aging and diets enriched in specific PUFAs lead to an increase in cellular PUFA levels which may sensitize cells to ferroptosis. Therefore, changes in dietary PUFAs or againg may impact the pathogenesis of diseases where ferroptosis is involved.
Collapse
Affiliation(s)
- Graeme I Lancaster
- Department of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Immunology, Monash University, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia.
| | - Andrew J Murphy
- Department of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Immunology, Monash University, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
35
|
Wang L, ChenLiu Z, Wang D, Tang D. Cross-talks of GSH, mitochondria, RNA m6A modification, NRF2, and p53 between ferroptosis and cuproptosis in HCC: A review. Int J Biol Macromol 2025; 302:140523. [PMID: 39894098 DOI: 10.1016/j.ijbiomac.2025.140523] [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: 11/11/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
Hepatocellular carcinoma (HCC) is a common malignant tumor with high morbidity and mortality, as well as poor prognosis. Therefore, it is imperative to explore alternative therapeutic targets for HCC treatment. Ferroptosis and cuproptosis have recently been identified as metal-dependent cell death mechanisms that play significant roles in HCC treatment. This study identified potential cross-talk between ferroptosis and cuproptosis, including the common hub glutathione, common site of occurrence, mitochondria, shared epigenetic modification mode, RNA N6 methyladenosine modification, mutual inhibitor, nuclear factor erythroid 2-related factor 2, and dual regulator, p53. These findings provide a theoretical foundation for the joint induction of HCC cell death and effective inhibition of HCC progression. However, some immune cells are susceptible to ferroptosis or cuproptosis, which may impair or enhance anti-cancer immune function. We propose strategies to target specific targets molecules such as tripartite motif containing 25, ferroptosis suppressor protein 1, and peroxisome proliferator-activated receptor gamma or exploit the unique acidic environment surrounding cancer cells to precisely induce ferroptosis in cancer cells. This approach aims to advance the development of precision medicine for HCC treatment.
Collapse
Affiliation(s)
- Leihan Wang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, People's Republic of China
| | - Zhenni ChenLiu
- Clinical Medical College, Yangzhou University, Yangzhou 225000, People's Republic of China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, The Yangzhou Clinical Medical College of Xuzhou Medical University, The Yangzhou School of Clinical Medicine of Dalian Medical University, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou 225000, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, The Yangzhou Clinical Medical College of Xuzhou Medical University, The Yangzhou School of Clinical Medicine of Dalian Medical University, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou 225000, China.
| |
Collapse
|
36
|
Guo Z, Wang X, Sun J, Chen Q, Chen L, Wu O, Jin Y, Lyu T, Morgan J, Li YM, Zhou H, Chen Y, You X, Zhou Y, Chen Y, Qian Q, Wu A. Injectable Nanocomposite Hydrogels for Intervertebral Disc Degeneration: Combating Oxidative Stress, Mitochondrial Dysfunction, and Ferroptosis. Adv Healthc Mater 2025; 14:e2403892. [PMID: 40079053 DOI: 10.1002/adhm.202403892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/24/2025] [Indexed: 03/14/2025]
Abstract
Intervertebral disc degeneration (IVDD) is a major cause of low back pain, where oxidative stress and mitochondrial dysfunction are key contributors. Additionally, ferroptosis, an iron-dependent form of cell death, is identified as a critical mechanism in IVDD pathogenesis. Herein, the therapeutic potential of gallic acid (GA)-derived PGA-Cu nanoparticles, enhanced with functional octapeptide (Cys-Lys-His-Gly-d-Arg-d-Tyr-Lys-Phe, SS08) to build the mitochondria-targeted nanoparticles (PGA-Cu@SS08), and embedded within a hydrogel matrix to form a nanocomposite hydrogel, is explored. The nanoparticles show targeted localization within mitochondria, effectively scavenging reactive oxygen species and preserving mitochondrial function. The abundant phenolic hydroxyl groups present on the nanoparticle surface, along with the histidine residue of the SS08 peptide, endow these entities with the capacity to chelate iron. Through RNA sequencing analysis, it is discovered that PGA-Cu@SS08 activates the NRF2 signaling pathway, mitigating ferroptosis. It also reduces iron overload by inhibiting the autophagy of iron storage proteins. Additionally, the nanocomposite hydrogels exhibit excellent biocompatibility and biodegradability, along with enhanced mechanical properties that improve intervertebral disc (IVD) performance. PGA-Cu@SS08 is continuously released from these hydrogels, restoring IVD height and maintaining tissue hydration levels, thus facilitating future applications for alleviating IVDD.
Collapse
Affiliation(s)
- Zhenyu Guo
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xinzhou Wang
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Jing Sun
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - QiZhu Chen
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200082, China
| | - Linjie Chen
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Ouqiang Wu
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yuxin Jin
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Taidong Lyu
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Jones Morgan
- Spine Unit, The Royal Orthopaedic Hospital, Bristol Road South, Northfield, Birmingham, B31 2AP, UK
| | - Yan Michael Li
- The Minimally Invasive Brain and Spine Institute, Department of Neurosurgery, State University of New York Upstate medical university, 475 Irving Ave, #402, Syracuse, NY, 13210, USA
| | - Hao Zhou
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
| | - Yongcheng Chen
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xiuling You
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yunlong Zhou
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Qiuping Qian
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
- CINBIO, University of Vigo, Campus University Lagoas Marcosende, Vigo, 36310, Spain
| | - Aimin Wu
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
37
|
Yin Z, Shen G, Fan M, Zheng P. Lipid metabolic reprogramming and associated ferroptosis in osteosarcoma: From molecular mechanisms to potential targets. J Bone Oncol 2025; 51:100660. [PMID: 39958756 PMCID: PMC11830322 DOI: 10.1016/j.jbo.2025.100660] [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: 12/01/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/18/2025] Open
Abstract
Osteosarcoma is a common bone tumor in adolescents, which is characterized by lipid metabolism disorders and plays a key role in tumorigenesis and disease progression. Ferroptosis is an iron-dependent form of programmed cell death associated with lipid peroxidation. This review provides an in-depth analysis of the complex relationship between lipid metabolic reprogramming and associated ferroptosis in OS from the perspective of metabolic enzymes and metabolites. We discussed the molecular basis of lipid uptake, synthesis, storage, lipolysis, and the tumor microenvironment, as well as their significance in OS development. Key enzymes such as adenosine triphosphate-citrate lyase (ACLY), acetyl-CoA synthetase 2 (ACSS2), fatty acid synthase (FASN) and stearoyl-CoA desaturase-1 (SCD1) are overexpressed in OS and associated with poor prognosis. Based on specific changes in metabolic processes, this review highlights potential therapeutic targets in the lipid metabolism and ferroptosis pathways, and in particular the HMG-CoA reductase inhibitor simvastatin has shown potential in inducing apoptosis and inhibiting OS metastasis. Targeting these pathways provides new strategies for the treatment of OS. However, challenges such as the complexity of drug development and metabolic interactions must be overcome. A comprehensive understanding of the interplay between dysregulation of lipid metabolism and ferroptosis is essential for the development of innovative and effective therapies for OS, with the ultimate goal of improving patient outcomes.
Collapse
Affiliation(s)
- Zhiyang Yin
- Department of Orthopaedics Surgery, Children’s Hospital of Nanjing Medical University, Nanjing 210000 Jiangsu Province, China
| | - Guanlu Shen
- School of Pharmacy, Jiangsu Ocean University, Lianyungang, Jiangsu, China
| | - Minjie Fan
- Department of Orthopaedics Surgery, Children’s Hospital of Nanjing Medical University, Nanjing 210000 Jiangsu Province, China
| | - Pengfei Zheng
- Department of Orthopaedics Surgery, Children’s Hospital of Nanjing Medical University, Nanjing 210000 Jiangsu Province, China
| |
Collapse
|
38
|
Zheng J, Conrad M. Ferroptosis: when metabolism meets cell death. Physiol Rev 2025; 105:651-706. [PMID: 39661331 DOI: 10.1152/physrev.00031.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/18/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024] Open
Abstract
We present here a comprehensive update on recent advancements in the field of ferroptosis, with a particular emphasis on its metabolic underpinnings and physiological impacts. After briefly introducing landmark studies that have helped to shape the concept of ferroptosis as a distinct form of cell death, we critically evaluate the key metabolic determinants involved in its regulation. These include the metabolism of essential trace elements such as selenium and iron; amino acids such as cyst(e)ine, methionine, glutamine/glutamate, and tryptophan; and carbohydrates, covering glycolysis, the citric acid cycle, the electron transport chain, and the pentose phosphate pathway. We also delve into the mevalonate pathway and subsequent cholesterol biosynthesis, including intermediate metabolites like dimethylallyl pyrophosphate, squalene, coenzyme Q (CoQ), vitamin K, and 7-dehydrocholesterol, as well as fatty acid and phospholipid metabolism, including the biosynthesis and remodeling of ester and ether phospholipids and lipid peroxidation. Next, we highlight major ferroptosis surveillance systems, specifically the cyst(e)ine/glutathione/glutathione peroxidase 4 axis, the NAD(P)H/ferroptosis suppressor protein 1/CoQ/vitamin K system, and the guanosine triphosphate cyclohydrolase 1/tetrahydrobiopterin/dihydrofolate reductase axis. We also discuss other potential anti- and proferroptotic systems, including glutathione S-transferase P1, peroxiredoxin 6, dihydroorotate dehydrogenase, glycerol-3-phosphate dehydrogenase 2, vitamin K epoxide reductase complex subunit 1 like 1, nitric oxide, and acyl-CoA synthetase long-chain family member 4. Finally, we explore ferroptosis's physiological roles in aging, tumor suppression, and infection control, its pathological implications in tissue ischemia-reperfusion injury and neurodegeneration, and its potential therapeutic applications in cancer treatment. Existing drugs and compounds that may regulate ferroptosis in vivo are enumerated.
Collapse
Affiliation(s)
- Jiashuo Zheng
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
- Translational Redox Biology, Technical University of Munich (TUM), TUM Natural School of Sciences, Garching, Germany
| |
Collapse
|
39
|
Kong S, Pan H, Zhang YW, Wang F, Chen J, Dong J, Yin C, Wu J, Zhou D, Peng J, Ma J, Zhou J, Ge D, Lu Y, Wei DD, Fang J, Han W, Shen C, Koeffler HP, Wang B, Jiang Y, Jiang YY. Targeting aldehyde dehydrogenase ALDH3A1 increases ferroptosis vulnerability in squamous cancer. Oncogene 2025; 44:1037-1050. [PMID: 39863749 DOI: 10.1038/s41388-025-03277-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 12/15/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Ferroptosis is a unique modality of regulated cell death induced by excessive lipid peroxidation, playing a crucial role in tumor suppression and providing potential therapeutic strategy for cancer treatment. Here, we find that aldehyde dehydrogenase-ALDH3A1 tightly links to ferroptosis in squamous cell carcinomas (SCCs). Functional assays demonstrate the enzymatic activity-dependent regulation of ALDH3A1 in protecting SCC cells against ferroptosis through catalyzing aldehydes and mitigating lipid peroxidation. Furthermore, a specific covalent inhibitor of ALDH3A1-EN40 significantly enhances the ferroptosis sensitivity induced by the ferroptosis inducer. The combination of EN40 and a ferroptosis inducer exhibits a synergistic effect, effectively inhibiting the proliferation of SCC cells/organoids and suppressing tumor growth both in vitro and in vivo. On mechanism, high expression of ALDH3A1 is transcriptionally governed by TP63, which binds to super-enhancer of ALDH3A1. Collectively, our findings reveal a yet-unrecognized function of ALDH3A1 exploited by SCC cells to evade ferroptosis, and targeting ALDH3A1 may enhance the effect of ferroptosis-induced therapy in SCCs.
Collapse
Affiliation(s)
- Shuai Kong
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Huaguang Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yuan-Wei Zhang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Fei Wang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Jian Chen
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Jinxiu Dong
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Chuntong Yin
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Jiaqi Wu
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Dan Zhou
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Jingyi Peng
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Junboya Ma
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Jianian Zhou
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
| | - Dianlong Ge
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Yan Lu
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Dan-Dan Wei
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Jinman Fang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Wei Han
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Chengyin Shen
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - H Phillip Koeffler
- Department of Medicine, Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Boshi Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China.
| | - Yuan Jiang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- University of Science and Technology of China, Hefei, 230026, China.
| | - Yan-Yi Jiang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
40
|
Song Y, Luo X, Yao L, Chen Y, Mao X. Exploring the Role of Ferroptosis-Related Circular RNAs in Subarachnoid Hemorrhage. Mol Biotechnol 2025; 67:1310-1320. [PMID: 38619799 DOI: 10.1007/s12033-024-01140-7] [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: 01/07/2024] [Accepted: 03/06/2024] [Indexed: 04/16/2024]
Abstract
Subarachnoid hemorrhage (SAH) is a devastating cerebrovascular event associated with high mortality and significant morbidity. Recent studies have highlighted the emerging role of ferroptosis, a novel form of regulated cell death, in the pathogenesis of SAH. Circular RNAs (circRNAs), have been found to play essential roles in various cellular processes, including gene regulation and disease pathogenesis. The expression profile of circRNAs in neural tissues, particularly in the brain, suggests their critical role in synaptic function and neurogenesis. Moreover, the interplay between circRNAs and ferroptosis-related pathways, such as iron metabolism and lipid peroxidation, is explored in the context of SAH. Understanding the functional roles of specific circRNAs in the context of SAH may provide potential therapeutic targets to attenuate ferroptosis-associated brain injury. Furthermore, the potential of circRNAs as diagnostic biomarkers for SAH severity, prognosis, and treatment response is discussed. Overall, this review highlights the significance of studying the intricate interplay between circRNAs and ferroptosis in the context of SAH. Unraveling the mechanisms by which circRNAs modulate ferroptotic cell death may pave the way for the development of novel therapeutic strategies and diagnostic approaches for SAH management, ultimately improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Yanju Song
- Department of Neurology, The Third Hospital of Changsha, Changsha, 410015, China
| | - Xin Luo
- Department of Neurology, The Third Hospital of Changsha, Changsha, 410015, China
| | - Liping Yao
- Department of Neurology, The Third Hospital of Changsha, Changsha, 410015, China
| | - Yinchao Chen
- Department of Neurology, The Third Hospital of Changsha, Changsha, 410015, China
| | - Xinfa Mao
- Department of Neurology, The Third Hospital of Changsha, Changsha, 410015, China.
| |
Collapse
|
41
|
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
|
42
|
Chia TY, Sadagopan NS, Miska J. Lipid peroxidation and immune activation: TRAF3's double-edged strategy against glioblastoma. J Clin Invest 2025; 135:e190471. [PMID: 40166928 PMCID: PMC11957689 DOI: 10.1172/jci190471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Glioblastoma (GBM), the most aggressive type of primary brain tumor, continues to defy therapeutic advances with its metabolic adaptability and resistance to treatment. In this issue of the JCI, Zeng et al. delve into a pivotal mechanism underpinning this adaptability. They identified an important role for TNF receptor-associated factor 3 (TRAF3) in regulating lipid metabolism through its interaction with enoyl-CoA hydratase 1 (ECH1). These findings elucidate a unique signaling axis that shields GBM cells from lipid peroxidation and antitumor immunity, advancing therapeutic strategies for GBM that may also carry over to other cancers with similar metabolic vulnerabilities.
Collapse
Affiliation(s)
- Tzu-Yi Chia
- Department of Neurological Surgery, and
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago Illinois, USA
| | - Nishanth S. Sadagopan
- Department of Neurological Surgery, and
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago Illinois, USA
| | - Jason Miska
- Department of Neurological Surgery, and
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago Illinois, USA
| |
Collapse
|
43
|
Cheng L, Wang Y, Zhang Y. Dying to survive: harnessing inflammatory cell death for better immunotherapy. Trends Cancer 2025; 11:376-402. [PMID: 39986988 DOI: 10.1016/j.trecan.2025.01.012] [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: 10/01/2024] [Revised: 01/20/2025] [Accepted: 01/24/2025] [Indexed: 02/24/2025]
Abstract
Immunotherapy has transformed cancer treatment paradigms, but its effectiveness depends largely on the immunogenicity of the tumor. Unfortunately, the high resemblance of cancer to normal tissues makes most tumors immunologically 'cold', with a poor response to immunotherapy. Danger signals are critical for breaking immune tolerance and mobilizing robust, long-lasting antitumor immunity. Recent studies have identified inflammatory cell death modalities and their power in providing danger signals to trigger optimal tumor suppression. However, key mediators of inflammatory cell death are preferentially silenced during early tumor immunoediting. Strategies to rejuvenate inflammatory cell death hold great promise for broadening immunotherapy-responsive tumors. In this review, we examine how inflammatory cell death enhances tumor immunogenicity, how it is suppressed during immunoediting, and the potential of harnessing it for improved immunotherapy.
Collapse
Affiliation(s)
- Long Cheng
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Yibo Wang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ying Zhang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
44
|
Liang Y, Zhao Y, Qi Z, Li X, Zhao Y. Ferroptosis: CD8 +T cells' blade to destroy tumor cells or poison for self-destruction. Cell Death Discov 2025; 11:128. [PMID: 40169575 PMCID: PMC11962101 DOI: 10.1038/s41420-025-02415-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/19/2025] [Accepted: 03/19/2025] [Indexed: 04/03/2025] Open
Abstract
Ferroptosis represents an emerging, iron-dependent form of cell death driven by lipid peroxidation. In recent years, it has garnered significant attention in the realm of cancer immunotherapy, particularly in studies involving immune checkpoint inhibitors. This form of cell death not only enhances our comprehension of the tumor microenvironment but is also considered a promising therapeutic strategy to address tumor resistance, investigate immune activation mechanisms, and facilitate the development of cancer vaccines. The combination of immunotherapy with ferroptosis provides innovative targets and fresh perspectives for advancing cancer treatment. Nevertheless, tumor cells appear to possess a wider array of ferroptosis evasion strategies compared to CD8+T cells, which have been conclusively shown to be more vulnerable to ferroptosis. Furthermore, ferroptosis in the TME can create a favorable environment for tumor survival and invasion. Under this premise, both inducing tumor cell ferroptosis and inhibiting T cell ferroptosis will impact antitumor immunity to some extent, and even make the final result run counter to our therapeutic purpose. This paper systematically elucidates the dual-edged sword role of ferroptosis in the antitumor process of T cells, briefly outlining the complexity of ferroptosis within the TME. It explores potential side effects associated with ferroptosis-inducing therapies and critically considers the combined application of ferroptosis-based therapies with ICIs. Furthermore, it highlights the current challenges faced by this combined therapeutic approach and points out future directions for development.
Collapse
Affiliation(s)
- Yuan Liang
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Yixin Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhaoyang Qi
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Xinru Li
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Yuguang Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
45
|
Qi L, Qian L, Yu X, Qiu K. SIRT6 mitigates oxidative stress and RSL3-induced ferroptosis in HTR-8/SVneo cells. Tissue Cell 2025; 93:102639. [PMID: 39642638 DOI: 10.1016/j.tice.2024.102639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/06/2024] [Accepted: 11/24/2024] [Indexed: 12/09/2024]
Abstract
Dysregulation in placental trophoblast cells frequently results in oxidative stress, culminating in pregnancy-related complications. While iron is essential for fetal development, cellular ferroptosis due to elevated iron levels might mediate the emergence of preeclampsia (PE), presenting significant risks during gestation. We found abnormally activated oxidative stress and increased iron concentration in the placental tissues of PE patients. Subsequently, we treated placental trophoblasts with hydrogen peroxide and RSL3 to induce oxidative stress and ferroptosis models. The results revealed that SIRT6 overexpression activates the Nrf2/HO-1 pathway, restores the oxidative imbalance of the cells, and protects the cells from ferroptosis. Meanwhile, activation of the Nrf2/HO-1 pathway alone showed similar results. Thus, we posit that SIRT6, via the Nrf2/HO-1 pathway, alleviates cellular oxidative stress and diminishes ferroptosis, offering a novel therapeutic avenue for PE.
Collapse
Affiliation(s)
- Lifang Qi
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China
| | - Liyan Qian
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China
| | - Xiaoting Yu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China
| | - Kan Qiu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China.
| |
Collapse
|
46
|
Li M, Jin S, Ma H, Yang X, Zhang Z. Reciprocal regulation between ferroptosis and STING-type I interferon pathway suppresses head and neck squamous cell carcinoma growth through dendritic cell maturation. Oncogene 2025:10.1038/s41388-025-03368-2. [PMID: 40164871 DOI: 10.1038/s41388-025-03368-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 02/23/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
Head and neck squamous cell carcinoma (HNSCC) presents a serious clinical challenge mainly due to its resistance to conventional therapies and its complex, immunosuppressive tumor microenvironment. While recent studies have identified ferroptosis as a new therapeutic option, its impact on the immune microenvironment in HNSCC remains controversial, which may hinder its translational application. Although the role of the stimulator of interferon genes (STING)-type I interferon (IFN-I) pathway in antitumor immune responses has been widely investigated, its relationship with ferroptosis in HNSCC has not been fully explored. In this study, we discovered that ferroptosis in HNSCC inhibited tumor growth, activated STING-IFN-I pathway and subsequently improved recruitment and maturation of dendritic cells. We further demonstrated that IFN-I could enhance ferroptosis by inhibiting xCT-glutathione peroxidase 4 (GPX4) antioxidant system. To harness this positive feedback loop, we treated HNSCC tumors with both ferroptosis inducer and STING agonist, resulting in significant tumor suppression, elevated ferroptosis levels and enhanced dendritic cell infiltration. Overall, our findings reveal a mutually regulatory relationship between ferroptosis and the intrinsic STING-IFN-I pathway, providing novel insights into immune-mediated tumor suppression and suggesting its potential as therapeutic approach in HNSCC.
Collapse
Affiliation(s)
- Mingyu Li
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, No. 639, Zhizaoju Rd, Shanghai, 200011, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology. No. 639, Zhizaoju Rd, Shanghai, 200011, China
- Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China
| | - Shufang Jin
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology. No. 639, Zhizaoju Rd, Shanghai, 200011, China
- Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China
- Department of Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Hailong Ma
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology. No. 639, Zhizaoju Rd, Shanghai, 200011, China.
- Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
| | - Xi Yang
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology. No. 639, Zhizaoju Rd, Shanghai, 200011, China.
- Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
| | - Zhiyuan Zhang
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology. No. 639, Zhizaoju Rd, Shanghai, 200011, China.
- Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
| |
Collapse
|
47
|
Sun D, Cui X, Yang W, Wei M, Yan Z, Zhang M, Yu W. Simvastatin inhibits PD-L1 via ILF3 to induce ferroptosis in gastric cancer cells. Cell Death Dis 2025; 16:208. [PMID: 40140647 PMCID: PMC11947124 DOI: 10.1038/s41419-025-07562-8] [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: 10/28/2024] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025]
Abstract
The treatment of gastric cancer remains challenging, with immunotherapy serving as a critical component of the holistic approach to its treatment. The results of this study indicated that statins could decrease the serum levels of interleukin-enhancing binding factor 3 (ILF3) and programmed cell death ligand 1(PD-L1) in GC patients and improve their prognosis. Functional experiments demonstrated that simvastatin induced ferroptosis by inhibiting ILF3 in GC cells and enhanced the killing effect of activated CD8+ T cells on GC cells. The CUT&Tag assay revealed that, mechanistically, simvastatin inhibited ILF3 expression by reducing the acetylation level at residue site H3K14 in ILF3. Next-generation sequencing and Kyoto Encyclopedia of Genes and Genomes analysis revealed that ILF3 regulated PD-L1 expression through the DEPTOR/mTOR signaling pathway. Overall, simvastatin induced ferroptosis in GC cells by inhibiting ILF3 expression while promoting the activation of CD8+ T cells to augment antitumor immune responses, thereby facilitating synergistic immunotherapy.
Collapse
Affiliation(s)
- Danping Sun
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Xiaohan Cui
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Wenshuo Yang
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Meng Wei
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Zhibo Yan
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Mingxiang Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Wenbin Yu
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China.
| |
Collapse
|
48
|
Yao S, Quan Y. Research progress of ferroptosis pathway and its related molecular ubiquitination modification in liver cancer. Front Oncol 2025; 15:1502673. [PMID: 40190567 PMCID: PMC11968660 DOI: 10.3389/fonc.2025.1502673] [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: 09/27/2024] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
As a new type of programmed cell death, ferroptosis is characterized by iron metabolism disorder and reactive oxygen species (ROS) accumulation, and is involved in regulating the occurrence and development of cancer cells. Especially in the field of liver cancer treatment, ferroptosis shows great potential because it can induce tumor cell death. Ubiquitination is a process of protein post-translational modification, which can affect the stability of proteins and regulate the progress of ferroptosis. This article reviews the research progress of ubiquitination modification of molecules related to ferroptosis pathway in the regulation of liver cancer, providing a new strategy for the treatment of liver cancer.
Collapse
Affiliation(s)
- Silin Yao
- The First Clinical Medical School, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yi Quan
- The First People’s Hospital of Zhaoqing, Guangdong Medical University, Zhaoqing, Guangdong, China
| |
Collapse
|
49
|
Ju Y, Lv Y, Liu X, Lu J, Shi Y, Guo H, Xu S, Tian J, Yang J, Zhong J. Role of long non-coding RNAs in the regulation of ferroptosis in tumors. Front Immunol 2025; 16:1568567. [PMID: 40191204 PMCID: PMC11968707 DOI: 10.3389/fimmu.2025.1568567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Normal cells begin to grow indefinitely and immortalize to form tumor cells after an external stimulus resulting in a genetic mutation. Effective killing of tumor cells is the basis of various cancer therapies. Ferroptosis is a class of cell death types dependent on iron and cellular lipid peroxidation. Tumors themselves are iron-dependent, and conventional radiotherapy also sensitizes cancer cells to ferroptosis. Increasing the sensitivity of tumor cells to ferroptosis may be a potential therapeutic strategy to overcome the resistance mechanisms of conventional cancer therapy. Long noncoding RNAs (LncRNAs) are a class of transcripts more than 200 nucleotides in length that regulate gene expression at multiple levels and are involved in biological processes such as cell differentiation, cell cycle arrest, and maintenance of tumor stemness. Recent studies have found that lncRNAs regulate ferroptosis of tumor cells through multiple mechanisms and may influence or ameliorate tumor resistance to chemotherapeutic agents. With the continuous maturation of nanomaterials technology, it may provide new means for cancer treatment by regulating the levels of ferroptosis-related lncRNAs inside tumors as well as increasing the levels of Fe2+ and ROS inside tumors. In this paper, we systematically introduce the regulatory mechanism of lncRNAs in ferroptosis, the role of ferroptosis in tumor immunotherapy and the application of lncRNAs combined with ferroptosis in nanomaterials, which provides new perspectives for tumor therapy.
Collapse
Affiliation(s)
- Ying Ju
- Department of Gynecology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yuanhao Lv
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Xu Liu
- Department of Anesthesia and Perioperative Medicine, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jing Lu
- Department of Gynecology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yashen Shi
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Huimin Guo
- Department of Gynecology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Siguang Xu
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jiaqi Tian
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jun Yang
- Department of Gynecology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jiateng Zhong
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Xinxiang Engineering Technology Research Center of Digestive Tumor Molecular Diagnosis, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
50
|
Sokol KH, Lee CJ, Rogers TJ, Waldhart A, Ellis AE, Madireddy S, Daniels SR, House RRJ, Ye X, Olesnavich M, Johnson A, Furness BR, Sheldon RD, Lien EC. Lipid availability influences ferroptosis sensitivity in cancer cells by regulating polyunsaturated fatty acid trafficking. Cell Chem Biol 2025; 32:408-422.e6. [PMID: 39442523 PMCID: PMC11928283 DOI: 10.1016/j.chembiol.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/09/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
Ferroptosis is a form of cell death caused by lipid peroxidation that is emerging as a target for cancer therapy, highlighting the need to identify factors that govern ferroptosis susceptibility. Lipid peroxidation occurs primarily on phospholipids containing polyunsaturated fatty acids (PUFAs). Here, we show that even though extracellular lipid limitation reduces cellular PUFA levels, lipid-starved cancer cells are paradoxically more sensitive to ferroptosis. Using mass spectrometry-based lipidomics with stable isotope fatty acid labeling, we show that lipid limitation induces a fatty acid trafficking pathway in which PUFAs are liberated from triglycerides to synthesize highly unsaturated PUFAs such as arachidonic and adrenic acid. These PUFAs then accumulate in phospholipids, including ether phospholipids, to promote ferroptosis sensitivity. Therefore, PUFA levels within cancer cells do not necessarily correlate with ferroptosis susceptibility. Rather, how cancer cells respond to extracellular lipid levels by trafficking PUFAs into proper phospholipid pools contributes to their sensitivity to ferroptosis.
Collapse
Affiliation(s)
- Kelly H Sokol
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Cameron J Lee
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Thomas J Rogers
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Althea Waldhart
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Abigail E Ellis
- Mass Spectrometry Core, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Sahithi Madireddy
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Samuel R Daniels
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA; Van Andel Institute Graduate School, Grand Rapids, MI 49503, USA
| | - Rachel Rae J House
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Xinyu Ye
- Van Andel Institute Graduate School, Grand Rapids, MI 49503, USA
| | - Mary Olesnavich
- Van Andel Institute Graduate School, Grand Rapids, MI 49503, USA
| | - Amy Johnson
- Mass Spectrometry Core, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Benjamin R Furness
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Ryan D Sheldon
- Mass Spectrometry Core, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Evan C Lien
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA.
| |
Collapse
|