1
|
Xue X, Wang M, Cui J, Yang M, Ma L, Kang R, Tang D, Wang J. Glutathione metabolism in ferroptosis and cancer therapy. Cancer Lett 2025; 621:217697. [PMID: 40189013 DOI: 10.1016/j.canlet.2025.217697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/16/2025]
Abstract
Glutathione (GSH), a non-enzymatic antioxidant in mammalian cells, plays an essential role in maintaining redox balance, mitigating oxidative stress, and preserving cellular homeostasis. Beyond its well-established function in detoxifying reactive oxygen species (ROS), GSH serves as a critical regulator of ferroptosis-an iron-dependent form of cell death marked by excessive lipid peroxidation. Serving as a cofactor for glutathione peroxidase 4 (GPX4), GSH catalyzes the conversion of lipid peroxides into non-toxic lipid alcohols, thereby preventing the accumulation of deleterious lipid oxidation products and halting the spread of oxidative damage. In cancer cells, upregulated GSH synthesis and GPX4 activity contribute to an enhanced antioxidant defense, countering oxidative stress provoked by increased metabolic demands and exposure to therapeutic agents such as chemotherapy, radiotherapy, and immunotherapy. This ability of cancer cells to modulate their ferroptosis susceptibility through GSH metabolism underscores its potential as a therapeutic target. Additionally, GSH influences several key oncogenic and tumor-suppressive signaling pathways, including NFE2L2/NRF2, TP53/p53, NF-κB, Hippo, and mTOR, which collectively regulate responses to oxidative stress, affect metabolic processes, and modulate sensitivity to ferroptosis in cancer cells. This review explores recent advancements in understanding GSH's multifaceted role in ferroptosis, emphasizing its implications for cancer biology and therapeutic interventions.
Collapse
Affiliation(s)
- Xiangfei Xue
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China
| | - Manyuan Wang
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, 200025, China
| | - Jiangtao Cui
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China
| | - Minying Yang
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, 200025, China
| | - Lifang Ma
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China; Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, 75390, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, 75390, USA.
| | - Jiayi Wang
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China; Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China; Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
2
|
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] [Download PDF] [Figures] [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
|
3
|
Guo Q, Yang W, Robinson G, Chaludiya K, Abdulkadir AN, Roy FG, Shivakumar D, Ahmad AN, Abdulkadir SA, Kirschner AN. Unlocking the Radiosensitizing Potential of MYC Inhibition in Neuroendocrine Malignancies. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00431-6. [PMID: 40354951 DOI: 10.1016/j.ijrobp.2025.04.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 04/04/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025]
Abstract
The MYC family of transcription factors-comprising c-MYC, N-MYC, and L-MYC-plays a pivotal role in oncogenesis, driving cancer progression and resistance to therapy. While MYC proteins have long been considered challenging drug targets due to their intricate structures, recent advances have led to the development of promising inhibitors. This review explores the role of MYC overexpression in promoting radiation therapy resistance in aggressive neuroendocrine malignancies through multiple mechanisms, including increased tumor cell invasion, enhanced DNA damage repair and oxidative stress management, prosurvival autophagy, survival of circulating tumor cells, angiogenesis, awakening from dormancy, and modulation of chronic inflammation and host immunity. Paradoxically, MYC overexpression can also enhance radiosensitivity in certain cancer cells by driving proapoptotic pathways, such as reactive oxygen species-induced DNA damage that overwhelms cellular repair mechanisms, ultimately leading to cell death. Additionally, we provide a comprehensive summary of direct MYC inhibitors, detailing their current stage of preclinical and clinical development as novel anticancer therapeutics. This review highlights the role of MYC in cancer metastasis and radiation therapy resistance while examining the potential of MYC inhibitors as radiosensitizers in adult and pediatric neuroendocrine malignancies, including small cell lung cancer, large cell neuroendocrine lung cancer, Merkel cell carcinoma, neuroendocrine-differentiated prostate cancer, neuroblastoma, central nervous system embryonal tumors, and medulloblastoma.
Collapse
Affiliation(s)
- Qianyu Guo
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, Florida; Department of Internal Medicine, Mayo Clinic, Jacksonville, Florida; Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida; Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - William Yang
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Guy Robinson
- Department of Internal Medicine, Mayo Clinic, Jacksonville, Florida; Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida
| | - Keyur Chaludiya
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Divya Shivakumar
- Kamineni Academy of Medical Science and Research Centre, Hyderabad, Telangana, India
| | - Ayesha N Ahmad
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Boonshoft School of Medicine, Wright State University, Fairborn, Ohio
| | - Sarki A Abdulkadir
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| | - Austin N Kirschner
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
4
|
Hylton-McComas HM, Cordes A, Floros KV, Faber A, Drapkin BJ, Miles WO. Myc family proteins: Molecular drivers of tumorigenesis and resistance in neuroendocrine tumors. Biochim Biophys Acta Rev Cancer 2025; 1880:189332. [PMID: 40280500 DOI: 10.1016/j.bbcan.2025.189332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
Neuroendocrine cancers are a diverse and poorly understood collection of malignancies derived from neuroendocrine cells throughout the body. These cancers uniquely exhibit properties of both the nervous and endocrine systems. Only a limited number of genetic driver mutations have been identified in neuroendocrine cancers, however the mechanisms of how these genetic aberrations alter tumor biology remain elusive. Recent studies have implicated the MYC family of transcription factors as important oncogenic factors in neuroendocrine tumors. We take a systematic approach to understand the roles of the MYC family (c-MYC, n-MYC, l-MYC) in the tumorigenesis of neuroendocrine cancers of the lung, GI tract, pancreas, kidney, prostate, pediatric neuroblastoma, and adrenal glands. Reflecting the complexity of neuroendocrine cancers, we highlight the roles of the MYC family in deregulating the cell cycle and transcriptional networks, invoking cellular plasticity, affecting proliferation capacity, aiding in chromatin remodeling, angiogenesis, metabolic changes, and resistance mechanisms. Depicting the diversity of neuroendocrine cancers, we suggest new approaches in understanding the underlying tumorigenic processes of neuroendocrine cancers from the perspective of MYC.
Collapse
Affiliation(s)
- Hannah M Hylton-McComas
- Department of Cancer Biology and Genetics, The Ohio State University, 460 West 12(th) Avenue, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, The Ohio State University, 460 West 12(th) Avenue, Columbus, OH 43210, USA
| | - Alyssa Cordes
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Konstantinos V Floros
- VCU Philips Institute, Virginia Commonwealth University School of Dentistry and Massey Comprehensive Cancer Center, Richmond, VA 23298, USA; Department of Pediatrics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Anthony Faber
- VCU Philips Institute, Virginia Commonwealth University School of Dentistry and Massey Comprehensive Cancer Center, Richmond, VA 23298, USA; Department of Pediatrics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Benjamin J Drapkin
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wayne O Miles
- Department of Cancer Biology and Genetics, The Ohio State University, 460 West 12(th) Avenue, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, The Ohio State University, 460 West 12(th) Avenue, Columbus, OH 43210, USA.
| |
Collapse
|
5
|
Zhou X, Zhou Z, Qin X, Cheng J, Fu Y, Wang Y, Wang J, Qin P, Zhang D. Amino Acid Metabolism Subtypes in Neuroblastoma Identifying Distinct Prognosis and Therapeutic Vulnerabilities. J Proteome Res 2025; 24:1560-1578. [PMID: 39442086 DOI: 10.1021/acs.jproteome.4c00554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Although amino acid (AA) metabolism is linked to tumor progression and could serve as an attractive intervention target, its association with neuroblastoma (NB) is unknown. Based on AA metabolism-related genes, we established three NB subtypes associated with distinct prognoses and specific functions, with C1 and C2 having better outcomes. The C1 displayed enhanced metabolic activity and recruited metabolism-associated cells. The C2 exhibited an activated immune microenvironment and was more vulnerable to immunotherapy. The C3, characterized by cell cycle peculiarity, possessed a dismal prognosis and high frequency of gene mutations and was susceptible to chemotherapy. Furthermore, single-cell RNA sequencing analysis revealed that the C3-associated Scissor+ cell subpopulation was characterized by notorious functional states and orchestrated an immunosuppressive microenvironment. Additionally, we identified that ALK and BIRC5 contributed to the shorter lifespan of C3 and their corresponding inhibitors were potential interventions. In conclusion, we identified three distinct subtypes of NB, which help us foster individualized therapeutic strategies to improve the prognosis of NB.
Collapse
Affiliation(s)
- Xing Zhou
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xiaohan Qin
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jian Cheng
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yongcheng Fu
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yuanyuan Wang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jingyue Wang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Pan Qin
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Da Zhang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| |
Collapse
|
6
|
Xu H, Qiao X, Liang J, Qiu L, Xue L, Fang Y, Xiang H, Li X, Chen Y, Ding H. Organomolecular Ferroelectric Nanocatalyst Augments Tumor Immunotherapy by Inducing Apoptosis and Ferroptosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2417422. [PMID: 39995362 DOI: 10.1002/adma.202417422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/06/2025] [Indexed: 02/26/2025]
Abstract
Immunogenic programmed cell death effectively triggers acute inflammatory responses, thereby enhancing antitumor immunity. The advancement of biodegradable nonmetallic dual inducers represents a promising strategy. Herein, a biodegradable organomolecular ferroelectric nanoplatform (C60-TCNQ, CT) is designed to facilitate effective ferroelectric catalysis, thereby augmenting tumor immunotherapy through apoptosis and ferroptosis. CT-mediated ultrasound-triggered ferroelectric catalysis promotes ferroelectric polarization and significantly increases the production of reactive oxygen species, leading to substantial tumor cell apoptosis. Moreover, the polycyano group of CT nanoparticles selectively reacts with cysteine under mild conditions, resulting in redox imbalances and the accumulation of lipid peroxides, which contribute to the induction of ferroptosis in tumor cells. Additionally, the apoptosis and ferroptosis induced by CT stimulate immunogenic cell death progression, eliciting robust immune responses. In vivo evaluation using a bilateral tumor model demonstrates the capacity of CT to sensitize anti-PD-L1 therapy under ultrasound irradiation, achieving an impressive antitumor response rate of 96.2% against malignant melanoma and an 80% inhibition of tumor metastasis. RNA sequencing analysis revealed that treatment with CT resulted in a downregulation of gene signatures associated with the immune-related Jak-Stat signaling pathway. This study opens a novel avenue to developing organomolecular ferroelectric nanomedicines for effective tumor immunotherapy.
Collapse
Affiliation(s)
- Huning Xu
- Department of Ultrasound National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xiaohui Qiao
- Department of Ultrasound National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jing Liang
- Department of Ultrasound National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Luping Qiu
- Department of Ultrasound National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Liyun Xue
- Department of Ultrasound National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yan Fang
- Department of Ultrasound National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Huijing Xiang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Xingguang Li
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Hong Ding
- Department of Ultrasound National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| |
Collapse
|
7
|
Mete M, Ojha A, Dhar P, Das D. Deciphering Ferroptosis: From Molecular Pathways to Machine Learning-Guided Therapeutic Innovation. Mol Biotechnol 2025; 67:1290-1309. [PMID: 38613722 DOI: 10.1007/s12033-024-01139-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/11/2024] [Indexed: 04/15/2024]
Abstract
Ferroptosis is a unique form of cell death reliant on iron and lipid peroxidation. It disrupts redox balance, causing cell death by damaging the plasma membrane, with inducers acting through enzymatic pathways or transport systems. In cancer treatment, suppressing ferroptosis or circumventing it holds significant promise. Beyond cancer, ferroptosis affects aging, organs, metabolism, and nervous system. Understanding ferroptosis mechanisms holds promise for uncovering novel therapeutic strategies across a spectrum of diseases. However, detection and regulation of this regulated cell death are still mired with challenges. The dearth of cell, tissue, or organ-specific biomarkers muted the pharmacological use of ferroptosis. This review covers recent studies on ferroptosis, detailing its properties, key genes, metabolic pathways, and regulatory networks, emphasizing the interaction between cellular signaling and ferroptotic cell death. It also summarizes recent findings on ferroptosis inducers, inhibitors, and regulators, highlighting their potential therapeutic applications across diseases. The review addresses challenges in utilizing ferroptosis therapeutically and explores the use of machine learning to uncover complex patterns in ferroptosis-related data, aiding in the discovery of biomarkers, predictive models, and therapeutic targets. Finally, it discusses emerging research areas and the importance of continued investigation to harness the full therapeutic potential of targeting ferroptosis.
Collapse
Affiliation(s)
- Megha Mete
- Department of Bioengineering, National Institute of Technology Agartala, Agartala, Tripura, 799046, India
| | - Amiya Ojha
- Department of Bioengineering, National Institute of Technology Agartala, Agartala, Tripura, 799046, India
| | - Priyanka Dhar
- CSIR-Indian Institute of Chemical Biology, Kolkata, 700032, India
| | - Deeplina Das
- Department of Bioengineering, National Institute of Technology Agartala, Agartala, Tripura, 799046, India.
| |
Collapse
|
8
|
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
|
9
|
Tan L, He G, Shen C, He S, Chen Y, Guo X. Construction of a ferroptosis-based prediction model for the prognosis of MYCN-amplified neuroblastoma and screening and verification of target sites. Hereditas 2025; 162:41. [PMID: 40108662 PMCID: PMC11921587 DOI: 10.1186/s41065-025-00413-8] [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: 02/10/2025] [Accepted: 03/09/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Neuroblastoma (NB) is a prevalent extracranial solid tumor in pediatric patients. Of these, the MYCN-amplified type has a poor treatment response and prognosis. To enhance therapeutic efficacy and prognostic outcomes, numerous research teams have undertaken extensive investigations through various pathways and directions. Among these, ferroptosis has recently emerged as a significant area of research focus.Ferroptosis, a type of iron-dependent cell death, is primarily caused by lipid peroxides. This study intends to develop a prognosis model based on MYCN-amplified NB and ferroptosis-related genes (FGs). METHODS Data for this study were sourced from the TARGET and FerrDb databases. Lasso regression algorithms and univariate COX analysis were leveraged to determine feature genes; multivariate COX analysis was employed to develop a prediction model and risk scores; and receiver operating characteristic (ROC) curves and Kaplan-Meier analysis were utilized to assess the predictive ability of the model. Furthermore, discrepancies in immune cell infiltration (ICI) between the high-risk (HR) and low-risk (LR) populations were assessed via CIBERSORT analysis. Finally, experiments were conducted on MYCN-amplified and MYCN non-amplified cells so as to validate the differential expression of the gene. RESULTS A prediction model was constructed and risk scores were calculated based on 4 genes (LIFR, TP53, NRAS, and OSBPL9). The HR group, which was stratified by the median score, had a lower overall survival rate than the LR group.The differences in expression of each gene between MYCN-amplified and MYCN non-amplified cells were further confirmed through cell experiments and qPCR. CONCLUSION The prediction model in this study can be employed to forecast the prognosis of MYCN-amplified NB. These genes may represent promising new ferroptosis-related intervention targets (FITs) in treating MYCN-amplified NB, with the potential to improve patient outcomes.
Collapse
Affiliation(s)
- Linjun Tan
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, No.149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, China
- Department of Pediatrics, Guizhou Children's Hospital, No.149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, China
| | - Guoqian He
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Section 3, South Renmin Road, Chengdu, 610041, China
| | - Chengqi Shen
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Section 3, South Renmin Road, Chengdu, 610041, China
| | - Sijia He
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Section 3, South Renmin Road, Chengdu, 610041, China
| | - Yan Chen
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, No.149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, China.
- Department of Pediatrics, Guizhou Children's Hospital, No.149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, China.
- Collaborative Innovation Center for Tissue Injury Repairand Regenerative Medicine of Zunyi Medical University, No.149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, China.
| | - Xia Guo
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, No.149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, China.
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Section 3, South Renmin Road, Chengdu, 610041, China.
| |
Collapse
|
10
|
Liu Y, Fleishman JS, Wang H, Huo L. Pharmacologically Targeting Ferroptosis and Cuproptosis in Neuroblastoma. Mol Neurobiol 2025; 62:3863-3876. [PMID: 39331355 PMCID: PMC11790790 DOI: 10.1007/s12035-024-04501-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024]
Abstract
Neuroblastoma is a deadly pediatric cancer that originates from the neural crest and frequently develops in the abdomen or adrenal gland. Although multiple approaches, including chemotherapy, radiotherapy, targeted therapy, and immunotherapy, are recommended for treating neuroblastoma, the tumor will eventually develop resistance, leading to treatment failure and cancer relapse. Therefore, a firm understanding of the molecular mechanisms underlying therapeutic resistance is vital for the development of new effective therapies. Recent research suggests that cancer-specific modifications to multiple subtypes of nonapoptotic regulated cell death (RCD), such as ferroptosis and cuproptosis, contribute to therapeutic resistance in neuroblastoma. Targeting these specific types of RCD may be viable novel targets for future drug discovery in the treatment of neuroblastoma. In this review, we summarize the core mechanisms by which the inability to properly execute ferroptosis and cuproptosis can enhance the pathogenesis of neuroblastoma. Therefore, we focus on emerging therapeutic compounds that can induce ferroptosis or cuproptosis, delineating their beneficial pharmacodynamic effects in neuroblastoma treatment. Cumulatively, we suggest that the pharmacological stimulation of ferroptosis and ferroptosis may be a novel and therapeutically viable strategy to target neuroblastoma.
Collapse
Affiliation(s)
- Ying Liu
- Department of Pediatrics, The Fourth Affiliated Hospital of China Medical University, Shenyang, 100012, China.
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Hongquan Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Liang Huo
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 11004, China.
| |
Collapse
|
11
|
Fernández-Acosta R, Vintea I, Koeken I, Hassannia B, Vanden Berghe T. Harnessing ferroptosis for precision oncology: challenges and prospects. BMC Biol 2025; 23:57. [PMID: 39988655 PMCID: PMC11849278 DOI: 10.1186/s12915-025-02154-6] [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: 11/28/2024] [Accepted: 02/12/2025] [Indexed: 02/25/2025] Open
Abstract
The discovery of diverse molecular mechanisms of regulated cell death has opened new avenues for cancer therapy. Ferroptosis, a unique form of cell death driven by iron-catalyzed peroxidation of membrane phospholipids, holds particular promise for targeting resistant cancer types. This review critically examines current literature on ferroptosis, focusing on its defining features and therapeutic potential. We discuss how molecular profiling of tumors and liquid biopsies can generate extensive multi-omics datasets, which can be leveraged through machine learning-based analytical approaches for patient stratification. Addressing these challenges is essential for advancing the clinical integration of ferroptosis-driven treatments in cancer care.
Collapse
Affiliation(s)
- Roberto Fernández-Acosta
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Iuliana Vintea
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Biobix, Lab of Bioinformatics and Computational Genomics, Department of Mathematical Modelling, Statistics and Bioinformatics, Ghent University, Ghent, Belgium
| | - Ine Koeken
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Behrouz Hassannia
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Tom Vanden Berghe
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
12
|
Wang X, Zhang T, Wang S, Shi H, Dong H, Huang Y, Lai W, Hu Y, Yue C. Bio-nanocomplexes impair iron homeostasis to induce non-canonical ferroptosis in cancer cells. J Nanobiotechnology 2025; 23:121. [PMID: 39972473 PMCID: PMC11837358 DOI: 10.1186/s12951-025-03117-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 01/13/2025] [Indexed: 02/21/2025] Open
Abstract
The targeted elevation of the labile iron pool (LIP) represents the most direct and effective strategy to induce ferroptosis in cancer cells. However, the efficiency of increasing LIP to induce ferroptosis via iron supplementation is controversial due to the iron homeostasis between LIP and storage iron pool, leading to poor effects and serious safety concerns. In this study, a bio-nanocomplex named AbDA-Lim, composed of albumin, polydopamine, and limonene, is prepared to promote LIP and induce non-canonical ferroptosis in cancer cells by destroying the iron balance. Albumin avidity drives AbDA-Lim entering cancer cells. Subsequently, the released polydopamine enhances the expression of HMOX1 to degrade haem and facilitate the transformation of Fe (III) to Fe (II). Meanwhile, limonene reduces glutathione (GSH) levels via inhibiting CBS, thereby, triggering the release of Fe (II) into LIP from its GSH-bound storage state. The augmentation of LIP ultimately triggers non-canonical ferroptosis in cancer cells. Furthermore, the photothermal property of polydopamine augments the synergistic anti-tumor efficiency of AbDA-Lim by incorporating photothermal therapy. This study presents a distinctive, cascading, and biotic strategy for promoting LIP non-canonically to induce ferroptosis.
Collapse
Affiliation(s)
- Xin Wang
- Department of General Surgery, Center of Nutrition and Metabolism of Cancer, Beijing Shijitan Hospital, Key Laboratory of Cancer FSMP for State Market Regulation, Capital Medical University, Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China
| | - Tianyi Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Drug R&D, Jiangsu Key Laboratory for Nano Technology, Medical School, School of Life Science, Nanjing University, Nanjing, 210093, China
| | - Shuai Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Drug R&D, Jiangsu Key Laboratory for Nano Technology, Medical School, School of Life Science, Nanjing University, Nanjing, 210093, China
| | - Hanping Shi
- Department of General Surgery, Center of Nutrition and Metabolism of Cancer, Beijing Shijitan Hospital, Key Laboratory of Cancer FSMP for State Market Regulation, Capital Medical University, Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, 100038, China
| | - Hong Dong
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Drug R&D, Jiangsu Key Laboratory for Nano Technology, Medical School, School of Life Science, Nanjing University, Nanjing, 210093, China
| | - Yanning Huang
- Office of International Cooperation and Exchanges, Central South University, Changsha, 410008, China
| | - Wenjia Lai
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, 100190, China.
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Drug R&D, Jiangsu Key Laboratory for Nano Technology, Medical School, School of Life Science, Nanjing University, Nanjing, 210093, China.
| | - Chunyan Yue
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Drug R&D, Jiangsu Key Laboratory for Nano Technology, Medical School, School of Life Science, Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
13
|
Berg SZ, Berg J. Microbes, macrophages, and melanin: a unifying theory of disease as exemplified by cancer. Front Immunol 2025; 15:1493978. [PMID: 39981299 PMCID: PMC11840190 DOI: 10.3389/fimmu.2024.1493978] [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: 09/10/2024] [Accepted: 12/03/2024] [Indexed: 02/22/2025] Open
Abstract
It is widely accepted that cancer mostly arises from random spontaneous mutations triggered by environmental factors. Our theory challenges the idea of the random somatic mutation theory (SMT). The SMT does not fit well with Charles Darwin's theory of evolution in that the same relatively few mutations would occur so frequently and that these mutations would lead to death rather than survival of the fittest. However, it would fit well under the theory of evolution, if we were to look at it from the vantage point of pathogens and their supporting microbial communities colonizing humans and mutating host cells for their own benefit, as it does give them an evolutionary advantage and they are capable of selecting genes to mutate and of inserting their own DNA or RNA into hosts. In this article, we provide evidence that tumors are actually complex microbial communities composed of various microorganisms living within biofilms encapsulated by a hard matrix; that these microorganisms are what cause the genetic mutations seen in cancer and control angiogenesis; that these pathogens spread by hiding in tumor cells and M2 or M2-like macrophages and other phagocytic immune cells and traveling inside them to distant sites camouflaged by platelets, which they also reprogram, and prepare the distant site for metastasis; that risk factors for cancer are sources of energy that pathogens are able to utilize; and that, in accordance with our previous unifying theory of disease, pathogens utilize melanin for energy for building and sustaining tumors and metastasis. We propose a paradigm shift in our understanding of what cancer is, and, thereby, a different trajectory for avenues of treatment and prevention.
Collapse
Affiliation(s)
- Stacie Z. Berg
- Department of Translational Biology, William Edwards LLC, Baltimore, MD, United States
| | - Jonathan Berg
- Department of Translational Biology, William Edwards LLC, Baltimore, MD, United States
| |
Collapse
|
14
|
Liu H, Xu Y, Liu Y, Han X, Zhao L, Liu Y, Zhang F, Fu Y. Identification of novel ferroptosis-related biomarkers associated with the oxidative stress pathways in ischemic cardiomyopathy. IJC HEART & VASCULATURE 2025; 56:101584. [PMID: 39807364 PMCID: PMC11726793 DOI: 10.1016/j.ijcha.2024.101584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025]
Abstract
Background Ferroptosis is a cell death process that depends on iron and reactive oxygen species. It significantly contributes to cardiovascular diseases. However, its exact role in ischemic cardiomyopathy (ICM) is still unclear. Methods Using bioinformatics methods, we identified new molecular targets associated with ferroptosis in ICM and conducted various analyses-including correlation analysis, pathway enrichment analysis, protein interaction network construction, and analysis of transcription factor and drug interactions, to reveal the potential mechanisms behind these genes. Results We evaluated two independent training sets of ICM, GSE57338 and GSE5406, comprising 203 ICM samples, and validation sets GSE76701 to examine differentially expressed genes (DEGs) related to ferroptosis. After extracting the intersection of the gene sets and ferroptosis-related genes, 53 DEGs were identified. Enrichment analyses showed that the alterations in ferroptosis-related DEGs were mainly enriched in oxidative stress response, and immune-related pathways. Furthermore, 11 hub genes were identified using protein-protein interaction network analysis. The key interactions between 11 hub genes were more pronounced in protein localization during ICM development. In addition, we construct a hub gene and transcription factor interaction network and a small molecule drug-gene interaction network. We found that among these hub genes, the N-acetylneuraminate outer membrane channel(NANC) gene is positively correlated with most of the small-molecule drugs used to treat ICM, and its high expression might increase resistance. Conclusions Ferroptosis exists in ICM and and is associated with oxidative stress. This association suggests that ferroptosis may facilitate the progression of ICM.
Collapse
Affiliation(s)
- Huilin Liu
- Department of Geriatrics, Peking University Third Hospital, Beijing 100191, PR China
| | - Yuan Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Yuanmei Liu
- Department of Geriatrics, Peking University Third Hospital, Beijing 100191, PR China
| | - XueJun Han
- Department of Orthopaedics, Jiayuguan Municipal First People’s Hospital, Jiayuguan 735100, PR China
| | - Liping Zhao
- Department of Ophthalmology, Jiayuguan Municipal First People’s Hospital, Jiayuguan 735100, PR China
| | - Yixuan Liu
- College of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China
| | - Fuchun Zhang
- Department of Geriatrics, Peking University Third Hospital, Beijing 100191, PR China
| | - Yicheng Fu
- Department of Geriatrics, Peking University Third Hospital, Beijing 100191, PR China
| |
Collapse
|
15
|
Würth R, Donato E, Michel LL, Saini M, Becker L, Cheytan T, Doncevic D, Messmer T, Gutjahr E, Weber R, Klein C, Alborzinia H, Yildiz U, Vogel V, Hlevnjak M, Kozyulina P, Neuberth SJ, Schwerd-Kleine P, Jakab S, Pfarr N, Angeles AK, Laut AK, Karpova D, Falcone M, Hardt O, Theek B, Wagner CV, Becker M, Wagner S, Haselmayr M, Schmitt A, Müller-Tidow C, Riethdorf S, Pantel K, Zapatka M, Sültmann H, Herrmann C, Thewes V, Lichter P, Schneeweiss A, Sprick MR, Trumpp A. Circulating tumor cell plasticity determines breast cancer therapy resistance via neuregulin 1-HER3 signaling. NATURE CANCER 2025; 6:67-85. [PMID: 39753722 PMCID: PMC11779641 DOI: 10.1038/s43018-024-00882-2] [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: 09/25/2023] [Accepted: 11/18/2024] [Indexed: 01/31/2025]
Abstract
Circulating tumor cells (CTCs) drive metastasis, the leading cause of death in individuals with breast cancer. Due to their low abundance in the circulation, robust CTC expansion protocols are urgently needed to effectively study disease progression and therapy responses. Here we present the establishment of long-term CTC-derived organoids from female individuals with metastatic breast cancer. Multiomics analysis of CTC-derived organoids along with preclinical modeling with xenografts identified neuregulin 1 (NRG1)-ERBB2 receptor tyrosine kinase 3 (ERBB3/HER3) signaling as a key pathway required for CTC survival, growth and dissemination. Genome-wide CRISPR activation screens revealed that fibroblast growth factor receptor 1 (FGFR1) signaling serves a compensatory function to the NRG1-HER3 axis and rescues NRG1 deficiency in CTCs. Conversely, NRG1-HER3 activation induced resistance to FGFR1 inhibition, whereas combinatorial blockade impaired CTC growth. The dynamic interplay between NRG1-HER3 and FGFR1 signaling reveals the molecular basis of cancer cell plasticity and clinically relevant strategies to target it. Our CTC organoid platform enables the identification and validation of patient-specific vulnerabilities and represents an innovative tool for precision medicine.
Collapse
MESH Headings
- Humans
- Neuregulin-1/metabolism
- Neuregulin-1/genetics
- Receptor, ErbB-3/metabolism
- Receptor, ErbB-3/genetics
- Female
- Breast Neoplasms/pathology
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/genetics
- Signal Transduction
- Animals
- Neoplastic Cells, Circulating/metabolism
- Neoplastic Cells, Circulating/pathology
- Neoplastic Cells, Circulating/drug effects
- Drug Resistance, Neoplasm
- Mice
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Organoids/metabolism
- Organoids/pathology
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Roberto Würth
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany.
| | - Elisa Donato
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Laura L Michel
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Massimo Saini
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Lisa Becker
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Tasneem Cheytan
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Daria Doncevic
- Department of Bioinformatics, Institute of Pharmacy and Molecular Biotechnology & BioQuant, Heidelberg University, Heidelberg, Germany
| | - Tobias Messmer
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Ewgenija Gutjahr
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Rebecca Weber
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Corinna Klein
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Hamed Alborzinia
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Umut Yildiz
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Vanessa Vogel
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Mario Hlevnjak
- Computational Oncology, Molecular Precision Oncology Program, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Polina Kozyulina
- Computational Oncology, Molecular Precision Oncology Program, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Sarah-Jane Neuberth
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Paul Schwerd-Kleine
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Sevinç Jakab
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Nicole Pfarr
- Institute of Pathology, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Arlou Kristina Angeles
- Division of Cancer Genome Research, German Cancer Research Center, National Center for Tumor Diseases, Heidelberg, Germany
| | - Astrid K Laut
- Division of Cancer Genome Research, German Cancer Research Center, National Center for Tumor Diseases, Heidelberg, Germany
| | - Darja Karpova
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Mattia Falcone
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Olaf Hardt
- Miltenyi Biotec, Bergisch Gladbach, Germany
| | | | - Celina V Wagner
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mirjam Becker
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sabine Wagner
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martina Haselmayr
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Sabine Riethdorf
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marc Zapatka
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Holger Sültmann
- Division of Cancer Genome Research, German Cancer Research Center, National Center for Tumor Diseases, Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Carl Herrmann
- Department of Bioinformatics, Institute of Pharmacy and Molecular Biotechnology & BioQuant, Heidelberg University, Heidelberg, Germany
| | - Verena Thewes
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University, Heidelberg, Germany
| | - Peter Lichter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University, Heidelberg, Germany
| | - Andreas Schneeweiss
- Gynecologic Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin R Sprick
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH)), Heidelberg, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
16
|
Cheng J, Dong X, Yang Y, Qin X, Zhou X, Zhang D. Synergistic machine learning models utilizing ferroptosis-related genes for improved neuroblastoma outcome prediction. Transl Pediatr 2024; 13:2164-2182. [PMID: 39822999 PMCID: PMC11732634 DOI: 10.21037/tp-24-323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/03/2024] [Indexed: 01/19/2025] Open
Abstract
Background Neuroblastoma (NB) is a highly heterogeneous and common pediatric malignancy with a poor prognosis. Ferroptosis, an iron-dependent cell death pathway, may play a crucial role in NB tumor progression and immune response. This study aimed to investigate ferroptosis in NB to identify potential therapeutic targets and develop predictive models for prognosis and recurrence. Methods Six datasets were accessed from the ArrayExpress database and Gene Expression Omnibus. Ferroptosis-related genes (FRGs) were selected from the FerrDb website. Unsupervised clustering, differential expression analysis, weighted correlation network analysis (WGCNA), and gene set enrichment analysis (GSEA) were adopted to investigate potential pathways associated with ferroptosis in NB and identify the key genes involved. We used the least absolute shrinkage and selection operator (LASSO) and multivariate Cox regression to develop the ferroptosis-related prognostic signatures (FRPS) while using machine learning (ML) algorithms to construct the recurrence model. Results Ribosome and cell cycle may be the potential pathways for ferroptosis involved in NB, with MYCN and RRM2 identified as key genes in this regulatory process. Five FRGs-ATG7 (-1.009), ELAVL1 (1.739), PPARA (0.493), RDX6 (1.457), and TERT (0.247)-were screed out for the FRPS, which showed excellent predictive performance in comparison with other published NB signatures. Eight FRGs-ALDH3A2 (48.597), TERT (23.398), ULK2 (21.034), AKR1C1 (20.699), MFN2 (12.575), SLC16A1 (12.342), TF (10.240), and DDR2 (7.598)-were selected based on the importance scores to construct the recurrence model. Among the models, utilizing random forest (RF), XGboost, support vector machine (SVM), K-nearest neighbors (KNN), and linear discriminant analysis (LDA), the RF model exhibited the highest performance. Conclusions We investigated the potential ferroptosis-related pathways and hub- FRGs in NB and developed prognosis and recurrence models, providing new potential targets for prognostic evaluation and treatment in NB patients.
Collapse
Affiliation(s)
- Jian Cheng
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiao Dong
- Department of Pediatrics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Yang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaohan Qin
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xing Zhou
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Da Zhang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
17
|
Sainero-Alcolado L, Sjöberg Bexelius T, Santopolo G, Yuan Y, Liaño-Pons J, Arsenian-Henriksson M. Defining neuroblastoma: From origin to precision medicine. Neuro Oncol 2024; 26:2174-2192. [PMID: 39101440 PMCID: PMC11630532 DOI: 10.1093/neuonc/noae152] [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: 03/09/2024] [Indexed: 08/06/2024] Open
Abstract
Neuroblastoma (NB), a heterogenous pediatric tumor of the sympathetic nervous system, is the most common and deadly extracranial solid malignancy diagnosed in infants. Numerous efforts have been invested in understanding its origin and in development of novel curative targeted therapies. Here, we summarize the recent advances in the identification of the cell of origin and the genetic alterations occurring during development that contribute to NB. We discuss current treatment regimens, present and future directions for the identification of novel therapeutic metabolic targets, differentiation agents, as well as personalized combinatory therapies as potential approaches for improving the survival and quality of life of children with NB.
Collapse
Affiliation(s)
- Lourdes Sainero-Alcolado
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum B7, Karolinska Institutet, Stockholm SE-17165, Sweden
| | - Tomas Sjöberg Bexelius
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm SE-17177, Sweden
- Paediatric Oncology Unit, Astrid Lindgren’s Children Hospital, Solna SE-17164, Sweden
| | - Giuseppe Santopolo
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum B7, Karolinska Institutet, Stockholm SE-17165, Sweden
| | - Ye Yuan
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum B7, Karolinska Institutet, Stockholm SE-17165, Sweden
| | - Judit Liaño-Pons
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum B7, Karolinska Institutet, Stockholm SE-17165, Sweden
| | - Marie Arsenian-Henriksson
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University, Lund SE-22381, Sweden
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum B7, Karolinska Institutet, Stockholm SE-17165, Sweden
| |
Collapse
|
18
|
Chen Z, Inague A, Kaushal K, Fazeli G, Schilling D, Xavier da Silva TN, Dos Santos AF, Cheytan T, Freitas FP, Yildiz U, Viviani LG, Lima RS, Pinz MP, Medeiros I, Iijima TS, Alegria TGP, Pereira da Silva R, Diniz LR, Weinzweig S, Klein-Seetharaman J, Trumpp A, Mañas A, Hondal R, Bartenhagen C, Fischer M, Shimada BK, Seale LA, Chillon TS, Fabiano M, Schomburg L, Schweizer U, Netto LE, Meotti FC, Dick TP, Alborzinia H, Miyamoto S, Friedmann Angeli JP. PRDX6 contributes to selenocysteine metabolism and ferroptosis resistance. Mol Cell 2024; 84:4645-4659.e9. [PMID: 39547224 DOI: 10.1016/j.molcel.2024.10.027] [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: 04/10/2024] [Revised: 08/02/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024]
Abstract
Selenocysteine (Sec) metabolism is crucial for cellular function and ferroptosis prevention and begins with the uptake of the Sec carrier, selenoprotein P (SELENOP). Following uptake, Sec released from SELENOP is metabolized via selenocysteine lyase (SCLY), producing selenide, a substrate for selenophosphate synthetase 2 (SEPHS2), which provides the essential selenium donor, selenophosphate (H2SePO3-), for the biosynthesis of the Sec-tRNA. Here, we discovered an alternative pathway in Sec metabolism mediated by peroxiredoxin 6 (PRDX6), independent of SCLY. Mechanistically, we demonstrate that PRDX6 can readily react with selenide and interact with SEPHS2, potentially acting as a selenium delivery system. Moreover, we demonstrate the functional significance of this alternative route in human cancer cells, revealing a notable association between elevated expression of PRDX6 and human MYCN-amplified neuroblastoma subtype. Our study sheds light on a previously unrecognized aspect of Sec metabolism and its implications in ferroptosis, offering further possibilities for therapeutic exploitation.
Collapse
Affiliation(s)
- Zhiyi Chen
- Rudolf Virchow Zentrum (RVZ), Center for Integrative and Translational Bioimaging, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Alex Inague
- Rudolf Virchow Zentrum (RVZ), Center for Integrative and Translational Bioimaging, University of Wuerzburg, 97080 Wuerzburg, Germany; Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil
| | - Kamini Kaushal
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Gholamreza Fazeli
- Rudolf Virchow Zentrum (RVZ), Center for Integrative and Translational Bioimaging, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Danny Schilling
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Thamara N Xavier da Silva
- Rudolf Virchow Zentrum (RVZ), Center for Integrative and Translational Bioimaging, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Ancely Ferreira Dos Santos
- Rudolf Virchow Zentrum (RVZ), Center for Integrative and Translational Bioimaging, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Tasneem Cheytan
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Florencio Porto Freitas
- Rudolf Virchow Zentrum (RVZ), Center for Integrative and Translational Bioimaging, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Umut Yildiz
- European Molecular Biology Laboratory, Genome Biology Unit, 69117 Heidelberg, Germany
| | - Lucas Gasparello Viviani
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil
| | - Rodrigo Santiago Lima
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil
| | - Mikaela Peglow Pinz
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil
| | - Isadora Medeiros
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil
| | - Thais Satie Iijima
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil
| | - Thiago Geronimo Pires Alegria
- Biosciences Institute, Department of Genetics and Evolutionary Biology, Universidade de São Paulo, 05508900 Sao Paulo, Brazil
| | - Railmara Pereira da Silva
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil
| | - Larissa Regina Diniz
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil
| | - Simon Weinzweig
- School of Molecular Sciences, Arizona State University, Phoenix, AZ 85281, USA
| | | | - Andreas Trumpp
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Adriana Mañas
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, IdiPAZ, Hospital Universitario La Paz, 28049 Madrid, Spain; IdiPAZ-CNIO Pediatric Onco-Hematology Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), 28049 Madrid, Spain
| | - Robert Hondal
- Department of Biochemistry, University of Vermont, Burlington, VT 05405, USA
| | - Christoph Bartenhagen
- Center for Molecular Medicine Cologne (CMMC) and Department of Experimental Pediatric Oncology, University Children's Hospital, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Matthias Fischer
- Center for Molecular Medicine Cologne (CMMC) and Department of Experimental Pediatric Oncology, University Children's Hospital, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Briana K Shimada
- Pacific Biosciences Research Center, University of Hawaii at Manoa, Honolulu, HI 96848, USA
| | - Lucia A Seale
- Pacific Biosciences Research Center, University of Hawaii at Manoa, Honolulu, HI 96848, USA
| | - Thilo Samson Chillon
- Institute for Experimental Endocrinology, CVK, Charité-Universtitätsmedizin Berlin, 10115 Berlin, Germany
| | - Marietta Fabiano
- Institut für Biochemie und Molekularbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany
| | - Lutz Schomburg
- Institute for Experimental Endocrinology, CVK, Charité-Universtitätsmedizin Berlin, 10115 Berlin, Germany
| | - Ulrich Schweizer
- Institut für Biochemie und Molekularbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany
| | - Luis E Netto
- Biosciences Institute, Department of Genetics and Evolutionary Biology, Universidade de São Paulo, 05508900 Sao Paulo, Brazil
| | - Flavia C Meotti
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil
| | - Tobias P Dick
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Hamed Alborzinia
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Sayuri Miyamoto
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil.
| | - José Pedro Friedmann Angeli
- Rudolf Virchow Zentrum (RVZ), Center for Integrative and Translational Bioimaging, University of Wuerzburg, 97080 Wuerzburg, Germany.
| |
Collapse
|
19
|
Fang F, Hua M, Yu G. Study of the mechanism of fibroblast-like synoviocytes-derived exosomes inducing macrophages M1 polarization and CD8 +T cells immune regulation ferroptosis and autophagy in rheumatoid arthritis. Immunol Lett 2024; 270:106936. [PMID: 39447763 DOI: 10.1016/j.imlet.2024.106936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/17/2024] [Accepted: 10/20/2024] [Indexed: 10/26/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that affects the joints. The pathogenesis of RA is complex, involving membrane lipid antioxidant systems, oxidative stress, and lipid peroxidation. In this study, it was found that cysteine dioxygenase 1 (CDO1) is significantly upregulated in RA fibroblast-like synoviocytes (RA-FLS) and that exosomes derived from these RA-FLS deliver CDO1 to promote M1 polarization of macrophages, thus facilitating RA progression. In the immune microenvironment, CD8+T cells play a role in immune regulation by producing cytokines such as interferon gamma (IFNγ) in various diseases. The results of this study suggested that in RA-FLS, CD8+T cells deliver IFNγ, which not only inhibits the viability of RA-FLS but also affects glutathione (GSH) through CDO1, regulating the GPX4 antioxidant signaling pathway to promote ferroptosis and autophagy in cells. It was also discovered that IFNγ enhances the expression of TRI69, ubiquitinates and degrades FSP1, thereby forming a cooperative regulation process of GPX4 and FSP1 in ferroptosis. These findings provide a new direction for the treatment of RA.
Collapse
Affiliation(s)
- Fang Fang
- Department of Immunology, School of Laboratory Medicine, Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, Anhui 233030, China.
| | - Mengqing Hua
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, China
| | - GenMing Yu
- Bengbu Medical College, Bengbu, Anhui 233030, China
| |
Collapse
|
20
|
Zhou Q, Meng Y, Le J, Sun Y, Dian Y, Yao L, Xiong Y, Zeng F, Chen X, Deng G. Ferroptosis: mechanisms and therapeutic targets. MedComm (Beijing) 2024; 5:e70010. [PMID: 39568772 PMCID: PMC11577302 DOI: 10.1002/mco2.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
Ferroptosis is a nonapoptotic form of cell death characterized by iron-dependent lipid peroxidation in membrane phospholipids. Since its identification in 2012, extensive research has unveiled its involvement in the pathophysiology of numerous diseases, including cancers, neurodegenerative disorders, organ injuries, infectious diseases, autoimmune conditions, metabolic disorders, and skin diseases. Oxidizable lipids, overload iron, and compromised antioxidant systems are known as critical prerequisites for driving overwhelming lipid peroxidation, ultimately leading to plasma membrane rupture and ferroptotic cell death. However, the precise regulatory networks governing ferroptosis and ferroptosis-targeted therapy in these diseases remain largely undefined, hindering the development of pharmacological agonists and antagonists. In this review, we first elucidate core mechanisms of ferroptosis and summarize its epigenetic modifications (e.g., histone modifications, DNA methylation, noncoding RNAs, and N6-methyladenosine modification) and nonepigenetic modifications (e.g., genetic mutations, transcriptional regulation, and posttranslational modifications). We then discuss the association between ferroptosis and disease pathogenesis and explore therapeutic approaches for targeting ferroptosis. We also introduce potential clinical monitoring strategies for ferroptosis. Finally, we put forward several unresolved issues in which progress is needed to better understand ferroptosis. We hope this review will offer promise for the clinical application of ferroptosis-targeted therapies in the context of human health and disease.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yu Meng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Jiayuan Le
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yating Dian
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Lei Yao
- Department of General Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yixiao Xiong
- Department of Dermatology Tongji Hospital Huazhong University of Science and Technology Wuhan Hubei China
| | - Furong Zeng
- Department of Oncology Xiangya Hospital Central South University Changsha Hunan Province China
| | - Xiang Chen
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Guangtong Deng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| |
Collapse
|
21
|
Wu H, Cao P, Wang H, Wang W, Yu H, You C, Shen T, Yang S, Hu Z, Zhou T, Wang J, Wang Q, Qian X, Zhang J, Wang X, Cao Y, Ning L, Lin F. Postoperative Injection of a Triptolide-Preloaded Hydrogel Prevents the Recurrence of Glioblastoma by Dual-Pathway Activation of Ferroptosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2406036. [PMID: 39375977 DOI: 10.1002/smll.202406036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/14/2024] [Indexed: 10/09/2024]
Abstract
Glioblastoma (GBM) recurrence leads to high mortality, which remains a major concern in clinical therapy. Herein, an injectable triptolide (TP)-preloaded hydrogel (TP@DNH) accompanied by a postoperative injection strategy is developed to prevent the recurrence of GBM. With a potential inhibitor of the NRF2/SLC7A11/GPX4 axis, it is demonstrated that TP can deactivate glutathione peroxidase 4 (GPX4) from the source of glutathione (GSH) biosynthesis, thereby activating ferroptosis in GBM cells by blocking the neutralization of intracellular lipid peroxide (LPO). Based on acid-sensitive Fe3+/tannic acid (TA) metal-phenolic networks (MPNs), the TP@DNH hydrogel can induce the effective generation of reactive oxygen species (ROS) through Fe3+/TA-mediated Fenton reaction and achieve controllable release of TP in resected GBM cavity. Due to ROS generation and GPX4 deactivation, postoperative injection of TP@DNH can achieve high-level ferroptosis through dual-pathway LPO accumulation, remarkably suppressing the growth of recurrent GBM and prolonging the overall survival in orthotopic GBM relapse mouse model. This work provides an alternative paradigm for regulating ferroptosis in the postoperative treatment of GBM.
Collapse
Affiliation(s)
- Hongshuai Wu
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
- Wuxi Key Laboratory of Biomaterials for Clinical Application, Key Laboratory for Multidisciplinary Intersection of Radiotherapy and Immunology for Gastrointestinal Tumor, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, Wuxi, Jiangsu, 214499, P. R. China
| | - Pingping Cao
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Haiyang Wang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
- Department of Digestive Endoscopy, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, P. R. China
| | - Wenhong Wang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, P. R. China
| | - Hanyang Yu
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Chaoqun You
- Jiangsu Key Lab for the Chemistry and Utilization of Agro-Forest Biomass, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, P. R. China
| | - Tianqi Shen
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Suisui Yang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Ziyi Hu
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Tingting Zhou
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Jing Wang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Qianghu Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, 211166, P. R. China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Xu Qian
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Junxia Zhang
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
- Department of Neurosurgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, P. R. China
| | - Xiuxing Wang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Yuandong Cao
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
- Department of Radiation Oncology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, P. R. China
| | - Like Ning
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Fan Lin
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, P. R. China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
- Department of Gastroenterology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan, 471023, P. R. China
| |
Collapse
|
22
|
Huang L, Zhu J, Wu G, Xiong W, Feng J, Yan C, Yang J, Li Z, Fan Q, Ren B, Li Y, Chen C, Yu X, Shen Z. A strategy of "adding fuel to the flames" enables a self-accelerating cycle of ferroptosis-cuproptosis for potent antitumor therapy. Biomaterials 2024; 311:122701. [PMID: 38981152 DOI: 10.1016/j.biomaterials.2024.122701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/12/2024] [Accepted: 07/06/2024] [Indexed: 07/11/2024]
Abstract
Cuproptosis in antitumor therapy faces challenges from copper homeostasis efflux mechanisms and high glutathione (GSH) levels in tumor cells, hindering copper accumulation and treatment efficacy. Herein, we propose a strategy of "adding fuel to the flames" for potent antitumor therapy through a self-accelerating cycle of ferroptosis-cuproptosis. Disulfiram (DSF) loaded hollow mesoporous copper-iron sulfide (HMCIS) nanoparticle with conjugation of polyethylene glycol (PEG) and folic acid (FA) (i.e., DSF@HMCIS-PEG-FA) was developed to swiftly release DSF, H2S, Cu2+, and Fe2+ in the acidic tumor microenvironment (TME). The hydrogen peroxide (H2O2) levels and acidity within tumor cells enhanced by the released H2S induce acceleration of Fenton (Fe2+) and Fenton-like (Cu2+) reactions, enabling the powerful tumor ferroptosis efficacy. The released DSF acts as a role of "fuel", intensifying catalytic effect ("flame") in tumor cells through the sustainable Fenton chemistry (i.e., "add fuel to the flames"). Robust ferroptosis in tumor cells is characterized by serious mitochondrial damage and GSH depletion, leading to excess intracellular copper that triggers cuproptosis. Cuproptosis disrupts mitochondria, compromises iron-sulfur (Fe-S) proteins, and elevates intracellular oxidative stress by releasing free Fe3+. These interconnected processes form a self-accelerating cycle of ferroptosis-cuproptosis with potent antitumor capabilities, as validated in both cancer cells and tumor-bearing mice.
Collapse
Affiliation(s)
- Lin Huang
- School of Biomedical Engineering, Southern Medical University, 1023 Sha-Tai South Road, Guangzhou, Guangdong, 510515, China
| | - Jiaoyang Zhu
- School of Biomedical Engineering, Southern Medical University, 1023 Sha-Tai South Road, Guangzhou, Guangdong, 510515, China
| | - Guochao Wu
- School of Biomedical Engineering, Southern Medical University, 1023 Sha-Tai South Road, Guangzhou, Guangdong, 510515, China
| | - Wei Xiong
- Medical Imaging Center, Nanfang Hospital, Southern Medical University, 1023 Sha-Tai South Road, Guangzhou, Guangdong, 510515, China
| | - Jie Feng
- Medical Imaging Center, Nanfang Hospital, Southern Medical University, 1023 Sha-Tai South Road, Guangzhou, Guangdong, 510515, China
| | - Chenggong Yan
- Medical Imaging Center, Nanfang Hospital, Southern Medical University, 1023 Sha-Tai South Road, Guangzhou, Guangdong, 510515, China
| | - Jing Yang
- School of Biomedical Engineering, Southern Medical University, 1023 Sha-Tai South Road, Guangzhou, Guangdong, 510515, China
| | - Zongheng Li
- School of Biomedical Engineering, Southern Medical University, 1023 Sha-Tai South Road, Guangzhou, Guangdong, 510515, China
| | - Qingdeng Fan
- School of Biomedical Engineering, Southern Medical University, 1023 Sha-Tai South Road, Guangzhou, Guangdong, 510515, China
| | - Bin Ren
- School of Biomedical Engineering, Southern Medical University, 1023 Sha-Tai South Road, Guangzhou, Guangdong, 510515, China
| | - Yan Li
- School of Biomedical Engineering, Southern Medical University, 1023 Sha-Tai South Road, Guangzhou, Guangdong, 510515, China
| | - Chaomin Chen
- School of Biomedical Engineering, Southern Medical University, 1023 Sha-Tai South Road, Guangzhou, Guangdong, 510515, China.
| | - Xiangrong Yu
- Department of Radiology, Zhuhai People's Hospital, Zhuhai Clinical Medical College of Jinan University, Zhuhai, 519000, China.
| | - Zheyu Shen
- School of Biomedical Engineering, Southern Medical University, 1023 Sha-Tai South Road, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
23
|
Sun N, Wang J, Qin J, Ma S, Luan J, Hou G, Zhang W, Gao M. Oncogenic RTKs sensitize cancer cells to ferroptosis via c-Myc mediated upregulation of ACSL4. Cell Death Dis 2024; 15:861. [PMID: 39604370 PMCID: PMC11603294 DOI: 10.1038/s41419-024-07254-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Alteration or abnormal activation of RTKs have been recurrently observed and recognized as an important driving factor in the progression of many human cancers. Ferroptosis, an iron-dependent form of regulated necrosis triggered by the accumulation of lethal lipid peroxides on cell membranes, has been implicated in various tumor types. Here we reported that oncogenic RTKs/RAS/RAF/c-Myc axis promotes cancer cells to ferroptosis. Mechanistically, c-Myc binds to the promoter region of ACSL4 and promotes the expression of ACSL4, thereby sensitizes cells to ferroptosis. We further showed that RTKs/RAS/RAF promote ferroptosis by upregulating c-Myc mediated expression of ACSL4 in cancer cells. Notably, overexpression of RTKs enhances the vulnerability of melanoma to the ferroptosis inducer in mouse xenograft model. These findings may provide an attractive intervention strategy to target cancers with oncogenic activation of RTKs via a ferroptosis-inducing approach.
Collapse
Affiliation(s)
- Na Sun
- The HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Jiawa Wang
- The HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Jianhua Qin
- The HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Shuang Ma
- The HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Jing Luan
- The HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Guoyuan Hou
- The HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Wei Zhang
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA.
| | - Minghui Gao
- The HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
24
|
Sun X, Liu X, Wang C, Ren Z, Yang X, Liu Y. Deciphering Mechanisms of Adipocyte Differentiation in Abdominal Fat of Broilers. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:25403-25413. [PMID: 39483088 PMCID: PMC11565640 DOI: 10.1021/acs.jafc.4c06867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 11/03/2024]
Abstract
The excessive deposition of abdominal fat tissue (AFT) in broilers has emerged as a major concern in the poultry industry. Despite some progress in recent years, the molecular mechanisms underlying AFT development remain ambiguous. The current study combined RNA-seq with transposase-accessible chromatin sequencing (ATAC-seq) to map the dynamic profiling of chromatin accessibility and transcriptional reprogramming in AFT adipocyte differentiation in broilers at day 3 (D3) and D14. Our results found that the levels of CDK1 and PCNA were down-regulated at D14, D28, and D42 compared to D3, while the levels of C/EBPα and FABP4 were up-regulated at D14 and D42 compared to D3. Meanwhile, PPARγ was significantly up-regulated at D28 and D42. RNA-seq of AFT identified 1705 up-regulated and 1112 down-regulated differential expression genes (DEGs) between D3 and D14. Pathways based on up-regulated DEGs mainly enriched some pathways related to adipocyte differentiation, while down-regulated DEGs pointed to DNA replication, cell cycle, and gap junction. Gene set enrichment analysis (GSEA) revealed that DNA replication and the cell cycle were down-regulated at D14, while the insulin signaling pathway was up-regulated. In the OA-induced immortalized chicken preadipocyte (ICP2) model, protein dynamic changes were consistent with AFT from D3 to D14. Same pathways were enriched in ICP2. In addition, based on overlapped DEGs from AFT and ICP2, enriched pathways related to adipocyte differentiation or proliferation mentioned above were all involved. A total of 1600 gain and 16727 loss differential peaks (DPs) were identified in ICP2 by ATAC-seq. Predicted genes from DPs at the promoter regions were enriched in glycerophospholipid metabolism, TGF-β signaling, FoxO signaling, and ubiquitin-mediated proteolysis. DNA motifs predicted 159 transcription factors (TFs) based on gain and loss peaks from the promoter regions, where 1 and 10 TFs were overlapped with up or down TFs from DEGs. Overall, this study presents a framework for the comprehension of the epigenetic regulatory mechanisms of adipocyte differentiation and identifies candidate genes and potential TFs involved in AFT adipocyte differentiation in broilers.
Collapse
Affiliation(s)
- Xi Sun
- College of Animal Science
and Technology, Northwest A&F University, Yangling 712100, China
| | - Xiaoying Liu
- College of Animal Science
and Technology, Northwest A&F University, Yangling 712100, China
| | - Chaohui Wang
- College of Animal Science
and Technology, Northwest A&F University, Yangling 712100, China
| | - Zhouzheng Ren
- College of Animal Science
and Technology, Northwest A&F University, Yangling 712100, China
| | - Xiaojun Yang
- College of Animal Science
and Technology, Northwest A&F University, Yangling 712100, China
| | - Yanli Liu
- College of Animal Science
and Technology, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
25
|
Lin YS, Tsai YC, Li CJ, Wei TT, Wang JL, Lin BW, Wu YN, Wu SR, Lin SC, Lin SC. Overexpression of NUDT16L1 sustains proper function of mitochondria and leads to ferroptosis insensitivity in colorectal cancer. Redox Biol 2024; 77:103358. [PMID: 39317106 PMCID: PMC11465047 DOI: 10.1016/j.redox.2024.103358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024] Open
Abstract
Cancer research is continuously exploring new avenues to improve treatments, and ferroptosis induction has emerged as a promising approach. However, the lack of comprehensive analysis of the ferroptosis sensitivity in different cancer types has limited its clinical application. Moreover, identifying the key regulator that influences the ferroptosis sensitivity during cancer progression remains a major challenge. In this study, we shed light on the role of ferroptosis in colorectal cancer and identified a novel ferroptosis repressor, NUDT16L1, that contributes to the ferroptosis insensitivity in this cancer type. Mechanistically, NUDT16L1 promotes ferroptosis insensitivity in colon cancer by enhancing the expression of key ferroptosis repressor and mitochondrial genes through direct binding to NAD-capped RNAs and the indirect action of MALAT1. Our findings also reveal that NUDT16L1 localizes to the mitochondria to maintain its proper function by preventing mitochondrial DNA leakage after treatment of ferroptosis inducer in colon cancer cells. Importantly, our orthotopic injection and Nudt16l1 transgenic mouse models of colon cancer demonstrated the critical role of NUDT16L1 in promoting tumor growth. Moreover, clinical specimens revealed that NUDT16L1 was overexpressed in colorectal cancer, indicating its potential as a therapeutic target. Finally, our study shows the therapeutic potential of a NUDT16L1 inhibitor in vitro, in vivo and ex vivo. Taken together, these findings provide new insights into the crucial role of NUDT16L1 in colorectal cancer and highlight its potential as a promising therapeutic target.
Collapse
Affiliation(s)
- Yi-Syuan Lin
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Chuan Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Jung Li
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Tzu-Tang Wei
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jui-Lin Wang
- National Laboratory Animal Center, National Applied Research Laboratories, Tainan, Taiwan
| | - Bo-Wen Lin
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Na Wu
- School of Dentistry and Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shang-Rung Wu
- School of Dentistry and Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shin-Chih Lin
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shih-Chieh Lin
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
26
|
Lai Z, Wei Y, He M, Lin C, Ouyang W, Liu X. Antimony trioxide nanoparticles promote ferroptosis in developing zebrafish (Danio rerio) by disrupting iron homeostasis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 949:175140. [PMID: 39084369 DOI: 10.1016/j.scitotenv.2024.175140] [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: 04/09/2024] [Revised: 06/25/2024] [Accepted: 07/28/2024] [Indexed: 08/02/2024]
Abstract
The widespread use of antimony trioxide (ATO) and ATO nanoparticles (nATO) has led to increasing ecological and health risks. However, there is relatively insufficient research on the aquatic ecotoxicology of nATO. This study revealed that nATO affects the development of zebrafish embryos and mainly induces ferroptosis through the dissolution of Sb(III). The size of nATO ranged from 50 to 250 nm, and it generated free radicals in water. It can be ingested and accumulate in zebrafish larvae and affects normal development. Compared with those in the control group, the levels of reactive oxygen species (ROS), cell apoptosis, mitochondrial damage and iron content in the group exposed to high concentrations of nATO were increased. The transcriptomics results indicated that nATO significantly altered the expression levels of key genes related to glutathione metabolism and ferroptosis. Quantitative polymerase chain reaction consistently demonstrated the reliability of the transcriptome data and revealed that nATO induced ferroptosis by disrupting iron homeostasis and the key factor is the dissolution of Sb(III). Furthermore, ferrostatin-1, an inhibitor of ferroptosis, decreased the levels of ROS, apoptosis and mitochondrial damage induced by nATO, which further prove that nATO can promote ferroptosis. This work deepens the understanding of the ecological toxicological effects of nATO in aquatic environments and its mechanisms, which is highly important for the development of antimony management strategies.
Collapse
Affiliation(s)
- Ziyang Lai
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing 100875, China
| | - Yihan Wei
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing 100875, China
| | - Mengchang He
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing 100875, China.
| | - Chunye Lin
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing 100875, China
| | - Wei Ouyang
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing 100875, China; Advanced Interdisciplinary Institute of Environment and Ecology, Beijing Normal University, Zhuhai 519087, China
| | - Xitao Liu
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
27
|
Chen C, Shen X, Shi S, Xu Y, Song H, Qu L, Du S, Gao Y, Han X. Biomimetic Fe3+ metal-phenolic networks enable DNAzyme and Cas9 RNP delivery for synergistic tumor ferroptosis-immunotherapy. CHEMICAL ENGINEERING JOURNAL 2024; 499:156050. [DOI: 10.1016/j.cej.2024.156050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
28
|
Ma Y, Su J, Ma C. Causal relationship between amino acids and ovarian cancer in the European population: A bidirectional Mendelian randomization study and meta-analysis. Medicine (Baltimore) 2024; 103:e40189. [PMID: 39470531 PMCID: PMC11521036 DOI: 10.1097/md.0000000000040189] [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: 07/05/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/30/2024] Open
Abstract
In recent years, an increasing number of observational studies have reported the impact of amino acids on ovarian cancer. However, Mendelian randomization studies have not yet been conducted to explore the causal relationship between them in the context of ovarian cancer. This study conducted Mendelian randomization (MR) analysis of 20 amino acids in relation to ovarian cancer data from 2 different sources within the European population, using a two-sample MR approach. The primary results from the inverse variance weighting analysis were then subjected to a meta-analysis, followed by multiple testing correction for the meta-analysis thresholds. Finally, reverse causality testing was performed on the positively associated amino acids and ovarian cancer. MR analyses were conducted for 20 amino acids with ovarian cancer data from both the Finngen R10 and Open genome-wide association study databases. The inverse variance weighted results from these 2 analyses were then combined through meta-analysis, with multiple corrections applied to the significance thresholds of the meta-analysis results. The findings showed that only cysteine had a significant association with ovarian cancer, with an (odds ratio) odds ratio value of 0.507 (95% confidence interval: 0.335-0.767, P = .025). The P-value of the combined MR and meta-analysis, after multiple testing correction, was 0.025, indicating statistical significance (P < .05). Additionally, cysteine did not show a reverse causal relationship with ovarian cancer in either data source. Cysteine is a protective factor for ovarian cancer, potentially reducing the risk of ovarian cancer and slowing the progression of the disease.
Collapse
Affiliation(s)
- Yingji Ma
- Jiaozhou Hospital of Tongji University Dongfang Hospital Qingdao, Shangdong, China
| | - Jiaqi Su
- Jiaozhou Hospital of Tongji University Dongfang Hospital Qingdao, Shangdong, China
| | - Changbo Ma
- Jiaozhou Hospital of Tongji University Dongfang Hospital Qingdao, Shangdong, China
| |
Collapse
|
29
|
Li Z, Wang Y, Liang S, Yuan T, Liu J. EIF2S1 Silencing Impedes Neuroblastoma Development Through GPX4 Inactivation and Ferroptosis Induction. Int J Genomics 2024; 2024:6594426. [PMID: 39465005 PMCID: PMC11512646 DOI: 10.1155/2024/6594426] [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: 06/24/2024] [Accepted: 09/20/2024] [Indexed: 10/29/2024] Open
Abstract
Background: Neuroblastoma (NB) is one of the most devastating malignancies in children, accounting for a high mortality rate due to limited treatment options. This study is aimed at elucidating the role of the ferroptosis-related EIF2S1 gene in NB pathogenesis and exploring its potential as a therapeutic target. Methods: We conducted comprehensive bioinformatics analyses utilizing the FerrDb database and NB-related transcriptomics data to investigate the role of EIF2S1 in NB. Changes in EIF2S1 expression were subsequently validated in NB tissues and cell lines. Loss-of-function experiments were performed in SK-N-SH and IMR-32 cell lines through shRNA-mediated EIF2S1 knockdown. The impact of EIF2S1 knockdown on the tumorigenesis of SK-N-SH cells was assessed in nude mice. Results: Bioinformatics analyses revealed a significant association between elevated EIF2S1 expression and poor prognosis in NB patients. The increased levels of EIF2S1 expression were confirmed in NB tissues and cancerous cell lines. Furthermore, EIF2S1 overexpression was linked to translational regulation and immune cell infiltration modulation. Silencing of EIF2S1 resulted in the suppression of cell proliferation, migration, and tumorigenicity in NB cells. Additionally, EIF2S1 knockdown led to an accumulation of iron and oxidative stress, as well as a reduction in GPX4 and SLC7A11 expression. Conclusion: Our findings indicate that EIF2S1 appears to facilitate the progression of NB by protecting tumor cells from ferroptosis through modulating GPX4 and SLC7A11 expression. Consequently, EIF2S1 may serve as a potential therapeutic target for the management of NB.
Collapse
Affiliation(s)
- Zhen Li
- Department of General & Pediatric Surgery, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Zhifu District, Yantai 264099, China
| | - Yunhui Wang
- Department of General & Pediatric Surgery, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Zhifu District, Yantai 264099, China
| | - Shubin Liang
- Department of General & Pediatric Surgery, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Zhifu District, Yantai 264099, China
| | - Tingdong Yuan
- Department of General & Pediatric Surgery, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Zhifu District, Yantai 264099, China
| | - Jing Liu
- Department of Pathology, Yantai Yuhuangding Hospital, No. 20 Yuhuangding East Road, Zhifu District, Yantai 264099, China
| |
Collapse
|
30
|
Zheng H, Chen H, Cai Y, Shen M, Li X, Han Y, Deng X, Cao H, Liu J, Li H, Liu B, Li G, Wang X, Chen H, Hou J, Lin SH, Zong L, Zhang Y. Hydrogen sulfide-mediated persulfidation regulates homocysteine metabolism and enhances ferroptosis in non-small cell lung cancer. Mol Cell 2024; 84:4016-4030.e6. [PMID: 39321805 DOI: 10.1016/j.molcel.2024.08.035] [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: 11/15/2023] [Revised: 06/15/2024] [Accepted: 08/30/2024] [Indexed: 09/27/2024]
Abstract
Hydrogen sulfide (H₂S), a metabolite of the transsulfuration pathway, has been implicated in ferroptosis, a unique form of cell death caused by lipid peroxidation. While the exact mechanisms controlling ferroptosis remain unclear, our study reveals that H₂S sensitizes human non-small cell lung cancer (NSCLC) cells to this process, particularly when cysteine levels are low. Combining H₂S with cystine depletion significantly enhances the effectiveness of ferroptosis-based cancer therapy. Mechanistically, H₂S persulfidates the 195th cysteine on S-adenosyl homocysteine hydrolase (SAHH), reducing its enzymatic activity. This leads to decreased homocysteine levels, subsequently lowering cysteine and glutathione concentrations under cystine depletion conditions. These changes ultimately increase the vulnerability of NSCLC cells to ferroptosis. Our findings establish H₂S as a key regulator of homocysteine metabolism and a critical factor in determining NSCLC cell susceptibility to ferroptosis. These results highlight the potential of H₂S-based therapies to improve the efficacy of ferroptosis-targeted cancer treatments for NSCLC.
Collapse
Affiliation(s)
- Hualei Zheng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Huidi Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Yunjie Cai
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Min Shen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Xilin Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Yi Han
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Xusheng Deng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Hongjie Cao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Junjia Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Hao Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Benchao Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Ganlin Li
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Xindong Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Hui Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Jingjing Hou
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Shu-Hai Lin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute for Data Science in Health and Medicine Engineering, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen 361102, Fujian, China
| | - Lili Zong
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Yongyou Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute for Data Science in Health and Medicine Engineering, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen 361102, Fujian, China.
| |
Collapse
|
31
|
Okano Y, Yamauchi T, Fukuzaki R, Tsuruta A, Yoshida Y, Tsurudome Y, Ushijima K, Matsunaga N, Koyanagi S, Ohdo S. Oncogenic accumulation of cysteine promotes cancer cell proliferation by regulating the translation of D-type cyclins. J Biol Chem 2024; 300:107890. [PMID: 39413876 DOI: 10.1016/j.jbc.2024.107890] [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: 05/04/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/18/2024] Open
Abstract
Malignant cells exhibit a high demand for amino acids to sustain their abnormal proliferation. Particularly, the intracellular accumulation of cysteine is often observed in cancer cells. Previous studies have shown that deprivation of intracellular cysteine in cancer cells results in the accumulation of lipid peroxides in the plasma membrane and induction of ferroptotic cell death, indicating that cysteine plays a critical role in the suppression of ferroptosis. Herein, we found that the oncogenic accumulation of cysteine also contributes to cancer cell proliferation by promoting the cell cycle progression, which is independent of its suppressive effect on ferroptosis. The growth ability of four types of cancer cells, including murine hepatocarcinoma cells, but not of primary hepatocytes, were dependent on the exogenous supply of cysteine. Deprivation of intracellular cysteine in cancer cells induced cell cycle arrest at the G0/G1 phase, accompanied by a decrease in the expression of cyclin D1 and D2 proteins. The cysteine deprivation-induced decrease in D-type cyclin expression was associated with the upregulation of eukaryotic translation initiation factor 4E binding protein 1, which represses the translation of cyclin D1 and D2 proteins by binding to eukaryotic translation initiation factor 4E. Similar results were observed in hepatocarcinoma cells treated with erastin, an inhibitor of cystine/glutamate antiporter, xCT. These findings reveal an unappreciated role of cysteine in regulating the growth of malignant cancer cells and deepen our understanding of the cytotoxic effect of xCT inhibitor to prevent cancer cell proliferation.
Collapse
Affiliation(s)
- Yumi Okano
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoaki Yamauchi
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Runa Fukuzaki
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Akito Tsuruta
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuya Yoshida
- Department of Clinical Pharmacokinetics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuya Tsurudome
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyoonoda, Yamaguchi, Japan
| | - Kentaro Ushijima
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyoonoda, Yamaguchi, Japan
| | - Naoya Matsunaga
- Department of Clinical Pharmacokinetics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Satoru Koyanagi
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.
| | - Shigehiro Ohdo
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
32
|
Lai Z, Wei Y, He M, Lin C, Ouyang W, Liu X. Toxicity and related molecular mechanisms of Sb(III) in the embryos and larvae of zebrafish (Danio rerio). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 359:124551. [PMID: 39004205 DOI: 10.1016/j.envpol.2024.124551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/16/2024]
Abstract
Antimony (Sb) pollution poses a severe threat to humans and ecosystems due to the extensive use of Sb in various fields. However, little is known about the toxic effects of Sb and its aquatic ecotoxicological mechanism. This study aimed to reveal the toxicity and related molecular mechanisms of trivalent Sb (Sb(III)) in zebrafish embryos/larvae. Sb(III) accumulated in larvae, which correlated with the exposure concentration. Although no significant lethal or teratogenic effects were observed, normal growth and development were affected. Exposure to 10 or 20 mg/L Sb(III) increased the levels of reactive oxygen species in the larvae while enhancing catalase activity and increasing cell apoptosis. Transcriptomic analysis revealed that Sb(III) promoted glutathione metabolism and the ferroptosis pathway. In addition, symptoms associated with ferroptosis, including mitochondrial damage, biochemical levels of related molecules and increased tissue iron content, were detected. Quantitative polymerase chain reaction (qPCR) analyses further confirmed that Sb(III) significantly altered the transcription levels of genes related to the ferroptosis pathway by disrupting iron homeostasis. Furthermore, ferrostatin-1 (Fer-1) mitigated the toxic effects induced by Sb(III) in zebrafish. Our research fills the gap in the literature on the toxicity and mechanism of Sb(III) in aquatic organisms, which is highly important for understanding the ecological risks associated with Sb.
Collapse
Affiliation(s)
- Ziyang Lai
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing, 100875, China
| | - Yihan Wei
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing, 100875, China
| | - Mengchang He
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing, 100875, China.
| | - Chunye Lin
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing, 100875, China
| | - Wei Ouyang
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing, 100875, China; Advanced Interdisciplinary Institute of Environment and Ecology, Beijing Normal University, Zhuhai, 519087, China
| | - Xitao Liu
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing, 100875, China
| |
Collapse
|
33
|
Bai J, Zhang X, Zhao Z, Sun S, Cheng W, Yu H, Chang X, Wang B. CuO Nanozymes Catalyze Cysteine and Glutathione Depletion Induced Ferroptosis and Cuproptosis for Synergistic Tumor Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400326. [PMID: 38813723 DOI: 10.1002/smll.202400326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 05/06/2024] [Indexed: 05/31/2024]
Abstract
The latest research identifies that cysteine (Cys) is one of the key factors in tumor proliferation, metastasis, and recurrence. The direct depletion of intracellular Cys shows a profound antitumor effect. However, using nanozymes to efficiently deplete Cys for tumor therapy has not yet attracted widespread attention. Here, a (3-carboxypropyl) triphenylphosphonium bromide-derived hyaluronic acid-modified copper oxide nanorods (denoted as MitCuOHA) are designed with cysteine oxidase-like, glutathione oxidase-like and peroxidase-like activities to realize Cys depletion and further induce cellular ferroptosis and cuproptosis for synergistic tumor therapy. MitCuOHA nanozymes can efficiently catalyze the depletion of Cys and glutathione (GSH), accompanied by the generation of H2O2 and the subsequent conversion into highly active hydroxyl radicals, thereby successfully inducing ferroptosis in cancer cells. Meanwhile, copper ions released by MitCuOHA under tumor microenvironment stimulation directly bind to lipoylated proteins of the tricarboxylic acid cycle, leading to the abnormal aggregation of lipoylated proteins and subsequent loss of iron-sulfur cluster proteins, which ultimately triggers proteotoxic stress and cell cuproptosis. Both in vitro and in vivo results show the drastically enhanced anticancer efficacy of Cys oxidation catalyzed by the MitCuOHA nanozymes, demonstrating the high feasibility of such catalytic Cys depletion-induced synergistic ferroptosis and cuproptosis therapeutic concept.
Collapse
Affiliation(s)
- Jinwei Bai
- State Key Laboratory of Applied Organic Chemistry and Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Xuan Zhang
- State Key Laboratory of Applied Organic Chemistry and Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Zhiwen Zhao
- State Key Laboratory of Applied Organic Chemistry and Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Shihao Sun
- State Key Laboratory of Applied Organic Chemistry and Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Wenyuan Cheng
- State Key Laboratory of Applied Organic Chemistry and Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Hongxiang Yu
- State Key Laboratory of Applied Organic Chemistry and Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Xinyue Chang
- State Key Laboratory of Applied Organic Chemistry and Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Baodui Wang
- State Key Laboratory of Applied Organic Chemistry and Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China
| |
Collapse
|
34
|
Lan W, Yang L, Tan X. Crosstalk between ferroptosis and macrophages: potential value for targeted treatment in diseases. Mol Cell Biochem 2024; 479:2523-2543. [PMID: 37880443 DOI: 10.1007/s11010-023-04871-4] [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: 06/26/2023] [Accepted: 10/05/2023] [Indexed: 10/27/2023]
Abstract
Ferroptosis is a newly identified form of programmed cell death that is connected to iron-dependent lipid peroxidization. It involves a variety of physiological processes involving iron metabolism, lipid metabolism, oxidative stress, and biosynthesis of nicotinamide adenine dinucleotide phosphate, glutathione, and coenzyme Q10. So far, it has been discovered to contribute to the pathological process of many diseases, such as myocardial infarction, acute kidney injury, atherosclerosis, and so on. Macrophages are innate immune system cells that regulate metabolism, phagocytize pathogens and dead cells, mediate inflammatory reactions, promote tissue repair, etc. Emerging evidence shows strong associations between macrophages and ferroptosis, which can provide us with a deeper comprehension of the pathological process of diseases and new targets for the treatments. In this review, we summarized the crosstalk between macrophages and ferroptosis and anatomized the application of this association in disease treatments, both non-neoplastic and neoplastic diseases. In addition, we have also addressed problems that remain to be investigated, in the hope of inspiring novel therapeutic strategies for diseases.
Collapse
Affiliation(s)
- Wanxin Lan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Lei Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China
| | - Xuelian Tan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
35
|
Nakamura T, Conrad M. Exploiting ferroptosis vulnerabilities in cancer. Nat Cell Biol 2024; 26:1407-1419. [PMID: 38858502 DOI: 10.1038/s41556-024-01425-8] [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: 02/09/2024] [Accepted: 04/17/2024] [Indexed: 06/12/2024]
Abstract
Ferroptosis is a distinct lipid peroxidation-dependent form of necrotic cell death. This process has been increasingly contemplated as a new target for cancer therapy because of an intrinsic or acquired ferroptosis vulnerability in difficult-to-treat cancers and tumour microenvironments. Here we review recent advances in our understanding of the molecular mechanisms that underlie ferroptosis and highlight available tools for the modulation of ferroptosis sensitivity in cancer cells and communication with immune cells within the tumour microenvironment. We further discuss how these new insights into ferroptosis-activating pathways can become new armouries in the fight against cancer.
Collapse
Affiliation(s)
- Toshitaka Nakamura
- Institute of Metabolism and Cell Death, Molecular Targets & Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets & Therapeutics Center, Helmholtz Munich, Neuherberg, Germany.
| |
Collapse
|
36
|
Mao C, Wang M, Zhuang L, Gan B. Metabolic cell death in cancer: ferroptosis, cuproptosis, disulfidptosis, and beyond. Protein Cell 2024; 15:642-660. [PMID: 38428031 PMCID: PMC11365558 DOI: 10.1093/procel/pwae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 02/19/2024] [Indexed: 03/03/2024] Open
Abstract
Cell death resistance represents a hallmark of cancer. Recent studies have identified metabolic cell death as unique forms of regulated cell death resulting from an imbalance in the cellular metabolism. This review discusses the mechanisms of metabolic cell death-ferroptosis, cuproptosis, disulfidptosis, lysozincrosis, and alkaliptosis-and explores their potential in cancer therapy. Our review underscores the complexity of the metabolic cell death pathways and offers insights into innovative therapeutic avenues for cancer treatment.
Collapse
Affiliation(s)
- Chao Mao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Min Wang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Li Zhuang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
37
|
Berndt C, Alborzinia H, Amen VS, Ayton S, Barayeu U, Bartelt A, Bayir H, Bebber CM, Birsoy K, Böttcher JP, Brabletz S, Brabletz T, Brown AR, Brüne B, Bulli G, Bruneau A, Chen Q, DeNicola GM, Dick TP, Distéfano A, Dixon SJ, Engler JB, Esser-von Bieren J, Fedorova M, Friedmann Angeli JP, Friese MA, Fuhrmann DC, García-Sáez AJ, Garbowicz K, Götz M, Gu W, Hammerich L, Hassannia B, Jiang X, Jeridi A, Kang YP, Kagan VE, Konrad DB, Kotschi S, Lei P, Le Tertre M, Lev S, Liang D, Linkermann A, Lohr C, Lorenz S, Luedde T, Methner A, Michalke B, Milton AV, Min J, Mishima E, Müller S, Motohashi H, Muckenthaler MU, Murakami S, Olzmann JA, Pagnussat G, Pan Z, Papagiannakopoulos T, Pedrera Puentes L, Pratt DA, Proneth B, Ramsauer L, Rodriguez R, Saito Y, Schmidt F, Schmitt C, Schulze A, Schwab A, Schwantes A, Soula M, Spitzlberger B, Stockwell BR, Thewes L, Thorn-Seshold O, Toyokuni S, Tonnus W, Trumpp A, Vandenabeele P, Vanden Berghe T, Venkataramani V, Vogel FCE, von Karstedt S, Wang F, Westermann F, Wientjens C, Wilhelm C, Wölk M, Wu K, Yang X, Yu F, Zou Y, Conrad M. Ferroptosis in health and disease. Redox Biol 2024; 75:103211. [PMID: 38908072 PMCID: PMC11253697 DOI: 10.1016/j.redox.2024.103211] [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: 03/21/2024] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 06/24/2024] Open
Abstract
Ferroptosis is a pervasive non-apoptotic form of cell death highly relevant in various degenerative diseases and malignancies. The hallmark of ferroptosis is uncontrolled and overwhelming peroxidation of polyunsaturated fatty acids contained in membrane phospholipids, which eventually leads to rupture of the plasma membrane. Ferroptosis is unique in that it is essentially a spontaneous, uncatalyzed chemical process based on perturbed iron and redox homeostasis contributing to the cell death process, but that it is nonetheless modulated by many metabolic nodes that impinge on the cells' susceptibility to ferroptosis. Among the various nodes affecting ferroptosis sensitivity, several have emerged as promising candidates for pharmacological intervention, rendering ferroptosis-related proteins attractive targets for the treatment of numerous currently incurable diseases. Herein, the current members of a Germany-wide research consortium focusing on ferroptosis research, as well as key external experts in ferroptosis who have made seminal contributions to this rapidly growing and exciting field of research, have gathered to provide a comprehensive, state-of-the-art review on ferroptosis. Specific topics include: basic mechanisms, in vivo relevance, specialized methodologies, chemical and pharmacological tools, and the potential contribution of ferroptosis to disease etiopathology and progression. We hope that this article will not only provide established scientists and newcomers to the field with an overview of the multiple facets of ferroptosis, but also encourage additional efforts to characterize further molecular pathways modulating ferroptosis, with the ultimate goal to develop novel pharmacotherapies to tackle the various diseases associated with - or caused by - ferroptosis.
Collapse
Affiliation(s)
- Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Hamed Alborzinia
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM GGmbH), Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Vera Skafar Amen
- Rudolf Virchow Zentrum, Center for Integrative and Translational Bioimaging - University of Würzburg, Germany
| | - Scott Ayton
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Australia
| | - Uladzimir Barayeu
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ) Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany; Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany; Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany; German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Hülya Bayir
- Department of Pediatrics, Columbia University, New York City, NY, USA
| | - Christina M Bebber
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany; CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Kivanc Birsoy
- Laboratory of Metabolic Regulation and Genetics, Rockefeller University, New York City, NY, USA
| | - Jan P Böttcher
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich (TUM), Germany
| | - Simone Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | - Ashley R Brown
- Department of Biological Sciences, Columbia University, New York City, NY, USA
| | - Bernhard Brüne
- Institute of Biochemistry1-Pathobiochemistry, Goethe-Universität, Frankfurt Am Main, Germany
| | - Giorgia Bulli
- Department of Physiological Genomics, Ludwig-Maximilians-University, Munich, Germany
| | - Alix Bruneau
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Quan Chen
- College of Life Sciences, Nankai University, Tianjin, China
| | - Gina M DeNicola
- Department of Metabolism and Physiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Tobias P Dick
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ) Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Ayelén Distéfano
- Instituto de Investigaciones Biológicas, CONICET, National University of Mar Del Plata, Argentina
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Jan B Engler
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Germany
| | | | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Germany
| | - José Pedro Friedmann Angeli
- Rudolf Virchow Zentrum, Center for Integrative and Translational Bioimaging - University of Würzburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Germany
| | - Dominic C Fuhrmann
- Institute of Biochemistry1-Pathobiochemistry, Goethe-Universität, Frankfurt Am Main, Germany
| | - Ana J García-Sáez
- Institute for Genetics, CECAD, University of Cologne, Germany; Max Planck Institute of Biophysics, Frankfurt/Main, Germany
| | | | - Magdalena Götz
- Department of Physiological Genomics, Ludwig-Maximilians-University, Munich, Germany; Institute of Stem Cell Research, Helmholtz Center Munich, Germany
| | - Wei Gu
- Institute for Cancer Genetics, And Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Linda Hammerich
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | | | - Xuejun Jiang
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Aicha Jeridi
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), Germany, Member of the German Center for Lung Research (DZL)
| | - Yun Pyo Kang
- College of Pharmacy and Research Institute of Pharmaceutical Science, Seoul National University, Republic of Korea
| | | | - David B Konrad
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Stefan Kotschi
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Peng Lei
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Marlène Le Tertre
- Center for Translational Biomedical Iron Research, Heidelberg University, Germany
| | - Sima Lev
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Deguang Liang
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany; Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
| | - Carolin Lohr
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Svenja Lorenz
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Axel Methner
- Institute of Molecular Medicine, Johannes Gutenberg-Universität Mainz, Germany
| | - Bernhard Michalke
- Research Unit Analytical Biogeochemistry, Helmholtz Center Munich, Germany
| | - Anna V Milton
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Junxia Min
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Eikan Mishima
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany
| | | | - Hozumi Motohashi
- Department of Gene Expression Regulation, Tohoku University, Sendai, Japan
| | | | - Shohei Murakami
- Department of Gene Expression Regulation, Tohoku University, Sendai, Japan
| | - James A Olzmann
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Gabriela Pagnussat
- Instituto de Investigaciones Biológicas, CONICET, National University of Mar Del Plata, Argentina
| | - Zijan Pan
- School of Life Sciences, Westlake University, Hangzhou, China
| | | | | | - Derek A Pratt
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Canada
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany
| | - Lukas Ramsauer
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich (TUM), Germany
| | | | - Yoshiro Saito
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Felix Schmidt
- Institute of Molecular Medicine, Johannes Gutenberg-Universität Mainz, Germany
| | - Carina Schmitt
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Almut Schulze
- Division of Tumour Metabolism and Microenvironment, DKFZ Heidelberg and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Annemarie Schwab
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | - Anna Schwantes
- Institute of Biochemistry1-Pathobiochemistry, Goethe-Universität, Frankfurt Am Main, Germany
| | - Mariluz Soula
- Laboratory of Metabolic Regulation and Genetics, Rockefeller University, New York City, NY, USA
| | - Benedikt Spitzlberger
- Department of Immunobiology, Université de Lausanne, Switzerland; Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Brent R Stockwell
- Department of Biological Sciences, Columbia University, New York City, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA; Department of Chemistry, Columbia University, New York, NY, USA
| | - Leonie Thewes
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | | | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan; Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan; Center for Integrated Sciences of Low-temperature Plasma Core Research (iPlasma Core), Tokai National Higher Education and Research System, Nagoya, Japan
| | - Wulf Tonnus
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany
| | - Andreas Trumpp
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM GGmbH), Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Peter Vandenabeele
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- Department of Biomedical Sciences, University of Antwerp, Belgium; VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Vivek Venkataramani
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Germany
| | - Felix C E Vogel
- Division of Tumour Metabolism and Microenvironment, DKFZ Heidelberg and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Silvia von Karstedt
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany; CECAD Cluster of Excellence, University of Cologne, Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Germany
| | - Fudi Wang
- School of Medicine, Zhejiang University, Hangzhou, China
| | | | - Chantal Wientjens
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, Germany
| | - Christoph Wilhelm
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, Germany
| | - Michele Wölk
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Germany
| | - Katherine Wu
- Department of Pathology, Grossman School of Medicine, New York University, NY, USA
| | - Xin Yang
- Institute for Cancer Genetics, And Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Fan Yu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Yilong Zou
- School of Life Sciences, Westlake University, Hangzhou, China; Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany.
| |
Collapse
|
38
|
Yao H, Jiang W, Liao X, Wang D, Zhu H. Regulatory mechanisms of amino acids in ferroptosis. Life Sci 2024; 351:122803. [PMID: 38857653 DOI: 10.1016/j.lfs.2024.122803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/19/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
Ferroptosis, an iron-dependent non-apoptotic regulated cell death process, is associated with the pathogenesis of various diseases. Amino acids, which are indispensable substrates of vital activities, significantly regulate ferroptosis. Amino acid metabolism is involved in maintaining iron and lipid homeostasis and redox balance. The regulatory effects of amino acids on ferroptosis are complex. An amino acid may exert contrasting effects on ferroptosis depending on the context. This review systematically and comprehensively summarized the distinct roles of amino acids in regulating ferroptosis and highlighted the emerging opportunities to develop clinical therapeutic strategies targeting amino acid-mediated ferroptosis.
Collapse
Affiliation(s)
- Heying Yao
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China
| | - Wei Jiang
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China
| | - Xiang Liao
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China
| | - Dongqing Wang
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Haitao Zhu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| |
Collapse
|
39
|
Alborzinia H, Friedmann Angeli JP. Iron: The Secret Ingredient Breaking PARPi Resistance. Cancer Discov 2024; 14:1372-1374. [PMID: 39091202 DOI: 10.1158/2159-8290.cd-24-0775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 08/04/2024]
Abstract
PARP inhibitors (PARPi) are used as a first-line treatment option for cancers with BRCA1/2 mutations, yet a significant number of patients show a limited response to these agents. In the present study, Lei and colleagues demonstrate that PARPi promote increased ferroptosis sensitivity and this can be exploited therapeutically to improve the response to PARPi, marking an important therapeutic concept to exploit ferroptosis-based strategies in clinical settings. See related article by Lei et al., p. 1476 (2).
Collapse
Affiliation(s)
- Hamed Alborzinia
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - José Pedro Friedmann Angeli
- Rudolf Virchow Zentrum, Center for Integrative and Translational Bioimaging, Julius-Maximilians-University Würzburg, Würzburg, Germany
| |
Collapse
|
40
|
Wang Y, Gan X, Cheng X, Jia Y, Wang G, Tang X, Du H, Li X, Liu X, Xing X, Ji J, Li Z. ABCC2 induces metabolic vulnerability and cellular ferroptosis via enhanced glutathione efflux in gastric cancer. Clin Transl Med 2024; 14:e1754. [PMID: 39095325 PMCID: PMC11296884 DOI: 10.1002/ctm2.1754] [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: 03/01/2024] [Revised: 05/30/2024] [Accepted: 06/16/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Although it is traditionally believed that ATP binding cassette subfamily C member 2 (ABCC2) is a multidrug resistance-associated protein correlated with a worse prognosis, our previous and several other studies demonstrated the contrary to be true in gastric cancer (GC). We aim to explore the underlying mechanism of this discovery. METHODS Our study utilized whole-exome sequencing (WES), RNA sequencing, and droplet digital PCR (ddPCR) analysis of 80 gastric cancer samples, along with comprehensive immunohistochemical (IHC) analysis of 1044 human GC tissue samples.By utilizing CRISPRCas9 to genetically modify cell lines with the ABCC2-24C > T (rs717620) point mutation and conducting dual-luciferase reporter assays, we identified that transcription factors SOX9 and ETS1 serve as negative regulators of ABCC2 expression. Seahorse assay and mass spectrometry were used to discover altered metabolic patterns. Gain and loss-of-function experiments in GC cell lines and preclinical models were carried out to validate ABCC2 biological function. RESULTS ABCC2 high expression correlated with better prognosis, and rs717620 can influence ABCC2 expression by disrupting the binding of ETS1 and SOX9. Gain and loss-of-function experiments in GC cell lines demonstrated amino acid deprivation reduces proliferation, migration, and drug resistance in ABCC2-high GC cells. ABCC2 leads to reduced intracellular amino acid pools and disruption of cellular energy metabolism. This phenomenon depended on ABCC2-mediated GSH extrusion, resulting in alterations in redox status, thereby increasing the cell's susceptibility to ferroptosis. Furthermore, patient-derived organoids and patient-derived tumor-like cell clusters were used to observe impact of ABCC2 on therapeutic effect. In the xenograft model with high ABCC2 expression, we observed that constricting amino acid intake in conjunction with GPX4 inactivation resulted in notable tumor regression. CONCLUSIONS Our findings demonstrate a significant role of ABCC2 in amino acid metabolism and ferroptosis by mediating GSH efflux in GC. This discovery underlines the potential of combining multiple ferroptosis targets as a promising therapeutic strategy for GC with high ABCC2 expression. HIGHLIGHTS ABCC2 plays a crucial role in inducing metabolic vulnerability and ferroptosis in gastric cancer through enhanced glutathione efflux. The ABCC2 24C > T polymorphism is a key factor influencing its expression. These results highlight the potential of ABCC2 as a predictive biomarker and therapeutic target in gastric cancer.
Collapse
Affiliation(s)
- Yiding Wang
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
- Department of Gastrointestinal Cancer CenterWard IPeking University Cancer Hospital & InstituteBeijingP.R. China
| | - Xuejun Gan
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
- Department of Gastrointestinal Cancer CenterWard IPeking University Cancer Hospital & InstituteBeijingP.R. China
| | - Xiaojing Cheng
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
| | - Yongning Jia
- Department of Gastrointestinal Cancer CenterWard IPeking University Cancer Hospital & InstituteBeijingP.R. China
| | - Gangjian Wang
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
- Department of Gastrointestinal Cancer CenterWard IPeking University Cancer Hospital & InstituteBeijingP.R. China
| | - Xiaohuan Tang
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
- Department of Gastrointestinal Cancer CenterWard IPeking University Cancer Hospital & InstituteBeijingP.R. China
| | - Hong Du
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
| | - Xiaomei Li
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
| | - Xijuan Liu
- Department of Central LaboratoryKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Peking University Cancer Hospital & InstituteBeijingChina
| | - Xiaofang Xing
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
| | - Jiafu Ji
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
- Department of Gastrointestinal Cancer CenterWard IPeking University Cancer Hospital & InstituteBeijingP.R. China
| | - Ziyu Li
- Department of Gastrointestinal Cancer Translational ResearchKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education)Peking University Cancer Hospital & InstituteBeijingP.R. China
- Department of Gastrointestinal Cancer CenterWard IPeking University Cancer Hospital & InstituteBeijingP.R. China
| |
Collapse
|
41
|
Dai Q, Wei X, Zhao J, Zhang D, Luo Y, Yang Y, Xiang Y, Liu X. Inhibition of FSP1: A new strategy for the treatment of tumors (Review). Oncol Rep 2024; 52:105. [PMID: 38940330 PMCID: PMC11228423 DOI: 10.3892/or.2024.8764] [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: 02/23/2024] [Accepted: 05/10/2024] [Indexed: 06/29/2024] Open
Abstract
Ferroptosis, a regulated form of cell death, is intricately linked to iron‑dependent lipid peroxidation. Recent evidence strongly supports the induction of ferroptosis as a promising strategy for treating cancers resistant to conventional therapies. A key player in ferroptosis regulation is ferroptosis suppressor protein 1 (FSP1), which promotes cancer cell resistance by promoting the production of the antioxidant form of coenzyme Q10. Of note, FSP1 confers resistance to ferroptosis independently of the glutathione (GSH) and glutathione peroxidase‑4 pathway. Therefore, targeting FSP1 to weaken its inhibition of ferroptosis may be a viable strategy for treating refractory cancer. This review aims to clarify the molecular mechanisms underlying ferroptosis, the specific pathway by which FSP1 suppresses ferroptosis and the effect of FSP1 inhibitors on cancer cells.
Collapse
Affiliation(s)
- Qiangfang Dai
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Xiaoli Wei
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Jumei Zhao
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Die Zhang
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Yidan Luo
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Yue Yang
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Yang Xiang
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
- College of Physical Education, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Xiaolong Liu
- School of Medicine, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| |
Collapse
|
42
|
Murray MB, Dixon SJ. Ferroptosis regulation by Cap'n'collar family transcription factors. J Biol Chem 2024; 300:107583. [PMID: 39025451 PMCID: PMC11387702 DOI: 10.1016/j.jbc.2024.107583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024] Open
Abstract
Ferroptosis is an iron-dependent cell death mechanism that may be important to prevent tumor formation and useful as a target for new cancer therapies. Transcriptional networks play a crucial role in shaping ferroptosis sensitivity by regulating the expression of transporters, metabolic enzymes, and other proteins. The Cap'n'collar (CNC) protein NFE2 like bZIP transcription factor 2 (NFE2L2, also known as NRF2) is a key regulator of ferroptosis in many cells and contexts. Emerging evidence indicates that the related CNC family members, BTB domain and CNC homolog 1 (BACH1) and NFE2 like bZIP transcription factor 1 (NFE2L1), also have roles in ferroptosis regulation. Here, we comprehensively review the role of CNC transcription factors in governing cellular sensitivity to ferroptosis. We describe how CNC family members regulate ferroptosis sensitivity through modulation of iron, lipid, and redox metabolism. We also use examples of ferroptosis regulation by CNC proteins to illustrate the flexible and highly context-dependent nature of the ferroptosis mechanism in different cells and conditions.
Collapse
Affiliation(s)
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, California, USA.
| |
Collapse
|
43
|
Zhang J, Wang J, Xu D, Gui Y, Bai F, Huo Y, Cao L, Gui Y. Promoting Glutathione Synthesis: A Possibility for Treating Cardiomyopathy Induced by a Maternal Western Diet. Nutrients 2024; 16:2520. [PMID: 39125400 PMCID: PMC11313981 DOI: 10.3390/nu16152520] [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: 06/05/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND The adverse effects of a Western diet on obesity and diabetes among reproductive-aged women pose a significant threat to the cardiovascular health of their offspring. Given the crucial role of glutathione metabolism and glutathione-related antioxidant defense systems in cardiovascular diseases through scavenging ROS and maintaining redox homeostasis, further exploration of their specific influence is imperative to develop therapeutic strategies for cardiomyopathy induced by a maternal Western diet. METHODS We developed a prenatal maternal Western diet exposure model in C57/B6 mice to investigate cardiac morphology and function through histological analysis and echocardiography. RNA sequencing and analysis were utilized to elucidate the mechanisms underlying the impact of a maternal Western diet and N-acetylcysteine treatment on cardiomyopathy. Additionally, ELISAs, transmission electron microscopy, and flow cytometry were employed to assess the antioxidant defense system and mitochondrial ROS levels in progenitor cardiomyocytes. RESULTS N-acetylcysteine significantly mitigated cardiomyocyte hypertrophy, myocardial interstitial fibrosis, collagen type I accumulation, and left ventricular remodeling induced by a maternal Western diet, particularly in male offspring. Furthermore, N-acetylcysteine reversed the increase in apoptosis and the increase in the β/α-MyHC ratio in the myocardium of offspring that results from a maternal Western diet. RNA sequencing and GSEA revealed that the beneficial effects of N-acetylcysteine were linked to its ability to modulate oxidative phosphorylation pathways. Additionally, N-acetylcysteine treatment during pregnancy can markedly elevate glutathione levels, augment glutathione peroxidase (GPx) activity, and mitigate the accumulation of mitochondrial ROS caused by a maternal Western diet. CONCLUSIONS N-acetylcysteine mitigated cardiomyopathy induced by a maternal Western diet by bolstering glutathione synthesis and enhancing GPx activity, thereby scavenging mitochondrial ROS and modulating oxidative phosphorylation pathways.
Collapse
Affiliation(s)
- Jialing Zhang
- Institute of Pediatrics, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 201102, China; (J.Z.)
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai 201102, China
| | - Jiayu Wang
- Institute of Pediatrics, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 201102, China; (J.Z.)
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai 201102, China
| | - Da Xu
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai 201102, China
- Cardiovascular Center, Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Yiting Gui
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai 201102, China
- Cardiovascular Center, Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Fan Bai
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai 201102, China
- Cardiovascular Center, Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Yu Huo
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai 201102, China
- Cardiovascular Center, Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Li Cao
- Ultrasound Department, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, China
| | - Yonghao Gui
- National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai 201102, China
- Cardiovascular Center, Children’s Hospital of Fudan University, Shanghai 201102, China
| |
Collapse
|
44
|
Ryl T, Afanasyeva E, Hartmann T, Schwermer M, Schneider M, Schröder C, Wagemanns M, Bister A, Kanber D, Steenpass L, Schramm K, Jones B, Jones DTW, Biewald E, Astrahantseff K, Hanenberg H, Rahmann S, Lohmann DR, Schramm A, Ketteler P. A MYCN-driven de-differentiation profile identifies a subgroup of aggressive retinoblastoma. Commun Biol 2024; 7:919. [PMID: 39079981 PMCID: PMC11289481 DOI: 10.1038/s42003-024-06596-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 07/17/2024] [Indexed: 08/02/2024] Open
Abstract
Retinoblastoma are childhood eye tumors arising from retinal precursor cells. Two distinct retinoblastoma subtypes with different clinical behavior have been described based on gene expression and methylation profiling. Using consensus clustering of DNA methylation analysis from 61 retinoblastomas, we identify a MYCN-driven cluster of subtype 2 retinoblastomas characterized by DNA hypomethylation and high expression of genes involved in protein synthesis. Subtype 2 retinoblastomas outside the MYCN-driven cluster are characterized by high expression of genes from mesodermal development, including NKX2-5. Knockdown of MYCN expression in retinoblastoma cell models causes growth arrest and reactivates a subtype 1-specific photoreceptor signature. These molecular changes suggest that removing the driving force of MYCN oncogenic activity rescues molecular circuitry driving subtype 1 biology. The MYCN-RB gene signature generated from the cell models better identifies MYCN-driven retinoblastoma than MYCN amplification and can identify cases that may benefit from MYCN-targeted therapy. MYCN drives tumor progression in a molecularly defined retinoblastoma subgroup, and inhibiting MYCN activity could restore a more differentiated and less aggressive tumor biology.
Collapse
Affiliation(s)
- Tatsiana Ryl
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Elena Afanasyeva
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Till Hartmann
- Algorithms for Reproducible Bioinformatics, Genome Informatics, Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Melanie Schwermer
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Markus Schneider
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Christopher Schröder
- Algorithms for Reproducible Bioinformatics, Genome Informatics, Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Maren Wagemanns
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Arthur Bister
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Deniz Kanber
- Institute of Human Genetics, University Hospital Essen, University Duisburg Essen, Essen, Germany
| | - Laura Steenpass
- Human and Animal Cell Lines, Leibniz Institute DSMZ German Collection of Microorganisms and Cell Cultures, 38124, Braunschweig, Germany
| | - Kathrin Schramm
- Division of Pediatric Glioma Research, Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Barbara Jones
- Division of Pediatric Glioma Research, Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - David T W Jones
- Division of Pediatric Glioma Research, Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Eva Biewald
- Department of Ophthalmology, Medical Faculty, University of Duisburg-Essen, 45147, Essen, Germany
| | - Kathy Astrahantseff
- Department of Pediatric Oncology and Hematology, Charité - University Medicine Berlin, Berlin, Germany
| | - Helmut Hanenberg
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Sven Rahmann
- Algorithmic Bioinformatics, Center for Bioinformatics Saar and Saarland University, Saarland Informatics Campus, Saarbrücken, Germany
| | - Dietmar R Lohmann
- Institute of Human Genetics, University Hospital Essen, University Duisburg Essen, Essen, Germany
| | - Alexander Schramm
- Laboratory for Molecular Oncology, Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Petra Ketteler
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany.
- Institute of Human Genetics, University Hospital Essen, University Duisburg Essen, Essen, Germany.
| |
Collapse
|
45
|
Li TQ, Liu Y, Feng C, Bai J, Wang ZR, Zhang XY, Wang XX. Saikosaponin A attenuates osteoclastogenesis and bone loss by inducing ferroptosis. Front Mol Biosci 2024; 11:1390257. [PMID: 39114369 PMCID: PMC11303733 DOI: 10.3389/fmolb.2024.1390257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/15/2024] [Indexed: 08/10/2024] Open
Abstract
To alleviate bone loss, most current drugs target osteoclasts. Saikosaponin A (Ssa), a triterpene saponin derived from Bupleurum falcatum (also known as Radix bupleuri), has immunoregulatory, neuromodulatory, antiviral, anticancer, anti-convulsant, anti-inflammatory, and anti-proliferative effects. Recently, modulation of bone homeostasis was shown to involve ferroptosis. Herein, we aimed to determine Ssa's inhibitory effects on osteoclastogenesis and differentiation, whether ferroptosis is involved, and the underlying mechanisms. Tartrate-resistant acid phosphatase (TRAP) staining, F-actin staining, and pit formation assays were conducted to confirm Ssa-mediated inhibition of RANKL-induced osteoclastogenesis in vitro. Ssa could promote osteoclast ferroptosis and increase mitochondrial damage by promoting lipid peroxidation, as measured by iron quantification, FerroOrange staining, Dichloro-dihydro-fluorescein diacetate, MitoSOX, malondialdehyde, glutathione, and boron-dipyrromethene 581/591 C11 assays. Pathway analysis showed that Ssa can promote osteoclasts ferroptosis by inhibiting the Nrf2/SCL7A11/GPX4 axis. Notably, we found that the ferroptosis inhibitor ferrostatin-1 and the Nrf2 activator tert-Butylhydroquinone reversed the inhibitory effects of Ssa on RANKL-induced osteoclastogenesis. In vivo, micro-computed tomography, hematoxylin and eosin staining, TRAP staining, enzyme-linked immunosorbent assays, and immunofluorescence confirmed that in rats with periodontitis induced by lipopolysaccharide, treatment with Ssa reduced alveolar bone resorption dose-dependently. The results suggested Ssa as a promising drug to treat osteolytic diseases.
Collapse
Affiliation(s)
- Tian-Qi Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, China
| | - Yan Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Chong Feng
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, China
| | - Jin Bai
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, China
| | - Zi-Rou Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Xiang-Yu Zhang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, China
| | - Xin-Xing Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| |
Collapse
|
46
|
Lai WY, Chuang TP, Borenäs M, Lind DE, Hallberg B, Palmer RH. Anaplastic Lymphoma Kinase signaling stabilizes SLC3A2 expression via MARCH11 to promote neuroblastoma cell growth. Cell Death Differ 2024; 31:910-923. [PMID: 38858548 PMCID: PMC11239919 DOI: 10.1038/s41418-024-01319-0] [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: 10/01/2023] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/12/2024] Open
Abstract
Solute Carrier Family 3, Member 2 (SLC3A2 or 4F2hc) is a multifunctional glycoprotein that mediates integrin-dependent signaling, acts as a trafficking chaperone for amino acid transporters, and is involved in polyamine transportation. We identified SLC3A2 as a potential Anaplastic Lymphoma Kinase (ALK) interacting partner in a BioID-proximity labeling screen in neuroblastoma (NB) cells. In this work we show that endogenous SLC3A2 and ALK interact in NB cells and that this SLC3A2:ALK interaction was abrogated upon treatment with the ALK inhibitor lorlatinib. We show here that loss of ALK activity leads to decreased SLC3A2 expression and reduced SLC3A2 protein stability in a panel of NB cell lines, while stimulation of ALK with ALKAL2 ligand resulted in increased SLC3A2 protein levels. We further identified MARCH11, an E3 ligase, as a regulator of SLC3A2 ubiquitination downstream of ALK. Further, knockdown of SLC3A2 resulted in inhibition of NB cell growth. To investigate the therapeutic potential of SLC3A2 targeting, we performed monotreatment of NB cells with AMXT-1501 (a polyamine transport inhibitor), which showed only moderate effects in NB cells. In contrast, a combination lorlatinib/AMXT-1501 treatment resulted in synergistic inhibition of cell growth in ALK-driven NB cell lines. Taken together, our results identify a novel role for the ALK receptor tyrosine kinase (RTK), working in concert with the MARCH11 E3 ligase, in regulating SLC3A2 protein stability and function in NB cells. The synergistic effect of combined ALK and polyamine transport inhibition shows that ALK/MARCH11/SLC3A2 regulation of amino acid transport is important for oncogenic growth and survival in NB cells.
Collapse
Affiliation(s)
- Wei-Yun Lai
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden
| | - Tzu-Po Chuang
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden
| | - Marcus Borenäs
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden
| | - Dan E Lind
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden
| | - Bengt Hallberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden.
| | - Ruth H Palmer
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530, Gothenburg, Sweden.
| |
Collapse
|
47
|
Zhang T, Yao C, Zhou X, Liu S, Qi L, Zhu S, Zhao C, Hu D, Shen W. Glutathione‑degrading enzymes in the complex landscape of tumors (Review). Int J Oncol 2024; 65:72. [PMID: 38847236 PMCID: PMC11173371 DOI: 10.3892/ijo.2024.5660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/24/2024] [Indexed: 06/12/2024] Open
Abstract
Glutathione (GSH)‑degrading enzymes are essential for starting the first stages of GSH degradation. These enzymes include extracellular γ‑glutamyl transpeptidase (GGT) and intracellular GSH‑specific γ‑glutamylcyclotransferase 1 (ChaC1) and 2. These enzymes are essential for cellular activities, such as immune response, differentiation, proliferation, homeostasis regulation and programmed cell death. Tumor tissue frequently exhibits abnormal expression of GSH‑degrading enzymes, which has a key impact on the development and spread of malignancies. The present review summarizes gene and protein structure, catalytic activity and regulation of GSH‑degrading enzymes, their vital roles in tumor development (including regulation of oxidative and endoplasmic reticulum stress, control of programmed cell death, promotion of inflammation and tumorigenesis and modulation of drug resistance in tumor cells) and potential role as diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Tianyi Zhang
- Department of Acupuncture, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Chongjie Yao
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
- Department of Rehabilitation, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Xu Zhou
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Shimin Liu
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai 200030, P.R. China
| | - Li Qi
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Shiguo Zhu
- School of Basic Medical Sciences, Center for Traditional Chinese Medicine and Immunology Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Chen Zhao
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Dan Hu
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Weidong Shen
- Department of Acupuncture, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| |
Collapse
|
48
|
Chu B, Chen Z, Wu X, Shi H, Jin X, Song B, Cui M, Zhao Y, Zhao Y, He Y, Wang H, Dong F. Photoactivated Gas-Generating Nanocontrast Agents for Long-Term Ultrasonic Imaging-Guided Combined Therapy of Tumors. ACS NANO 2024; 18:15590-15606. [PMID: 38847586 DOI: 10.1021/acsnano.4c01041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2024]
Abstract
To date, long-term and continuous ultrasonic imaging for guiding the puncture biopsy remains a challenge. In order to address this issue, a multimodality imaging and therapeutic method was developed in the present study to facilitate long-term ultrasonic and fluorescence imaging-guided precision diagnosis and combined therapy of tumors. In this regard, certain types of photoactivated gas-generating nanocontrast agents (PGNAs), capable of exhibiting both ultrasonic and fluorescence imaging ability along with photothermal and sonodynamic function, were designed and fabricated. The advantages of these fabricated PGNAs were then utilized against tumors in vivo, and high therapeutic efficacy was achieved through long-term ultrasonic imaging-guided treatment. In particular, the as-prepared multifunctional PGNAs were applied successfully for the fluorescence-based determination of patient tumor samples collected through puncture biopsy in clinics, and superior performance was observed compared to the clinically used SonoVue contrast agents that are incapable of specifically distinguishing the tumor in ex vivo tissues.
Collapse
Affiliation(s)
- Binbin Chu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123 China
| | - Zhiming Chen
- Department of Ultrasound, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Xiaofeng Wu
- Department of Ultrasound, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Haoliang Shi
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123 China
| | - Xiangbowen Jin
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123 China
| | - Bin Song
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123 China
| | - Mingyue Cui
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123 China
| | - Yadan Zhao
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123 China
| | - Yingying Zhao
- Department of Ultrasound, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Yao He
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123 China
- Macao Translational Medicine Center, Macau University of Science and Technology, Taipa, 999078 Macau SAR, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, 999078 Macau SAR, China
| | - Houyu Wang
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123 China
| | - Fenglin Dong
- Department of Ultrasound, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| |
Collapse
|
49
|
Abstract
Ferroptosis is a non-apoptotic cell death mechanism characterized by iron-dependent membrane lipid peroxidation. Here, we review what is known about the cellular mechanisms mediating the execution and regulation of ferroptosis. We first consider how the accumulation of membrane lipid peroxides leads to the execution of ferroptosis by altering ion transport across the plasma membrane. We then discuss how metabolites and enzymes that are distributed in different compartments and organelles throughout the cell can regulate sensitivity to ferroptosis by impinging upon iron, lipid and redox metabolism. Indeed, metabolic pathways that reside in the mitochondria, endoplasmic reticulum, lipid droplets, peroxisomes and other organelles all contribute to the regulation of ferroptosis sensitivity. We note how the regulation of ferroptosis sensitivity by these different organelles and pathways seems to vary between different cells and death-inducing conditions. We also highlight transcriptional master regulators that integrate the functions of different pathways and organelles to modulate ferroptosis sensitivity globally. Throughout this Review, we highlight open questions and areas in which progress is needed to better understand the cell biology of ferroptosis.
Collapse
Affiliation(s)
- Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA.
| | - James A Olzmann
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA.
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA.
| |
Collapse
|
50
|
Thomas M, Nguyen TH, Drnevich J, D’Souza AM, de Alarcon PA, Gnanamony M. Hu14.18K.322A Causes Direct Cell Cytotoxicity and Synergizes with Induction Chemotherapy in High-Risk Neuroblastoma. Cancers (Basel) 2024; 16:2064. [PMID: 38893185 PMCID: PMC11171330 DOI: 10.3390/cancers16112064] [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: 04/03/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
The disialoganglioside, GD2, is a promising therapeutic target due to its overexpression in certain tumors, particularly neuroblastoma (NB), with limited expression in normal tissues. Despite progress, the intricate mechanisms of action and the full spectrum of the direct cellular responses to anti-GD2 antibodies remain incompletely understood. In this study, we examined the direct cytotoxic effects of the humanized anti-GD2 antibody hu14.18K322A (hu14) on NB cell lines, by exploring the associated cell-death pathways. Additionally, we assessed the synergy between hu14 and conventional induction chemotherapy drugs. Our results revealed that hu14 treatment induced direct cytotoxic effects in CHLA15 and SK-N-BE1 cell lines, with a pronounced impact on proliferation and colony formation. Apoptosis emerged as the predominant cell-death pathway triggered by hu14. Furthermore, we saw a reduction in GD2 surface expression in response to hu14 treatment. Hu14 demonstrated synergy with induction chemotherapy drugs with alterations in GD2 expression. Our comprehensive investigation provides valuable insights into the multifaceted effects of hu14 on NB cells, shedding light on its direct cytotoxicity, cell-death pathways, and interactions with induction chemotherapy drugs. This study contributes to the evolving understanding of anti-GD2 antibody therapy and its potential synergies with conventional treatments in the context of NB.
Collapse
Affiliation(s)
- Maria Thomas
- Department of Pediatrics, University of Illinois College of Medicine Peoria, One Illini Drive, Peoria, IL 61605, USA; (M.T.); (T.H.N.); (A.M.D.); (P.A.d.A.)
| | - Thu Hien Nguyen
- Department of Pediatrics, University of Illinois College of Medicine Peoria, One Illini Drive, Peoria, IL 61605, USA; (M.T.); (T.H.N.); (A.M.D.); (P.A.d.A.)
| | - Jenny Drnevich
- Roy J. Carver Biotechnology Center, The University of Illinois at Urbana-Champaign, 1206 W. Gregory Drive, Urbana, IL 61801, USA;
| | - Amber M. D’Souza
- Department of Pediatrics, University of Illinois College of Medicine Peoria, One Illini Drive, Peoria, IL 61605, USA; (M.T.); (T.H.N.); (A.M.D.); (P.A.d.A.)
| | - Pedro A. de Alarcon
- Department of Pediatrics, University of Illinois College of Medicine Peoria, One Illini Drive, Peoria, IL 61605, USA; (M.T.); (T.H.N.); (A.M.D.); (P.A.d.A.)
| | - Manu Gnanamony
- Department of Pediatrics, University of Illinois College of Medicine Peoria, One Illini Drive, Peoria, IL 61605, USA; (M.T.); (T.H.N.); (A.M.D.); (P.A.d.A.)
| |
Collapse
|