1
|
Song L, Gao F, Man J. Ferroptosis: the potential key roles in idiopathic pulmonary fibrosis. Eur J Med Res 2025; 30:341. [PMID: 40296070 PMCID: PMC12036158 DOI: 10.1186/s40001-025-02623-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/21/2025] [Indexed: 04/30/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease characterized by recurrent injury to alveolar epithelial cells, epithelial-mesenchymal transition, and fibroblast activation, which leads to excessive deposition of extracellular matrix (ECM) proteins. However, effective preventative and therapeutic interventions are currently lacking. Ferroptosis, a unique form of iron-dependent lipid peroxidation-induced cell death, exhibits distinct morphological, physiological, and biochemical features compared to traditional programmed cell death. Recent studies have revealed a close relationship between iron homeostasis and the pathogenesis of pulmonary interstitial fibrosis. Ferroptosis exacerbates tissue damage and plays a crucial role in regulating tissue repair and the pathological processes involved. It leads to recurrent epithelial injury, where dysregulated epithelial cells undergo epithelial-mesenchymal transition via multiple signaling pathways, resulting in the excessive release of cytokines and growth factors. This dysregulated environment promotes the activation of pulmonary fibroblasts, ultimately culminating in pulmonary fibrosis. This review summarizes the latest advancements in ferroptosis research and its role in the pathogenesis and treatment of IPF, highlighting the significant potential of targeting ferroptosis for IPF management. Importantly, despite the rapid developments in this emerging research field, ferroptosis studies continue to face several challenges and issues. This review also aims to propose solutions to these challenges and discusses key concepts and pressing questions for the future exploration of ferroptosis.
Collapse
Affiliation(s)
- Longfei Song
- Department of Rehabilitation Medicine, Affiliated Hospital of Shandong Second Medical University, No. 2428 Yuhe Road, Kuiwen District, Weifang City, 261041, Shandong Province, China
| | - Fusheng Gao
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Shandong Second Medical University, No. 2428, Yuhe Road, Kuiwen District, Weifang City, 261041, Shandong Province, China
| | - Jun Man
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Shandong Second Medical University, No. 2428, Yuhe Road, Kuiwen District, Weifang City, 261041, Shandong Province, China.
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, No. 4948, Shengli East Street, Kuiwen District, Weifang City, 261041, Shandong Province, China.
| |
Collapse
|
2
|
Mishima E, Nakamura T, Doll S, Proneth B, Fedorova M, Pratt DA, Friedmann Angeli JP, Dixon SJ, Wahida A, Conrad M. Recommendations for robust and reproducible research on ferroptosis. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00843-2. [PMID: 40204928 DOI: 10.1038/s41580-025-00843-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2025] [Indexed: 04/11/2025]
Abstract
Ferroptosis is a necrotic, non-apoptotic cell death modality triggered by unrestrained iron-dependent lipid peroxidation. By unveiling the regulatory mechanisms of ferroptosis and its relevance to various diseases, research over the past decade has positioned ferroptosis as a promising therapeutic target. The rapid growth of this research field presents challenges, associated with potentially inadequate experimental approaches that may lead to misinterpretations in the assessment of ferroptosis. Typical examples include assessing whether an observed phenotype is indeed linked to ferroptosis, and selecting appropriate animal models and small-molecule modulators of ferroptotic cell death. This Expert Recommendation outlines state-of-the-art methods and tools to reliably study ferroptosis and increase the reproducibility and robustness of experimental results. We present highly validated compounds and animal models, and discuss their advantages and limitations. Furthermore, we provide an overview of the regulatory mechanisms and the best-studied players in ferroptosis regulation, such as GPX4, FSP1, SLC7A11 and ACSL4, discussing frequent pitfalls in experimental design and relevant guidance. These recommendations are intended for researchers at all levels, including those entering the expanding and exciting field of ferroptosis research.
Collapse
Affiliation(s)
- Eikan Mishima
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toshitaka Nakamura
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Sebastian Doll
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Dresden, Germany
| | - Derek A Pratt
- Department of Chemistry and Biomolecular Science, University of Ottawa, Ottawa, Ontario, Canada
| | - José Pedro Friedmann Angeli
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Adam Wahida
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany.
- Translational Redox Biology, TUM Natural School of Sciences, Technical University of Munich, Garching, Germany.
| |
Collapse
|
3
|
Satkanov M, Nurbekova Z, Bilyalov A, Tazhibay D, Zhaksylyk M, Kulatayeva M, Wang Z, Cui J, Alikulov Z. Biochemical properties of molybdenum cofactor isolated from fish liver. FISH PHYSIOLOGY AND BIOCHEMISTRY 2025; 51:62. [PMID: 40053255 DOI: 10.1007/s10695-025-01473-3] [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: 11/30/2024] [Accepted: 02/23/2025] [Indexed: 05/01/2025]
Abstract
Recent studies have demonstrated that the fish liver protein fraction extract obtained by gel filtration exhibits nitric oxide synthase (NOS)-independent NO synthase from nitrates and nitrites. This activity was attributed to the molybdenum enzymes (Mo-enzymes) group which was already demonstrated in mammals. However, the evidence that NOS-independent NO synthase activity can be classified as a fish Mo-enzyme has been poorly demonstrated. In mammals, Mo-enzymes NOS-independent NO synthase activity occurs at the molybdenum center. We studied the ability of molybdenum cofactor (Mo-co) isolated from the protein fraction of fish liver extract to restore the NADPH-nitrate reductase (NADPH-NR) activity from Neurospora crassa nit-1. Our results demonstrated that Mo-co from the extract from fish liver was able to recover NADPH-NR activity in the extract of N. crassa nit-1, thereby possessing the ability to reduce nitrogen compounds. However, the oxidation of Mo-co from fish liver destabilizes molybdenum, leading to its inactivation. However, the results obtained under anaerobic conditions with dithionite indicate that Mo remains bound to Mo-co under highly reducing conditions. This may also indicate that the availability of Mo is not the sole factor affecting the activity of Mo-enzymes, also oxygen content after the synthesis of mature Mo-co may play a role in cofactor inactivation.
Collapse
Affiliation(s)
- Mereke Satkanov
- Department of Biotechnology and Microbiology, L.N. Gumilyov, Eurasian National University, Astana, 010000, Kazakhstan
- Higher School of Living Systems, Immanuel Kant Baltic Federal University, Kaliningrad, 236041, Russia
- Department of General Biology and Genomics, L.N. Gumilyov, Eurasian National University, Astana, 010000, Kazakhstan
| | - Zhadyrassyn Nurbekova
- Department of Biotechnology and Microbiology, L.N. Gumilyov, Eurasian National University, Astana, 010000, Kazakhstan
| | - Alikhan Bilyalov
- Department of Biotechnology and Microbiology, L.N. Gumilyov, Eurasian National University, Astana, 010000, Kazakhstan
| | - Diana Tazhibay
- Department of Biotechnology and Microbiology, L.N. Gumilyov, Eurasian National University, Astana, 010000, Kazakhstan
| | - Masalimov Zhaksylyk
- Department of Biotechnology and Microbiology, L.N. Gumilyov, Eurasian National University, Astana, 010000, Kazakhstan
| | - Maral Kulatayeva
- Department of Biotechnology and Microbiology, L.N. Gumilyov, Eurasian National University, Astana, 010000, Kazakhstan
| | - Zhaoqi Wang
- CAS Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology Chinese Academy of Sciences, Chengdu, 610041, China
| | - Junfang Cui
- Institute of Mountain Hazards and Environment, Chinese Academy of Sciences, Chengdu, 610299, China
| | - Zerekbay Alikulov
- Department of Biotechnology and Microbiology, L.N. Gumilyov, Eurasian National University, Astana, 010000, Kazakhstan.
- Department of General Biology and Genomics, L.N. Gumilyov, Eurasian National University, Astana, 010000, Kazakhstan.
| |
Collapse
|
4
|
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
|
5
|
Lu Y, Li T, Shu Y, Lu C, Luo Z, Wang J, Xiong H, Li W. Lipid peroxidation and sarcopenia: molecular mechanisms and potential therapeutic approaches. Front Med (Lausanne) 2025; 12:1525205. [PMID: 39963429 PMCID: PMC11831367 DOI: 10.3389/fmed.2025.1525205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 01/02/2025] [Indexed: 02/20/2025] Open
Abstract
Sarcopenia is an age-related condition characterized by the progressive loss of skeletal muscle mass and strength. With the global aging population, its incidence is rapidly increasing. Lipid peroxidation is a critical biochemical process that generates reactive oxygen species (ROS), leading to the destruction of muscle cell structure and function. It plays a pivotal role in the onset and progression of sarcopenia. This review summarizes the mechanisms by which lipid peroxidation contributes to sarcopenia, with a focus on its regulatory effects on cell membrane damage, mitochondrial dysfunction, and cell death. In addition, we discuss the protective role of antioxidant factors such as GPX4 (glutathione peroxidase 4) and antioxidant peptides like SS peptides in mitigating lipid peroxidation and delaying the progression of sarcopenia. Finally, the potential of various strategies, including natural compounds, supplements, natural extracts, and lifestyle interventions, in inhibiting lipid peroxidation and promoting muscle health is explored.
Collapse
Affiliation(s)
- Yifan Lu
- Department of Orthopedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
- Department of Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Tiao Li
- Department of Respiratory Medicine, Xiangya Hospital of Central South University, Changsha, China
| | - Yang Shu
- Department of Graduate School, Hunan University of Chinese Medicine, Changsha, China
- Department of Orthopedics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chengyin Lu
- Department of Orthopedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
- Department of Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Zhiqiang Luo
- Department of Orthopedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
- Department of Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Jingrui Wang
- Department of Graduate School, Hunan University of Chinese Medicine, Changsha, China
- Department of Orthopedics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Hui Xiong
- Department of Orthopedics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wangyang Li
- Department of Orthopedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
- Department of Graduate School, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
6
|
Lange M, Wölk M, Doubravsky CE, Hendricks JM, Kato S, Otoki Y, Styler B, Nakagawa K, Fedorova M, Olzmann JA. FSP1-mediated lipid droplet quality control prevents neutral lipid peroxidation and ferroptosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631537. [PMID: 39829838 PMCID: PMC11741373 DOI: 10.1101/2025.01.06.631537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Lipid droplets (LDs) are organelles that store and supply lipids based on cellular needs. While mechanisms preventing oxidative damage to membrane phospholipids are established, the vulnerability of LD neutral lipids to peroxidation and protective mechanisms are unknown. Here, we identify LD-localized Ferroptosis Suppressor Protein 1 (FSP1) as a critical regulator that prevents neutral lipid peroxidation by recycling coenzyme Q10 (CoQ10) to its lipophilic antioxidant form. Lipidomics reveal that FSP1 loss leads to the accumulation of oxidized triacylglycerols and cholesteryl esters, and biochemical reconstitution of FSP1 with CoQ10 and NADH suppresses triacylglycerol peroxidation in vitro. Notably, polyunsaturated fatty acid (PUFA)-rich triacylglycerols enhance cancer cell sensitivity to FSP1 loss and inducing PUFA-rich LDs triggers triacylglycerol peroxidation and LD-initiated ferroptosis when FSP1 activity is impaired. These findings uncover the first LD lipid quality control pathway, wherein LD-localized FSP1 maintains neutral lipid integrity to prevent the buildup of oxidized lipids and induction of ferroptosis.
Collapse
Affiliation(s)
- Mike Lange
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Michele Wölk
- Center of Membrane Biochemistry and Lipid Research, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden Dresden, Germany
| | - Cody E. Doubravsky
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Joseph M. Hendricks
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Shunji Kato
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Yurika Otoki
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Benjamin Styler
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kiyotaka Nakagawa
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden Dresden, Germany
| | - James A. Olzmann
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
7
|
Wang M, Prachyathipsakul T, Wisniewski CA, Xiong C, Goel S, Goel HL, Karner ER, Mukhopadhyay D, Gupta P, Majee A, Thayumanavan S, Mercurio AM. Therapeutic induction of ferroptosis in tumors using PD-L1 targeting antibody nanogel conjugates. Cell Chem Biol 2024; 31:2039-2051.e6. [PMID: 39603241 DOI: 10.1016/j.chembiol.2024.10.014] [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: 03/18/2024] [Revised: 08/16/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024]
Abstract
Although programmed cell death ligand 1 (PD-L1) is best known for its role in immune suppression, tumor-intrinsic functions are emerging. Here, we report that tumor cells that express PD-L1 are sensitive to ferroptosis inducers such as imidazole ketone erastin (IKE). PD-L1 promotes ferroptosis sensitivity because it suppresses SLC7A11 expression and diminishes glutathione levels. Although the use of anti-PD-L1 antibody drug conjugates (ADCs) could be effective for the delivery of ferroptosis inducers to specific tumor populations, the chemistry of most ferroptosis inducers precludes their incorporation in ADCs. To overcome this challenge, we synthesized an antibody nanogel conjugate (ANC) comprised of an anti-PD-L1 antibody conjugated to a nanogel encapsulated with IKE. This ANC targets PD-L1-expressing cells in vitro and in vivo and induces ferroptosis, resulting in tumor suppression. Importantly, this approach is superior to systemic administration of IKE because it enables enhanced delivery of IKE specifically to tumor cells and it requires lower drug doses for efficacy.
Collapse
Affiliation(s)
- Mengdie Wang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | - Christi A Wisniewski
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Choua Xiong
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Shivam Goel
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Hira Lal Goel
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Emmet R Karner
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Dimpi Mukhopadhyay
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Prachi Gupta
- Department of Chemistry, University of Massachusetts, Amherst, MA, USA
| | - Aniket Majee
- Department of Chemistry, University of Massachusetts, Amherst, MA, USA
| | - S Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, MA, USA; Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA.
| | - Arthur M Mercurio
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
8
|
Kim C, Gabriel KR, Boone D, Brown MR, Oppenheimer K, Kost-Alimova M, Pablo JLB, Greka A. FAF2 is a bifunctional regulator of peroxisomal homeostasis and saturated lipid responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.12.628015. [PMID: 39763943 PMCID: PMC11702540 DOI: 10.1101/2024.12.12.628015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Exposure to saturated fatty acids (SFAs), such as palmitic acid, can lead to cellular metabolic dysfunction known as lipotoxicity. Although canonical adaptive metabolic processes like lipid storage or desaturation are known cellular responses to saturated fat exposure, the link between SFA metabolism and organellar biology remains an area of active inquiry. We performed a genome-wide CRISPR knockout screen in human epithelial cells to identify modulators of SFA toxicity. The screen revealed peroxisomal proteins, especially those that impact ether lipid synthesis, as important regulators of lipotoxicity. We identified Fas-associated factor family member 2 (FAF2) as a critical bifunctional co-regulator of peroxisomal and fatty acid biology. We further uncovered a new biological function for the ubiquitin-regulatory X (UBX) and UAS thioredoxin-like domains of FAF2, demonstrating their requirement for peroxisomal protein abundance and SFA-induced cellular stress. Our work highlights the role of FAF2 in regulating peroxisomal abundance and function, and the peroxisome as a key organelle in the cellular response to SFAs.
Collapse
Affiliation(s)
- Choah Kim
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, Mass General Brigham, Boston, MA 02115, USA
| | - Katlyn R. Gabriel
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Department of Medicine, Mass General Brigham, Boston, MA 02115, USA
| | - Dylan Boone
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Katherine Oppenheimer
- Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, Mass General Brigham, Boston, MA 02115, USA
| | | | | | - Anna Greka
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, Mass General Brigham, Boston, MA 02115, USA
| |
Collapse
|
9
|
Ito J, Nakamura T, Toyama T, Chen D, Berndt C, Poschmann G, Mourão ASD, Doll S, Suzuki M, Zhang W, Zheng J, Trümbach D, Yamada N, Ono K, Yazaki M, Kawai Y, Arisawa M, Ohsaki Y, Shirakawa H, Wahida A, Proneth B, Saito Y, Nakagawa K, Mishima E, Conrad M. PRDX6 dictates ferroptosis sensitivity by directing cellular selenium utilization. Mol Cell 2024; 84:4629-4644.e9. [PMID: 39547222 DOI: 10.1016/j.molcel.2024.10.028] [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: 04/10/2024] [Revised: 07/29/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024]
Abstract
Selenium-dependent glutathione peroxidase 4 (GPX4) is the guardian of ferroptosis, preventing unrestrained (phospho)lipid peroxidation by reducing phospholipid hydroperoxides (PLOOH). However, the contribution of other phospholipid peroxidases in ferroptosis protection remains unclear. We show that cells lacking GPX4 still exhibit substantial PLOOH-reducing capacity, suggesting a contribution of alternative PLOOH peroxidases. By scrutinizing potential candidates, we found that although overexpression of peroxiredoxin 6 (PRDX6), a thiol-specific antioxidant enzyme with reported PLOOH-reducing activity, failed to prevent ferroptosis, its genetic loss sensitizes cancer cells to ferroptosis. Mechanistically, we uncover that PRDX6, beyond its known peroxidase activity, acts as a selenium-acceptor protein, facilitating intracellular selenium utilization and efficient selenium incorporation into selenoproteins, including GPX4. Its physiological significance was demonstrated by reduced GPX4 expression in Prdx6-deficient mouse brains and increased sensitivity to ferroptosis in PRDX6-deficient tumor xenografts in mice. Our study highlights PRDX6 as a critical player in directing cellular selenium utilization and dictating ferroptosis sensitivity.
Collapse
Affiliation(s)
- Junya Ito
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Bavaria 85764, Germany; Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi 980-8572, Japan
| | - Toshitaka Nakamura
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Bavaria 85764, Germany
| | - Takashi Toyama
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan
| | - Deng Chen
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Bavaria 85764, Germany
| | - Carsten Berndt
- Department of Neurology, University Hospital and Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf 40225, Germany
| | - Gereon Poschmann
- Institute of Molecular Medicine, Proteome research, University Hospital and Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf 40225, Germany
| | | | - Sebastian Doll
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Bavaria 85764, Germany
| | - Mirai Suzuki
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi 980-8572, Japan
| | - Weijia Zhang
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Bavaria 85764, Germany
| | - Jiashuo Zheng
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Bavaria 85764, Germany
| | - Dietrich Trümbach
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Bavaria 85764, Germany
| | - Naoya Yamada
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Bavaria 85764, Germany
| | - Koya Ono
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Bavaria 85764, Germany
| | - Masana Yazaki
- Department of Bioscience and Biotechnology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Yasutaka Kawai
- Department of Bioscience and Biotechnology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Mieko Arisawa
- Department of Bioscience and Biotechnology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Yusuke Ohsaki
- Laboratory of Nutrition, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi 980-8572, Japan
| | - Hitoshi Shirakawa
- Laboratory of Nutrition, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi 980-8572, Japan
| | - Adam Wahida
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Bavaria 85764, Germany
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Bavaria 85764, Germany
| | - Yoshiro Saito
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan
| | - Kiyotaka Nakagawa
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi 980-8572, Japan
| | - Eikan Mishima
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Bavaria 85764, Germany; Division of Nephrology, Rheumatology and Endocrinology, Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8574, Japan.
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Bavaria 85764, Germany.
| |
Collapse
|
10
|
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
|
11
|
Pezacki AT, Gonciarz RL, Okamura T, Matier CD, Torrente L, Cheng K, Miller SG, Ralle M, Ward NP, DeNicola GM, Renslo AR, Chang CJ. A tandem activity-based sensing and labeling strategy reveals antioxidant response element regulation of labile iron pools. Proc Natl Acad Sci U S A 2024; 121:e2401579121. [PMID: 38968123 PMCID: PMC11252945 DOI: 10.1073/pnas.2401579121] [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: 01/23/2024] [Accepted: 05/29/2024] [Indexed: 07/07/2024] Open
Abstract
Iron is an essential element for life owing to its ability to participate in a diverse array of oxidation-reduction reactions. However, misregulation of iron-dependent redox cycling can also produce oxidative stress, contributing to cell growth, proliferation, and death pathways underlying aging, cancer, neurodegeneration, and metabolic diseases. Fluorescent probes that selectively monitor loosely bound Fe(II) ions, termed the labile iron pool, are potentially powerful tools for studies of this metal nutrient; however, the dynamic spatiotemporal nature and potent fluorescence quenching capacity of these bioavailable metal stores pose challenges for their detection. Here, we report a tandem activity-based sensing and labeling strategy that enables imaging of labile iron pools in live cells through enhancement in cellular retention. Iron green-1 fluoromethyl (IG1-FM) reacts selectively with Fe(II) using an endoperoxide trigger to release a quinone methide dye for subsequent attachment to proximal biological nucleophiles, providing a permanent fluorescent stain at sites of elevated labile iron. IG1-FM imaging reveals that degradation of the major iron storage protein ferritin through ferritinophagy expands the labile iron pool, while activation of nuclear factor-erythroid 2-related factor 2 (NRF2) antioxidant response elements (AREs) depletes it. We further show that lung cancer cells with heightened NRF2 activation, and thus lower basal labile iron, have reduced viability when treated with an iron chelator. By connecting labile iron pools and NRF2-ARE activity to a druggable metal-dependent vulnerability in cancer, this work provides a starting point for broader investigations into the roles of transition metal and antioxidant signaling pathways in health and disease.
Collapse
Affiliation(s)
- Aidan T. Pezacki
- Department of Chemistry, University of California, Berkeley, CA94720
| | - Ryan L. Gonciarz
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA94158
| | - Toshitaka Okamura
- Department of Chemistry, University of California, Berkeley, CA94720
| | - Carson D. Matier
- Department of Chemistry, University of California, Berkeley, CA94720
| | - Laura Torrente
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL33612
| | - Ke Cheng
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA94158
| | - Sophia G. Miller
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR97239
| | - Martina Ralle
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR97239
| | - Nathan P. Ward
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL33612
| | - Gina M. DeNicola
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL33612
| | - Adam R. Renslo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA94158
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA94158
| | - Christopher J. Chang
- Department of Chemistry, University of California, Berkeley, CA94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA94720
| |
Collapse
|
12
|
Qian Z, Zhang X, Huang J, Niu X, Zhu C, Tai Z, Zhu Q, Chen Z, Zhu T, Wu G. ROS-responsive MSC-derived Exosome Mimetics Carrying MHY1485 Alleviate Renal Ischemia Reperfusion Injury through Multiple Mechanisms. ACS OMEGA 2024; 9:24853-24863. [PMID: 38882096 PMCID: PMC11170644 DOI: 10.1021/acsomega.4c01624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 06/18/2024]
Abstract
Renal ischemia reperfusion (IR) injury is a prevalent inflammatory nephropathy in surgeries such as renal transplantation or partial nephrectomy, damaging renal function through inducing inflammation and cell death in renal tubules. Mesenchymal stromal/stem cell (MSC)-based therapies, common treatments to attenuate inflammation in IR diseases, fail to exhibit satisfying effects on cell death in renal IR. In this study, we prepared MSC-derived exosome mimetics (EMs) carrying the mammalian target of the rapamycin (mTOR) agonist to protect kidneys in proinflammatory environments under IR conditions. The thioketal-modified EMs carried the mTOR agonist and bioactive molecules in MSCs and responsively released them in kidney IR areas. MSC-derived EMs and mTOR agonists protected kidneys synergistically from IR through alleviating inflammation, apoptosis, and ferroptosis. The current study indicates that MSC-TK-MHY1485 EMs (MTM-EM) are promising therapeutic biomaterials for renal IR injury.
Collapse
Affiliation(s)
- Zhiyu Qian
- Department of Urology, Zhongshan Hospital Fudan University, 170 Fenglin Road, Shanghai 200030, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200030, China
| | - Xinyue Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China
| | - Jiahua Huang
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Shanghai 201500, China
| | - Xinhao Niu
- Department of Urology, Zhongshan Hospital Fudan University, 170 Fenglin Road, Shanghai 200030, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200030, China
| | - Cuisong Zhu
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Shanghai 201500, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China
| | - Tongyu Zhu
- Department of Urology, Zhongshan Hospital Fudan University, 170 Fenglin Road, Shanghai 200030, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200030, China
| | - Guoyi Wu
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Shanghai 201500, China
| |
Collapse
|
13
|
Mehmood AH, Chang J, Wang Y, Li S, Ma J, Dong B, Liu H. A D-π-A-type ratiometric fluorescent probe to detect polarity changes and inhibition effect during ferroptosis. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:3486-3491. [PMID: 38804096 DOI: 10.1039/d4ay00519h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
To thoroughly understand ferroptosis's biological functions in living cells, it is crucial to investigate the polarity variations that occur during this unique Fe(II)-facilitated oxidative type of cell death. In this work, we report the development of a ratiometric probe (Po-P) to visualize the polarity changes in living cells and the inhibition effect during ferroptosis. The polarity-responsive fluorophore utilized by Po-P has a D-π-A-type structure. Based on theoretical calculations, ICT was proposed as the basis for Po-P's polarity-responsive mechanism. According to cell imaging results, Po-P had a desirable capacity for monitoring polarity fluctuations and erastin-induced ferroptosis. Furthermore, inhibition imaging revealed that dihydrolipoic acid (DHLA) could potentially prevent polarity changes that occur during erastin-induced ferroptosis, just as vitamin E (VE). We anticipate that the probe Po-P could be a valuable tool to quickly monitor polarity fluctuations and inhibition effects during ferroptosis and create new medications for treating disorders related to ferroptosis.
Collapse
Affiliation(s)
- Abdul Hadi Mehmood
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China.
| | - Jia Chang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China.
| | - Yan Wang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China.
| | - Shijing Li
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China.
| | - Jiale Ma
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China.
| | - Baoli Dong
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China.
| | - Hong Liu
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China.
| |
Collapse
|
14
|
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
|
15
|
Li X, Gao L, Li X, Xia J, Pan Y, Bai C. Autophagy, Pyroptosis and Ferroptosis are Rising Stars in the Pathogenesis of Diabetic Nephropathy. Diabetes Metab Syndr Obes 2024; 17:1289-1299. [PMID: 38505538 PMCID: PMC10949337 DOI: 10.2147/dmso.s450695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 03/01/2024] [Indexed: 03/21/2024] Open
Abstract
Diabetic nephropathy (DN) is one of the most common microvascular complications in diabetes and can potentially develop into end-stage renal disease. Its pathogenesis is complex and not fully understood. Podocytes, glomerular endothelial cells (GECs), glomerular mesangial cells (GMCs) and renal tubular epithelial cells (TECs) play important roles in the normal function of glomerulus and renal tubules, and their injury is involved in the progression of DN. Although our understanding of the mechanisms leading to DN has substantially improved, we still need to find more effective therapeutic targets. Autophagy, pyroptosis and ferroptosis are programmed cell death processes that are associated with inflammation and are closely related to a variety of diseases. Recently, a growing number of studies have reported that autophagy, pyroptosis and ferroptosis regulate the function of podocytes, GECs, GMCs and TECs. This review highlights the contributions of autophagy, pyroptosis, and ferroptosis to DN injury in these cells, offering potential therapeutic targets for DN treatment.
Collapse
Affiliation(s)
- Xiudan Li
- Department of Endocrinology, Affiliated Hospital of Chifeng University, Chifeng, 024000, China
- Inner Mongolia Key Laboratory of Human Genetic Disease Research, Chifeng University, Chifeng, 024000, China
| | - Lifeng Gao
- Inner Mongolia Key Laboratory of Human Genetic Disease Research, Chifeng University, Chifeng, 024000, China
| | - Xuyang Li
- Inner Mongolia Key Laboratory of Human Genetic Disease Research, Chifeng University, Chifeng, 024000, China
| | - Jingdong Xia
- Department of Endocrinology, Affiliated Hospital of Chifeng University, Chifeng, 024000, China
| | - Yurong Pan
- Department of Endocrinology, Affiliated Hospital of Chifeng University, Chifeng, 024000, China
| | - Chunying Bai
- Inner Mongolia Key Laboratory of Human Genetic Disease Research, Chifeng University, Chifeng, 024000, China
| |
Collapse
|
16
|
Mathiowetz AJ, Olzmann JA. Lipid droplets and cellular lipid flux. Nat Cell Biol 2024; 26:331-345. [PMID: 38454048 PMCID: PMC11228001 DOI: 10.1038/s41556-024-01364-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 01/24/2024] [Indexed: 03/09/2024]
Abstract
Lipid droplets are dynamic organelles that store neutral lipids, serve the metabolic needs of cells, and sequester lipids to prevent lipotoxicity and membrane damage. Here we review the current understanding of the mechanisms of lipid droplet biogenesis and turnover, the transfer of lipids and metabolites at membrane contact sites, and the role of lipid droplets in regulating fatty acid flux in lipotoxicity and cell death.
Collapse
Affiliation(s)
- Alyssa J Mathiowetz
- 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
| | - 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
|