1
|
Zhou Y, Wen R, Song T, Long B, Deng GJ. Efficient unsymmetric disulfide formation by molecular-scale tailoring of ortho-polyquinone-based polymer photocatalyst. J Colloid Interface Sci 2025; 678:1022-1030. [PMID: 39232475 DOI: 10.1016/j.jcis.2024.08.224] [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: 07/31/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024]
Abstract
Disulfide bonds, especially unsymmetric disulfide bonds, have important applications in bioactivity and drug molecules, but the synthesis of unsymmetric disulfide bonds remains challenging due to efficiency and selectivity issues. Herein, this work utilizes anthraquinone (AQ) and cyclictriphosphonononitrile through a nucleophilic substitution reaction to synthesize an organic polymer (ANTH-AMI) that incorporates an ortho-polyquinone (o-polyquinone) redox center. The anthraquinone molecule functions as a redox center, capable of accepting photoinduced electrons and subsequently transferring them to initiate an electron-coupled hydrogenation reaction (AQ to AQH). Moreover, the proximity of the o-polyquinone redox sites facilitates the catalysis of unsymmetric disulfide bond formation. Consequently, the ANTH-AMI photocatalysts demonstrate exceptional yields (up to 82 %), substrate versatility, cycling stability, and scalable preparation in promoting unsymmetric coupling reactions of thiol. This work offers a solution for designing organic polymer photocatalysts with adjacent multiple redox centers for cross-coupling reactions.
Collapse
Affiliation(s)
- Yazheng Zhou
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Application of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, PR China
| | - Richang Wen
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Application of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, PR China
| | - Ting Song
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Application of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, PR China.
| | - Bei Long
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Application of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, PR China
| | - Guo-Jun Deng
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Application of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, PR China; School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, PR China.
| |
Collapse
|
2
|
Alves F, Lane D, Nguyen TPM, Bush AI, Ayton S. In defence of ferroptosis. Signal Transduct Target Ther 2025; 10:2. [PMID: 39746918 PMCID: PMC11696223 DOI: 10.1038/s41392-024-02088-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/10/2024] [Accepted: 11/29/2024] [Indexed: 01/04/2025] Open
Abstract
Rampant phospholipid peroxidation initiated by iron causes ferroptosis unless this is restrained by cellular defences. Ferroptosis is increasingly implicated in a host of diseases, and unlike other cell death programs the physiological initiation of ferroptosis is conceived to occur not by an endogenous executioner, but by the withdrawal of cellular guardians that otherwise constantly oppose ferroptosis induction. Here, we profile key ferroptotic defence strategies including iron regulation, phospholipid modulation and enzymes and metabolite systems: glutathione reductase (GR), Ferroptosis suppressor protein 1 (FSP1), NAD(P)H Quinone Dehydrogenase 1 (NQO1), Dihydrofolate reductase (DHFR), retinal reductases and retinal dehydrogenases (RDH) and thioredoxin reductases (TR). A common thread uniting all key enzymes and metabolites that combat lipid peroxidation during ferroptosis is a dependence on a key cellular reductant, nicotinamide adenine dinucleotide phosphate (NADPH). We will outline how cells control central carbon metabolism to produce NADPH and necessary precursors to defend against ferroptosis. Subsequently we will discuss evidence for ferroptosis and NADPH dysregulation in different disease contexts including glucose-6-phosphate dehydrogenase deficiency, cancer and neurodegeneration. Finally, we discuss several anti-ferroptosis therapeutic strategies spanning the use of radical trapping agents, iron modulation and glutathione dependent redox support and highlight the current landscape of clinical trials focusing on ferroptosis.
Collapse
Affiliation(s)
- Francesca Alves
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Darius Lane
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | | | - Ashley I Bush
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia.
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia.
| | - Scott Ayton
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia.
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
3
|
Hong X, Zhang Y, Fu W, Wang L. [Research progress on the role of ferroptosis in aortic dissection]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:726-734. [PMID: 39694526 PMCID: PMC11736346 DOI: 10.3724/zdxbyxb-2024-0186] [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: 05/09/2024] [Accepted: 10/15/2024] [Indexed: 12/20/2024]
Abstract
Recent studies have shown that iron metabolism dysregulation and lipid peroxidation-induced ferroptosis, triggered by oxidative stress, play a key role in the development of aortic dissection. Dysregulated iron metabolism leads to excessive production of hydroxyl radicals due to abnormal iron levels and heme metabolism, while lipid peroxidation is linked to system Xc- dysfunction and accumulation of phospholipid hydroperoxides. These factors synergistically disrupt aortic homeostasis and drive ferroptosis in vascular cells, including endothelial and smooth muscle cells. Furthermore, disruptions in ferroptosis-related genes, along with risk factors such as smoking, epigenetic modifications such as protein methylation, and abnormalities in immune cells, particularly T cells, are closely linked to aortic dissection. Several small molecules and nanomaterials have shown potential in inhibiting ferroptosis in this context. This review elucidates the roles of ferroptosis in aortic dissection and proposes strategies for its targeted prevention and treatment.
Collapse
Affiliation(s)
- Xiang Hong
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen 361015, Fujian Province, China.
- Xiamen Municipal Vascular Disease Precise Diagnose & Treatment Lab, Xiamen 361015, Fujian Province, China.
| | - Yuchong Zhang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Vascular Surgery Institute of Fudan University, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China]
| | - Weiguo Fu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen 361015, Fujian Province, China
- Xiamen Municipal Vascular Disease Precise Diagnose & Treatment Lab, Xiamen 361015, Fujian Province, China
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Vascular Surgery Institute of Fudan University, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China]
| | - Lixin Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen 361015, Fujian Province, China.
- Xiamen Municipal Vascular Disease Precise Diagnose & Treatment Lab, Xiamen 361015, Fujian Province, China.
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Vascular Surgery Institute of Fudan University, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China].
| |
Collapse
|
4
|
Jin Z, Mollica F, Huang Y, Guernelli S, Baschieri A, Diquigiovanni C, Rizzardi N, Valenti F, Pincigher L, Bergamini C, Amorati R. Pro-aromatic Natural Terpenes as Unusual "Slingshot" Antioxidants with Promising Ferroptosis Inhibition Activity. Chemistry 2024; 30:e202403320. [PMID: 39392313 DOI: 10.1002/chem.202403320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 10/11/2024] [Indexed: 10/12/2024]
Abstract
Ferroptosis is a cell death mechanism based on extensive cellular membrane peroxidation, implicated in neurodegenerative and other diseases. The essential oil component γ-terpinene, a natural monoterpene with a unique highly oxidizable pro-aromatic 1,4-cyclohexadiene skeleton, inhibits peroxidation of polyunsaturated lipid in model heterogeneous systems (micelles and liposomes). Upon H-atom abstraction, an unstable γ-terpinene-derived peroxyl radical is formed, that aromatizes to p-cymene generating HOO⋅ radicals. As HOO⋅ are small and hydrophilic radicals, they quickly diffuse outside the lipid core, blocking the radical chain propagation of polyunsaturated lipids. This unprecedented antioxidant "slingshot" mechanism explains why γ-terpinene shows a protective activity against ferroptosis, being effective at submicromolar concentrations in human neuroblastoma (SH-SY5Y) cells.
Collapse
Affiliation(s)
- Zongxin Jin
- Department of Chemistry, "G. Ciamician" University of Bologna, Via Gobetti 83, 40129, Bologna, Italy
| | - Fabio Mollica
- Department of Chemistry, "G. Ciamician" University of Bologna, Via Gobetti 83, 40129, Bologna, Italy
| | - Yeqin Huang
- Department of Chemistry, "G. Ciamician" University of Bologna, Via Gobetti 83, 40129, Bologna, Italy
| | - Susanna Guernelli
- Department of Chemistry, "G. Ciamician" University of Bologna, Via Gobetti 83, 40129, Bologna, Italy
| | - Andrea Baschieri
- Institute for Organic Synthesis and Photoreactivity, (ISOF) National Research Council of Italy (CNR), Via Gobetti 101, 40129, Bologna, Italy
| | - Chiara Diquigiovanni
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Nicola Rizzardi
- Department of Pharmacy and Biotechnology, FABIT, University of Bologna, Via Irnerio 48, 40126, Bologna, Italy
| | - Francesca Valenti
- Department of Pharmacy and Biotechnology, FABIT, University of Bologna, Via Irnerio 48, 40126, Bologna, Italy
| | - Luca Pincigher
- Department of Pharmacy and Biotechnology, FABIT, University of Bologna, Via Irnerio 48, 40126, Bologna, Italy
| | - Christian Bergamini
- Department of Pharmacy and Biotechnology, FABIT, University of Bologna, Via Irnerio 48, 40126, Bologna, Italy
| | - Riccardo Amorati
- Department of Chemistry, "G. Ciamician" University of Bologna, Via Gobetti 83, 40129, Bologna, Italy
| |
Collapse
|
5
|
Zhou M, Xu K, Ge J, Luo X, Wu M, Wang N, Zeng J. Targeting Ferroptosis in Parkinson's Disease: Mechanisms and Emerging Therapeutic Strategies. Int J Mol Sci 2024; 25:13042. [PMID: 39684753 DOI: 10.3390/ijms252313042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/30/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra and the accumulation of α-synuclein in the brain. Ferroptosis, a recently identified form of regulated cell death, is critical in PD pathogenesis due to its association with iron deposition, overproduction of reactive oxygen species, iron-dependent lipid peroxidation and impaired lipid peroxidation clearance. This cell death mechanism is closely linked to several pathogenic processes in PD, including α-synuclein aggregation, oxidative stress, mitochondrial dysfunction, microglia-induced neuroinflammation, and neuromelanin accumulation. Given the significant role of ferroptosis in these mechanisms, there is increasing interest in targeting ferroptosis for PD treatment. Several drugs have shown potential in alleviating PD symptoms by inhibiting ferroptosis. This review aims to consolidate current knowledge on ferroptosis in PD and assess the therapeutic potential of anti-ferroptosis drugs, highlighting promising directions for future research and clinical applications.
Collapse
Affiliation(s)
- Minghao Zhou
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Keyang Xu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jianxian Ge
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Xingnian Luo
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Mengyao Wu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Ning Wang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jianfeng Zeng
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| |
Collapse
|
6
|
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
|
7
|
Papadimitriou‐Tsantarliotou A, Avgeros C, Konstantinidou M, Vizirianakis IS. Analyzing the role of ferroptosis in ribosome-related bone marrow failure disorders: From pathophysiology to potential pharmacological exploitation. IUBMB Life 2024; 76:1011-1034. [PMID: 39052023 PMCID: PMC11580388 DOI: 10.1002/iub.2897] [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: 04/16/2024] [Accepted: 06/04/2024] [Indexed: 07/27/2024]
Abstract
Within the last decade, the scientific community has witnessed the importance of ferroptosis as a novel cascade of molecular events leading to cellular decisions of death distinct from apoptosis and other known forms of cell death. Notably, such non- apoptotic and iron-dependent regulated cell death has been found to be intricately linked to several physiological processes as well as to the pathogenesis of various diseases. To this end, recent data support the notion that a potential molecular connection between ferroptosis and inherited bone marrow failure (IBMF) in individuals with ribosomopathies may exist. In this review, we suggest that in ribosome-related IBMFs the identified mutations in ribosomal proteins lead to changes in the ribosome composition of the hematopoietic progenitors, changes that seem to affect ribosomal function, thus enhancing the expression of some mRNAs subgroups while reducing the expression of others. These events lead to an imbalance inside the cell as some molecular pathways are promoted while others are inhibited. This disturbance is accompanied by ROS production and lipid peroxidation, while an additional finding in most of them is iron accumulation. Once lipid peroxidation and iron accumulation are the two main characteristics of ferroptosis, it is possible that this mechanism plays a key role in the manifestation of IBMF in this type of disease. If this molecular mechanism is further confirmed, new pharmacological targets such as ferroptosis inhibitors that are already exploited for the treatment of other diseases, could be utilized to improve the treatment of ribosomopathies.
Collapse
Affiliation(s)
| | - Chrysostomos Avgeros
- Laboratory of Pharmacology, School of PharmacyAristotle University of ThessalonikiThessalonikiGreece
| | - Maria Konstantinidou
- Laboratory of Pharmacology, School of PharmacyAristotle University of ThessalonikiThessalonikiGreece
| | - Ioannis S. Vizirianakis
- Laboratory of Pharmacology, School of PharmacyAristotle University of ThessalonikiThessalonikiGreece
- Department of Health Sciences, School of Life and Health SciencesUniversity of NicosiaNicosiaCyprus
| |
Collapse
|
8
|
Donohoe MN, Upadhyay A, Pratt DA. Ligand-Based Radical Reactivity of Metal Thiosemicarbazones Prompts the Identification of Platinum(II)-Based Cytoprotectants. J Am Chem Soc 2024; 146:31307-31320. [PMID: 39494512 DOI: 10.1021/jacs.4c12713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
CuATSM, a copper(II) complex of a bis(thiosemicarbazone) of diacetyl, prevents oxidative cell death and acts as a neuroprotectant in vivo, prompting its evaluation to treat amyotrophic lateral sclerosis and other neurodegenerative conditions in the clinic. We recently demonstrated that CuATSM functions as a potent radical-trapping antioxidant (RTA), inhibiting lipid peroxidation and associated ferroptotic cell death by a noncanonical mechanism based on radical addition to the ligand backbone. Herein we report our investigations of the generality of this reactivity, which include studies of corresponding complexes of various other metals, including Co, Ru, Ni, Pd, Pt, and Au. Inhibited autoxidations of styrene and dioxane reveal that most of these complexes exhibit RTA activity, consistent with ligand-based reactivity, but the identity of the metal atom nevertheless plays a role. In particular, analyses of the electronic structures of the complexes of metals within the same group (i.e., the group 10 metals Ni, Pd and Pt) highlight how the metal atom can modulate the ligand-based reactivity by enabling spin delocalization to the other thiosemicarbazone moiety. The RTA activity determined in organic solution largely translates to phospholipid bilayers and mammalian cells, where most complexes inhibited lipid peroxidation and associated ferroptotic cell death. A preliminary structure-activity study revealed Pt complexes with potencies eclipsing those of archetype ferroptosis inhibitors ferrostatin-1 and liproxstatin-1, suggesting that Pt (and to a lesser extent Ni) bis(thiosemicarbazone)s may be better suited to optimization for therapeutic development than those based on Cu.
Collapse
Affiliation(s)
- Michael N Donohoe
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa K1N 6N5, Ontario, Canada
| | - Aditya Upadhyay
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa K1N 6N5, Ontario, Canada
| | - Derek A Pratt
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa K1N 6N5, Ontario, Canada
| |
Collapse
|
9
|
Wang LY, Zhang L, Bai XY, Qiang RR, Zhang N, Hu QQ, Cheng JZ, Yang YL, Xiang Y. The Role of Ferroptosis in Amyotrophic Lateral Sclerosis Treatment. Neurochem Res 2024; 49:2653-2667. [PMID: 38864944 DOI: 10.1007/s11064-024-04194-w] [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: 03/25/2024] [Revised: 05/17/2024] [Accepted: 06/06/2024] [Indexed: 06/13/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a rare neurodegenerative disease with a challenging treatment landscape, due to its complex pathogenesis and limited availability of clinical drugs. Ferroptosis, an iron-dependent form of programmed cell death (PCD), stands distinct from apoptosis, necrosis, autophagy, and other cell death mechanisms. Recent studies have increasingly highlighted the role of iron deposition, reactive oxygen species (ROS) accumulation, oxidative stress, as well as systemic Xc- and glutamate accumulation in the antioxidant system in the pathogenesis of amyotrophic lateral sclerosis. Therefore, targeting ferroptosis emerges as a promising strategy for amyotrophic lateral sclerosis treatment. This review introduces the regulatory mechanism of ferroptosis, the relationship between amyotrophic lateral sclerosis and ferroptosis, and the drugs used in the clinic, then discusses the current status of amyotrophic lateral sclerosis treatment, hoping to provide new directions and targets for its treatment.
Collapse
Affiliation(s)
- Le Yi Wang
- Yan 'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Lei Zhang
- Yan 'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Xin Yue Bai
- Yan 'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Rong Rong Qiang
- Yan 'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Ning Zhang
- Yan 'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Qian Qian Hu
- Yan 'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Jun Zhi Cheng
- Yan 'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Yan Ling Yang
- Yan 'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Yang Xiang
- College of Physical Education, Yan'an University, Shaanxi, 716000, China.
| |
Collapse
|
10
|
Chen W, Xu J, Rao W, Shen SS, Yang ZY, Ackermann L, Wang SY. Copper(0)-Catalyzed Reductive Coupling of Disulfurating Reagents and (Hetero)aryl/Alkyl Halides. Org Lett 2024. [PMID: 39291854 DOI: 10.1021/acs.orglett.4c03032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Herein, we reported a copper(0)-catalyzed reductive coupling of disulfurating reagents and (hetero)aryl/alkyl halides. Copper(0) can be directly inserted into tetrasulfide and then undergoes reductive coupling with (hetero)aryl Iodides to construct disulfide. The method features the unprecedented use of copper(0)-catalyzed disulfurating reagents (tetrasulfides) in cross-coupling chemistry and is convenient with broad substrate scopes, even applicable to different halogenated hydrocarbons. It is worth noting that the methodology is practical with the late-stage modification of bioactive scaffolds of pharmaceuticals. In the meantime, the synthesis of disulfides is successfully achieved on a gram scale, indicating the approach is highly valuable.
Collapse
Affiliation(s)
- Wang Chen
- Key Laboratory of Organic Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China
| | - Jiuwen Xu
- Key Laboratory of Organic Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China
| | - Weidong Rao
- Key Laboratory of Biomass-Based Green Fuels and Chemicals, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Shu-Su Shen
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, No. 99, Xuefu Road, Huqiu District, Suzhou 215009, P. R. China
| | - Zhao-Ying Yang
- Soochow College, Soochow University, Suzhou, 215123, China
| | - Lutz Ackermann
- Wöhler Research Institute for Sustainable Chemistry, Georg-August-Universität Göttingen, Tammannstraße 2, 37077 Göttingen, Germany
| | - Shun-Yi Wang
- Key Laboratory of Organic Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China
| |
Collapse
|
11
|
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
|
12
|
Jiao T, Chen Y, Sun H, Yang L. Targeting ferroptosis as a potential prevention and treatment strategy for aging-related diseases. Pharmacol Res 2024; 208:107370. [PMID: 39181344 DOI: 10.1016/j.phrs.2024.107370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Ferroptosis, an emerging paradigm of programmed cellular necrosis posited in recent years, manifests across a spectrum of maladies with profound implications for human well-being. Numerous investigations substantiate that modulating ferroptosis, whether through inhibition or augmentation, plays a pivotal role in the etiology and control of numerous age-related afflictions, encompassing neurological, circulatory, respiratory, and other disorders. This paper not only summarizes the regulatory mechanisms of ferroptosis, but also discusses the impact of ferroptosis on the biological processes of aging and its role in age-related diseases. Furthermore, it scrutinizes recent therapeutic strides in addressing aging-related conditions through the modulation of ferroptosis. The paper consolidates the existing knowledge on potential applications of ferroptosis-related pharmacotherapies and envisages the translational prospects of ferroptosis-targeted interventions in clinical paradigms.
Collapse
Affiliation(s)
- Taiwei Jiao
- Department of Gastroenterology and Endoscopy, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China
| | - Yiman Chen
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China
| | - Haiyan Sun
- Department of Endodontics, School of Stomatology, China Medical University, Shenyang, Liaoning 110001, PR China.
| | - Lina Yang
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China; Department of International Physical Examination Center, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China.
| |
Collapse
|
13
|
Jeong HJ, Picou C, Jeong K, Chung JK. Oxidation Kinetics of Fluorescent Membrane Lipid Peroxidation Indicators. ACS Chem Biol 2024; 19:1786-1793. [PMID: 39037001 DOI: 10.1021/acschembio.4c00269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
The oxidation of the cellular membrane through lipid peroxidation (LPO) is linked to aging and disease. Despite the physiological importance, the chemical mechanisms underlying LPO and oxidative reactions in membranes in general remain incompletely understood, and challenges exist in translating LPO inhibitor efficacies from in vitro to in vivo. The complexity of LPO, including multiple oxidation reactions in complex membrane environments and the difficulty in quantifying reaction kinetics, underlies these difficulties. In this work, we developed a robust and straightforward method for quantifying the oxidation rate kinetics of fluorescent molecules and determined the oxidation kinetics of widely fluorophores used as indicators of membrane LPO, diphenylhexatriene (DPH), BODIPY-C11, and Liperfluo. The measurement is initiated by lipoxygenase, which provides chemical specificity and enables a straightforward interpretation of oxidation kinetics. Our results reveal that the membrane composition significantly impacts the observed kinetics oxidation in DPH and BODIPY-C11 but not Liperfluo. Reaction mechanisms for their lipid peroxide-induced oxidation are proposed. This work provides a foundation for the quantitative analysis of LPO with fluorescence and extricating the complexity of oxidation reactions within membranes.
Collapse
Affiliation(s)
- Hye Jin Jeong
- Department of Chemistry, Colorado State University Fort Collins, Fort Collins, Colorado 80523, United States
| | - Cyrus Picou
- Department of Chemistry, Colorado State University Fort Collins, Fort Collins, Colorado 80523, United States
| | - Keunhong Jeong
- Department of Chemistry, Colorado State University Fort Collins, Fort Collins, Colorado 80523, United States
| | - Jean K Chung
- Department of Chemistry, Colorado State University Fort Collins, Fort Collins, Colorado 80523, United States
| |
Collapse
|
14
|
Lu Y, Shen Z, Xu Y, Lin H, Shen L, Jin Y, Guo Y, Lu J, Li L, Zhuang Y, Jin Y, Zhuang W, Huang W, Dong X, Dai H, Che J. Discovery of New Phenyltetrazolium Derivatives as Ferroptosis Inhibitors for Treating Ischemic Stroke: An Example Development from Free Radical Scavengers. J Med Chem 2024; 67:11712-11731. [PMID: 38996382 DOI: 10.1021/acs.jmedchem.4c00211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Ferroptosis is a promising therapeutic target for injury-related diseases, yet diversity in ferroptosis inhibitors remains limited. In this study, initial structure optimization led us to focus on the bond dissociation enthalpy (BDE) of the N-H bond and the residency time of radical scavengers in a phospholipid bilayer, which may play an important role in ferroptosis inhibition potency. This led to the discovery of compound D1, exhibiting potent ferroptosis inhibition, high radical scavenging, and moderate membrane permeability. D1 demonstrated significant neuroprotection in an oxygen glucose deprivation/reoxygenation (OGD/R) model and reduced infarct volume in an in vivo stroke model upon intravenous treatment. Further screening based on this strategy identified NecroX-7 and Eriodictyol-7-O-glucoside as novel ferroptosis inhibitors with highly polar structural characteristics. This approach bridges the gap between free radical scavengers and ferroptosis inhibitors, providing a foundation for research and insights into novel ferroptosis inhibitor development.
Collapse
Affiliation(s)
- Yang Lu
- Department of Pharmacy, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zexu Shen
- Department of Pharmacy, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yaping Xu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Haoran Lin
- Department of Pharmacy, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Liteng Shen
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yizhen Jin
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yu Guo
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jialiang Lu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Linjie Li
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuxin Zhuang
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuheng Jin
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Weihao Zhuang
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wenhai Huang
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Hangzhou Medical College, Hangzhou 310013, PR China
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Xiaowu Dong
- Department of Pharmacy, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Haibin Dai
- Department of Pharmacy, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jinxin Che
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
15
|
Wei Y, Zhong S, Yang H, Wang X, Lv B, Bian Y, Pei Y, Xu C, Zhao Q, Wu Y, Luo D, Wang F, Sun H, Chen Y. Current therapy in amyotrophic lateral sclerosis (ALS): A review on past and future therapeutic strategies. Eur J Med Chem 2024; 272:116496. [PMID: 38759454 DOI: 10.1016/j.ejmech.2024.116496] [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: 01/11/2024] [Revised: 05/11/2024] [Accepted: 05/11/2024] [Indexed: 05/19/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that affects the first and second motoneurons (MNs), associated with muscle weakness, paralysis and finally death. The exact etiology of the disease still remains unclear. Currently, efforts to develop novel ALS treatments which target specific pathomechanisms are being studied. The mechanisms of ALS pathogenesis involve multiple factors, such as protein aggregation, glutamate excitotoxicity, oxidative stress, mitochondrial dysfunction, apoptosis, inflammation etc. Unfortunately, to date, there are only two FDA-approved drugs for ALS, riluzole and edavarone, without curative treatment for ALS. Herein, we give an overview of the many pathways and review the recent discovery and preclinical characterization of neuroprotective compounds. Meanwhile, drug combination and other therapeutic approaches are also reviewed. In the last part, we analyze the reasons of clinical failure and propose perspective on the treatment of ALS in the future.
Collapse
Affiliation(s)
- Yuqing Wei
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Sheng Zhong
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Huajing Yang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xueqing Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Bingbing Lv
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yaoyao Bian
- Jiangsu Provincial Engineering Center of TCM External Medication Researching and Industrializing, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yuqiong Pei
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chunlei Xu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qun Zhao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yulan Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Daying Luo
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Fan Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
16
|
Zhang L, Luo YL, Xiang Y, Bai XY, Qiang RR, Zhang X, Yang YL, Liu XL. Ferroptosis inhibitors: past, present and future. Front Pharmacol 2024; 15:1407335. [PMID: 38846099 PMCID: PMC11153831 DOI: 10.3389/fphar.2024.1407335] [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: 03/26/2024] [Accepted: 05/06/2024] [Indexed: 06/09/2024] Open
Abstract
Ferroptosis is a non-apoptotic mode of programmed cell death characterized by iron dependence and lipid peroxidation. Since the ferroptosis was proposed, researchers have revealed the mechanisms of its formation and continue to explore effective inhibitors of ferroptosis in disease. Recent studies have shown a correlation between ferroptosis and the pathological mechanisms of neurodegenerative diseases, as well as diseases involving tissue or organ damage. Acting on ferroptosis-related targets may provide new strategies for the treatment of ferroptosis-mediated diseases. This article specifically describes the metabolic pathways of ferroptosis and summarizes the reported mechanisms of action of natural and synthetic small molecule inhibitors of ferroptosis and their efficacy in disease. The paper also describes ferroptosis treatments such as gene therapy, cell therapy, and nanotechnology, and summarises the challenges encountered in the clinical translation of ferroptosis inhibitors. Finally, the relationship between ferroptosis and other modes of cell death is discussed, hopefully paving the way for future drug design and discovery.
Collapse
Affiliation(s)
- Lei Zhang
- School of Medicine, Yan’an University, Yan’an, China
| | - Yi Lin Luo
- School of Medicine, Yan’an University, Yan’an, China
| | - Yang Xiang
- College of Physical Education, Yan’an University, Yan’an, China
| | - Xin Yue Bai
- School of Medicine, Yan’an University, Yan’an, China
| | | | - Xin Zhang
- School of Medicine, Yan’an University, Yan’an, China
| | - Yan Ling Yang
- School of Medicine, Yan’an University, Yan’an, China
| | - Xiao Long Liu
- School of Medicine, Yan’an University, Yan’an, China
| |
Collapse
|
17
|
Zhao Q, Ma L, Chen S, Huang L, She G, Sun Y, Shi W, Mu L. Tracking mitochondrial Cu(I) fluctuations through a ratiometric fluorescent probe in AD model cells: Towards understanding how AβOs induce mitochondrial Cu(I) dyshomeostasis. Talanta 2024; 271:125716. [PMID: 38301373 DOI: 10.1016/j.talanta.2024.125716] [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: 12/11/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/03/2024]
Abstract
Mitochondrial copper signaling pathway plays a role in Alzheimer's disease (AD), especially in relevant Amyloid-β oligomers (AβOs) neurotoxicity and mitochondrial dysfunction. Clarifying the relationship between mitochondrial copper homeostasis and both of mitochondrial dysfunction and AβOs neurotoxicity is important for understanding AD pathogenesis. Herein, we designed and synthesized a ratiometric fluorescent probe CHC-NS4 for Cu(I). CHC-NS4 possesses excellent ratiometric response, high selectivity to Cu(I) and specific ability to target mitochondria. Under mitochondrial dysfunction induced by oligomycin, mitochondrial Cu(I) levels gradually increased, which may be related to inhibition of ATP7A-mediated Cu(I) exportation and/or high expression of COX. On this basis, CHC-NS4 was further utilized to visualize the fluctuations of mitochondrial Cu(I) levels during progression of AD model cells induced by AβOs. It was found that mitochondrial Cu(I) levels were gradually elevated during the AD progression, which depended on not only AβOs concentration but also incubation time. Moreover, endocytosis maybe served as a prime pathway mode for mitochondrial Cu(I) dyshomeostasis induced by AβOs during AD progression. These results have provided a novel inspiration into mitochondrial copper biology in AD pathogenesis.
Collapse
Affiliation(s)
- Qiaowen Zhao
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liyi Ma
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Siwei Chen
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| | - Lushan Huang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guangwei She
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Yongan Sun
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| | - Wensheng Shi
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Lixuan Mu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
18
|
Chen Y, Wu Z, Li S, Chen Q, Wang L, Qi X, Tian C, Yang M. Mapping the Research of Ferroptosis in Parkinson's Disease from 2013 to 2023: A Scientometric Review. Drug Des Devel Ther 2024; 18:1053-1081. [PMID: 38585257 PMCID: PMC10999190 DOI: 10.2147/dddt.s458026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024] Open
Abstract
Methods Related studies on PD and ferroptosis were searched in Web of Science Core Collection (WOSCC) from inception to 2023. VOSviewer, CiteSpace, RStudio, and Scimago Graphica were employed as bibliometric analysis tools to generate network maps about the collaborations between authors, countries, and institutions and to visualize the co-occurrence and trends of co-cited references and keywords. Results A total of 160 original articles and reviews related to PD and ferroptosis were retrieved, produced by from 958 authors from 162 institutions. Devos David was the most prolific author, with 9 articles. China and the University of Melbourne had leading positions in publication volume with 84 and 12 publications, respectively. Current hot topics focus on excavating potential new targets for treating PD based on ferroptosis by gaining insight into specific molecular mechanisms, including iron metabolism disorders, lipid peroxidation, and imbalanced antioxidant regulation. Clinical studies aimed at treating PD by targeting ferroptosis remain in their preliminary stages. Conclusion A continued increase was shown in the literature within the related field over the past decade. The current study suggested active collaborations among authors, countries, and institutions. Research into the pathogenesis and treatment of PD based on ferroptosis has remained a prominent topic in the field in recent years, indicating that ferroptosis-targeted therapy is a potential approach to halting the progression of PD.
Collapse
Affiliation(s)
- Yingfan Chen
- Medical School of Chinese People’s Liberation Army, Beijing, People’s Republic of China
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Zhenhui Wu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, People’s Republic of China
| | - Shaodan Li
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Qi Chen
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Liang Wang
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Xiaorong Qi
- Medical School of Chinese People’s Liberation Army, Beijing, People’s Republic of China
| | - Chujiao Tian
- Medical School of Chinese People’s Liberation Army, Beijing, People’s Republic of China
| | - Minghui Yang
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
19
|
Chhillar B, Kadian R, Kumar M, Yadav M, Sodhi N, Xavier da Silva TN, Friedmann Angeli JP, Singh VP. Aminic Organoselenium Compounds as Glutathione Peroxidase Mimics and Inhibitors of Ferroptosis. Chembiochem 2024; 25:e202400074. [PMID: 38293899 DOI: 10.1002/cbic.202400074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/03/2023] [Accepted: 01/29/2024] [Indexed: 02/01/2024]
Abstract
The synthesis of diarylamine-based organoselenium compounds via the nucleophilic substitution reactions has been described. Symmetrical monoselenides and diselenides were conveniently synthesized by the reduction of their corresponding selenocyanates using sodium borohydride. Selenocyanates were obtained from 2-chloro acetamides by the nucleophilic displacement with potassium selenocyanate. Selenides were synthesized by treating the 2-chloro acetamides with in situ generated sodium butyl selenolate as nucleophile. Further, the newly synthesized organoselenium compounds were evaluated for their glutathione peroxidase (GPx)-like activity in thiophenol assay. This study revealed that the methoxy-substituted organoselenium compounds showed significant effect on the GPx-like activity. The catalytic parameters for the most efficient catalysts were also determined. The anti-ferroptotic activity for all GPx-mimics evaluated in a 4-OH-tamoxifen (TAM) inducible GPx4 knockout cell line using liproxstatin as standard.
Collapse
Affiliation(s)
- Babli Chhillar
- Department of Chemistry & Centre of Advanced Studies in Chemistry, Panjab University, Sector-14, Chandigarh, 160 014, India
| | - Rajni Kadian
- Department of Chemistry & Centre of Advanced Studies in Chemistry, Panjab University, Sector-14, Chandigarh, 160 014, India
| | - Manish Kumar
- Department of Chemistry & Centre of Advanced Studies in Chemistry, Panjab University, Sector-14, Chandigarh, 160 014, India
| | - Manisha Yadav
- Department of Chemistry & Centre of Advanced Studies in Chemistry, Panjab University, Sector-14, Chandigarh, 160 014, India
| | - Nikhil Sodhi
- Department of Chemistry & Centre of Advanced Studies in Chemistry, Panjab University, Sector-14, Chandigarh, 160 014, India
| | - Thamara Nishida Xavier da Silva
- Rudolf Virchow Zentrum, Centre for Integrative and Translational Bioimaging, Julius-Maximillian, University of Wurzburg, 97080, Wurzburg, Germany
| | - Jose Pedro Friedmann Angeli
- Rudolf Virchow Zentrum, Centre for Integrative and Translational Bioimaging, Julius-Maximillian, University of Wurzburg, 97080, Wurzburg, Germany
| | - Vijay P Singh
- Department of Chemistry & Centre of Advanced Studies in Chemistry, Panjab University, Sector-14, Chandigarh, 160 014, India
| |
Collapse
|
20
|
Liddell JR, Hilton JBW, Kysenius K, Billings JL, Nikseresht S, McInnes LE, Hare DJ, Paul B, Mercer SW, Belaidi AA, Ayton S, Roberts BR, Beckman JS, McLean CA, White AR, Donnelly PS, Bush AI, Crouch PJ. Microglial ferroptotic stress causes non-cell autonomous neuronal death. Mol Neurodegener 2024; 19:14. [PMID: 38317225 PMCID: PMC10840184 DOI: 10.1186/s13024-023-00691-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 11/28/2023] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Ferroptosis is a form of regulated cell death characterised by lipid peroxidation as the terminal endpoint and a requirement for iron. Although it protects against cancer and infection, ferroptosis is also implicated in causing neuronal death in degenerative diseases of the central nervous system (CNS). The precise role for ferroptosis in causing neuronal death is yet to be fully resolved. METHODS To elucidate the role of ferroptosis in neuronal death we utilised co-culture and conditioned medium transfer experiments involving microglia, astrocytes and neurones. We ratified clinical significance of our cell culture findings via assessment of human CNS tissue from cases of the fatal, paralysing neurodegenerative condition of amyotrophic lateral sclerosis (ALS). We utilised the SOD1G37R mouse model of ALS and a CNS-permeant ferroptosis inhibitor to verify pharmacological significance in vivo. RESULTS We found that sublethal ferroptotic stress selectively affecting microglia triggers an inflammatory cascade that results in non-cell autonomous neuronal death. Central to this cascade is the conversion of astrocytes to a neurotoxic state. We show that spinal cord tissue from human cases of ALS exhibits a signature of ferroptosis that encompasses atomic, molecular and biochemical features. Further, we show the molecular correlation between ferroptosis and neurotoxic astrocytes evident in human ALS-affected spinal cord is recapitulated in the SOD1G37R mouse model where treatment with a CNS-permeant ferroptosis inhibitor, CuII(atsm), ameliorated these markers and was neuroprotective. CONCLUSIONS By showing that microglia responding to sublethal ferroptotic stress culminates in non-cell autonomous neuronal death, our results implicate microglial ferroptotic stress as a rectifiable cause of neuronal death in neurodegenerative disease. As ferroptosis is currently primarily regarded as an intrinsic cell death phenomenon, these results introduce an entirely new pathophysiological role for ferroptosis in disease.
Collapse
Affiliation(s)
- Jeffrey R Liddell
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - James B W Hilton
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Kai Kysenius
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jessica L Billings
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Sara Nikseresht
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Lachlan E McInnes
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Dominic J Hare
- Atomic Medicine Initiative, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Bence Paul
- School of Earth Science, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Stephen W Mercer
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Abdel A Belaidi
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Scott Ayton
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Blaine R Roberts
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Joseph S Beckman
- Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA
| | - Catriona A McLean
- Anatomical Pathology, Alfred Hospital, Melbourne, VIC, 3005, Australia
| | - Anthony R White
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Ashley I Bush
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Peter J Crouch
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
21
|
Zhou YC, Zhao TK, Tao SM, Wang P, Guan YC, Yang KP, Chen SQ, Pu XY. Recent Progress in Ferroptosis Induced Tumor Cell Death by Anti-tumor Metallic complexes. Chem Asian J 2024; 19:e202301020. [PMID: 38149729 DOI: 10.1002/asia.202301020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/21/2023] [Accepted: 12/25/2023] [Indexed: 12/28/2023]
Abstract
Metal complexes represented by platinum complexes play a very important role in cancer treatment due to their diverse chemical structures and anti-tumor activities. Recently, ferroptosis has emerged as a newly occurring cell death form in the anti-tumor process. It has been reported that metal complexes could inhibit the proliferation and metastasis of tumors and combat chemotherapy resistance by targeting ferroptosis. In this review, we briefly describe ferroptosis as a fundamental process for tumor suppression and triggering anti-tumor immune responses. We summarize recent developments on metal complexes that induce ferroptosis. Finally, we outline the prospects for the application of metal complexes to the treatment of tumors based on ferroptosis and the associated problems that need to be solved, and discussed other potential research directions of metal complexes.
Collapse
Affiliation(s)
- Yong-Chang Zhou
- College of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730050, P.R. China
| | - Tian-Kun Zhao
- College of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730050, P.R. China
| | - Si-Man Tao
- College of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730050, P.R. China
| | - Peng Wang
- School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Yi-Chen Guan
- College of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730050, P.R. China
| | - Ke-Pei Yang
- College of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730050, P.R. China
| | - Sheng-Qiang Chen
- College of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730050, P.R. China
| | - Xiu-Ying Pu
- College of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730050, P.R. China
| |
Collapse
|
22
|
Wu Z, Vlaming R, Donohoe M, Pratt DA. Interrupted Homolytic Substitution Enables Organoboron Compounds to Inhibit Radical Chain Reactions Rather than Initiate Them. J Am Chem Soc 2024; 146:1153-1166. [PMID: 38156607 DOI: 10.1021/jacs.3c12438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
The reactions of organoboranes with peroxyl radicals are key to their use as radical initiators for a vast array of radical chain reactions, particularly at low temperatures where high stereoselectivity or regioselectivity is desired. Whereas these reactions generally proceed via concerted homolytic substitution (SH2) mechanisms, organoboranes that bear groups that can stabilize tetracoordinate boron radical "ate" complexes (e.g., catecholboranes) undergo this reaction via a stepwise addition/fragmentation sequence and serve as useful stoichiometric alkyl radical precursors. Here we show that arylboronic esters and amides derived from catecholborane and diaminonaphthaleneborane, respectively, are potent radical-trapping antioxidants (RTAs). Mechanistic studies reveal that this is because the radical "ate" complexes derived from peroxyl radical addition to boron are sufficiently persistent to trap another radical in an interrupted SH2 reaction. Remarkably, the reactivity of these organoboranes as inhibitors of autoxidation was shown to translate from simple hydrocarbons to the phospholipids of biological membranes such that they can inhibit ferroptosis, the cell death modality driven by lipid autoxidation and relevant in neurodegeneration and other major pathologies. The unique mechanism of these organoboranes is one of only a handful of RTA mechanisms that are not based on H-atom transfer processes and provide a new dimension to boron chemistry and its applications.
Collapse
Affiliation(s)
- Zijun Wu
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Robynne Vlaming
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Michael Donohoe
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Derek A Pratt
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
23
|
Ying D, Shen X, Wang S, Chen J, Wu Z, Chen W, Wang F, Min J, Yu Y. Discovery of 4-hydroxyl pyrazole derivatives as potent ferroptosis inhibitors. Eur J Med Chem 2024; 263:115913. [PMID: 37950965 DOI: 10.1016/j.ejmech.2023.115913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/11/2023] [Accepted: 10/24/2023] [Indexed: 11/13/2023]
Abstract
Ferroptosis, an iron-dependent form of regulated cell death, has been well recognized as a pathogenic mechanism in driving many diseases, such as neurodegenerative disorders, ischemia-reperfusion (I/R) injury. Blocking ferroptosis has been emerging as a feasible therapeutic strategy for the prevention and treatment of these diseases. However, novel potent ferroptosis inhibitors remain to be developed for further clinical applications. In this study, we screened our in-house compound libraries by phenotypic assays and identified a 4-hydroxyl pyrazole derivative HW-3 with good ferroptosis inhibitory activity (EC50 = 120.1 ± 3.5 nM). Based on the structure of HW-3, a series of 4-hydroxyl pyrazole derivatives were further designed and synthesized. Among these compounds, compound 25 could significantly inhibit RSL3-induced ferroptosis with an EC50 value of 8.6 ± 2.2 nM in HT-1080 cells, which was 3-fold more potent than the classical ferroptosis inhibitor ferrostatin-1 (Fer-1) (EC50 = 23.4 ± 1.3 nM). The potent ferroptosis inhibitory activity of compound 25 was further validated in multiple additional cell lines. Our mechanistic study revealed that compound 25 inhibited ferroptosis via intrinsic radical-trapping antioxidative capacity. Taken together, the findings of our study demonstrate 4-hydroxyl pyrazole derivative 25 is a potent ferroptosis inhibitor, which holds a great therapeutic potential for further development.
Collapse
Affiliation(s)
- Danzhi Ying
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xin Shen
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Shuqi Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Junyi Chen
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zhenying Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wenteng Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Fudi Wang
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Junxia Min
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Yongping Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
24
|
Pope LE, Dixon SJ. Regulation of ferroptosis by lipid metabolism. Trends Cell Biol 2023; 33:1077-1087. [PMID: 37407304 PMCID: PMC10733748 DOI: 10.1016/j.tcb.2023.05.003] [Citation(s) in RCA: 113] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 07/07/2023]
Abstract
Ferroptosis is an iron-dependent lethal mechanism that can be activated in disease and is a proposed target for cancer therapy. Ferroptosis is defined by the overwhelming accumulation of membrane lipid peroxides. Ferroptotic lipid peroxidation is initiated on internal membranes and then appears at the plasma membrane, triggering lethal ion imbalances and membrane permeabilization. Sensitivity to ferroptosis is governed by the levels of peroxidizable polyunsaturated lipids and associated lipid metabolic enzymes. A different network of enzymes and endogenous metabolites restrains lipid peroxidation by interfering with the initiation or propagation of this process. This emerging understanding is informing new approaches to treat disease by modulating lipid metabolism to enhance or inhibit ferroptosis.
Collapse
Affiliation(s)
- Lauren E Pope
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
25
|
Scarpellini C, Klejborowska G, Lanthier C, Hassannia B, Vanden Berghe T, Augustyns K. Beyond ferrostatin-1: a comprehensive review of ferroptosis inhibitors. Trends Pharmacol Sci 2023; 44:902-916. [PMID: 37770317 DOI: 10.1016/j.tips.2023.08.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/24/2023] [Accepted: 08/26/2023] [Indexed: 09/30/2023]
Abstract
Ferroptosis is an iron-catalysed form of regulated cell death, which is critically dependent on phospholipid peroxidation of cellular membranes. Ferrostatin 1 was one of the first synthetic radical-trapping antioxidants (RTAs) reported to block ferroptosis and it is widely used as reference compound. Ferroptosis has been linked to multiple diseases and the use of its inhibitors could have therapeutic potential. Although, novel biochemical pathways provide insights for different pharmacological targets, the use of lipophilic RTAs to block ferroptosis remains superior. In this Review, we provide a comprehensive overview of the different classes of ferroptosis inhibitors, focusing on endogenous and synthetic RTAs. A thorough analysis of their chemical, pharmacokinetic, and pharmacological properties and potential for in vivo use is provided.
Collapse
Affiliation(s)
- Camilla Scarpellini
- Laboratory of Medicinal Chemistry, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Greta Klejborowska
- Laboratory of Medicinal Chemistry, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Caroline Lanthier
- Laboratory of Medicinal Chemistry, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Behrouz Hassannia
- Ferroptosis and Inflammation Research Team, VIB-UGent Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium; Pathophysiology Lab, Department of Biomedical Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Tom Vanden Berghe
- Ferroptosis and Inflammation Research Team, VIB-UGent Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium; Pathophysiology Lab, Department of Biomedical Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Koen Augustyns
- Laboratory of Medicinal Chemistry, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium.
| |
Collapse
|
26
|
Zhang D, Jia X, Lin D, Ma J. Melatonin and ferroptosis: Mechanisms and therapeutic implications. Biochem Pharmacol 2023; 218:115909. [PMID: 37931663 DOI: 10.1016/j.bcp.2023.115909] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/08/2023]
Abstract
Ferroptosis, a regulated form of cell death, is characterized by iron-dependent lipid peroxidation leading to oxidative damage to cell membranes. Cell sensitivity to ferroptosis is influenced by factors such as iron overload, lipid metabolism, and the regulation of the antioxidant system. Melatonin, with its demonstrated capacity to chelate iron, modulate iron metabolism proteins, regulate lipid peroxidation, and regulate antioxidant systems, has promise as a potential therapeutic agent in mediating ferroptosis. The availability of approved drugs targeting ferroptosis is limited; therefore, melatonin is a candidate for broad application due to its safety and efficacy in attenuating ferroptosis in noncancerous diseases. Melatonin has been demonstrated to attenuate ferroptosis in cellular and animal models of noncancerous diseases, showcasing effectiveness in organs such as the heart, brain, lung, liver, kidney, and bone. This review outlines the molecular mechanisms of ferroptosis, investigates melatonin's potential effects on ferroptosis, and discusses melatonin's therapeutic potential as a promising intervention against diseases associated with ferroptosis. Through this discourse, we aim to lay a strong foundation for developing melatonin as a therapeutic strategy to modulate ferroptosis in a variety of disease contexts.
Collapse
Affiliation(s)
- Dongni Zhang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Xiaotong Jia
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.
| | - Duomao Lin
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.
| | - Jun Ma
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.
| |
Collapse
|
27
|
Ryan SK, Ugalde CL, Rolland AS, Skidmore J, Devos D, Hammond TR. Therapeutic inhibition of ferroptosis in neurodegenerative disease. Trends Pharmacol Sci 2023; 44:674-688. [PMID: 37657967 DOI: 10.1016/j.tips.2023.07.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 09/03/2023]
Abstract
Iron accumulation has been associated with the etiology and progression of multiple neurodegenerative diseases (NDDs). The exact role of iron in these diseases is not fully understood, but an iron-dependent form of regulated cell death called ferroptosis could be key. Although there is substantial preclinical and clinical evidence that ferroptosis plays a role in NDD, there are still questions regarding how to target ferroptosis therapeutically, including which proteins to target, identification of clinically relevant biomarkers, and which patients might benefit most. Clinical trials of iron- and ferroptosis-targeted therapies are beginning to provide some answers, but there is growing interest in developing new ferroptosis inhibitors. We describe newly identified ferroptosis targets, opportunities, and challenges in NDD, as well as key considerations for progressing new therapeutics to the clinic.
Collapse
Affiliation(s)
- Sean K Ryan
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA, USA
| | - Cathryn L Ugalde
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, UK
| | - Anne-Sophie Rolland
- Department of Medical Pharmacology, Expert Center of Parkinson's Disease, ALS, and Neurogenetics, University of Lille, LilNCog, Lille Neuroscience and Cognition, INSERM UMR S1172, CHU de Lille, LICEND, COEN, Center, NS-PARK Network, France
| | - John Skidmore
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, UK
| | - David Devos
- Department of Medical Pharmacology, Expert Center of Parkinson's Disease, ALS, and Neurogenetics, University of Lille, LilNCog, Lille Neuroscience and Cognition, INSERM UMR S1172, CHU de Lille, LICEND, COEN, Center, NS-PARK Network, France
| | | |
Collapse
|
28
|
Sun S, Shen J, Jiang J, Wang F, Min J. Targeting ferroptosis opens new avenues for the development of novel therapeutics. Signal Transduct Target Ther 2023; 8:372. [PMID: 37735472 PMCID: PMC10514338 DOI: 10.1038/s41392-023-01606-1] [Citation(s) in RCA: 108] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/24/2023] [Accepted: 08/11/2023] [Indexed: 09/23/2023] Open
Abstract
Ferroptosis is an iron-dependent form of regulated cell death with distinct characteristics, including altered iron homeostasis, reduced defense against oxidative stress, and abnormal lipid peroxidation. Recent studies have provided compelling evidence supporting the notion that ferroptosis plays a key pathogenic role in many diseases such as various cancer types, neurodegenerative disease, diseases involving tissue and/or organ injury, and inflammatory and infectious diseases. Although the precise regulatory networks that underlie ferroptosis are largely unknown, particularly with respect to the initiation and progression of various diseases, ferroptosis is recognized as a bona fide target for the further development of treatment and prevention strategies. Over the past decade, considerable progress has been made in developing pharmacological agonists and antagonists for the treatment of these ferroptosis-related conditions. Here, we provide a detailed overview of our current knowledge regarding ferroptosis, its pathological roles, and its regulation during disease progression. Focusing on the use of chemical tools that target ferroptosis in preclinical studies, we also summarize recent advances in targeting ferroptosis across the growing spectrum of ferroptosis-associated pathogenic conditions. Finally, we discuss new challenges and opportunities for targeting ferroptosis as a potential strategy for treating ferroptosis-related diseases.
Collapse
Affiliation(s)
- Shumin Sun
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Shen
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianwei Jiang
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Fudi Wang
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, The Second Affiliated Hospital, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
29
|
Li W, Li S, Zhang Z, Xu G, Man X, Yang F, Liang H. Developing a Multitargeted Anticancer Palladium(II) Agent Based on the His-242 Residue in the IIA Subdomain of Human Serum Albumin. J Med Chem 2023. [PMID: 37321208 DOI: 10.1021/acs.jmedchem.3c00248] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
To obtain next-generation metal drugs that can overcome the deficiencies of platinum (Pt) drugs and treat cancer more effectively, we proposed to develop a multitargeted palladium (Pd) agent to the tumor microenvironment (TME) based on the specific residue(s) of human serum albumin (HSA). To this end, we optimized a series of Pd(II) 2-benzoylpyridine thiosemicarbazone compounds to obtain a Pd agent (5b) with significant cytotoxicity. The HSA-5b complex structure revealed that 5b bound to the hydrophobic cavity in the HSA IIA subdomain and then His-242 replaced a leaving group (Cl) of 5b, coordinating with the Pd center. The in vivo results showed that the 5b/HSA-5b complex had significant capacity of inhibiting tumor growth, and HSA optimized the therapeutic behavior of 5b. In addition, we confirmed that the 5b/HSA-5b complex inhibited tumor growth through multiple actions on different components of TME: killing cancer cells, inhibiting tumor angiogenesis, and activating T cells.
Collapse
Affiliation(s)
- Wenjuan Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, Guangxi, China
| | - Shanhe Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, Guangxi, China
| | - Zhenlei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, Guangxi, China
| | - Gang Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, Guangxi, China
| | - Xueyu Man
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, Guangxi, China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, Guangxi, China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, Guangxi, China
| |
Collapse
|
30
|
Tong J, Lan XT, Zhang Z, Liu Y, Sun DY, Wang XJ, Ou-Yang SX, Zhuang CL, Shen FM, Wang P, Li DJ. Ferroptosis inhibitor liproxstatin-1 alleviates metabolic dysfunction-associated fatty liver disease in mice: potential involvement of PANoptosis. Acta Pharmacol Sin 2023; 44:1014-1028. [PMID: 36323829 PMCID: PMC10104837 DOI: 10.1038/s41401-022-01010-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 09/25/2022] [Indexed: 11/07/2022] Open
Abstract
Ferroptosis is a new form of regulated cell death characterized by excessive iron accumulation and uncontrollable lipid peroxidation. The role of ferroptosis in metabolic dysfunction-associated fatty liver disease (MAFLD) is not fully elucidated. In this study we compared the therapeutic effects of ferroptosis inhibitor liproxstatin-1 (LPT1) and iron chelator deferiprone (DFP) in MAFLD mouse models. This model was established in mice by feeding a high-fat diet with 30% fructose in water (HFHF) for 16 weeks. The mice then received LPT1 (10 mg·kg-1·d-1, ip) or DFP (100 mg·kg-1·d-1, ig) for another 2 weeks. We showed that both LPT1 and DFP treatment blocked the ferroptosis markers ACSL4 and ALOX15 in MAFLD mice. Furthermore, LPT1 treatment significantly reduced the liver levels of triglycerides and cholesterol, lipid peroxidation markers 4-hydroxynonenal (4-HNE) and malondialdehyde (MDA), and ameliorated the expression of lipid synthesis/oxidation genes (Pparα, Scd1, Fasn, Hmgcr and Cpt1a), insulin resistance, mitochondrial ROS content and liver fibrosis. Importantly, LPT1 treatment potently inhibited hepatic apoptosis (Bax/Bcl-xL ratio and TUNEL+ cell number), pyroptosis (cleavages of Caspase-1 and GSDMD) and necroptosis (phosphorylation of MLKL). Moreover, LPT1 treatment markedly inhibited cleavages of PANoptosis-related caspase-8 and caspase-6 in MAFLD mouse liver. In an in vitro MAFLD model, treatment with LPT1 (100 nM) prevented cultured hepatocyte against cell death induced by pro-PANoptosis molecules (TNF-α, LPS and nigericin) upon lipid stress. On the contrary, DFP treatment only mildly attenuated hepatic inflammation but failed to alleviate lipid deposition, insulin resistance, apoptosis, pyroptosis and necroptosis in MAFLD mice. We conclude that ferroptosis inhibitor LPT1 protects against steatosis and steatohepatitis in MAFLD mice, which may involve regulation of PANoptosis, a coordinated cell death pathway that involves apoptosis, pyroptosis and necroptosis. These results suggest a potential link between ferroptosis and PANoptosis.
Collapse
Affiliation(s)
- Jie Tong
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Xiu-Ting Lan
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Zhen Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yi Liu
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Di-Yang Sun
- School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, 200433, China
| | - Xu-Jie Wang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Shen-Xi Ou-Yang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Chun-Lin Zhuang
- School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, 200433, China
| | - Fu-Ming Shen
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Pei Wang
- School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, 200433, China.
| | - Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200072, China.
| |
Collapse
|
31
|
Jiang X, Wu K, Ye XY, Xie T, Zhang P, Blass BE, Bai R. Novel druggable mechanism of Parkinson's disease: Potential therapeutics and underlying pathogenesis based on ferroptosis. Med Res Rev 2023. [PMID: 36924451 DOI: 10.1002/med.21939] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/07/2023] [Accepted: 02/26/2023] [Indexed: 03/18/2023]
Abstract
Genetics, age, environmental factors, and oxidative stress have all been implicated in the development of Parkinson's disease (PD); however, a complete understanding of its pathology remains elusive. At present, there is no cure for PD, and currently available therapeutics are insufficient to meet patient needs. Ferroptosis, a distinctive iron-dependent cell death mode characterized by lipid peroxidation and oxidative stress, has pathophysiological features similar to those of PD, including iron accumulation, reactive oxygen species-induced oxidative damage, and mitochondrial dysfunction. Ferroptosis has been identified as a specific pathway of neuronal death and is closely related to the pathogenesis of PD. Despite the similarities in the biological targets involved in PD pathogenesis and ferroptosis, the relationship between novel targets in PD and ferroptosis has been neglected in the literature. In this review, the mechanism of ferroptosis is discussed, and the potential therapeutic targets implicated in both PD and ferroptosis are compared. Furthermore, the anti-PD effects of several ferroptosis inhibitors, as well as clinical studies thereof, and the identification of novel lead compounds for the treatment of PD and the inhibition of ferroptosis are reviewed. It is hoped that this review can promote research to further elucidate the relationship between ferroptosis and PD and provide new strategies for the development of novel ferroptosis-targeting PD therapy.
Collapse
Affiliation(s)
- Xiaoying Jiang
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, People's Republic of China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, People's Republic of China.,Department of Chemistry, College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology, Hangzhou Normal University, Hangzhou, People's Republic of China
| | - Kaiyu Wu
- Department of Chemistry, College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology, Hangzhou Normal University, Hangzhou, People's Republic of China
| | - Xiang-Yang Ye
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, People's Republic of China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, People's Republic of China
| | - Tian Xie
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, People's Republic of China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, People's Republic of China
| | - Pengfei Zhang
- Department of Chemistry, College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology, Hangzhou Normal University, Hangzhou, People's Republic of China
| | - Benjamin E Blass
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, Pennsylvania, USA
| | - Renren Bai
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, People's Republic of China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, People's Republic of China
| |
Collapse
|
32
|
Morrow JP, Pizzi D, Mazrad ZAI, Bush AI, Kempe K. Bioactive poly(2-oxazoline)-based nanomaterials bearing arylalkylamine and benzamide motifs possess intrinsic radical trapping and anti-ferroptosis properties. Biomater Sci 2023; 11:3159-3171. [PMID: 36919797 DOI: 10.1039/d2bm02087d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Radical trapping agents such as Ferrostatin-1 (Fer-1) are capable of rescuing cells from ferroptosis, an iron-dependent form of cell death. Previously, poly(2-oxazoline)-Fer-1 (POx-Fer-1) conjugates were reported, which possess increased water-solubility and remain active after covalent conjugation of Fer-1. In this study, we break down the structural and functional layers of POx-Fer-1 conjugates and reveal that drug-free POx containing arylalkylamine and benzamide motifs show anti-ferroptosis properties. Intriguingly, even the basic construct poly(2-methyl-2-oxazoline-grad-2-phenyl-2-oxazoline) P(MeOx-grad-PhOx) was found to be active. Therefore, P(MeOx-grad-PhOx) of varying compositions were prepared, characterized by 1H NMR spectroscopy and size exclusion chromatography and investigated with regard to their self-assembly in aqueous solution and activity in an in vitro ferroptosis model. These findings were further explored for the design of defined and bioactive core-crosslinked micelles with intrinsic anti-ferroptosis behaviour. Cellular interaction studies involving C11-BODIPY assays and confocal microscopy investigations revealed lysosomal processing of the nanomaterials and perturbation of ferroptotic cell death through reducing lipid-peroxidation. This study highlights new drug/cargo-free anti-ferroptotic nanomaterials as proof of concept that hold potential for therapy of ferroptosis-associated diseases and highlights the role of nanocarriers in a therapeutic context.
Collapse
Affiliation(s)
- Joshua P Morrow
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - David Pizzi
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Zihnil A I Mazrad
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Kristian Kempe
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia.,Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
33
|
Ji QX, Zeng FY, Zhou J, Wu WB, Wang XJ, Zhang Z, Zhang GY, Tong J, Sun DY, Zhang JB, Cao WX, Shen FM, Lu JJ, Li DJ, Wang P. Ferroptotic stress facilitates smooth muscle cell dedifferentiation in arterial remodelling by disrupting mitochondrial homeostasis. Cell Death Differ 2023; 30:457-474. [PMID: 36477078 PMCID: PMC9950429 DOI: 10.1038/s41418-022-01099-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Smooth muscle cell (SMC) phenotypic switch from a quiescent 'contractile' phenotype to a dedifferentiated and proliferative state underlies the development of cardiovascular diseases (CVDs); however, our understanding of the mechanism is still incomplete. In the present study, we explored the potential role of ferroptosis, a novel nonapoptotic form of cell death, in SMC phenotypic switch and related neointimal formation. We found that ferroptotic stress was triggered in cultured dedifferentiated SMCs and arterial neointimal tissue of wire-injured mice. Moreover, pro-ferroptosis stress was activated in arterial neointimal tissue of clinical patients who underwent carotid endarterectomy. Blockade of ferroptotic stress via administration of a pharmacological inhibitor or by global genetic overexpression of glutathione peroxidase-4 (GPX4), a well-established anti-ferroptosis molecule, delayed SMC phenotype switch and arterial remodelling. Conditional SMC-specific gene delivery of GPX4 using adreno-associated virus in the carotid artery inhibited ferroptosis and prevented neointimal formation. Conversely, ferroptosis stress directly triggered dedifferentiation of SMCs. Transcriptomics analysis demonstrated that inhibition of ferroptotic stress mainly targets the mitochondrial respiratory chain and oxidative phosphorylation. Mechanistically, ferroptosis inhibition corrected the disrupted mitochondrial homeostasis in dedifferentiated SMCs, including enhanced mitochondrial ROS production, dysregulated mitochondrial dynamics, and mitochondrial hyperpolarization, and ultimately inhibited SMC phenotypic switch and growth. Copper-diacetyl-bisN4-methylthiosemicarbazone (CuATSM), an agent used for clinical molecular imaging and that potently inhibits ferroptosis, prevented SMC phenotypic switch, neointimal formation and arterial inflammation in mice. These results indicate that pro-ferroptosis stress is likely to promote SMC phenotypic switch during neointimal formation and imply that inhibition of ferroptotic stress may be a promising translational approach to treat CVDs with SMC phenotype switch.
Collapse
Affiliation(s)
- Qing-Xin Ji
- Department of Pharmacology, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fei-Yan Zeng
- Department of Pharmacology, Shanghai Forth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jian Zhou
- Department of Cardiac Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen-Bin Wu
- Department of Pharmacology, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Xu-Jie Wang
- Department of Pharmacology, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhen Zhang
- Department of Pharmacology, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guo-Yan Zhang
- Department of Pharmacology, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jie Tong
- Department of Pharmacology, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Di-Yang Sun
- Department of Pharmacology, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Jia-Bao Zhang
- Department of Pharmacology, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Wen-Xiang Cao
- Department of Pharmacology, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Fu-Ming Shen
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China.
| | - Pei Wang
- Department of Pharmacology, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China.
| |
Collapse
|
34
|
Wu Z, Sun J, Liao Z, Qiao J, Chen C, Ling C, Wang H. An update on the therapeutic implications of long-chain acyl-coenzyme A synthetases in nervous system diseases. Front Neurosci 2022; 16:1030512. [PMID: 36507355 PMCID: PMC9731139 DOI: 10.3389/fnins.2022.1030512] [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: 08/29/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Long-chain acyl-coenzyme A synthetases (ACSLs) are a family of CoA synthetases that activate fatty acid (FA) with chain lengths of 12-20 carbon atoms by forming the acyl-AMP derivative in an isozyme-specific manner. This family mainly includes five members (ACSL1, ACSL3, ACSL4, ACSL5, and ACSL6), which are thought to have specific and different functions in FA metabolism and oxidative stress of mammals. Accumulating evidence shows that the dysfunction of ACSLs is likely to affect cell proliferation and lead to metabolic diseases in multiple organs and systems through different signaling pathways and molecular mechanisms. Hence, a central theme of this review is to emphasize the therapeutic implications of ACSLs in nervous system disorders.
Collapse
Affiliation(s)
- Zhimin Wu
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jun Sun
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhi Liao
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jia Qiao
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chuan Chen
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Cong Ling
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hui Wang
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China,*Correspondence: Hui Wang,
| |
Collapse
|
35
|
Xi J, Zhang Z, Wang Z, Wu Q, He Y, Xu Y, Ding Z, Zhao H, Da H, Zhang F, Zhao H, Fang J. Hinokitiol functions as a ferroptosis inhibitor to confer neuroprotection. Free Radic Biol Med 2022; 190:202-215. [PMID: 35985562 DOI: 10.1016/j.freeradbiomed.2022.08.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 12/22/2022]
Abstract
The intrinsic link of ferroptosis to neurodegeneration, such as Parkinson's disease and Alzheimer's disease, has set promises to apply ferroptosis inhibitors for treatment of neurodegenerative disorders. Herein, we report that the natural small molecule hinokitiol (Hino) functions as a potent ferroptosis inhibitor to rescue neuronal damages in vitro and in vivo. The action mechanisms of Hino involve chelating irons and activating cytoprotective transcription factor Nrf2 to upregulate the antioxidant genes including solute carrier family 7 member 11, glutathione peroxidase 4 and Heme oxygenase-1. In vivo studies demonstrate that Hino rescues the deficits of locomotor activity and neurodevelopment in zebrafishes. In addition, Hino shows the efficient blood-brain barrier permeability in mice, supporting the application of Hino for brain disorders. Paclitaxel is one of the most widely used broad-spectrum antineoplastic agents. However, its neurotoxic side effect is a severe concern. We demonstrate that the neurotoxicity of paclitaxel is ferroptosis-related and Hino also alleviates the paclitaxel-induced neurotoxicity without compromising its cytotoxicity to cancer cells. Hino also salvages the neurobehavioral impairment by paclitaxel in zebrafishes. Collectively, the discovery of Hino as a novel ferroptosis inhibitor and disclosure of its action mechanisms establish a foundation for the further development of Hino as a neuroprotective agent.
Collapse
Affiliation(s)
- Junmin Xi
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China; School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, 730000, China
| | - Zhijun Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Zuo Wang
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, 730000, China
| | - Qingfeng Wu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Ying He
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Yanyi Xu
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, 730000, China
| | - Zhenjiang Ding
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Huanhuan Zhao
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Honghong Da
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Fang Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Haiyu Zhao
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, 730000, China.
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China; School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing, 210094, China.
| |
Collapse
|
36
|
Wu Z, Khodade VS, Chauvin JPR, Rodriguez D, Toscano JP, Pratt DA. Hydropersulfides Inhibit Lipid Peroxidation and Protect Cells from Ferroptosis. J Am Chem Soc 2022; 144:15825-15837. [PMID: 35977425 DOI: 10.1021/jacs.2c06804] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hydropersulfides (RSSH) are believed to serve important roles in vivo, including as scavengers of damaging oxidants and electrophiles. The α-effect makes RSSH not only much better nucleophiles than thiols (RSH), but also much more potent H-atom transfer agents. Since HAT is the mechanism of action of the most potent small-molecule inhibitors of phospholipid peroxidation and associated ferroptotic cell death, we have investigated their reactivity in this context. Using the fluorescence-enabled inhibited autoxidation (FENIX) approach, we have found RSSH to be highly reactive toward phospholipid-derived peroxyl radicals (kinh = 2 × 105 M-1 s-1), equaling the most potent ferroptosis inhibitors identified to date. Related (poly)sulfide products resulting from the rapid self-reaction of RSSH under physiological conditions (e.g., disulfide, trisulfide, H2S) are essentially unreactive, but combinations from which RSSH can be produced in situ (i.e., polysulfides with H2S or thiols with H2S2) are effective. In situ generation of RSSH from designed precursors which release RSSH via intramolecular substitution or hydrolysis improve the radical-trapping efficiency of RSSH by minimizing deleterious self-reactions. A brief survey of structure-reactivity relationships enabled the design of new precursors that are more efficient. The reactivity of RSSH and their precursors translates from (phospho)lipid bilayers to cell culture (mouse embryonic fibroblasts), where they were found to inhibit ferroptosis induced by inactivation of glutathione peroxidase-4 (GPX4) or deletion of the gene encoding it. These results suggest that RSSH and the pathways responsible for their biosynthesis may act as a ferroptosis suppression system alongside the recently discovered FSP1/ubiquinone and GCH1/BH4/DHFR systems.
Collapse
Affiliation(s)
- Zijun Wu
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ONK1N 6N5, Canada
| | - Vinayak S Khodade
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland21218, United States
| | - Jean-Philippe R Chauvin
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ONK1N 6N5, Canada
| | - Deborah Rodriguez
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland21218, United States
| | - John P Toscano
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland21218, United States
| | - Derek A Pratt
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ONK1N 6N5, Canada
| |
Collapse
|
37
|
Sun Y, Xia X, Basnet D, Zheng JC, Huang J, Liu J. Mechanisms of Ferroptosis and Emerging Links to the Pathology of Neurodegenerative Diseases. Front Aging Neurosci 2022; 14:904152. [PMID: 35837484 PMCID: PMC9273851 DOI: 10.3389/fnagi.2022.904152] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/03/2022] [Indexed: 12/15/2022] Open
Abstract
Neurodegenerative diseases are a diverse class of diseases attributed to chronic progressive neuronal degeneration and synaptic loss in the brain and/or spinal cord, including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis and multiple sclerosis. The pathogenesis of neurodegenerative diseases is complex and diverse, often involving mitochondrial dysfunction, neuroinflammation, and epigenetic changes. However, the pathogenesis of neurodegenerative diseases has not been fully elucidated. Recently, accumulating evidence revealed that ferroptosis, a newly discovered iron-dependent and lipid peroxidation-driven type of programmed cell death, provides another explanation for the occurrence and progression of neurodegenerative diseases. Here, we provide an overview of the process and regulation mechanisms of ferroptosis, and summarize current research progresses that support the contribution of ferroptosis to the pathogenesis of neurodegenerative diseases. A comprehensive understanding of the emerging roles of ferroptosis in neurodegenerative diseases will shed light on the development of novel therapeutic technologies and strategies for slowing down the progression of these diseases.
Collapse
Affiliation(s)
- Yiyan Sun
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Xiaohuan Xia
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Shanghai Frontiers Science Center of Nanocatalytic Medicine, Shanghai, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Diksha Basnet
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Jialin C. Zheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Shanghai Frontiers Science Center of Nanocatalytic Medicine, Shanghai, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, China
- *Correspondence: Jialin C. Zheng,
| | - Jian Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
- Jian Huang,
| | - Jianhui Liu
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Jianhui Liu,
| |
Collapse
|
38
|
Du H, Ren X, Bai J, Yang W, Gao Y, Yan S. Research Progress of Ferroptosis in Adiposity-Based Chronic Disease (ABCD). OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1052699. [PMID: 35502211 PMCID: PMC9056228 DOI: 10.1155/2022/1052699] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/27/2022] [Accepted: 03/30/2022] [Indexed: 12/14/2022]
Abstract
Ferroptosis is a multistep regulated cell death process induced by iron accumulation and lipid peroxidation. Classical GPX4-dependent pathway and GPX4-independent pathways can independently and synergistically inhibit ferroptosis and jointly maintain the oxidative balance of the body. WHO defines obesity as "a condition of abnormal or excessive fat accumulation in adipose tissue, to the extent that health may be impaired," and obesity is also defined as an adiposity-based chronic disease (ABCD). Obesity is a systemic disease that leads to metabolic abnormalities in various systems, resulting in a series of complications including obesity cardiomyopathy, atherosclerosis, nonalcoholic fatty liver disease, and diabetes mellitus. Emerging evidence shows that ferroptosis is closely associated with the occurrence and progression of various diseases. In recent years, ferroptosis has been found to play critical roles in obesity and its complications. This review discusses the mechanisms of how ferroptosis is initiated and controlled and discusses the research progress of ferroptosis in obesity and its complications.
Collapse
Affiliation(s)
- Huijun Du
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang, China
| | - Xiaoying Ren
- Department of Endocrinology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang, China
| | - Juncai Bai
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang, China
| | - Wei Yang
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang, China
| | - Yunan Gao
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang, China
| | - Shuang Yan
- Department of Endocrinology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang, China
| |
Collapse
|