1
|
Gao Q, Zhou Y, Chen Y, Hu W, Jin W, Zhou C, Yuan H, Li J, Lin Z, Lin W. Role of iron in brain development, aging, and neurodegenerative diseases. Ann Med 2025; 57:2472871. [PMID: 40038870 PMCID: PMC11884104 DOI: 10.1080/07853890.2025.2472871] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 02/03/2025] [Accepted: 02/10/2025] [Indexed: 03/06/2025] Open
Abstract
It is now understood that iron crosses the blood-brain barrier via a complex metabolic regulatory network and participates in diverse critical biological processes within the central nervous system, including oxygen transport, energy metabolism, and the synthesis and catabolism of myelin and neurotransmitters. During brain development, iron is distributed throughout the brain, playing a pivotal role in key processes such as neuronal development, myelination, and neurotransmitter synthesis. In physiological aging, iron can selectively accumulate in specific brain regions, impacting cognitive function and leading to intracellular redox imbalance, mitochondrial dysfunction, and lipid peroxidation, thereby accelerating aging and associated pathologies. Furthermore, brain iron accumulation may be a primary contributor to neurodegenerative diseases such as Alzheimer's and Parkinson's diseases. Comprehending the role of iron in brain development, aging, and neurodegenerative diseases, utilizing iron-sensitive Magnetic Resonance Imaging (MRI) technology for timely detection or prediction of abnormal neurological states, and implementing appropriate interventions may be instrumental in preserving normal central nervous system function.
Collapse
Affiliation(s)
- Qiqi Gao
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yiyang Zhou
- Department of Urology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Yu Chen
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Hu
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenwen Jin
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chunting Zhou
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hao Yuan
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianshun Li
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhenlang Lin
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Lin
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
2
|
Mavaddatiyan L, Naeini S, Khodabandeh S, Hosseini F, Skelton RP, Azizi V, Talkhabi M. Exploring the association between aging, ferroptosis, and common age-related diseases. Arch Gerontol Geriatr 2025; 135:105877. [PMID: 40339241 DOI: 10.1016/j.archger.2025.105877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 04/14/2025] [Accepted: 04/26/2025] [Indexed: 05/10/2025]
Abstract
Aging is a natural biological process that is characterized by the progressive decline in physiological functions and an increased vulnerability to age-related diseases. The aging process is driven by different cell and molecular mechanisms. It has recently been shown that aging is associated with heightened vulnerability to ferroptosis (an intracellular iron-dependent form of programmed cell death). This susceptibility arises from various factors including oxidative stress, impaired antioxidant defences, and dysregulated iron homeostasis. The progressive decline in cellular antioxidant capacity and the accumulation of damaged components contribute to the increased susceptibility of aging cells to ferroptosis. Dysregulation of key regulators involved in ferroptosis, such as glutathione peroxidase 4 (GPX4), iron regulatory proteins, and lipid metabolism enzymes, further exacerbates this vulnerability. The decline in cellular defence mechanisms against ferroptosis during aging contributes to the accumulation of damaged cells and tissues, ultimately resulting in the manifestation of age-related diseases. Understanding the intricate relevance between aging and ferroptosis holds significant potential for developing strategies to counteract the detrimental effects of aging and age-related diseases. This will subsequently act to mitigate the negative consequences of aging and improving overall health in the elderly population. This review aims to clarify the relationship between aging and ferroptosis, and explores the underlying mechanisms and implications for age-related disorders, including neurodegenerative, cardiovascular, and neoplastic diseases. We also discuss the accumulating evidence suggesting that the imbalance of redox homeostasis and perturbations in iron metabolism contribute to the age-associated vulnerability to ferroptosis.
Collapse
Affiliation(s)
- Laleh Mavaddatiyan
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - SaghiHakimi Naeini
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Sara Khodabandeh
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Fatemeh Hosseini
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - RhysJ P Skelton
- Flinders Medical Centre, Department of Ophthalmology, Bedford Park, Australia
| | - Vahid Azizi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Mahmood Talkhabi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
3
|
Zeng H, Jin Z. The role of ferroptosis in Alzheimer's disease: Mechanisms and therapeutic potential (Review). Mol Med Rep 2025; 32:192. [PMID: 40341407 PMCID: PMC12076055 DOI: 10.3892/mmr.2025.13557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/27/2025] [Indexed: 05/10/2025] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by insidious onset and progressive symptom deterioration. It extends beyond a simple aging process, involving irreversible and progressive neurological degeneration that impairs brain function through multiple etiologies. Iron dysregulation is implicated in the pathophysiology of AD; however, the precise mechanisms remain unclear. Additionally, vitamin E and selenium are key in regulating ferroptosis through their antioxidant properties. The present review examined the mechanistic pathways by which ferroptosis contributes to AD, the regulatory roles of vitamin E, selenium, ferrostatin‑1, N‑acetylcysteine and curcumin, and their potential as therapeutic agents to mitigate neurodegeneration.
Collapse
Affiliation(s)
- Heng Zeng
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhaohui Jin
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
4
|
Lin Z, Ying C, Si X, Xue N, Liu Y, Zheng R, Chen Y, Pu J, Zhang B. NOX4 exacerbates Parkinson's disease pathology by promoting neuronal ferroptosis and neuroinflammation. Neural Regen Res 2025; 20:2038-2052. [PMID: 38993139 PMCID: PMC11691449 DOI: 10.4103/nrr.nrr-d-23-01265] [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: 07/29/2023] [Revised: 11/18/2023] [Accepted: 03/18/2024] [Indexed: 07/13/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202507000-00026/figure1/v/2024-09-09T124005Z/r/image-tiff Parkinson's disease is primarily caused by the loss of dopaminergic neurons in the substantia nigra compacta. Ferroptosis, a novel form of regulated cell death characterized by iron accumulation and lipid peroxidation, plays a vital role in the death of dopaminergic neurons. However, the molecular mechanisms underlying ferroptosis in dopaminergic neurons have not yet been completely elucidated. NADPH oxidase 4 is related to oxidative stress, however, whether it regulates dopaminergic neuronal ferroptosis remains unknown. The aim of this study was to determine whether NADPH oxidase 4 is involved in dopaminergic neuronal ferroptosis, and if so, by what mechanism. We found that the transcriptional regulator activating transcription factor 3 increased NADPH oxidase 4 expression in dopaminergic neurons and astrocytes in an 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine-induced Parkinson's disease model. NADPH oxidase 4 inhibition improved the behavioral impairments observed in the Parkinson's disease model animals and reduced the death of dopaminergic neurons. Moreover, NADPH oxidase 4 inhibition reduced lipid peroxidation and iron accumulation in the substantia nigra of the Parkinson's disease model animals. Mechanistically, we found that NADPH oxidase 4 interacted with activated protein kinase C α to prevent ferroptosis of dopaminergic neurons. Furthermore, by lowering the astrocytic lipocalin-2 expression, NADPH oxidase 4 inhibition reduced 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine-induced neuroinflammation. These findings demonstrate that NADPH oxidase 4 promotes ferroptosis of dopaminergic neurons and neuroinflammation, which contribute to dopaminergic neuron death, suggesting that NADPH oxidase 4 is a possible therapeutic target for Parkinson's disease.
Collapse
Affiliation(s)
- Zhihao Lin
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Changzhou Ying
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Xiaoli Si
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Naijia Xue
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yi Liu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Ran Zheng
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Ying Chen
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jiali Pu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Baorong Zhang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
5
|
Zhang J, Cai R, Ning C, Zhou Z, Zhang Y, Wang S, Li Y, Guo X. A novel integrated diagnostic and therapeutic ferroptosis inhibitor based on a phenothiazine scaffold with ROS-Responsive strategy. Bioorg Med Chem 2025; 124:118195. [PMID: 40252566 DOI: 10.1016/j.bmc.2025.118195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/21/2025]
Abstract
Ferroptosis is a newly discovered form of cell death that is closely related to the occurrence of various diseases, such as neurodegenerative diseases, cardiovascular and cerebrovascular ischemic damage, and organ fibrosis. Therefore, the discovery of new active compounds with ferroptosis inhibitory activity is regarded as a new strategy for the clinical treatment of these diseases. In this study, a multifunctional prodrug molecule PNX-B2 with a phenoxazine structure was designed based on the oxidative microenvironment characteristic of ferroptosis. PNX-B2 can recognize the ferroptosis-associated oxidative conditions and simultaneously release compounds with ferroptosis-inhibitory activity. Moreover, it integrates diagnostic and therapeutic functions and offers a fluorescent indication of the ferroptosis microenvironment. PNX-B2 has demonstrated excellent ferroptosis-inhibitory activity with an EC50 value of 1.7 nM. This intelligent multifunctional compound shows great potential as a novel clinical agent for ferroptosis inhibition and presents broad prospects for future development.
Collapse
Affiliation(s)
- Jiangye Zhang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Rui Cai
- Instrumental Analysis Center, Dalian University of Technology, Dalian 116024, China.
| | - Changxu Ning
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Zhongxiang Zhou
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yibo Zhang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Shisheng Wang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; Ningbo Institute of Dalian University of Technology, Ningbo 315016, China
| | - Yueqing Li
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; Ningbo Institute of Dalian University of Technology, Ningbo 315016, China
| | - Xiuhan Guo
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; Ningbo Institute of Dalian University of Technology, Ningbo 315016, China.
| |
Collapse
|
6
|
Li Y, Guo F, Suo R, Wu X, Jin S, Zhou J, Zhang C, Li S, Qian W, Huan Ling, Huang S, Chen H, Wu B. A caged luciferin analogue generating near-infrared bioluminescence for activity-sensing of labile iron. Biosens Bioelectron 2025; 278:117290. [PMID: 40020638 DOI: 10.1016/j.bios.2025.117290] [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: 09/28/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Iron plays a vital role in physiological processes due to its high oxygen affinity and efficient redox capability. However, perturbations in iron homeostasis, particularly in its labile forms that drive oxidative stress, have been implicated in a spectrum of pathologies, including infectious diseases, malignancies, and neurodegenerative disorders. Despite the critical importance of detecting labile Fe2+, conventional fluorescent and bioluminescent probes are constrained by inherent limitations, such as suboptimal sensitivity, elevated background noise, and inadequate tissue penetration depth. To overcome these challenges, we report the development of a novel caged luciferin analogue, O-Akalumine (O-Aka), designed with an Fe2+-specific switchable N-oxide bond to enable turn-on near-infrared (NIR) bioluminescence imaging of labile Fe2+. The bioluminescence emitted by O-Aka in the presence of native firefly luciferase is centered in the NIR spectrum (λmax = 677 nm), substantially improving signal penetration through biological tissues. Exhibiting low intrinsic background noise, high sensitivity, and deep tissue imaging capability, O-Aka effectively visualized exogenous Fe2+ in cellular models and a murine breast cancer model, as well as endogenous Fe2+ in an acute cardiac injury model. These results underscore the utility of O-Aka as a robust bioluminescent probe for elucidating the physiological and pathological roles of Fe2+ and exploring its potential anticancer mechanisms.
Collapse
Affiliation(s)
- Yi Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China; College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430062, PR China
| | - Fangliang Guo
- Department of Neurology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, PR China
| | - Ruiyang Suo
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Xinze Wu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430062, PR China
| | - Shiqi Jin
- School of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062, PR China
| | - Jun Zhou
- Interventional Diagnostic and Therapeutic Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Caiju Zhang
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Shuqi Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Wang Qian
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Huan Ling
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China
| | - Shiwen Huang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430062, PR China
| | - Huaixia Chen
- School of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062, PR China.
| | - Bo Wu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, PR China.
| |
Collapse
|
7
|
O'Hare MA, Rust C, Malan-Müller S, Pirovano W, Lowry CA, Ramaboli M, van den Heuvel LL, Seedat S, PGC‐PTSD Microbiome Workgroup, Hemmings SMJ. Preliminary Insights Into the Relationship Between the Gut Microbiome and Host Genome in Posttraumatic Stress Disorder. GENES, BRAIN, AND BEHAVIOR 2025; 24:e70025. [PMID: 40492293 DOI: 10.1111/gbb.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 05/18/2025] [Accepted: 05/22/2025] [Indexed: 06/11/2025]
Abstract
Posttraumatic stress disorder (PTSD) may develop following trauma exposure; however, not all trauma-exposed individuals develop PTSD, suggesting the presence of susceptibility and resilience factors. The gut microbiome and host genome, which are interconnected, have been implicated in the aetiology of PTSD. However, their interaction has yet to be investigated in a South African population. Using genome-wide genotype data and 16S rRNA (V4) gene amplicon sequencing data from 53 trauma-exposed controls and 74 PTSD cases, we observed no significant association between the host genome and summed abundance of Mitsuokella, Odoribacter, Catenibacterium and Olsenella, previously reported as associated with PTSD status in this cohort. However, PROM2 rs2278067 T-allele was significantly positively associated with the summed relative abundance of these genera, but only in individuals with PTSD and not trauma-exposed controls (p < 0.014). Polygenic risk scores generated using genome-wide association study summary statistics from the PGC-PTSD Overall Freeze 2 were not predictive of gut microbial composition in this cohort. These preliminary results suggest a potential role for the interaction between genetic variation and gut microbial composition in the context of PTSD, underscoring the need for further investigation.
Collapse
Affiliation(s)
- Michaela A O'Hare
- Department of Biomedical Sciences, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Unit, Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Carlien Rust
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Unit, Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Stefanie Malan-Müller
- Department of Pharmacology and Toxicology, Faculty of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Biomedical Network Research Center of Mental Health (CIBERSAM), Institute of Health Carlos III, Madrid, Spain
- Neurochemistry Research Institute UCM, Hospital 12 de Octubre Research Institute (Imas12), Madrid, Spain
| | - Walter Pirovano
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Christopher A Lowry
- Departments of Integrative Physiology and Psychology and Neuroscience, Center for Neuroscience and Center for Microbial Exploration, University of Colorado Boulder, Boulder, Colorado, USA
| | - Matsepo Ramaboli
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Unit, Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Leigh L van den Heuvel
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Unit, Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Soraya Seedat
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Unit, Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | | | - Sian M J Hemmings
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Unit, Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
8
|
Dai L, Peng J, Zhang M, Hu Y, Gao Z, Wang J, Zhang H, Li S. Gypenosides Attenuates CORT-Induced Ferroptosis via Inhibiting TNF-α/NF-κB Signaling Pathway in PC12 Cells. Molecules 2025; 30:2103. [PMID: 40430276 PMCID: PMC12114596 DOI: 10.3390/molecules30102103] [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: 04/02/2025] [Revised: 05/04/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Chronic stress can lead to nervous system dysfunction and depression-like behaviors in animals. Gypenosides can improve chronic stress-induced neuronal damage, but the protective mechanism remains poorly understood. This study aims to investigate the effect and mechanism of gypenosides on chronic stress-induced neuronal ferroptosis. Therefore, we established a chronic stress-induced neuronal damage model in vitro using corticosterone to induce PC12 cell injury. We demonstrated that ferroptosis inhibitors DFO and Ferrostatin-1 alleviated corticosterone-induced cell death in PC12 cells by reducing iron accumulation, lipid peroxidation, and increasing cell viability. Meanwhile, gypenosides attenuated ferroptosis agonist Erastin-induced ferroptosis in PC12 cells. Then, gypenosides ameliorated corticosterone-induced ferroptosis in PC12 cells. In terms of molecular mechanisms, gypenosides decreased the expression of Hepcidin and DMT1, and increased the expression of Ferritin and FPN1, thereby improving corticosterone-induced iron homeostasis disorders and iron accumulation. Moreover, gypenosides improved corticosterone-induced lipid peroxidation by inhibiting GLS2 expression, upregulating the expression of SLC7A11 and glutathione peroxidase 4, and reducing glutamate accumulation and GSH depletion. Gypenosides also reduced corticosterone-induced release of inflammatory cytokines, the expression of TNFR1, and the phosphorylation of NF-κB and p53 in PC12 cells. These findings indicate that gypenosides attenuate corticosterone-induced ferroptosis by inhibiting TNF-α/NF-κB signaling pathway in PC12 cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Haiyang Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Shoujun Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
9
|
Wang F, Zhang W, Yin L, Huang C, Zhao H. Exogenous Maresin1 attenuates doxorubicin-induced cardiomyocyte ferroptosis and mitochondrial impairment via NRF2/GPX4 axis. Free Radic Biol Med 2025; 235:335-346. [PMID: 40348287 DOI: 10.1016/j.freeradbiomed.2025.05.388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/06/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Doxorubicin (Dox)-induced cardiotoxicity in patients with cancer, mediated primarily through cardiomyocyte ferroptosis and mitochondrial dysfunction, presents both life-threatening risks and significant limitations to Dox chemotherapeutic efficacy. The study investigated the therapeutic potential and mechanistic role of Maresin1-a novel proinflammatory regression mediator (SPM) -in Dox-induced cardiomyocyte ferroptosis. We employed both in vitro and in vivo models: H9C2 cells and human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) for in vitro ferroptosis modeling and C57BL/6 mice for in vivo validation. Our key findings showed that Maresin1 in Dox-treated cardiomyocytes attenuated lipid peroxidation, upregulated the anti-ferroptosis-related protein expression via the NRF2/GPX4 axis and mitigated mitochondrial structural and functional impairment. However, inhibition of NRF2 signaling abolished the cardioprotective effects of Maresin1 against Dox-induced ferroptosis. In conclusion, Maresin1 preserved cardiac function by preventing Dox-associated cardiomyocyte ferroptosis and mitochondrial impairment through the NRF2/GPX4 axis activation.
Collapse
Affiliation(s)
- Fengyuan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| | - Wei Zhang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang, 443000, PR China
| | - Lin Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China
| | - Hongyi Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, 430060, PR China.
| |
Collapse
|
10
|
Zeng J, Yuan L, Chen G, Qi Y, Qie X, Jin Y, Chen Y, Li H. The ferroptosis of sertoli cells inducing blood-testis barrier damage is produced by oxidative stress in cryptorchidism. Free Radic Biol Med 2025; 232:97-106. [PMID: 40032029 DOI: 10.1016/j.freeradbiomed.2025.02.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/17/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
Oxidative Stress (OS) is the main cause of damage to the Blood-Testis Barrier (BTB) in cryptorchidism, which seriously endangers male reproductive health. It is well known that the OS induced ferroptosis is an important cause of dysfunction in the body. However, it is still unknown whether BTB damage in cryptorchidism leads to ferroptosis of Sertoli cells. We establishing the cryptorchidism model through surgery to avoid the complex effects of drugs on the model animals, combined with in vitro culture of the primary Sertoli cells for validation, and the methods of immunofluorescence staining, Western blotting and Prussian blue staining were used to study the oxidative stress in cryptorchidism. The effects of ferroptosis of Sertoli cells inducing BTB damage caused by OS in cryptorchidism were analyzed. We found that the inhibition of Nrf-2/keap-1/HO-1 pathway resulted in decreased expression levels of Glutathione Peroxidase 4 (GPX4), Ferroportin 1 (FPN1), and increased expression of Ferritin light chain (FTL) protein. Our research further confirms that inhibiting ferroptosis reduced BTB damage by reflecting a decrease expression of Zonula Occludens protein 1 (ZO-1), Occludin and Claudin-11 protein caused by OS. In addition, we found that the testosterone (T) secretion disorders and the supplementation of T can alleviate the damage of the BTB in cryptorchidism, and this effect is achieved through the Androgen Receptor (AR). In conclusion, our study found that the inhibition of Nrf-2/keap-1/HO-1 pathway in testis and the reduction of Tight junction proteins (TJs) ZO-1, Occludin and Claudin-11 protein expression levels in cryptorchidic mice, indicated that the cryptorchidism triggering a serious reproductive disorder, and one of the important reasons is the OS induced ferroptosis of Sertoli cells, which ultimately leads to the damage of the BTB. This findings may have important implications in the field of male reproductive disorders.
Collapse
Affiliation(s)
- Jianlin Zeng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, China
| | - Ligang Yuan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, China.
| | - Guojuan Chen
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Huangzhong District Animal Disease Prevention and Control Center, Xining, 811600, China
| | - Yumei Qi
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, China
| | - Xiaolong Qie
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, China
| | - Yajuan Jin
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, China
| | - Yulu Chen
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, China
| | - Haijun Li
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, China
| |
Collapse
|
11
|
Khodadadi H, Łuczyńska K, Winiarczyk D, Leszczyński P, Taniguchi H. NFE2L1 as a central regulator of proteostasis in neurodegenerative diseases: interplay with autophagy, ferroptosis, and the proteasome. Front Mol Neurosci 2025; 18:1551571. [PMID: 40375958 PMCID: PMC12078313 DOI: 10.3389/fnmol.2025.1551571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 03/14/2025] [Indexed: 05/18/2025] Open
Abstract
Maintaining proteostasis is critical for neuronal health, with its disruption underpinning the progression of neurodegenerative diseases such as Alzheimer's, Parkinson's, and Huntington's diseases. Nuclear Factor Erythroid 2-Related Factor 1 (NFE2L1) has emerged as a key regulator of proteostasis, integrating proteasome function, autophagy, and ferroptosis to counteract oxidative stress and protein misfolding. This review synthesizes current knowledge on the role of NFE2L1 in maintaining neuronal homeostasis, focusing on its mechanisms for mitigating proteotoxic stress and supporting cellular health, offering protection against neurodegeneration. Furthermore, we discuss the pathological implications of NFE2L1 dysfunction and explore its potential as a therapeutic target. By highlighting gaps in the current understanding and presenting future research directions, this review aims to elucidate NFE2L1's role in advancing treatment strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Hossein Khodadadi
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Poland
| | - Kamila Łuczyńska
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Poland
- The Second Department of Psychiatry, Institute of Psychiatry and Neurology in Warsaw, Warsaw, Poland
| | - Dawid Winiarczyk
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Poland
| | - Paweł Leszczyński
- Department of Stem Cell Bioengineering Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Hiroaki Taniguchi
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Poland
- African Genome Center, University Mohammed VI Polytechnic (UM6P), Ben Guerir, Morocco
| |
Collapse
|
12
|
Liu YJ, Jia GR, Zhang SH, Guo YL, Ma XZ, Xu HM, Xie JX. The role of microglia in neurodegenerative diseases: from the perspective of ferroptosis. Acta Pharmacol Sin 2025:10.1038/s41401-025-01560-4. [PMID: 40307457 DOI: 10.1038/s41401-025-01560-4] [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: 12/17/2024] [Accepted: 04/06/2025] [Indexed: 05/02/2025]
Abstract
Iron plays a pivotal role in numerous fundamental biological processes in the brain. Among the various cell types in the central nervous system, microglia are recognized as the most proficient cells in accumulating and storing iron. Nonetheless, iron overload can induce inflammatory phenotype of microglia, leading to the production of proinflammatory cytokines and contributing to neurodegeneration. A growing body of evidence shows that disturbances in iron homeostasis in microglia is associated with a range of neurodegenerative disorders. Recent research has revealed that microglia are highly sensitive to ferroptosis, a form of iron-dependent cell death. How iron overload influences microglial function? Whether disbiosis in iron metabolism and ferroptosis in microglia are involved in neurodegenerative disorders and the underlying mechanisms remain to be elucidated. In this review we focus on the recent advances in research on microglial iron metabolism as well as ferroptosis in microglia. Meanwhile, we provide a comprehensive overview of the involvement of microglial ferroptosis in neurodegenerative disorders from the perspective of crosstalk between microglia and neuron, with a focus on Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Ying-Juan Liu
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China
| | - Guo-Rui Jia
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Sheng-Han Zhang
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Yun-Liang Guo
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China
| | - Xi-Zhen Ma
- College of Life Sciences and Health, University of Health and Rehabilitation Science, Qingdao, 266113, China.
| | - Hua-Min Xu
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China.
- Department of Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| | - Jun-Xia Xie
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
13
|
Abdukarimov N, Kokabi K, Kunz J. Ferroptosis and Iron Homeostasis: Molecular Mechanisms and Neurodegenerative Disease Implications. Antioxidants (Basel) 2025; 14:527. [PMID: 40427409 PMCID: PMC12108473 DOI: 10.3390/antiox14050527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Iron dysregulation has emerged as a pivotal factor in neurodegenerative pathologies, especially through its capacity to promote ferroptosis, a unique form of regulated cell death driven by iron-catalyzed lipid peroxidation. This review synthesizes current evidence on the molecular underpinnings of ferroptosis, focusing on how disruptions in iron homeostasis interact with key antioxidant defenses, such as the system Xc--glutathione-GPX4 axis, to tip neurons toward lethal oxidative damage. Building on these mechanistic foundations, we explore how ferroptosis intersects with hallmark pathologies in Alzheimer's disease (AD) and Parkinson's disease (PD) and examine how iron accumulation in vulnerable brain regions may fuel disease-specific protein aggregation and neurodegeneration. We further surveyed the distinct components of ferroptosis, highlighting the role of lipid peroxidation enzymes, mitochondrial dysfunction, and recently discovered parallel pathways that either exacerbate or mitigate neuronal death. Finally, we discuss how these insights open new avenues for neuroprotective strategies, including iron chelation and lipid peroxidation inhibitors. By highlighting open questions, this review seeks to clarify the current state of knowledge and proposes directions to harness ferroptosis modulation for disease intervention.
Collapse
Affiliation(s)
| | | | - Jeannette Kunz
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan; (N.A.); (K.K.)
| |
Collapse
|
14
|
Zhao N, Li S, Wu H, Wei D, Pu N, Wang K, Liu Y, Tao Y, Song Z. Ferroptosis: An Energetic Villain of Age-Related Macular Degeneration. Biomedicines 2025; 13:986. [PMID: 40299661 PMCID: PMC12024642 DOI: 10.3390/biomedicines13040986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/01/2025] [Accepted: 04/14/2025] [Indexed: 05/01/2025] Open
Abstract
Iron homeostasis plays an important role in maintaining cellular homeostasis; however, excessive iron can promote the production of reactive oxygen species (ROS). Ferroptosis is iron-dependent programmed cell death that is characterized by excessive iron accumulation, elevated lipid peroxides, and the overproduction of ROS. The maintenance of iron homeostasis is contingent upon the activity of the transferrin receptor (TfR), ferritin (Ft), and ferroportin (FPn). In the retina, iron accumulation and lipid peroxidation can contribute to the development of age-related macular degeneration (AMD). This phenomenon can be explained by the occurrence of the Fenton reaction, in which the interaction between divalent iron and hydrogen peroxide leads to the generation of highly reactive hydroxyl radicals. The hydroxyl radicals exhibit a propensity to attack proteins, lipids, nucleic acids, and carbohydrates, thereby instigating oxidative damage and promoting lipid peroxidation. Ultimately, these processes culminate in cell death and retinal degeneration. In this context, a comprehensive understanding of the exact mechanisms underlying ferroptosis may hold significant importance for developing therapeutic interventions. This review summarizes recent findings on iron metabolism, cellular ferroptosis, and lipid metabolism in the aging retina. We also introduce developments in the therapeutic strategies using iron chelating agents. Further refinements of these knowledges would deepen our comprehension of the pathophysiology of AMD and advance the clinical management of degenerative retinopathy. A comprehensive search strategy was employed to identify relevant studies on the role of ferroptosis in AMD. We performed systematic searches of the PubMed and Web of Science electronic databases from inception to the current date. The keywords used in the search included "ferroptosis", "AMD", "age-related macular degeneration", "iron metabolism", "oxidative stress", and "ferroptosis pathways". Peer-reviewed articles, including original research, reviews, meta-analyses, and clinical studies, were included in this paper, with a focus on the molecular mechanisms of ferroptosis in AMDs. Studies not directly related to ferroptosis, iron metabolism, or oxidative stress in the context of AMD were excluded. Furthermore, articles that lacked sufficient data or were not peer-reviewed (e.g., conference abstracts, editorials, or opinion pieces) were not considered.
Collapse
Affiliation(s)
- Na Zhao
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Siyu Li
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Hao Wu
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Dong Wei
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Ning Pu
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Kexin Wang
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
| | - Yashuang Liu
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
| | - Ye Tao
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
| | - Zongming Song
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
| |
Collapse
|
15
|
Kaftan Öcal G, Armagan G. Induction of Ferroptotic Cell Death by Neuromelanin Pigments in Dopaminergic Cells. ACS Chem Neurosci 2025; 16:1500-1510. [PMID: 40145657 PMCID: PMC12006986 DOI: 10.1021/acschemneuro.5c00029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/24/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
Neuromelanin (NM) is an iron-rich, insoluble brown or black pigment that exhibits protective properties. However, its accumulation over time may render it a source of free radicals. In Parkinson's disease, dopaminergic neurons with the highest NM levels and increased iron content are preferentially vulnerable to degeneration. Considering NM's iron binding capacity and the critical role of iron in ferroptosis, we aimed to investigate the interplay between neuromelanin and ferroptosis in dopaminergic cells. We prepared two NM pigments: iron-free NM (ifNM) and iron-containing NM (Fe3+NM) and, exposed to cells. After verifying NM accumulation, cell viability was assessed in the absence or presence of antioxidants (NAC (1 mM), Trolox (100 μM)) and specific inhibitors of cell death types. Ferroptosis-related parameters, including lipid peroxidation byproducts (4-HNE), lipid ROS, glutathione, intracellular iron, GPX4, and ACSL4, and cellular iron metabolism-related proteins (TfR1, ferroportin, ferritin, IREB2) were evaluated following ifNM and Fe3+NM treatments, with or without Ferrostatin-1, Liproxstatin-1 and deferoxamine. Both NMs induced cell death via distinct mechanisms. Ferroptotic cell death by ifNM and Fe3+NM was reversed by ferrostatin-1 and NAC (p < 0.05). Significant alterations in lipid peroxidation, GPX4 levels, and iron metabolism were observed independent of NM's iron composition (p < 0.05). Ferritin levels increased following ifNM treatment, reflecting an adaptive response to iron overload, while Fe3+NM treatment led to ferritin depletion, possibly via ferritinophagy. Our findings reveal a distinct role of iron-rich and iron-free neuromelanin in modulating ferroptotic pathways, highlighting the potential of targeting neuromelanin-iron interactions as a therapeutic strategy to mitigate neuronal ferroptosis in Parkinson's disease.
Collapse
Affiliation(s)
- Gizem Kaftan Öcal
- Biochemistry
PhD Programme, Graduate School of Health Sciences, Ege University, Izmir 35100, Türkiye
- Department
of Biochemistry, Faculty of Pharmacy, Ayfonkarahisar
Health Sciences University, Afyonkarahisar 03218, Türkiye
| | - Güliz Armagan
- Department
of Biochemistry, Faculty of Pharmacy, Ege
University, Izmir 35100, Türkiye
| |
Collapse
|
16
|
Ollivier T, Devos D, Kuchcinski G, Defebvre L, Huin V, Mutez E. Conservative iron chelation for VAC14: Two-year clinical-radiological follow-up. JOURNAL OF PARKINSON'S DISEASE 2025:1877718X251331820. [PMID: 40221969 DOI: 10.1177/1877718x251331820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
There is a distinct lack of consensus on the most effective treatments for neurodegeneration with brain iron accumulation. This is due to the rarity of the disease, its phenotypic variability, and the multiplicity of pathophysiological mechanisms. Our team has already proposed the use of conservative iron chelation in cases of neuroferritinopathy, with interesting results. However, no mention has yet been made of the treatment of parkinsonism-dystonia related to VAC14 gene mutations. The case reported here illustrates clinical stability after 2 years of conservative iron chelation, with an improvement in radiological images.
Collapse
Affiliation(s)
- Thomas Ollivier
- Neurology and Movement Disorders Department, Expert Center for Parkinson's Disease, Lille University Hospital, Lille, France
- Univ. Lille, INSERM, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - David Devos
- Univ. Lille, INSERM, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
- Department of Medical Pharmacology, Univ. Lille, Inserm, CHU Lille, Lille Neuroscience and Cognition INSERM, U1172, LiCEND, NS-Park/F-CRIN Network, Lille, France
| | - Gregory Kuchcinski
- Univ. Lille, INSERM, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
- Department of Neuroradiology, Univ. Lille, INSERM, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Luc Defebvre
- Neurology and Movement Disorders Department, Expert Center for Parkinson's Disease, Lille University Hospital, Lille, France
- Univ. Lille, INSERM, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Vincent Huin
- Univ. Lille, INSERM, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
- Toxicology and Genopathies Department, UF Neurobiology, Lille University Hospital, Lille, France
| | - Eugenie Mutez
- Neurology and Movement Disorders Department, Expert Center for Parkinson's Disease, Lille University Hospital, Lille, France
- Univ. Lille, INSERM, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| |
Collapse
|
17
|
Soni P, Sharma SM, Pieper AA, Paul BD, Thomas B. Nrf2/Bach1 signaling axis: A promising multifaceted therapeutic strategy for Alzheimer's disease. Neurotherapeutics 2025; 22:e00586. [PMID: 40199685 PMCID: PMC12047509 DOI: 10.1016/j.neurot.2025.e00586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia, which continues to elude effective treatment despite decades of research and numerous clinical trials. While existing therapeutic strategies have primarily targeted neuropathological hallmarks such as amyloid plaques and tau tangles, they have failed to halt disease progression, leaving patients with limited options. This persistent failure reveals a critical gap in our understanding of AD and calls for a fresh perspective - one that goes beyond the traditional targets and dives deeper into the fundamental cellular processes that drive neurodegeneration. Recent advances in molecular biology underscore the significance of nuclear factor E2-related factor 2 (Nrf2), often termed the "guardian of redox homeostasis," in the pathophysiology of AD. Nrf2 orchestrates cellular responses to oxidative stress and neuroinflammation - two interlinked pathological features of AD. In the brains of AD patients, Nrf2 activity is diminished, weakening the brain's ability to counteract oxidative damage. Additionally, the BTB and CNC homology 1 (Bach1) protein, a transcriptional repressor of Nrf2, has emerged as a potential therapeutic target. Here, we review the current landscape of clinical trials in AD and identify the limitations of the conventional approaches. We then explore the prospects of a novel approach that combines Nrf2 activation with Bach1 inhibition to achieve a multipronged defense against oxidative stress, neuroinflammation, and other molecular culprits driving AD. This innovative strategy holds promise for synergistically modulating multiple neuroprotective pathways to advance AD treatment.
Collapse
Affiliation(s)
- Priyanka Soni
- Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC, USA; Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Sudarshana M Sharma
- Department of Biochemistry and Molecular Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Andrew A Pieper
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Bindu D Paul
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Bobby Thomas
- Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC, USA; Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA; Department of Drug Discovery, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
18
|
Selvaraj NR, Nandan D, Nair BG, Nair VA, Venugopal P, Aradhya R. Oxidative Stress and Redox Imbalance: Common Mechanisms in Cancer Stem Cells and Neurodegenerative Diseases. Cells 2025; 14:511. [PMID: 40214466 PMCID: PMC11988017 DOI: 10.3390/cells14070511] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 04/14/2025] Open
Abstract
Oxidative stress (OS) is an established hallmark of cancer and neurodegenerative disorders (NDDs), which contributes to genomic instability and neuronal loss. This review explores the contrasting role of OS in cancer stem cells (CSCs) and NDDs. Elevated levels of reactive oxygen species (ROS) contribute to genomic instability and promote tumor initiation and progression in CSCs, while in NDDs such as Alzheimer's and Parkinson's disease, OS accelerates neuronal death and impairs cellular repair mechanisms. Both scenarios involve disruption of the delicate balance between pro-oxidant and antioxidant systems, which leads to chronic oxidative stress. Notably, CSCs and neurons display alterations in redox-sensitive signaling pathways, including Nrf2 and NF-κB, which influence cell survival, proliferation, and differentiation. Mitochondrial dynamics further illustrate these differences: enhanced function in CSCs supports adaptability and survival, whereas impairments in neurons heighten vulnerability. Understanding these common mechanisms of OS-induced redox imbalance may provide insights for developing interventions, addressing aging hallmarks, and potentially mitigating or preventing both cancer and NDDs.
Collapse
Affiliation(s)
| | | | | | | | - Parvathy Venugopal
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India; (N.R.S.); (D.N.); (B.G.N.); (V.A.N.)
| | - Rajaguru Aradhya
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India; (N.R.S.); (D.N.); (B.G.N.); (V.A.N.)
| |
Collapse
|
19
|
Jiang C, Yan Y, Long T, Xu J, Chang C, Kang M, Wang X, Chen Y, Qiu J. Ferroptosis: a potential therapeutic target in cardio-cerebrovascular diseases. Mol Cell Biochem 2025:10.1007/s11010-025-05262-7. [PMID: 40148662 DOI: 10.1007/s11010-025-05262-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
Cardio-cerebrovascular diseases (CCVDs) are the leading cause of global mortality, yet effective treatment options remain limited. Ferroptosis, a novel form of regulated cell death, has emerged as a critical player in various CCVDs, including atherosclerosis, myocardial infarction, ischemia-reperfusion injury, cardiomyopathy, and ischemic/hemorrhagic strokes. This review highlights the core mechanisms of ferroptosis, its pathological implications in CCVDs, and the therapeutic potential of targeting this process. Additionally, it explores the role of Chinese herbal medicines (CHMs) in mitigating ferroptosis, offering novel therapeutic strategies for CCVDs management. Ferroptosis is regulated by several key pathways. The GPX4-GSH-System Xc- axis is central to ferroptosis execution, involving GPX4 using GSH to neutralize lipid peroxides, with system Xc- being crucial for GSH synthesis. The NAD(P)H/FSP1/CoQ10 axis involves FSP1 regenerating CoQ10 via NAD(P)H, inhibiting lipid peroxidation independently of GPX4. Lipid peroxidation, driven by PUFAs and enzymes like ACSL4 and LPCAT3, and iron metabolism, regulated by proteins like TfR1 and ferritin, are also crucial for ferroptosis. Inhibiting ferroptosis shows promise in managing CCVDs. In atherosclerosis, ferroptosis inhibitors reduce iron accumulation and lipid peroxidation. In myocardial infarction, inhibitors protect cardiomyocytes by preserving GPX4 and SLC7A11 levels. In ischemia-reperfusion injury, targeting ferroptosis reduces myocardial and cerebral damage. In diabetic cardiomyopathy, Nrf2 activators alleviate oxidative stress and iron metabolism irregularities. CHMs offer natural compounds that mitigate ferroptosis. They possess antioxidant properties, chelate iron, and modulate signaling pathways like Nrf2 and AMPK. For example, Salvia miltiorrhiza and Astragalus membranaceus reduce oxidative stress, while some CHMs chelate iron, reducing its availability for ferroptosis. In conclusion, ferroptosis plays a pivotal role in CCVDs, and targeting it offers novel therapeutic avenues. CHMs show promise in reducing ferroptosis and improving patient outcomes. Future research should explore combination therapies and further elucidate the molecular interactions in ferroptosis.
Collapse
Affiliation(s)
- Chenlong Jiang
- School of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
| | - Yang Yan
- Department of Cardiology, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China
| | - Tianlin Long
- Department of Neurosurgery, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China
| | - Jiawei Xu
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
| | - Cuicui Chang
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
- Department of Cardiology, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China
| | - Meili Kang
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
| | - Xuanqi Wang
- Department of Cardiology, First Hospital of Northwestern University, Northwest University, No. 512 Xianning East Road, Xi'an, 710043, Shaanxi, China.
| | - Yuhua Chen
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China.
- Department of Neurosurgery, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China.
- School of Life and Health Science, Hainan University, No. 58 People's Avenue, Haikou, 570100, Hainan, China.
| | - Junlin Qiu
- Department of Cardiology, First Hospital of Northwestern University, Northwest University, No. 512 Xianning East Road, Xi'an, 710043, Shaanxi, China.
| |
Collapse
|
20
|
Bjørklund G, Wallace DR, Hangan T, Butnariu M, Gurgas L, Peana M. Cerebral iron accumulation in multiple sclerosis: Pathophysiology and therapeutic implications. Autoimmun Rev 2025; 24:103741. [PMID: 39756528 DOI: 10.1016/j.autrev.2025.103741] [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: 12/14/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disorder of the central nervous system characterized by demyelination, neuroinflammation, and neurodegeneration. Recent studies highlight the role of cerebral iron (Fe) accumulation in exacerbating MS pathophysiology. Fe, essential for neural function, contributes to oxidative stress and inflammation when dysregulated, particularly in the brain's gray matter and demyelinated lesions. Advanced imaging techniques, including susceptibility-weighted and quantitative susceptibility mapping, have revealed abnormal Fe deposition patterns in MS patients, suggesting its involvement in disease progression. Iron's interaction with immune cells, such as microglia, releases pro-inflammatory cytokines, further amplifying neuroinflammation and neuronal damage. These findings implicate Fe dysregulation as a significant factor in MS progression, contributing to clinical manifestations like cognitive impairment. Therapeutic strategies targeting Fe metabolism, including Fe chelation therapies, show promise in reducing Fe-related damage, instilling optimism about the future of MS treatment. However, challenges such as crossing the blood-brain barrier and maintaining Fe homeostasis remain. Emerging approaches, such as Fe-targeted nanotherapeutics and biologics, offer new possibilities for personalized treatments. However, the journey is far from over. Continued research into the molecular mechanisms of Fe-induced neuroinflammation and oxidative damage is essential. Through this research, we can develop effective interventions that could slow MS progression and improve patient outcomes.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway.
| | - David R Wallace
- Department of Pharmacology, Oklahoma State University Center for Health Sciences, Tulsa, OK, United States
| | - Tony Hangan
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
| | - Monica Butnariu
- University of Life Sciences "King Mihai I" from Timisoara, Timis, Romania; CONEM Romania Biotechnology and Environmental Sciences Group, University of Life Sciences "King Mihai I" from Timisoara, Timis, Romania
| | - Leonard Gurgas
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
| | - Massimiliano Peana
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, Italy
| |
Collapse
|
21
|
Li Q, Yang X, Li T. Natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in central nervous system diseases: current preclinical evidence and future perspectives. Front Pharmacol 2025; 16:1570069. [PMID: 40196367 PMCID: PMC11973303 DOI: 10.3389/fphar.2025.1570069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 02/24/2025] [Indexed: 04/09/2025] Open
Abstract
Flavonoids are a class of important polyphenolic compounds, renowned for their antioxidant properties. However, recent studies have uncovered an additional function of these natural flavonoids: their ability to inhibit ferroptosis. Ferroptosis is a key mechanism driving cell death in central nervous system (CNS) diseases, including both acute injuries and chronic neurodegenerative disorders, characterized by iron overload-induced lipid peroxidation and dysfunction of the antioxidant defense system. This review discusses the therapeutic potential of natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in CNS diseases, focusing on their molecular mechanisms, summarizing findings from preclinical animal models, and providing insights for clinical translation. We specifically highlight natural flavonoids such as Baicalin, Baicalein, Chrysin, Vitexin, Galangin, Quercetin, Isoquercetin, Eriodictyol, Proanthocyanidin, (-)-epigallocatechin-3-gallate, Dihydromyricetin, Soybean Isoflavones, Calycosin, Icariside II, and Safflower Yellow, which have shown promising results in animal models of acute CNS injuries, including ischemic stroke, cerebral ischemia-reperfusion injury, intracerebral hemorrhage, subarachnoid hemorrhage, traumatic brain injury, and spinal cord injury. Among these, Baicalin and its precursor Baicalein stand out due to extensive research and favorable outcomes in acute injury models. Mechanistically, these flavonoids not only regulate the Nrf2/ARE pathway and activate GPX4/GSH-related antioxidant pathways but also modulate iron metabolism proteins, thereby alleviating iron overload and inhibiting ferroptosis. While flavonoids show promise as ferroptosis inhibitors for CNS diseases, especially in acute injury settings, further studies are needed to evaluate their efficacy, safety, pharmacokinetics, and blood-brain barrier penetration for clinical application.
Collapse
Affiliation(s)
- Qiuhe Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaohang Yang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Tiegang Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
22
|
Alves Fernandes TA, Tourville A, Ciss I, Ribeiro Silva R, Andretto de Mattos B, Dos Santos Pereira M, Oblaza M, Brunel JM, Ferrié L, Raisman-Vozari R, Figadère B, Del-Bel E, Michel PP. Oxytetracycline and its Non-Antibiotic Derivative DOT Protect Midbrain Dopamine Neurons from Iron-Driven Oxidative Damage. Neurotox Res 2025; 43:16. [PMID: 40119187 DOI: 10.1007/s12640-025-00742-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/24/2025]
Abstract
This study aimed to investigate the neuroprotective potential of the tetracycline (TC) antibiotic oxytetracycline (OT) and its non-antibiotic derivative 4-dedimethylamino 12a-deoxy-oxytetracycline (DOT), in experimental conditions that mimic the gradual loss of dopamine (DA) neurons in Parkinson's disease (PD). Specifically, we established a model system of mouse midbrain cultures where DA neurons progressively die when exposed to an iron-containing medium. We found that OT (EC50 = 0.25µM) and DOT (EC50 = 0.34µM) efficiently protected DA neurons from degeneration, with these effects observable until advanced stages of neurodegeneration. The reference antibiotic TC doxycycline (DOX) also exhibited protective effects in this context. Importantly, DA neurons rescued by OT, DOT, and DOX retained their capacity to accumulate and release DA, indicating full functional integrity. Additionally, molecules with iron-chelating properties (apotransferrin, desferoxamine), as well as inhibitors of lipid peroxidation and ferroptosis (Trolox, Liproxstatin-1), could replicate the rescue of DA neurons provided by OT, DOT, and DOX. Live-cell imaging studies showed that test TCs and other neuroprotective molecules prevented the emission of intracellular reactive oxygen species and the associated disruption of the mitochondrial membrane potential. However, neither OT, DOT, nor DOX could protect DA neurons from selective mitochondrial poisoning by 1-methyl-4-phenylpyridinium. This suggests that test TCs may be protective against iron-mediated damage through a mechanism not directly involving mitochondria. Overall, we demonstrate that OT and DOT possess promising properties that could be useful for combating PD neurodegeneration. However, the absence of antimicrobial activity makes DOT a better candidate drug compared to its parent compound OT.
Collapse
Affiliation(s)
- Thaís Antonia Alves Fernandes
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, 75013, France
- Department of Basic and Oral Biology, Ribeirão Preto School of Dentistry, University of São Paulo (USP), Ribeirão Preto, SP, 14049-904, Brazil
- Department of Physiology, Faculty of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Aurore Tourville
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, 75013, France
| | - Ismaila Ciss
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, 75013, France
- Université Paris-Saclay, BioCIS, CNRS, Orsay, 91190, France
| | - Rafaela Ribeiro Silva
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, 75013, France
| | - Bianca Andretto de Mattos
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, 75013, France
- Department of Basic and Oral Biology, Ribeirão Preto School of Dentistry, University of São Paulo (USP), Ribeirão Preto, SP, 14049-904, Brazil
- Department of Physiology, Faculty of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Maurício Dos Santos Pereira
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, 75013, France
- Stanley Center, Broad Institute, Cambridge, MA, 02142, USA
| | - Maxime Oblaza
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, 75013, France
- Université Paris-Saclay, BioCIS, CNRS, Orsay, 91190, France
| | - Jean-Michel Brunel
- Aix Marseille Univ, Inserm, Membranes et Cibles Thérapeutiques, Service de Santé des Armées, Marseille, 13385, France
| | - Laurent Ferrié
- Université Paris-Saclay, BioCIS, CNRS, Orsay, 91190, France
| | - Rita Raisman-Vozari
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, 75013, France
| | - Bruno Figadère
- Université Paris-Saclay, BioCIS, CNRS, Orsay, 91190, France
| | - Elaine Del-Bel
- Department of Basic and Oral Biology, Ribeirão Preto School of Dentistry, University of São Paulo (USP), Ribeirão Preto, SP, 14049-904, Brazil.
| | - Patrick Pierre Michel
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, 75013, France.
| |
Collapse
|
23
|
White JB, Sanchez KL, Currais A, Soriano-Castell D, Maher P, Soriano S. Ferroptosis and Charcot-Marie-Tooth Disease 1A: Emerging Evidence for a Pathogenic Association. Antioxidants (Basel) 2025; 14:331. [PMID: 40227266 PMCID: PMC11939244 DOI: 10.3390/antiox14030331] [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: 02/12/2025] [Revised: 03/08/2025] [Accepted: 03/08/2025] [Indexed: 04/15/2025] Open
Abstract
Charcot-Marie-Tooth disease (CMT) is the most common hereditary peripheral neuropathy worldwide, presenting clinically as muscle weakness that progresses to impaired ambulation or quadriplegia with age. CMT1A, the most common subtype, is caused by a duplication in PMP22, encoding an essential membrane protein for Schwann cell myelin integrity. While the mechanisms of peripheral neurodegeneration in CMT1A are poorly understood, excessive oxidative stress, particularly lipid peroxidation, is a known pathological feature, and antioxidant therapy has reversed the CMT1A phenotype in a mouse model. For the first time, we define the pathogenic link between CMT1A and ferroptosis, a form of regulated cell death caused by excessive lipid peroxidation and hindered antioxidant defenses. Human-derived CMT1A fibroblasts showed greater susceptibility to RSL3, a pro-ferroptosis agent, compared with controls, alongside several ferroptosis markers, including elevated lipid peroxides and depleted GPX4, a critical anti-ferroptosis repressor. Similarly, transcriptomic analysis of human iPSC-derived Schwann cells revealed elevated ferroptosis activation and cellular stress markers in CMT1A. We propose that chronic, sublethal ferroptotic stress, mediated by lipid peroxide accumulation, depletes antioxidant defenses in CMT1A Schwann cells, leading to decompensation with age, manifesting as symptomatic disease. These results emphasize ferroptosis as a driver of CMT1A pathology, potentially revealing a new therapeutic path.
Collapse
Affiliation(s)
- Jacob B. White
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (J.B.W.); (K.L.S.)
| | - Kayla L. Sanchez
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (J.B.W.); (K.L.S.)
| | - Antonio Currais
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; (A.C.); (D.S.-C.)
| | - David Soriano-Castell
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; (A.C.); (D.S.-C.)
| | - Pamela Maher
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; (A.C.); (D.S.-C.)
| | - Salvador Soriano
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (J.B.W.); (K.L.S.)
| |
Collapse
|
24
|
Gutierre RC, Rocha PR, Graciani AL, Coppi AA, Arida RM. Tau, amyloid, iron, oligodendrocytes ferroptosis, and inflammaging in the hippocampal formation of aged rats submitted to an aerobic exercise program. Brain Res 2025; 1850:149419. [PMID: 39725376 DOI: 10.1016/j.brainres.2024.149419] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
Alzheimer's disease is a progressive neurodegenerative disease affecting memory, language, and thinking with no curative treatment. Symptoms appear gradually, and pathological brain changes may occur twenty years before the physical and psychological signs, pointing to the urgent development of preventive interventions. Physical activity has been investigated as a preventive tool to defeat the main biological features of AD: pathological amyloid protein plaques, tau tangles, myelin degeneration, and iron deposits in the brain. This work quantifies tau tangles, amyloid, iron, and ferroptosis in oligodendrocytes in the aged rat hippocampal formation and statistically correlates neuron-neuron, neuron-glia, and glia-glia crosstalk and the effect of physical exercise on it. Our results indicate that iron overload in the oligodendrocytes is an inducer of ferroptosis; physical exercise reduces inflammaging, and improves axon-myelin volume relations; tau, amyloid, iron, and hippocampal formation cells present statistical correlations. Our data suggest the beneficial effects of physical exercise in AD and a mathematical relationship between the hippocampal formation cells in sedentary and active individuals, which should be considered in human and animal studies as a guide to a better understanding of crosstalk physiology.
Collapse
Affiliation(s)
- R C Gutierre
- Almeria Institute of Integrative Science, São Paulo, Brazil.
| | - P R Rocha
- Federal University of São Paulo, Paulista School of Medicine, Department of Physiology, Laboratory of Neurophysiology, São Paulo, Brazil
| | - A L Graciani
- Federal University of São Paulo, Paulista School of Medicine, Department of Physiology, Laboratory of Neurophysiology, São Paulo, Brazil
| | - A A Coppi
- University of Bristol, Faculty of Health and Life Sciences, Bristol, United Kingdom
| | - R M Arida
- Federal University of São Paulo, Paulista School of Medicine, Department of Physiology, Laboratory of Neurophysiology, São Paulo, Brazil
| |
Collapse
|
25
|
Luyken AK, Lappe C, Viard R, Löhle M, Kleinlein HR, Kuchcinski G, Langner S, Wenzel AM, Walter M, Weber MA, Storch A, Devos D, Walter U. High correlation of quantitative susceptibility mapping and echo intensity measurements of nigral iron overload in Parkinson's disease. J Neural Transm (Vienna) 2025; 132:407-417. [PMID: 39485510 PMCID: PMC11870917 DOI: 10.1007/s00702-024-02856-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024]
Abstract
Quantitative susceptibility mapping (QSM) and transcranial sonography (TCS) offer proximal evaluations of iron load in the substantia nigra. Our prospective study aimed to investigate the relationship between QSM and TCS measurements of nigral iron content in patients with Parkinson's disease (PD). In secondary analyses, we wanted to explore the correlation of substantia nigra imaging data with clinical and laboratory findings. Eighteen magnetic resonance imaging and TCS examinations were performed in 15 PD patients at various disease stages. Susceptibility measures of substantia nigra were calculated from referenced QSM maps. Echogenicity of substantia nigra on TCS was measured planimetrically (echogenic area) and by digitized analysis (echo-intensity). Iron-related blood serum parameters were measured. Clinical assessments included the Unified PD Rating Scale and non-motor symptom scales. Substantia nigra susceptibility correlated with echogenic area (Pearson correlation, r = 0.53, p = 0.001) and echo-intensity (r = 0.78, p < 0.001). Individual asymmetry indices correlated between susceptibility and echogenic area measurements (r = 0.50, p = 0.042) and, more clearly, between susceptibility and echo-intensity measurements (r = 0.85, p < 0.001). Substantia nigra susceptibility (individual mean of bilateral measurements) correlated with serum transferrin saturation (Spearman test, r = 0.78, p < 0.001) and, by trend, with serum iron (r = 0.69, p = 0.004). Nigral echogenicity was not clearly related to serum values associated with iron metabolism. Susceptibility and echogenicity measurements were unrelated to PD duration, motor subtype, and severity of motor and non-motor symptoms. The present results support the assumption that iron accumulation is involved in the increase of nigral echogenicity in PD. Nigral echo-intensity probably reflects ferritin-bound iron, e.g. stored in microglia.
Collapse
Affiliation(s)
- Adrian Konstantin Luyken
- Department of Neurology, Rostock University Medical Center, Gehlsheimer Str. 20, 18147, Rostock, Germany
| | - Chris Lappe
- Institute of Diagnostic and Interventional Radiology, Pediatric Radiology and Neuroradiology, University Medical Center Rostock, Rostock, Germany
- German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Network of Centers of Excellence in Neurodegeneration (CoEN) Center Rostock, Rostock, Germany
| | - Romain Viard
- UAR 2014 - US 41 - PLBS - Plateformes Lilloises en Biologie & Santé, University of Lille, Lille, France
- INSERM, Centre Hospitalier Universitaire (CHU) de Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, LICEND, University of Lille, Lille, France
| | - Matthias Löhle
- Department of Neurology, Rostock University Medical Center, Gehlsheimer Str. 20, 18147, Rostock, Germany
- German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Network of Centers of Excellence in Neurodegeneration (CoEN) Center Rostock, Rostock, Germany
| | - Hanna Rebekka Kleinlein
- Department of Neurology, Rostock University Medical Center, Gehlsheimer Str. 20, 18147, Rostock, Germany
| | - Grégory Kuchcinski
- UAR 2014 - US 41 - PLBS - Plateformes Lilloises en Biologie & Santé, University of Lille, Lille, France
- INSERM, Centre Hospitalier Universitaire (CHU) de Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, LICEND, University of Lille, Lille, France
- Department of Neuroradiology, Centre Hospitalier Universitaire (CHU) de Lille, Lille, France
| | - Sönke Langner
- Institute of Diagnostic and Interventional Radiology, Pediatric Radiology and Neuroradiology, University Medical Center Rostock, Rostock, Germany
| | - Anne-Marie Wenzel
- Institute of Diagnostic and Interventional Radiology, Pediatric Radiology and Neuroradiology, University Medical Center Rostock, Rostock, Germany
- German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Network of Centers of Excellence in Neurodegeneration (CoEN) Center Rostock, Rostock, Germany
| | - Michael Walter
- Institute of Clinical Chemistry and Laboratory Medicine, Rostock University Medical Center, Rostock, Germany
| | - Marc-André Weber
- Institute of Diagnostic and Interventional Radiology, Pediatric Radiology and Neuroradiology, University Medical Center Rostock, Rostock, Germany
| | - Alexander Storch
- Department of Neurology, Rostock University Medical Center, Gehlsheimer Str. 20, 18147, Rostock, Germany
- German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Network of Centers of Excellence in Neurodegeneration (CoEN) Center Rostock, Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - David Devos
- INSERM, Centre Hospitalier Universitaire (CHU) de Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, LICEND, University of Lille, Lille, France
- Neurology and Movement Disorders Department, Reference Center for Parkinson's Disease, Lille Center of Excellence for Neurodegenerative Disorders (LiCEND), Network of Centers of Excellence in Neurodegeneration (CoEN) Center, Centre Hospitalier Universitaire (CHU) de Lille, Lille, France
- Department of Pharmacology, Centre Hospitalier Universitaire (CHU) de Lille, Lille, France
| | - Uwe Walter
- Department of Neurology, Rostock University Medical Center, Gehlsheimer Str. 20, 18147, Rostock, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Network of Centers of Excellence in Neurodegeneration (CoEN) Center Rostock, Rostock, Germany.
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany.
| |
Collapse
|
26
|
Feng Y, Wang X, Li P, Shi X, Prokosch V, Liu H. Exogenous hydrogen sulfide and NOX2 inhibition mitigate ferroptosis in pressure-induced retinal ganglion cell damage. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167705. [PMID: 39914725 DOI: 10.1016/j.bbadis.2025.167705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 01/17/2025] [Accepted: 01/30/2025] [Indexed: 02/09/2025]
Abstract
Glaucoma, a leading cause of irreversible blindness worldwide, is characterized by the progressive degeneration of retinal ganglion cells (RGCs). While elevated intraocular pressure (IOP) significantly contributes to disease progression, managing IOP alone does not completely halt it. The mechanisms underlying RGCs loss in glaucoma remain unclear, but ferroptosis-an iron-dependent form of oxidative cell death-has been implicated, particularly in IOP-induced RGCs loss. There is an urgent need for neuroprotective treatments. Our previous research showed that hydrogen sulfide (H2S) protects RGCs against glaucomatous injury. This study aims to investigate the interplay between elevated pressure, mitochondrial dysfunction, iron homeostasis, and ferroptosis in RGCs death, focusing on how H2S may mitigate pressure-induced ferroptosis and protect RGCs. We demonstrate alterations in iron metabolism and mitochondrial function in a subacute IOP elevation model in vivo. In vitro, we confirm that elevated pressure, iron overload, and mitochondrial dysfunction lead to RGCs loss, increased retinal ferrous iron and total iron content, and heightened reactive oxygen species (ROS). Notably, pressure increases NADPH oxidase 2 (NOX2) and decreases glutathione peroxidase 4 (GPX4), a key regulator of ferroptosis. NOX2 deletion or inhibition by H2S prevents pressure-induced RGCs loss and ferroptosis. Our findings reveal that H2S chelates iron, regulates iron metabolism, reduces oxidative stress, and mitigates ferroptosis, positioning slow-releasing H2S donors are positioning as a promising multi-target therapy for glaucoma, with NOX2 emerging as a key regulator of ferroptosis.
Collapse
Affiliation(s)
- Yuan Feng
- Department of Ophthalmology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany
| | - Xiaosha Wang
- Department of Ophthalmology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany
| | - Panpan Li
- Department of Ophthalmology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany
| | - Xin Shi
- Department of Ophthalmology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany
| | - Verena Prokosch
- Department of Ophthalmology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany
| | - Hanhan Liu
- Department of Ophthalmology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany.
| |
Collapse
|
27
|
Xie Y, Tao Z, Wang B, Zhao Y, Chen X, Li B, Wang J, Chen G, Hu X. Ferroptosis-related genes as prognostic markers for survival and immunotherapy in triple-negative breast cancer: analysis of public databases and a single institution. Ther Adv Med Oncol 2025; 17:17588359251322291. [PMID: 40034604 PMCID: PMC11873862 DOI: 10.1177/17588359251322291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/05/2025] [Indexed: 03/05/2025] Open
Abstract
Background Ferroptosis plays a vital role in cancer development and treatment. The relationship between ferroptosis-related genes and breast cancer prognosis, as well as immunotherapy outcomes, remains unknown. Objectives To evaluate the prognostic value of ferroptosis-related genes in breast cancer. Methods We conducted differential expressions and prognostic analysis for ferroptosis-related genes on public databases and breast cancer patients in our center and analyzed their predictive value for immunotherapy of breast cancer patients. Results We identified prognostic ferroptosis-related genes, constructed a nomogram, and validated key genes using patient data from our center. We also investigated ferroptosis-related genes significantly associated with immune infiltration and identified FTH1 as a promising biomarker for triple-negative breast cancer immunotherapy. Conclusion Ferroptosis-related genes had potential prognostic value and predictive value for breast cancer immunotherapy.
Collapse
Affiliation(s)
- Yizhao Xie
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhonghua Tao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Biyun Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yannan Zhao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinyu Chen
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bin Li
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jinyan Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guangliang Chen
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xichun Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 270 Dongan Road, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
28
|
Li X, Pei R, Fei Z, Chen Z, Lin F, Sun P, Cao H. Could Blood Transfusion Increase the Risk of Alzheimer's Disease? A Narrative Review. Healthcare (Basel) 2025; 13:452. [PMID: 40077014 PMCID: PMC11898722 DOI: 10.3390/healthcare13050452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/05/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Alzheimer's disease (AD) is the most common progressive neurodegenerative disease, and its pathogenesis is complex. In addition to amyloid-β and phosphorylated tau, inflammation and microbial infections also play a role in the development of AD. Currently, there is no effective clinical intervention to cure AD or completely halt its progression. Blood transfusion, a critical life-saving medical procedure widely employed in modern healthcare, faces growing demand due to global population aging. However, whether blood transfusion could increase the risk of AD is still not clear. Aβ and tau play major roles in the pathogenesis of AD and may possess the potential for transmission through blood transfusion. Iron overload and chronic inflammation, which can independently influence AD pathogenesis, may result from repeated transfusions. Additionally, herpesvirus, known to accelerate AD progression, can also be potentially transmitted by blood transfusion. In this study, recent advances in the associations between blood transfusion and the occurrence and development of AD were reviewed, and whether blood transfusion could increase the risk of AD was discussed. Furthermore, the related proposals for blood management and future research were advanced to provide references for the prevention and control of AD.
Collapse
Affiliation(s)
| | | | | | | | | | - Pan Sun
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu 610052, China; (X.L.); (R.P.); (Z.F.); (Z.C.); (F.L.)
| | - Haijun Cao
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu 610052, China; (X.L.); (R.P.); (Z.F.); (Z.C.); (F.L.)
| |
Collapse
|
29
|
Li J, Tao L, Zhou Y, Zhu Y, Li C, Pan Y, Yao P, Qian X, Liu J. Identification of biomarkers in Alzheimer's disease and COVID-19 by bioinformatics combining single-cell data analysis and machine learning algorithms. PLoS One 2025; 20:e0317915. [PMID: 39965013 PMCID: PMC11835241 DOI: 10.1371/journal.pone.0317915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 01/07/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Since its emergence in 2019, COVID-19 has become a global epidemic. Several studies have suggested a link between Alzheimer's disease (AD) and COVID-19. However, there is little research into the mechanisms underlying these phenomena. Therefore, we conducted this study to identify key genes in COVID-19 associated with AD, and evaluate their correlation with immune cells characteristics and metabolic pathways. METHODS Transcriptome analyses were used to identify common biomolecular markers of AD and COVID-19. Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were performed on gene chip datasets (GSE213313, GSE5281, and GSE63060) from AD and COVID-19 patients to identify genes associated with both conditions. Gene ontology (GO) enrichment analysis identified common molecular mechanisms. The core genes were identified using machine learning. Subsequently, we evaluated the relationship between these core genes and immune cells and metabolic pathways. Finally, our findings were validated through single-cell analysis. RESULTS The study identified 484 common differentially expressed genes (DEGs) by taking the intersection of genes between AD and COVID-19. The black module, containing 132 genes, showed the highest association between the two diseases according to WGCNA. GO enrichment analysis revealed that these genes mainly affect inflammation, cytokines, immune-related functions, and signaling pathways related to metal ions. Additionally, a machine learning approach identified eight core genes. We identified links between these genes and immune cells and also found a association between EIF3H and oxidative phosphorylation. CONCLUSION This study identifies shared genes, pathways, immune alterations, and metabolic changes potentially contributing to the pathogenesis of both COVID-19 and AD.
Collapse
Affiliation(s)
- Juntu Li
- Department of Critical Care Medicine and Emergency, The Affiliated Suzhou Hospital of Nanjing Medical University (Suzhou Municipal Hospital), Gusu School, Nanjing Medical University, Suzhou Clinical Medical Center of Critical Care Medicine, Suzhou, Jiangsu, China
| | - Linfeng Tao
- Department of Critical Care Medicine and Emergency, The Affiliated Suzhou Hospital of Nanjing Medical University (Suzhou Municipal Hospital), Gusu School, Nanjing Medical University, Suzhou Clinical Medical Center of Critical Care Medicine, Suzhou, Jiangsu, China
| | - Yanyou Zhou
- Department of Critical Care Medicine and Emergency, The Affiliated Suzhou Hospital of Nanjing Medical University (Suzhou Municipal Hospital), Gusu School, Nanjing Medical University, Suzhou Clinical Medical Center of Critical Care Medicine, Suzhou, Jiangsu, China
| | - Yue Zhu
- Department of Breast and Thyroid Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University (Suzhou Municipal Hospital), Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Chao Li
- Department of Critical Care Medicine and Emergency, The Affiliated Suzhou Hospital of Nanjing Medical University (Suzhou Municipal Hospital), Gusu School, Nanjing Medical University, Suzhou Clinical Medical Center of Critical Care Medicine, Suzhou, Jiangsu, China
| | - Yiyuan Pan
- Department of Critical Care Medicine and Emergency, The Affiliated Suzhou Hospital of Nanjing Medical University (Suzhou Municipal Hospital), Gusu School, Nanjing Medical University, Suzhou Clinical Medical Center of Critical Care Medicine, Suzhou, Jiangsu, China
| | - Ping Yao
- Department of Critical Care Medicine and Emergency, The Affiliated Suzhou Hospital of Nanjing Medical University (Suzhou Municipal Hospital), Gusu School, Nanjing Medical University, Suzhou Clinical Medical Center of Critical Care Medicine, Suzhou, Jiangsu, China
| | - Xuefeng Qian
- Department of Critical Care Medicine and Emergency, The Affiliated Suzhou Hospital of Nanjing Medical University (Suzhou Municipal Hospital), Gusu School, Nanjing Medical University, Suzhou Clinical Medical Center of Critical Care Medicine, Suzhou, Jiangsu, China
| | - Jun Liu
- Department of Critical Care Medicine and Emergency, The Affiliated Suzhou Hospital of Nanjing Medical University (Suzhou Municipal Hospital), Gusu School, Nanjing Medical University, Suzhou Clinical Medical Center of Critical Care Medicine, Suzhou, Jiangsu, China
| |
Collapse
|
30
|
Feng W, Weng Y, Shi W, Liang S, Liao X, Chu R, Ai Q, Mai K, Wan M. Aquatic high iron induces hepatic ferroptosis in zebrafish (Danio rerio) via interleukin-22 signaling pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 367:125627. [PMID: 39746632 DOI: 10.1016/j.envpol.2024.125627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/26/2024] [Accepted: 12/31/2024] [Indexed: 01/04/2025]
Abstract
Iron is one of the indispensable trace elements in living organisms. However, excessive iron deposition in organisms is prone to induce dysfunction of the liver and other vital organs. The present study aimed to investigate the mechanism how aquatic high iron affects iron transport and induces hepatic injury in zebrafish. Our results showed that the iron levels in zebrafish liver and serum were significantly increased after the fish treated with aquatic high iron (200 mg/L ferric ammonium citrate, FAC) for 21 days. Meanwhile, hepatic fibrosis was observed in zebrafish with high iron treatment. Furthermore, the expression of hepcidin, a key factor in the regulation of iron homeostasis, as well as other factors related to iron transport, was significantly influenced by high iron treatment. Nonetheless, different tissues, such as liver, gill and gut, diversely responded to high iron in water. Interestingly, our results identified that the expression of IL-22, instead of IL-6, was significantly elevated after high iron treatment. Moreover, high iron triggered STAT3 phosphorylation via IL-22, leading to the augmented expression of hepcidin and hepatic iron accumulation. As a result, the iron overload in fish liver induced hepatic ferroptosis, marked as the repressed activity of glutathione peroxidase (GPx) and elevated lipid peroxidation. Further studies confirmed that, unlike wild-type (WT) zebrafish, the expression of hepcidin and iron content in the liver of il22-deficient zebrafish was unaffected upon to high iron treatment. At the meantime, hepatic ferroptosis and fibrosis induced by high iron was significantly alleviated in il22-deficient zebrafish. In summary, aquatic high iron induced hepcidin expression in zebrafish by activating the IL-22/STAT3 signaling pathway, which in turn regulated hepatic iron transport and ferroptosis in zebrafish. The present study identified for the first time that IL-22 may be a potential regulatory target for iron overload-induced liver injury.
Collapse
Affiliation(s)
- Wei Feng
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Yizhuo Weng
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Wenkai Shi
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Shufei Liang
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Xinmeng Liao
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Ruixia Chu
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong, 266237, China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong, 266237, China
| | - Min Wan
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong, 266237, China.
| |
Collapse
|
31
|
Cilleros-Holgado P, Gómez-Fernández D, Piñero-Pérez R, Romero-Domínguez JM, Reche-López D, Álvarez-Córdoba M, Romero-González A, López-Cabrera A, De Oliveira MC, Rodríguez-Sacristán A, González-Granero S, García-Verdugo JM, Sánchez-Alcázar JA. Polydatin and Nicotinamide Prevent Iron Accumulation and Lipid Peroxidation in Cellular Models of Mitochondrial Diseases. Antioxidants (Basel) 2025; 14:215. [PMID: 40002401 PMCID: PMC11851670 DOI: 10.3390/antiox14020215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Ferroptosis, an iron-dependent form of non-apoptotic cell death, is regulated by a complex network involving lipid metabolism, iron homeostasis, and the oxidative-reductive system, with iron accumulation and lipid peroxidation as key drivers. Mitochondrial dysfunction and ROS overproduction often underlie the pathogenesis of mitochondrial diseases, for which treatment options are limited, emphasizing the need for novel therapies. In this study, we investigated whether polydatin and nicotinamide could reverse ferroptosis-related pathological features in cellular models derived from patients with pathogenic GFM1 variants. Mutant fibroblasts showed increased iron and lipofuscin accumulation, altered expression of iron metabolism-related proteins, elevated lipid peroxidation, and heightened susceptibility to erastin-induced ferroptosis. Treatment with polydatin and nicotinamide effectively corrected these alterations and reduced iron accumulation and lipid peroxidation in induced neurons. Furthermore, chloramphenicol treatment in control cells mimicked the mutant phenotype, suggesting that these pathological changes are linked to the mitochondrial protein synthesis defect characteristic of pathogenic GFM1 variants. Notably, adding vitamin E to the polydatin and nicotinamide co-treatment resulted in a reduction in the minimum effective concentration, suggesting potential benefits of its inclusion. In conclusion, the combination of polydatin, nicotinamide, and vitamin E could represent a promising therapeutic option for patients with mitochondrial disorders caused by pathogenic GFM1 variants.
Collapse
Affiliation(s)
- Paula Cilleros-Holgado
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - David Gómez-Fernández
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Rocío Piñero-Pérez
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - José Manuel Romero-Domínguez
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Diana Reche-López
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Mónica Álvarez-Córdoba
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Ana Romero-González
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Alejandra López-Cabrera
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Marta Castro De Oliveira
- Neuropediatria, Neurolinkia, C. Jardín de la Isla, 8, Local 4 y 5, 41014 Sevilla, Spain;
- FEA Pediatría, Centro Universitario Hospitalar de Faro, R. Leão Penedo, 8000-386 Faro, Portugal
| | - Andrés Rodríguez-Sacristán
- Neuropediatría, Servicio de Pediatría, Hospital Universitario Virgen Macarena, 41009 Sevilla, Spain;
- Departamento de Farmacología, Radiología y Pediatría de la Facultad de Medicina de la Universidad de Sevilla, 41009 Sevilla, Spain
| | - Susana González-Granero
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia and CIBERNED-ISCIII, 46980 Valencia, Spain; (S.G.-G.); (J.M.G.-V.)
| | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia and CIBERNED-ISCIII, 46980 Valencia, Spain; (S.G.-G.); (J.M.G.-V.)
| | - José Antonio Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| |
Collapse
|
32
|
Fan S, Hu Y, Shi J. Role of ferroptosis in atrial fibrillation: a review. Front Pharmacol 2025; 16:1362060. [PMID: 39981174 PMCID: PMC11839810 DOI: 10.3389/fphar.2025.1362060] [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: 12/27/2023] [Accepted: 01/13/2025] [Indexed: 02/22/2025] Open
Abstract
Cardiovascular disease remains the leading cause of mortality, with atrial fibrillation emerging as one of the most common conditions encountered in clinical practice. However, its underlying mechanisms remain poorly understood, prompting ongoing research. Ferroptosis, a recently discovered form of regulated cell death characterized by lipid peroxidation and disrupted cellular redox balance leading to cell death due to iron overload, has attracted significant attention. Since its identification, ferroptosis has been extensively studied in various contexts, including cancer, stroke, myocardial ischemia/reperfusion injury, and heart failure. Growing evidence suggests that ferroptosis may also play a critical role in the onset and progression of atrial fibrillation, though research in this area is still limited. This article provides a concise overview of the potential mechanisms by which ferroptosis may contribute to the pathogenesis of atrial fibrillation.
Collapse
Affiliation(s)
- Shaowei Fan
- Lugouqiao Second Community Health Service Center, China Aerospace Science & Industry Corporation 731 Hospital, Beijing, China
| | - Yuanhui Hu
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Jingjing Shi
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| |
Collapse
|
33
|
Wang W, Chen J, Zhan L, Zou H, Wang L, Guo M, Gao H, Xu J, Wu W. Iron and ferroptosis in kidney disease: molecular and metabolic mechanisms. Front Immunol 2025; 16:1531577. [PMID: 39975561 PMCID: PMC11835690 DOI: 10.3389/fimmu.2025.1531577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
Maintaining iron homeostasis is necessary for kidney functioning. There is more and more research indicating that kidney disease is often caused by iron imbalance. Over the past decade, ferroptosis' role in mediating the development and progression of renal disorders, such as acute kidney injury (renal ischemia-reperfusion injury, drug-induced acute kidney injury, severe acute pancreatitis induced acute kidney injury and sepsis-associated acute kidney injury), chronic kidney disease (diabetic nephropathy, renal fibrosis, autosomal dominant polycystic kidney disease) and renal cell carcinoma, has come into focus. Thus, knowing kidney iron metabolism and ferroptosis regulation may enhance disease therapy. In this review, we discuss the metabolic and molecular mechanisms of iron signaling and ferroptosis in kidney disease. We also explore the possible targets of ferroptosis in the therapy of renal illness, as well as their existing limitations and future strategies.
Collapse
Affiliation(s)
- Wenjie Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jingdi Chen
- Department of orthopedics, The Airborne Military Hospital, Wuhan, Hubei, China
| | - Liying Zhan
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Handong Zou
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lu Wang
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Mengmeng Guo
- The First Clinical College of Wuhan University, Wuhan, Hubei, China
| | - Hang Gao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jing Xu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wei Wu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
34
|
Chen L, Shen Q, Liu Y, Zhang Y, Sun L, Ma X, Song N, Xie J. Homeostasis and metabolism of iron and other metal ions in neurodegenerative diseases. Signal Transduct Target Ther 2025; 10:31. [PMID: 39894843 PMCID: PMC11788444 DOI: 10.1038/s41392-024-02071-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/24/2024] [Accepted: 11/12/2024] [Indexed: 02/04/2025] Open
Abstract
As essential micronutrients, metal ions such as iron, manganese, copper, and zinc, are required for a wide range of physiological processes in the brain. However, an imbalance in metal ions, whether excessive or insufficient, is detrimental and can contribute to neuronal death through oxidative stress, ferroptosis, cuproptosis, cell senescence, or neuroinflammation. These processes have been found to be involved in the pathological mechanisms of neurodegenerative diseases. In this review, the research history and milestone events of studying metal ions, including iron, manganese, copper, and zinc in neurodegenerative diseases such as Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD), will be introduced. Then, the upstream regulators, downstream effector, and crosstalk of mental ions under both physiologic and pathologic conditions will be summarized. Finally, the therapeutic effects of metal ion chelators, such as clioquinol, quercetin, curcumin, coumarin, and their derivatives for the treatment of neurodegenerative diseases will be discussed. Additionally, the promising results and limitations observed in clinical trials of these metal ion chelators will also be addressed. This review will not only provide a comprehensive understanding of the role of metal ions in disease development but also offer perspectives on their modulation for the prevention or treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Leilei Chen
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Qingqing Shen
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Yingjuan Liu
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Yunqi Zhang
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Liping Sun
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Xizhen Ma
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Ning Song
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Junxia Xie
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China.
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China.
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China.
| |
Collapse
|
35
|
Cornelis MC, Fazlollahi A, Bennett DA, Schneider JA, Ayton S. Genetic Markers of Postmortem Brain Iron. J Neurochem 2025; 169:e16309. [PMID: 39918201 PMCID: PMC11804167 DOI: 10.1111/jnc.16309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025]
Abstract
Brain iron (Fe) dyshomeostasis is implicated in neurodegenerative diseases. Genome-wide association studies (GWAS) have identified plausible loci correlated with peripheral levels of Fe. Systemic organs and the brain share several Fe regulatory proteins but there likely exist different homeostatic pathways. We performed the first GWAS of inductively coupled plasma mass spectrometry measures of postmortem brain Fe from 635 Rush Memory and Aging Project (MAP) participants. Sixteen single nucleotide polymorphisms (SNPs) associated with Fe in at least one of four brain regions were measured (p < 5 × 10-8). Promising SNPs (p < 5 × 10-6) were followed up for replication in published GWAS of blood, spleen, and brain imaging Fe traits and mapped to candidate genes for targeted cortical transcriptomic and epigenetic analysis of postmortem Fe in MAP. Results for SNPs previously associated with other Fe traits were also examined. Ninety-eight SNPs associated with postmortem brain Fe were at least nominally (p < 0.05) associated with one or more related Fe traits. Most novel loci identified had no direct links to Fe regulatory pathways but rather endoplasmic reticulum-Golgi trafficking (SORL1, SORCS2, MARCH1, CLTC), heparan sulfate (HS3ST4, HS3ST1), and coenzyme A (SLC5A6, PANK3); supported by nearest gene function and omic analyses. We replicated (p < 0.05) several previously published Fe loci mapping to candidate genes in cellular and systemic Fe regulation. Finally, novel loci (BMAL, COQ5, SLC25A11) and replication of prior loci (PINK1, PPIF, LONP1) lend support to the role of circadian rhythms and mitochondria function in Fe regulation more generally. In summary, we provide support for novel loci linked to pathways that may have greater relevance to brain Fe accumulation; some of which are implicated in neurodegeneration. However, replication of a subset of prior loci for blood Fe suggests that genetic determinants or biological pathways underlying Fe accumulation in the brain are not completely distinct from those of Fe circulating in the periphery.
Collapse
Affiliation(s)
- Marilyn C. Cornelis
- Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Amir Fazlollahi
- Department of Radiology, Royal Melbourne HospitalUniversity of MelbourneMelbourneVictoriaAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQueenslandAustralia
| | | | | | - Scott Ayton
- The Florey Institute of Neuroscience and Mental HealthMelbourneVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
36
|
Naderi S, Khodagholi F, Janahmadi M, Motamedi F, Torabi A, Batool Z, Heydarabadi MF, Pourbadie HG. Ferroptosis and cognitive impairment: Unraveling the link and potential therapeutic targets. Neuropharmacology 2025; 263:110210. [PMID: 39521042 DOI: 10.1016/j.neuropharm.2024.110210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/24/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Neurodegenerative disorders, such as Alzheimer's and Parkinson's diseases, share key characteristics, notably cognitive impairment and significant cell death in specific brain regions. Cognition, a complex mental process allowing individuals to perceive time and place, is disrupted in these conditions. This consistent disruption suggests the possibility of a shared underlying mechanism across all neurodegenerative diseases. One potential common factor is the activation of pathways leading to cell death. Despite significant progress in understanding cell death pathways, no definitive treatments have emerged. This has shifted focus towards less-explored mechanisms like ferroptosis, which holds potential due to its involvement in oxidative stress and iron metabolism. Unlike apoptosis or necrosis, ferroptosis offers a novel therapeutic avenue due to its distinct biochemical and genetic underpinnings, making it a promising target in neurodegenerative disease treatment. Ferroptosis is distinguished from other cellular death mechanisms, by distinctive characteristics such as an imbalance of iron hemostasis, peroxidation of lipids in the plasma membrane, and dysregulated glutathione metabolism. In this review, we discuss the potential role of ferroptosis in cognitive impairment. We then summarize the evidence linking ferroptosis biomarkers to cognitive impairment brought on by neurodegeneration while highlighting recent advancements in our understanding of the molecular and genetic mechanisms behind the condition. Finally, we discuss the prospective therapeutic implications of targeting ferroptosis for the treatment of cognitive abnormalities associated with neurodegeneration, including natural and synthetic substances that suppress ferroptosis via a variety of mechanisms. Promising therapeutic candidates, including antioxidants and iron chelators, are being explored to inhibit ferroptosis and mitigate cognitive decline.
Collapse
Affiliation(s)
- Soudabeh Naderi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahyar Janahmadi
- Neuroscience Research Center, Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereshteh Motamedi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolfazl Torabi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zehra Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | | | - Hamid Gholami Pourbadie
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
37
|
Zhou Y, Li J, Yuan Y, Zhang H, Luo X, Wang F, Tao Y, Yue J, Huang L, Wu L, Cao Y, Yu Q, He Q. Metrnl/C-KIT Axis Attenuates Early Brain Injury Following Subarachnoid Hemorrhage by Inhibiting Neuronal Ferroptosis. CNS Neurosci Ther 2025; 31:e70286. [PMID: 39981761 PMCID: PMC11843251 DOI: 10.1111/cns.70286] [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: 11/05/2024] [Revised: 01/14/2025] [Accepted: 02/07/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND AND PURPOSE Ferroptosis is a distinct form of cell death characterized by iron-dependent lipid peroxidation and plays a crucial role in the early brain injury (EBI) following subarachnoid hemorrhage (SAH). As a newly discovered endogenous ligand for the C-KIT receptor tyrosine kinase, meteorin-like protein (Metrnl) exerts regulatory functions in oxidative stress and protects against various diseases. However, the specific role of the Metrnl/C-KIT axis in neuronal ferroptosis during EBI following SAH remains to be elucidated. METHODS Sprague Dawley rats were used to establish the SAH model through endovascular perforation. r-Metrnl was administered intranasally 1 h after SAH. Metrnl shRNA, C-KIT inhibitor ISCK03, AMPK inhibitor dorsomorphin, and Nrf2 inhibitor ML385 were administered intracerebroventricularly or intraperitoneally before r-Metrnl treatment to explore the underlying mechanisms. Neurobehavioral assessments, immunofluorescence, western blot, ELISA, Fluoro-Jade C staining, transmission electron microscopy, and Nissl staining were conducted to evaluate the effects. Additionally, primary neuron culture with hemoglobin (Hb) stimulation was used for in vitro studies. RESULTS Phosphorylated C-KIT and endogenous Metrnl levels were upregulated after SAH. Knockdown of Metrnl aggravated neurobehavioral deficits and neuronal ferroptosis, whereas r-Metrnl treatment showed a protective effect. Mechanistically, r-Metrnl significantly increased the protein levels of SLC7A11, GPX4, FTH, FSP1, and GSH, whereas it decreased the levels of ACSL4, 4HNE, and MDA in the ipsilateral hemisphere 24 h after SAH. Also, r-Metrnl reduced mitochondrial shrinkage, increased mitochondrial crista, and decreased membrane density. However, the beneficial effects of r-Metrnl were partially reversed by ISCK03, dorsomorphin, or ML385 treatment both in vivo and in vitro. CONCLUSIONS Our study demonstrated that r-Metrnl reduced neuronal ferroptosis and improved neurological outcomes after SAH by modulating the C-KIT/AMPK/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- You Zhou
- Department of Critical Care Medicine, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Jiani Li
- Department of Neurology, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Ye Yuan
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Xu Luo
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Feng Wang
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Yihao Tao
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Jianhe Yue
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Luyi Huang
- Key Laboratory of Molecular Biology for Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Lei Wu
- Department of NeurologyGuangdong Second Provincial General HospitalGuangzhouGuangdongChina
| | - Yunxing Cao
- Department of Critical Care Medicine, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Qian Yu
- Department of Neurosurgery, School of Medicine, The Second Affiliated HospitalZhejiang UniversityHangzhouZhejiangChina
| | - Qiuguang He
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| |
Collapse
|
38
|
Liu L, Jia P, Liu T, Liang J, Dang Y, Rastegar-Kashkooli Y, Li Q, Liu J, Man J, Zhao T, Xing N, Wang F, Chen X, Zhang J, Jiang C, Zille M, Zhang Z, Fan X, Wang J, Wang J. Metabolic dysfunction contributes to mood disorders after traumatic brain injury. Ageing Res Rev 2025; 104:102652. [PMID: 39746403 DOI: 10.1016/j.arr.2024.102652] [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: 09/05/2024] [Revised: 12/15/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
Traumatic brain injury (TBI) presents significant risks concerning mortality and morbidity. Individuals who suffer from TBI may exhibit mood disorders, including anxiety and depression. Both preclinical and clinical research have established correlations between TBI and disturbances in the metabolism of amino acids, lipids, iron, zinc, and copper, which are implicated in the emergence of mood disorders post-TBI. The purpose of this review is to elucidate the impact of metabolic dysfunction on mood disorders following TBI and to explore potential strategies for mitigating anxiety and depression symptoms. We researched the PubMed and Web of Science databases to delineate the mechanisms by which metabolic dysfunction contributes to mood disorders in the context of TBI. Particular emphasis was placed on the roles of glutamate, kynurenine, lipids, iron, zinc, and copper metabolism. Metabolic dysfunction is linked to mood disorders post-TBI through multiple pathways, encompassing the glutamatergic system, the kynurenine pathway, endocannabinoids, iron deposition, iron-related ferroptosis, zinc deficiency, and copper dysregulation. Furthermore, this review addresses the influence of metabolic dysfunction on mood disorders in the elderly demographic following TBI. Targeting metabolic dysfunction for therapeutic intervention appears promising in alleviating symptoms of anxiety and depression that arise after TBI. While further investigation is warranted to delineate the underlying pathophysiologic mechanisms of mood disorders post-TBI, current evidence underscores the potential contribution of metabolic dysfunction to these conditions. Therefore, rectifying metabolic dysfunction represents a viable and strategic approach to addressing mood disorders following TBI.
Collapse
Affiliation(s)
- Lang Liu
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Peijun Jia
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Tongzhou Liu
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jiaxin Liang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Yijia Dang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Qiang Li
- Department of Neurology, Shanghai Gongli Hospital of Pudong New Area, Shanghai 200135, China.
| | - Jingqi Liu
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jiang Man
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Ting Zhao
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Fushun Wang
- Department of Psychology, Sichuan Normal University, Chengdu, Sichuan 610060, China.
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jiewen Zhang
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Chao Jiang
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Marietta Zille
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna 1090, Austria.
| | - Zhenhua Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
39
|
Balk S, Panier F, Brandner S, Coras R, Blümcke I, Ekici AB, Sembill JA, Schwab S, Huttner HB, Sprügel MI. Intracerebral Hemorrhage-Associated Iron Release Leads to Pericyte-Dependent Cerebral Capillary Function Disruption. Biomolecules 2025; 15:164. [PMID: 40001467 PMCID: PMC11852616 DOI: 10.3390/biom15020164] [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: 11/30/2024] [Revised: 01/11/2025] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
Intracerebral hemorrhage leads to immediate brain injury due to local mechanical damage, on which current treatment approaches are focused, but it also induces secondary brain injury. The purpose of this study is to characterize blood components, degradation products and their effects in secondary brain injury. Immunocyto- and immunohistochemistry, Fluorescence-Activated Cell Sorting, WST-1 assays and RNA sequencing were applied using human cell cultures and human ex vivo brain tissue slices. Brain tissue was immediately collected, cooled and sliced during neurosurgical operations to perform experiments on living tissue slices of the human brain. Among the blood degradation products, free iron (Fe2+ and Fe3+), but not hemoglobin, induced detrimental effects on pericyte function and survival (78.5% vs. 94.3%; p-value < 0.001). RNA sequencing revealed ferroptosis as the underlining cellular mechanism, mediated via GPX-4 (log2 fold change > 1.0, p-value < 1.08 × 10-30) in pathway analysis and eventually resulting in oxidative cell death. Pericytes located at cerebral capillary branching sites were specifically affected by ferroptosis, leading to capillary disruption and vasoconstriction, which were partially prevented by ferrostatin-1. Free iron induces the pericyte-dependent disruption of cerebral capillary function and represents a therapeutic target to attenuate secondary injury after intracerebral hemorrhage.
Collapse
Affiliation(s)
- Stefanie Balk
- Department of Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Franziska Panier
- Department of Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Sebastian Brandner
- Department of Neurosurgery, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany
- Department of Neurosurgery, Fürth Hospital, Jakob-Henle-Straße 1, 90766 Fürth, Germany
| | - Roland Coras
- Department of Neuropathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Ingmar Blümcke
- Department of Neuropathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Arif B. Ekici
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaulallee 4, 91054 Erlangen, Germany
| | - Jochen A. Sembill
- Department of Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Stefan Schwab
- Department of Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Hagen B. Huttner
- Department of Neurology, Justus-Liebig-University Giessen, Klinikstraße 33, 35392 Gießen, Germany
| | - Maximilian I. Sprügel
- Department of Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany
| |
Collapse
|
40
|
Kuziak A, Heczko P, Pietrzyk A, Strus M. Iron Homeostasis Dysregulation, Oro-Gastrointestinal Microbial Inflammatory Factors, and Alzheimer's Disease: A Narrative Review. Microorganisms 2025; 13:122. [PMID: 39858890 PMCID: PMC11767265 DOI: 10.3390/microorganisms13010122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/23/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
Alzheimer's disease (AD), the most common form of dementia, is a progressive neurodegenerative disorder that profoundly impacts cognitive function and the nervous system. Emerging evidence highlights the pivotal roles of iron homeostasis dysregulation and microbial inflammatory factors in the oral and gut microbiome as potential contributors to the pathogenesis of AD. Iron homeostasis disruption can result in excessive intracellular iron accumulation, promoting the generation of reactive oxygen species (ROS) and oxidative damage. Additionally, inflammatory agents produced by pathogenic bacteria may enter the body via two primary pathways: directly through the gut or indirectly via the oral cavity, entering the bloodstream and reaching the brain. This infiltration disrupts cellular homeostasis, induces neuroinflammation, and exacerbates AD-related pathology. Addressing these mechanisms through personalized treatment strategies that target the underlying causes of AD could play a critical role in preventing its onset and progression.
Collapse
Affiliation(s)
- Agata Kuziak
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, św. Łazarza 16 Street, 31-008 Cracow, Poland;
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Cracow, Poland; (P.H.); (A.P.)
| | - Piotr Heczko
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Cracow, Poland; (P.H.); (A.P.)
| | - Agata Pietrzyk
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Cracow, Poland; (P.H.); (A.P.)
| | - Magdalena Strus
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18 Street, 31-121 Cracow, Poland; (P.H.); (A.P.)
| |
Collapse
|
41
|
Zhao H, Ji QH, Jia ZZ, Shen LH. Association between deep gray matter iron deposition and clinical symptoms in Parkinson's disease: a quantitative susceptibility mapping study. Front Neurol 2025; 15:1442903. [PMID: 39835146 PMCID: PMC11743366 DOI: 10.3389/fneur.2024.1442903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025] Open
Abstract
Purpose This study aimed to assess the association between motor and non-motor symptoms of Parkinson's disease (PD) and iron accumulation within the deep gray matter of the brain by Quantitative Susceptibility Mapping (QSM). Methods Fifty-six PD patients and twenty-nine healthy controls were recruited in this study. According to the Hoehn and Yahr (H-Y) stage score, PD patients were divided into early stage (H-Y ≤ 2) and advanced stage (H-Y > 2) groups. Specifically, the Regions of Interest (ROIs) encompassed the substantia nigra (SN), red nucleus (RN), caudate nucleus (CN), globus pallidus (GP) and putamen (PT). Meanwhile, various rating scales were used to assess the clinical symptoms of PD. Results Compared to healthy controls (HCs), PD patients showed a significant increase in magnetic susceptibility values (MSVs) within the SN and GP. Further comparisons indicated that the MSVs of the SN, PT, GP and CN are all higher in advanced stages than in early stages. Significant positive correlations were observed between the MSVs of the SN and scores on the UPDRS-III, HAMA, and HAMD (r = 0.310, p = 0.020; r = 0.273, p = 0.042; r = 0.342, p = 0.010, respectively). Likewise, the MSVs of the GP demonstrated notable correlations with HAMA and HAMD scores (r = 0.275, p = 0.040; r = 0.415, p = 0.001). Additionally, a significant correlation was found between the MSVs of the PT and HAMD scores (r = 0.360, p = 0.006). Furthermore, we identified a significant negative correlation between MMSE scores and the MSVs of both the PT and GP (r = -0.268, p = 0.046; r = -0.305, p = 0.022). Conclusion Our study revealed that QSM possesses the capability to serve as a biomarker for PD. Significant correlations were found between clinical features and the iron deposition in the nigrostriatal system.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Neurology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Department of Neurology, Affiliated Rudong Hospital of Xinglin College, Nantong University, Nantong, China
| | - Qiu-Hong Ji
- Department of Neurology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Zhong-Zheng Jia
- Department of Medical Imaging, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Li-Hua Shen
- Department of Neurology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| |
Collapse
|
42
|
Shibata H, Uchida Y, Kan H, Sakurai K, Madokoro Y, Iwano S, Maurya SK, Muñoz-González Á, Ardakani I, Yamada K, Matsukawa N. Deep-learning assessment of hippocampal magnetic susceptibility in Alzheimer's disease. J Alzheimers Dis 2025; 103:293-304. [PMID: 39584366 DOI: 10.1177/13872877241300278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
BACKGROUND Quantitative susceptibility mapping (QSM) is pivotal for analyzing neurodegenerative diseases. However, accurate hippocampal segmentation remains a challenge. OBJECTIVE This study introduces a method for extracting hippocampal magnetic susceptibility values using a convolutional neural network (CNN) model referred to as 3D residual UNET. METHODS The model was pre-trained on whole QSM images and hippocampal segmentations from 3D T1-weighted images of 297 patients with Alzheimer's disease and mild cognitive impairment. Fine-tuning was conducted through manually annotated hippocampal segmentations from the QSM images of 60 patients. The performance was assessed using the Dice similarity coefficient (DSC) and Pearson correlation coefficient. RESULTS The developed model was applied to another 98 patients, 49 with AD and 49 with mild cognitive impairment (MCI), and the correlation between the hippocampal magnetic susceptibility and volume was evaluated. The mean DSC for the hippocampal segmentation model was 0.716 ± 0.045. The correlation coefficient between the magnetic susceptibility values derived from manual segmentation and the CNN model was 0.983. The Pearson correlation coefficient between magnetic susceptibility and hippocampal volume from the CNN model was -0.252 (p = 0.012) on the left side and -0.311 (p = 0.002) on the right. CONCLUSIONS The 3D residual UNET model enhances hippocampal analysis precision using QSM, which is capable of accurately extracting magnetic susceptibility.
Collapse
Affiliation(s)
- Haruto Shibata
- Department of Neurology, Nagoya City University East Medical Center, Chikusa-ku, Nagoya, Japan
- Department of Neurology, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya, Japan
| | - Yuto Uchida
- Department of Neurology, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya, Japan
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hirohito Kan
- Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Keita Sakurai
- Department of Radiology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yuta Madokoro
- Department of Neurology, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya, Japan
| | | | | | | | | | - Kentaro Yamada
- Department of Neurology, Nagoya City University East Medical Center, Chikusa-ku, Nagoya, Japan
| | - Noriyuki Matsukawa
- Department of Neurology, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya, Japan
| |
Collapse
|
43
|
Zhang B, Li L, Wang N, Zhu Z, Wang M, Tan WP, Liu J, Zhou S. A new pathway for ferroptosis regulation: The PRMTs. Int J Biol Macromol 2025; 285:138143. [PMID: 39622375 DOI: 10.1016/j.ijbiomac.2024.138143] [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: 09/28/2024] [Revised: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024]
Abstract
Protein arginine methyltransferases (PRMTs) play an essential role in the regulation of ferroptosis, a form of programmed cell death characterized by abnormal iron ion metabolism, lipid peroxidation, and DNA damage. Through methylation, PRMTs modify specific proteins, thereby altering their activity, localizations, or interactions with other molecules to control the ferroptosis process. This study was conducted to provide a comprehensive overview of the relationship between PRMTs and ferroptosis, with a focus on the mechanisms by which PRMTs regulate ferroptosis and their effect on this cell death pathway. Currently, only a few studies have been conducted on the regulation of ferroptosis by PRMTs. However, this review provides insights into the effects of PRMTs on ferroptosis regulators, suggesting that the regulation of ferroptosis by PRMTs holds potential as a new therapeutic target for related diseases.
Collapse
Affiliation(s)
- Bei Zhang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin 541199, China; Basic Medical College, Guilin Medical College, Guilin 541199, China
| | - Luyao Li
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin 541199, China; Basic Medical College, Guilin Medical College, Guilin 541199, China
| | - Nan Wang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin 541199, China; Basic Medical College, Guilin Medical College, Guilin 541199, China
| | - Zixuan Zhu
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin 541199, China; Basic Medical College, Guilin Medical College, Guilin 541199, China
| | - Mingyang Wang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin 541199, China; Basic Medical College, Guilin Medical College, Guilin 541199, China
| | - Wu Peng Tan
- Department of Gynaecology, Maternal and Child Health Hospital of Hengyang, Hengyang 421001, China
| | - Jianfeng Liu
- Department of Pediatrics, The Second Affiliated Hospital of South China University, Hengyang 421001, China
| | - Shouhong Zhou
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin 541199, China; Basic Medical College, Guilin Medical College, Guilin 541199, China.
| |
Collapse
|
44
|
Zhao Q, Maimaitiaili S, Bi Y, Li M, Li X, Li Q, Shen X, Wu M, Fu L, Zhu Z, Zhang X, Chen J, Hu A, Zhang Z, Zhang W, Zhang B. Brain Iron Deposition Alterations in Type 2 Diabetes Mellitus Patients With Mild Cognitive Impairment Based on Quantitative Susceptibility Mapping. J Diabetes 2025; 17:e70052. [PMID: 39843980 PMCID: PMC11753919 DOI: 10.1111/1753-0407.70052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 12/12/2024] [Accepted: 12/31/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Iron is one of the most important elements in brain that may has a direct impact on the stability of central nervous system. The current study devoted to explore the alterations of iron distribution across the whole brain in type 2 diabetes mellitus (T2DM) patients with mild cognitive impairment (MCI). METHODS The quantitative susceptibility mapping (QSM) technique was used to quantify the intracranial iron content of 74 T2DM patients with MCI and 86 T2DM patients with normal cognition (NC). The group comparison was performed by a voxel-based analysis. Then we evaluated the relationships between cognitive indicators and magnetic susceptibility value (MSV) measured by QSM of the significant brain areas, which were set as the regions of interest (ROIs). In addition, we analyzed the moderation effects of grey matter volume (GMV) of the related brain areas and several metabolic and cerebrovascular factors on the associations between MSV of ROIs and cognitive characteristics. RESULTS T2DM patients with MCI exhibited a lower MSV in the right middle temporal gyrus (MTG) compared to NC group. And in the MCI group, there were significantly negative correlations between MSV of the right MTG and several memory indexes. Furthermore, the moderation effects of GMV of the whole brain and the bilateral MTG on the relationship between MSV of the right MTG and scores of list recognition were significant. CONCLUSIONS T2DM patients with MCI had a temporary decreased iron content in the right MTG, which may partially compensate for cognitive impairment. TRIAL REGISTRATION The study was registered at Clinicaltrials.gov (NCT02738671).
Collapse
Affiliation(s)
- Qiuyue Zhao
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Subinuer Maimaitiaili
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Yan Bi
- Department of Endocrinology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Ming Li
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Xin Li
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Qian Li
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Xinyi Shen
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Min Wu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Linqing Fu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Zhengyang Zhu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Xin Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Jiu Chen
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Anning Hu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Zhou Zhang
- Department of Endocrinology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Wen Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| | - Bing Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjing City, Jiangsu ProvinceChina
- Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjing City, Jiangsu ProvinceChina
| |
Collapse
|
45
|
Streit WJ, Phan L, Bechmann I. Ferroptosis and pathogenesis of neuritic plaques in Alzheimer disease. Pharmacol Rev 2025; 77:100005. [PMID: 39952690 DOI: 10.1124/pharmrev.123.000823] [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: 12/14/2023] [Revised: 07/25/2024] [Accepted: 09/13/2024] [Indexed: 10/09/2024] Open
Abstract
Neuritic plaques are pathognomonic and terminal lesions of Alzheimer disease (AD). They embody AD pathogenesis because they harbor in one space critical pathologic features of the disease: amyloid deposits, neurofibrillary degeneration, neuroinflammation, and iron accumulation. Neuritic plaques are thought to arise from the conversion of diffuse extracellular deposits of amyloid-β protein (Aβ), and it is believed that during conversion, amyloid toxicity creates the dystrophic neurites of neuritic plaques, as well as neurofibrillary tangles However, recent evidence from human postmortem studies suggests a much different mechanism of neuritic plaque formation, where the first step in their creation is neuronal degeneration driven by iron overload and ferroptosis. Similarly, neurofibrillary tangles represent the corpses of iron-laden neurons that develop independently of Aβ deposits. In this review, we will focus on the role of free redox-active iron in the development of typical AD pathology, as determined largely by evidence obtained in the human temporal lobe during early, preclinical stages of AD. The findings have allowed the construction of a scheme of AD pathogenesis where brain iron is center stage and is involved in every step of the sequence of events that produce characteristic AD pathology. We will discuss how the study of preclinical AD has produced a fresh and revised assessment of AD pathogenesis that may be important for reconsidering current therapeutic efforts and guiding future ones. SIGNIFICANCE STATEMENT: This review offers a novel perspective on Alzheimer disease pathogenesis where elevated brain iron plays a central role and is involved throughout the development of lesions. Herein, we review arguments against the amyloid cascade theory and explain how recent findings in humans during early preclinical disease support iron-mediated cell death and endogenous iron containment mechanisms as critical components of neuritic plaque formation and ensuing dementia.
Collapse
Affiliation(s)
- Wolfgang J Streit
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, Florida.
| | - Leah Phan
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, Florida
| | - Ingo Bechmann
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| |
Collapse
|
46
|
Zhang W, Yan Y, Yi C, Jiang X, Guo L, Huang S, Xia T, Huang F, Jiao Y, Li H, Yu B, Dai Y. Targeting ferroptosis in the neurovascular unit: A promising approach for treating diabetic cognitive impairment. Int Immunopharmacol 2024; 142:113146. [PMID: 39298819 DOI: 10.1016/j.intimp.2024.113146] [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: 05/02/2024] [Revised: 08/12/2024] [Accepted: 09/08/2024] [Indexed: 09/22/2024]
Abstract
The cognitive decline associated with chronic metabolic disease diabetes has garnered extensive scrutiny, yet its pathogenesis remains incompletely understood, and the advancement of targeted therapeutics has posed a persistent challenge. Ferroptosis, a novel form of cell death characterized by intracellular lipid peroxidation and iron overload, has recently emerged as a significant factor. Numerous contemporary studies have corroborated that ferroptosis within the neurovascular unit is intimately associated with the onset of diabetes-induced cognitive impairment. Numerous contemporary studies have corroborated that ferroptosis within the neurovascular unit is intimately associated with the onset of diabetic cognitive impairment (DCI). This article initially conducts a profound analysis of the mechanism of ferroptosis, followed by a detailed elucidation of the specific manifestations of neurovascular unit ferroptosis in the context of diabetic cognitive function impairment. Furthermore, an exhaustive review of pertinent literature from April 2020 to March 2024 has been undertaken, resulting in the selection of 31 documents of significant reference value. These documents encompass studies on 11 distinct drugs, all of which are centered around investigating methods to inhibit the ferroptosis pathway as a potential treatment for DCI. Simultaneously, we conducted a review of 12 supplementary literary sources that presented 10 pharmacological agents with anti-ferroptosis properties in other neurodegenerative disorders. This article critically examines the potential influence of neurovascular unit ferroptosis on the progression of cognitive impairment in diabetes, from the three aforementioned perspectives, and organizes the existing and potential therapeutic drugs. It is our aspiration that this article will serve as a theoretical foundation for scholars in related disciplines when conceptualizing, investigating, and developing novel clinical drugs for DCI.
Collapse
Affiliation(s)
- Wenlan Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yijing Yan
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chunmei Yi
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lin Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shanshan Huang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Tong Xia
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Fayin Huang
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yike Jiao
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Huhu Li
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Bin Yu
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yongna Dai
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
47
|
Ferretti S, Zanella I. The Underestimated Role of Iron in Frontotemporal Dementia: A Narrative Review. Int J Mol Sci 2024; 25:12987. [PMID: 39684697 DOI: 10.3390/ijms252312987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
The term frontotemporal dementia (FTD) comprises a group of neurodegenerative disorders characterized by the progressive degeneration of the frontal and temporal lobes of the brain with language impairment and changes in cognitive, behavioral and executive functions, and in some cases motor manifestations. A high proportion of FTD cases are due to genetic mutations and inherited in an autosomal-dominant manner with variable penetrance depending on the implicated gene. Iron is a crucial microelement that is involved in several cellular essential functions in the whole body and plays additional specialized roles in the central nervous system (CNS) mainly through its redox-cycling properties. Such a feature may be harmful under aerobic conditions, since it may lead to the generation of highly reactive hydroxyl radicals. Dysfunctions of iron homeostasis in the CNS are indeed involved in several neurodegenerative disorders, although it is still challenging to determine whether the dyshomeostasis of this essential but harmful metal is a direct cause of neurodegeneration, a contributor factor or simply a consequence of other neurodegenerative mechanisms. Unlike many other neurodegenerative disorders, evidence of the dysfunction in brain iron homeostasis in FTD is still scarce; nonetheless, the recent literature intriguingly suggests its possible involvement. The present review aims to summarize what is currently known about the contribution of iron dyshomeostasis in FTD based on clinical, imaging, histological, biochemical and molecular studies, further suggesting new perspectives and offering new insights for future investigations on this underexplored field of research.
Collapse
Affiliation(s)
- Sara Ferretti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Isabella Zanella
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Medical Genetics Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| |
Collapse
|
48
|
Zou Z, Yu Q, Yang Y, Wang F, Zhu P, Zhang X, Zhang J. Cytoglobin attenuates melanoma malignancy but protects melanoma cells from ferroptosis. Mol Med Rep 2024; 30:219. [PMID: 39370785 PMCID: PMC11465429 DOI: 10.3892/mmr.2024.13343] [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: 07/31/2024] [Accepted: 08/23/2024] [Indexed: 10/08/2024] Open
Abstract
Cutaneous malignant melanoma is the most aggressive and the deadliest form of skin cancer. There are two types of limitations which universally exist in current melanoma therapy: Adverse effects and reduced efficiency. Cytoglobin (CYGB), an iron hexacoordinated globin, is highly enriched in melanocytes and frequently epigenetically silenced during melanoma genesis. The present study aimed to explore its potential role as a biomarker for ferroptosis treatment. It was observed that B16F10 and A375 melanoma cells with loss of CYGB expression were highly sensitive to ferroptosis inducers RSL3 and erastin, whereas G361 melanoma cells with highly enriched CYGB were resistant to RSL3 or erastin. Ectopically overexpressed CYGB rendered B16F10 and A375 cells resistant to RSL3 or erastin, accompanied by decreased proliferation and epithelial‑mesenchymal transition (EMT). By contrast, knockdown of CYGB expression made G361 cells sensitive to ferroptosis induction but induced proliferation and EMT progression of G361 cells. Mechanistically, CYGB‑induced resistance of melanoma cells to ferroptosis may have been associated, in part, with i) Suppression of EMT; ii) upregulation of glutathione peroxidase 4 expression; iii) decrease of labile iron pool. In vivo study also demonstrated that CYGB overexpression rendered xenograft melanoma much more resist to RSL3 treatment. Based on these findings, CYGB is a potential therapeutic biomarker to screen the melanoma patients who are most likely benefit from ferroptosis treatment.
Collapse
Affiliation(s)
- Zuquan Zou
- Department of Health, Beilun District Center for Disease Control and Prevention, Ningbo, Zhejiang 315899, P.R. China
- Zhejiang Key Laboratory of Pathophysiology, Medical School, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Qingyao Yu
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang 315832, P.R. China
| | - Yong Yang
- Department of Clinical Laboratory of The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315020, P.R. China
| | - Feng Wang
- Department of Laboratory Medicine, Lihuili Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Pan Zhu
- Department of Health, Beilun District Center for Disease Control and Prevention, Ningbo, Zhejiang 315899, P.R. China
| | - Xiaohong Zhang
- Zhejiang Key Laboratory of Pathophysiology, Medical School, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jinjie Zhang
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang 315832, P.R. China
| |
Collapse
|
49
|
Tian M, Huang X, Li M, Lou P, Ma H, Jiang X, Zhou Y, Liu Y. Ferroptosis in diabetic cardiomyopathy: from its mechanisms to therapeutic strategies. Front Endocrinol (Lausanne) 2024; 15:1421838. [PMID: 39588340 PMCID: PMC11586197 DOI: 10.3389/fendo.2024.1421838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/15/2024] [Indexed: 11/27/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is defined as structural and functional cardiac abnormalities in diabetes, and cardiomyocyte death is the terminal event of DCM. Ferroptosis is iron-dependent oxidative cell death. Evidence has indicated that iron overload and ferroptosis play important roles in the pathogenesis of DCM. Mitochondria, an important organelle in iron homeostasis and ROS production, play a crucial role in cardiomyocyte ferroptosis in diabetes. Studies have shown some anti-diabetic medicines, plant extracts, and ferroptosis inhibitors might improve DCM by alleviating ferroptosis. In this review, we systematically reviewed the evidence of ferroptosis in DCM. Anti-ferroptosis might be a promising therapeutic strategy for the treatment of DCM.
Collapse
Affiliation(s)
- Meimei Tian
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xinli Huang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Min Li
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Pingping Lou
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huijie Ma
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, China
| | - Xinli Jiang
- Department of Ophthalmology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yaru Zhou
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yan Liu
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
50
|
Wu J, Wu J, Chen T, Cai J, Ren R. Protein aggregation and its affecting mechanisms in neurodegenerative diseases. Neurochem Int 2024; 180:105880. [PMID: 39396709 DOI: 10.1016/j.neuint.2024.105880] [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: 07/22/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/15/2024]
Abstract
Protein aggregation serves as a critical pathological marker in a spectrum of neurodegenerative diseases (NDs), including the formation of amyloid β (Aβ) and Tau neurofibrillary tangles in Alzheimer's disease, as well as α-Synuclein (α-Syn) aggregates in Parkinson's disease, Parkinson's disease-related dementia (PDD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). A significant proportion of patients with amyotrophic lateral sclerosis (ALS) exhibit TDP-43 aggregates. Moreover, a confluence of brain protein pathologies, such as Aβ, Tau, α-Syn, and TDP-43, has been identified in individual NDs cases, highlighting the intricate interplay among these proteins that is garnering heightened scrutiny. Importantly, protein aggregation is modulated by an array of factors, with burgeoning evidence suggesting that it frequently results from perturbations in protein homeostasis, influenced by the cellular membrane milieu, metal ion concentrations, post-translational modifications, and genetic mutations. This review delves into the pathological underpinnings of protein aggregation across various NDs and elucidates the intercommunication among disparate proteins within the same disease context. Additionally, we examine the pathogenic mechanisms by which diverse factors impinge upon protein aggregation, offering fresh perspectives for the future therapeutic intervention of NDs.
Collapse
Affiliation(s)
- Junyun Wu
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Jianan Wu
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Tao Chen
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Jing Cai
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China.
| | - Reng Ren
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|