1
|
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
|
2
|
Tang K, Ye T, He Y, Ba X, Xia D, Peng E, Chen Z, Ye Z, Yang X. Ferroptosis, necroptosis, and pyroptosis in calcium oxalate crystal-induced kidney injury. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167791. [PMID: 40086520 DOI: 10.1016/j.bbadis.2025.167791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/24/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Kidney stones represent a highly prevalent urological disorder worldwide, with high incidence and recurrence rates. Calcium oxalate (CaOx) crystal-induced kidney injury serves as the foundational mechanism for the formation and progression of CaOx stones. Regulated cell death (RCD) such as ferroptosis, necroptosis, and pyroptosis are essential in the pathophysiological process of kidney injury. Ferroptosis, a newly discovered RCD, is characterized by its reliance on iron-mediated lipid peroxidation. Necroptosis, a widely studied programmed necrosis, initiates with a necrotic phenotype that resembles apoptosis in appearance. Pyroptosis, a type of RCD that involves the gasdermin protein, is accompanied by inflammation and immune response. In recent years, increasing amounts of evidence has demonstrated that ferroptosis, necroptosis, and pyroptosis are significant pathophysiological processes involved in CaOx crystal-induced kidney injury. Herein, we summed up the roles of ferroptosis, necroptosis, and pyroptosis in CaOx crystal-induced kidney injury. Furthermore, we delved into the curative potential of ferroptosis, necroptosis, and pyroptosis in CaOx crystal-induced kidney injury.
Collapse
Affiliation(s)
- Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Ye
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Xia
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ejun Peng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqi Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
3
|
Sahu M, Jain U. Activation, interaction and intimation of Nrf2 pathway and their mutational studies causing Nrf2 associated cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167764. [PMID: 40088576 DOI: 10.1016/j.bbadis.2025.167764] [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: 09/09/2024] [Revised: 02/15/2025] [Accepted: 02/25/2025] [Indexed: 03/17/2025]
Abstract
Responses against infection trigger several signaling pathways that lead to the production of cytokines, these cytokines release ROS and RNS, damaging DNA and proteins turn into various diseases including cancer. To combat these harmful cytokines, the Nrf2 pathway is activated. The gene NFE2L2 encodes Nrf2, which is divided into seven conserved domains (Neh1-7). The DLG and ETGE motifs, conserved sequences of amino acid in the Neh2 domain of Nrf2, bind to the BTB domain of Keap1. BTB domain promotes Keap1's homodimerization resulting in Cul3 recruitment providing scaffold formation to E2 ubiquitin ligase to form ubiquitin complex. Under normal conditions, this complex regularly degrades Nrf2. However, once the cell is exposed to oxidative stress by ROS interaction with Keap1 resulting in conformational changes that stabilize the Nrf2. Nrf2 further concentrates on the nucleus where it binds with the transcriptional factor to perform the desired genes transcription for synthesizing SOD, GSH, CAT, and various other proteins which reduce the ROS levels preventing certain diseases. To prevent cells from oxidative stress, molecular hydrogen activates the Nrf2 pathway. To activate the Nrf2 pathway, molecular hydrogen oxidizes the iron porphyrin which acts as an electrophile and interacts with Keap1's cysteine residue.
Collapse
Affiliation(s)
- Mridul Sahu
- School of Health Sciences and Technology (SoHST), UPES, Bidholi, Dehradun - 248007, India
| | - Utkarsh Jain
- School of Health Sciences and Technology (SoHST), UPES, Bidholi, Dehradun - 248007, India.
| |
Collapse
|
4
|
Gu D, Sun Y, Wang J, Sun J, Lou H, Kang W. Metformin regulates ferroptosis in Skin cutaneous melanoma via ATF3/NRF2 axis. Cancer Genet 2025; 294-295:136-144. [PMID: 40318300 DOI: 10.1016/j.cancergen.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/24/2025] [Accepted: 04/08/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND To explore the effects of metformin on the proliferation and ferroptosis of skin cutaneous melanoma (SKCM) and its potential molecular mechanisms, providing a new theoretical basis and strategy for the treatment of cutaneous melanoma. METHODS The CCK-8 experiment was used to detect the effect of metformin on the proliferation of skin cutaneous melanoma cells. Kits were used to detect glutathione (GSH) content, reactive oxygen species (ROS), lipid peroxide (LPO), and malondialdehyde (MDA) levels to evaluate ferroptosis-related indicators. RNA-seq sequencing and related analyses were used to screen differentially expressed genes and explore their involved biological functions and signaling pathways. Western blot was used to detect the expression levels of ATF3 and NRF2 proteins and analyze the regulatory effect of metformin on the ATF3/NRF2 axis. RESULTS Metformin significantly reduced the proliferation ability of skin cutaneous melanoma cells. The treated cells showed a decrease in GSH content and an accumulation of ROS, LPO, and MDA, suggesting that ferroptosis was regulated. RNA-seq analysis found 2068 differentially expressed genes, of which 897 were up-regulated and 1171 were down-regulated. The related pathways such as iron metabolism disorders and ferroptosis were activated. After metformin treatment, the expression of ATF3 mRNA in cells increased and was positively correlated with the concentration, while the expression in SKCM tissues decreased. At the same time, the expression of ATF3 protein increased and the expression of NRF2 protein decreased, suggesting that metformin may induce ferroptosis through the ATF3/NRF2 axis. CONCLUSION Metformin can induce ferroptosis by regulating ATF3/NRF2 axis, which may be a novel strategy for improving the treatment of skin cutaneous melanoma.
Collapse
Affiliation(s)
- Da Gu
- Department of Plastic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, PR China
| | - Yulin Sun
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, PR China
| | - Jianghui Wang
- Department of Orthopedic Surgery, The Second People's Hospital of Dongying, Dongying, Shandong 257000, PR China
| | - Jinpeng Sun
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, PR China
| | - Huanmin Lou
- Department of Plastic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, PR China.
| | - Weiting Kang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, PR China.
| |
Collapse
|
5
|
Ge T, Wang Y, Han Y, Bao X, Lu C. Exploring the Updated Roles of Ferroptosis in Liver Diseases: Mechanisms, Regulators, and Therapeutic Implications. Cell Biochem Biophys 2025; 83:1445-1464. [PMID: 39543068 DOI: 10.1007/s12013-024-01611-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2024] [Indexed: 11/17/2024]
Abstract
Ferroptosis, a newly discovered mode of cell death, is a type of iron-dependent regulated cell death characterized by intracellular excessive lipid peroxidation and imbalanced redox. As the liver is susceptible to oxidative damage and the abnormal iron accumulation is a major feature of most liver diseases, studies on ferroptosis in the field of liver diseases are of great interest. Studies show that targeting the key regulators of ferroptosis can effectively alleviate or even reverse the deterioration process of liver diseases. System Xc- and glutathione peroxidase 4 are the main defense regulators of ferroptosis, while acyl-CoA synthetase long chain family member 4 is a key enzyme causing peroxidation in ferroptosis. Generally speaking, ferroptosis should be suppressed in alcoholic liver disease, non-alcoholic fatty liver disease, and drug-induced liver injury, while it should be induced in liver fibrosis and hepatocellular carcinoma. In this review, we summarize the main regulators involved in ferroptosis and then the mechanisms of ferroptosis in different liver diseases. Treatment options of drugs targeting ferroptosis are further concluded. Determining different triggers of ferroptosis can clarify the mechanism of ferroptosis occurs at both physiological and pathological levels.
Collapse
Affiliation(s)
- Ting Ge
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yang Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yiwen Han
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Xiaofeng Bao
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Chunfeng Lu
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
6
|
Liu Y, Cai Y, Wei X, Gao K, Jin G, Zhou X, Zhao Z. Molecular Mechanisms of Potentilla Discolor Bunge in Regulating Ferroptosis to Alleviate DKD via the Nrf2 Signaling Pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025:120035. [PMID: 40414576 DOI: 10.1016/j.jep.2025.120035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 05/15/2025] [Accepted: 05/23/2025] [Indexed: 05/27/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Potentilla Discolor Bunge (PDB), a plant belonging to the Rosaceae family, is often used in traditional folk medicine to treat diabetes and prevent related complications. Additionally, fresh and tender PDB stems could be consumed as a vegetable or used to make tea. AIM OF THE STUDY To explore the potential targets and mechanisms of PDB in treating Diabetic Kidney Disease (DKD) through network pharmacology, and to investigate its role in modulating the Nrf2 signaling pathway to inhibit ferroptosis using in vivo and in vitro experiments, thereby presenting a potential therapeutic avenue for DKD. MATERIALS AND METHODS Targets of PDB compounds were screened using the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database, while PDB action targets were screened using the DrugBank, OMIM, GeneCards, DisGeNET, and TTD databases. The intersecting targets were subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. Meanwhile, the binding modes of PDB core monomers to key targets were analysed using Molecular Docking (MD) and Surface Plasmon Resonance (SPR) experiments. Experimental validation was conducted using a High-Fat/Sucrose Diet (HFD) combined with Streptozotocin (STZ)-induced DKD rat model and an Advanced Glycation End Products (AGEs)-damaged human renal proximal tubular cell (HK-2) model. RESULTS As predicted through network pharmacology, PDB exerted therapeutic effects against DKD through multiple pathways, including AGE/RAGE, Nrf2 signaling, Oxidative Stress, and apoptosis. Furthermore, PDB's primary active constituents were Quercetin, Kaempferol, and β-sitosterol, with MD analyses suggesting strong binding affinities to the Nrf2 and Ho-1 proteins. In vivo experiments further revealed that PDB treatment reduced the 24 h urinary protein, Serum Creatinine (SCr), and urea levels. It also downregulated Malondialdehyde (MDA), Fe2+, and Reactive Oxygen Species (ROS) accumulation in renal tissues. Additionally, PDB alleviated the histopathological damage in DKD-afflicted rats and significantly upregulated Nrf2, Ho-1, Gpx4, and Slc7a11 in renal tissues. Moreover, Quercetin, Kaempferol, and β-sitosterol significantly upregulated Nrf2, Ho-1, and Gpx4, increased intracellular Glutathione (GSH) levels, reduced ROS and MDA content, and mitigated mitochondrial damage in the AGEs-exposed HK-2 cell injury model. CONCLUSION We predicted the mechanism of action of natural botanical PDB against DKD through network pharmacology, revealing that it significantly upregulated the Nrf2 signaling pathway and inhibited ferroptosis initiation, thus decelerating DKD progression. These findings were further validated through in vivo and in vitro experiments.
Collapse
Affiliation(s)
- Yunhua Liu
- International Acupuncture and Moxibustion Innovation Institute, School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yanmo Cai
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoyu Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Kun Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ge Jin
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xin Zhou
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zongjiang Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
7
|
Choudhary D, Nasiruddin Khan MD, Khan Z, Mehan S, Gupta GD, Narula AS, Samant R. Navigating the complexities of neuronal signaling and targets in neurological disorders: From pathology to therapeutics. Eur J Pharmacol 2025; 995:177417. [PMID: 40010482 DOI: 10.1016/j.ejphar.2025.177417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 02/28/2025]
Abstract
Neurological disorders arising from structural and functional disruptions in the nervous system present major global health challenges. This review examines the intricacies of various cellular signaling pathways, including Nrf2/Keap1/HO-1, SIRT-1, JAK/STAT3/mTOR, and BACE-1/gamma-secretase/MAPT, which play pivotal roles in neuronal health and pathology. The Nrf2-Keap1 pathway, a key antioxidant response mechanism, mitigates oxidative stress, while SIRT-1 contributes to mitochondrial integrity and inflammation control. Dysregulation of these pathways has been identified in neurodegenerative and neuropsychiatric disorders, including Alzheimer's and Parkinson's diseases, characterized by inflammation, protein aggregation, and mitochondrial dysfunction. Additionally, the JAK/STAT3 signaling pathway emphasizes the connection between cytokine responses and neuroinflammation, further compounding disease progression. This review explores the crosstalk among these signaling networks, elucidating how their disruption leads to neuronal decline. It also addresses the dual roles of these pathways, presenting challenges in targeting them for therapeutic purposes. Despite the potential benefits of activating neuroprotective pathways, excessive stimulation may cause deleterious effects, including tumorigenesis. Future research should focus on designing multi-targeted therapies that enhance the effectiveness and safety of treatments, considering individual variabilities and the obstacles posed by the blood-brain barrier to drug delivery. Understanding these complex signaling interactions is crucial for developing innovative and effective neuroprotective strategies that could significantly improve the management of neurological disorders.
Collapse
Affiliation(s)
- Divya Choudhary
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - M D Nasiruddin Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India.
| | | | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC, 27516, USA
| | | |
Collapse
|
8
|
Chen J, Fu Y, Weng S, He J, Dong C. Vitamin C Inhibits Scale Drop Disease Virus Infectivity by Targeting Nrf2 to Reduce Ferroptosis. Antioxidants (Basel) 2025; 14:576. [PMID: 40427458 PMCID: PMC12108497 DOI: 10.3390/antiox14050576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 05/04/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Scale drop disease virus (SDDV) poses an escalating threat to global aquaculture, prompting an urgent need for research. Our study found that SDDV infection upregulates genes related to iron, oxidative stress, and lipid metabolism, causing iron overload, reactive oxygen species (ROS) accumulation, and ultimately ferroptosis. Among the tested antioxidants, vitamin C (VC) demonstrated the most potent inhibitory effect in mandarin fish, reducing SDDV-induced mortality by 37.5%. qPCR and IFA results showed that VC effectively suppressed SDDV infection; decreased ROS, lipid peroxidation (LPO), and iron levels; and enhanced glutathione peroxidase 4 (GPX4) expression in infected cells. Mechanistically, VC's inhibitory effect was reversed by the nuclear factor erythroid 2-related factor 2 (Nrf2) inhibitor ML-385, indicating an Nrf2-dependent pathway. VC promoted Nrf2 nuclear translocation and activated downstream antioxidant genes. Moreover, VC modulated inflammation by regulating pro- and anti-inflammatory factors. These findings suggest VC as a promising therapeutic for SDDV infection.
Collapse
Affiliation(s)
- Jiaming Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (J.C.); (Y.F.); (S.W.); (J.H.)
- Institute of Aquatic Economic Animals, Guangdong Provincial Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou 510275, China
| | - Yuting Fu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (J.C.); (Y.F.); (S.W.); (J.H.)
- Institute of Aquatic Economic Animals, Guangdong Provincial Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou 510275, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Shaoping Weng
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (J.C.); (Y.F.); (S.W.); (J.H.)
- Institute of Aquatic Economic Animals, Guangdong Provincial Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou 510275, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Jianguo He
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (J.C.); (Y.F.); (S.W.); (J.H.)
- Institute of Aquatic Economic Animals, Guangdong Provincial Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou 510275, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Chuanfu Dong
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (J.C.); (Y.F.); (S.W.); (J.H.)
- Institute of Aquatic Economic Animals, Guangdong Provincial Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou 510275, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| |
Collapse
|
9
|
Wei X, Fan X, Chai W, Xiao J, Zhao J, He A, Tang X, Li F, Guo S. Dietary limonin ameliorates heart failure with preserved ejection fraction by targeting ferroptosis via modulation of the Nrf2/SLC7A11/GPX4 axis: an integrated transcriptomics and metabolomics analysis. Food Funct 2025; 16:3553-3574. [PMID: 40230319 DOI: 10.1039/d5fo00475f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex syndrome characterized by hypertension, metabolic disorders, and impaired diastolic function, with limited therapeutic options. Recent studies have highlighted the role of ferroptosis in the pathogenesis of HFpEF, and the inhibition of ferroptosis occurrence can significantly improve cardiac function. Limonin, a bioactive ingredient derived from citrus fruits, has been confirmed to exert potential anti-inflammatory and antioxidant effects in some cardiovascular diseases. This study aims to investigate the therapeutic effects of limonin on HFpEF and the underlying mechanisms of inhibiting ferroptosis. HFpEF mice were established by a combination of Nω-nitro-L-arginine methyl ester and a high-fat diet for 6 weeks. Subsequently, the HFpEF mice were treated with empagliflozin or limonin via oral gavage for an additional 6 weeks. Limonin curbed body weight gain and improved metabolic disorders and hypertension. Limonin also ameliorated concentric cardiac hypertrophy and diastolic dysfunction. Transcriptomics and metabolomics analyses revealed that limonin regulated ferroptosis-related pathways and lipid peroxidation. In vivo, limonin improved mitochondrial morphology, reduced cardiac Fe2+ levels and ferroptosis markers such as ROS, 4-HNE and MDA, and increased GSH levels, thereby enhancing antioxidant capacity. Mechanistically, limonin regulated the P53/SLC7A11/GPX4 signaling pathway, promoted the nuclear translocation of Nrf2 (its upstream signaling molecule), and subsequently activated its downstream antioxidant elements, ultimately inhibiting ferroptosis. Furthermore, limonin decreased the expressions of ACSL4, COX2, and ALOXs, which reduced the accumulation of lipid peroxides. These results demonstrate that limonin ameliorates HFpEF by targeting ferroptosis via modulation of the Nrf2/SLC7A11/GPX4 axis, providing a novel strategy for HFpEF treatment.
Collapse
Affiliation(s)
- Xiaoqi Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Xinyi Fan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Wangjing Chai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jinling Xiao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Jiong Zhao
- Shenzhen Hospital of Beijing University of Chinese Medicine (Longgang), Shenzhen 518116, China.
| | - Aolong He
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Xianwen Tang
- Shenzhen Hospital of Beijing University of Chinese Medicine (Longgang), Shenzhen 518116, China.
| | - Fanghe Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Shuzhen Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
10
|
Liu K, Wang Z, Guo X, Luo J, Wu X, Wang F, Mei Y. The glutamine starvation-induced lncRNA FERRIN suppresses ferroptosis via the stabilization of SLC7A11 mRNA. Int J Biol Macromol 2025; 308:142388. [PMID: 40127798 DOI: 10.1016/j.ijbiomac.2025.142388] [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: 09/20/2024] [Revised: 11/19/2024] [Accepted: 03/20/2025] [Indexed: 03/26/2025]
Abstract
As an essential nutrient for cancer cell survival, glutamine plays both promoting and inhibitory roles in ferroptosis; however, the underlying mechanisms remain obscure. Emerging evidence suggests that long noncoding RNAs (lncRNAs) are crucial regulators of ferroptosis. Nevertheless, it remains unclear whether lncRNAs are involved in glutamine-regulated ferroptosis. In this study, we report that the lncRNA FERRIN is induced by the transcription factor ATF4 under glutamine starvation conditions. FERRIN functions as an inhibitor of ferroptosis by upregulating SLC7A11 expression. Mechanistically, FERRIN interacts with the RNA binding protein hnRNPK, facilitating its binding to SLC7A11 mRNA and leading to the stabilization of SLC7A11 mRNA. Consistent with its inhibitory role in ferroptosis, FERRIN promotes in vitro cancer cell proliferation and in vivo xenograft tumor growth through its modulation of SLC7A11. Collectively, these findings establish FERRIN as a critical negative regulator of ferroptosis and suggest that FERRIN may represent an important link between glutamine availability and ferroptosis.
Collapse
Affiliation(s)
- Kaiyue Liu
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhongyu Wang
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaorui Guo
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jingjing Luo
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xianning Wu
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| | - Fang Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China.
| | - Yide Mei
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
11
|
Xie Y, Liang B, Meng Z, Guo R, Liu C, Yuan Y, Mu W, Wang Y, Cao J. Downregulation of HSPB1 and MGST1 Promotes Ferroptosis and Impacts Immune Infiltration in Diabetic Cardiomyopathy. Cardiovasc Toxicol 2025; 25:719-734. [PMID: 40053272 DOI: 10.1007/s12012-025-09982-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 02/26/2025] [Indexed: 03/12/2025]
Abstract
Diabetic cardiomyopathy (DCM) is a leading cause of death in diabetic patients. Current therapies do not adequately resolve this problem and focus only on the optimal level of blood glucose for patients. Ferroptosis plays an important role in diabetes mellitus and cardiovascular diseases. However, the role of ferroptosis in DCM remains unclear. Differentially expressed ferroptosis-related genes (DE-FRGs) were identified by intersection of the GSE26887 dataset and the Ferroptosis Database. The associations between the DE-FRGs and immune cells in DCM, estimated via the CIBERSORTx algorithm, were analysed. Flow cytometry (FCM) was used to evaluate the infiltration of immune cells in myocardial tissues. The expression of DE-FRGs, glutathione peroxidase 4 and solute carrier family 7 member 11 was examined via real-time quantitative PCR and Western blotting. Three DE-FRGs were identified: heat shock protein family B (small) member 1 (HSPB1), microsomal glutathione S-transferase 1 (MGST1) and solute carrier family 40 member 1 (SLC40A1), which are closely linked to immune cells in DCM. In vivo, the levels of CD8 + T cells, B cells and regulatory T (Treg) cells were significantly decreased in the DCM group, whereas the levels of CD4 + T cells, M1 cells, M2 cells and monocytes were increased. Diabetes significantly decreased HSPB1 and MGST1 levels and increased ferroptosis compared with the Normal group. Furthermore, the ferroptosis inhibitor ferrostatin-1 (Fer-1) alleviated high-fat diet (HFD)-induced cardiomyocyte injury and rescued ferroptosis. These findings suggest that the ferroptosis-related genes HSPB1 and MGST1 are closely related to immune cell infiltration and may be therapeutic targets for DCM.
Collapse
MESH Headings
- Ferroptosis/drug effects
- Animals
- Diabetic Cardiomyopathies/immunology
- Diabetic Cardiomyopathies/genetics
- Diabetic Cardiomyopathies/pathology
- Diabetic Cardiomyopathies/enzymology
- Diabetic Cardiomyopathies/metabolism
- Male
- Down-Regulation
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/immunology
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Disease Models, Animal
- Molecular Chaperones/genetics
- Molecular Chaperones/metabolism
- Humans
- Signal Transduction
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/immunology
- HSP27 Heat-Shock Proteins/genetics
- HSP27 Heat-Shock Proteins/metabolism
- Mice, Inbred C57BL
- Heat-Shock Proteins/genetics
- Heat-Shock Proteins/metabolism
- Databases, Genetic
Collapse
Affiliation(s)
- Yaoli Xie
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Bin Liang
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Zhijun Meng
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Rui Guo
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Caihong Liu
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Yi Yuan
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Wei Mu
- Department of Interventional Therapy for Tumor and Vascular Interventional Therapy, The Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, China
| | - Yajing Wang
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Jimin Cao
- MOE Key Laboratory of Cellular Physiology and the Department of Physiology, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
12
|
Cominelli G, Sulas F, Pinto D, Rinaldi F, Favero G, Rezzani R. Neuro-Nutritional Approach to Neuropathic Pain Management: A Critical Review. Nutrients 2025; 17:1502. [PMID: 40362812 PMCID: PMC12073121 DOI: 10.3390/nu17091502] [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: 03/04/2025] [Revised: 04/15/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Pain is a significant global public health issue that can interfere with daily activities, sleep, and interpersonal relationships when it becomes chronic or worsens, ultimately impairing quality of life. Despite ongoing efforts, the efficacy of pain treatments in improving outcomes for patients remains limited. At present, the challenge lies in developing a personalized care and management plan that helps to maintain patient activity levels and effectively manages pain. Neuropathic pain is a chronic condition resulting from damage to the somatosensory nervous system, significantly impacting quality of life. It is partly thought to be caused by inflammation and oxidative stress, and clinical research has suggested a link between this condition and diet. However, these links are not yet well understood and require further investigation to evaluate the pathways involved in neuropathic pain. Specifically, the question remains whether supplementation with dietary antioxidants, such as melatonin, could serve as a potential adjunctive treatment for neuropathic pain modulation. Melatonin, primarily secreted by the pineal gland but also produced by other systems such as the digestive system, is known for its anti-inflammatory, antioxidant, and anti-aging properties. It is found in various fruits and vegetables, and its presence alongside other polyphenols in these foods may enhance melatonin intake and contribute to improved health. The aim of this review is to provide an overview of neuropathic pain and examine the potential role of melatonin as an adjunctive treatment in a neuro-nutritional approach to pain management.
Collapse
Affiliation(s)
- Giorgia Cominelli
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.C.); (F.S.); (G.F.)
| | - Francesca Sulas
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.C.); (F.S.); (G.F.)
| | - Daniela Pinto
- Human Microbiome Advanced Project Institute, 20129 Milan, Italy; (D.P.); (F.R.)
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs-(ARTO)”, University of Brescia, 25123 Brescia, Italy
| | - Fabio Rinaldi
- Human Microbiome Advanced Project Institute, 20129 Milan, Italy; (D.P.); (F.R.)
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs-(ARTO)”, University of Brescia, 25123 Brescia, Italy
| | - Gaia Favero
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.C.); (F.S.); (G.F.)
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs-(ARTO)”, University of Brescia, 25123 Brescia, Italy
| | - Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.C.); (F.S.); (G.F.)
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs-(ARTO)”, University of Brescia, 25123 Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale–SISDO), 25123 Brescia, Italy
| |
Collapse
|
13
|
Yang X, Liu Y, Cao J, Wu C, Tang L, Bian W, Chen Y, Yu L, Wu Y, Li S, Shen Y, Xia J, Du J. Targeting epigenetic and post-translational modifications of NRF2: key regulatory factors in disease treatment. Cell Death Discov 2025; 11:189. [PMID: 40258841 PMCID: PMC12012105 DOI: 10.1038/s41420-025-02491-z] [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: 01/14/2025] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/23/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a key transcription factor involved in regulating cellular antioxidant defense and detoxification mechanisms. It mitigates oxidative stress and xenobiotic-induced damage by inducing the expression of cytoprotective enzymes, including HO-1 and NQO1. NRF2 also modulates inflammatory responses by inhibiting pro-inflammatory genes and mediates cell death pathways, including apoptosis and ferroptosis. Targeting NRF2 offers potential therapeutic avenues for treating various diseases. NRF2 is regulated through two principal mechanisms: post-translational modifications (PTMs) and epigenetic alterations. PTMs, including phosphorylation, ubiquitination, and acetylation, play a pivotal role in modulating NRF2's stability, activity, and subcellular localization, thereby precisely controlling its function in the antioxidant response. For instance, ubiquitination can lead to NRF2 degradation and reduced antioxidant activity, while deubiquitination enhances its stability and function. Epigenetic modifications, such as DNA methylation, histone modifications, and interactions with non-coding RNAs (e.g., MALAT1, PVT1, MIR4435-2HG, and TUG1), are essential for regulating NRF2 expression by modulating chromatin architecture and gene accessibility. This paper systematically summarizes the molecular mechanisms by which PTMs and epigenetic alterations regulate NRF2, and elucidates its critical role in cellular defense and disease. By analyzing the impact of PTMs, such as phosphorylation, ubiquitination, and acetylation, as well as DNA methylation, histone modifications, and non-coding RNA interactions on NRF2 stability, activity, and expression, the study reveals the complex cellular protection network mediated by NRF2. Furthermore, the paper explores how these regulatory mechanisms affect NRF2's roles in oxidative stress, inflammation, and cell death, identifying novel therapeutic targets and strategies. This provides new insights into the treatment of NRF2-related diseases, such as cancer, neurodegenerative disorders, and metabolic syndrome. This research deepens our understanding of NRF2's role in cellular homeostasis and lays the foundation for the development of NRF2-targeted therapies.
Collapse
Affiliation(s)
- Xinyi Yang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yingchao Liu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jinghao Cao
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Cuiyun Wu
- Cancer Center, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lusheng Tang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Wenxia Bian
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yuhan Chen
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Lingyan Yu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yunyi Wu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Sainan Li
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yuhuan Shen
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Jun Xia
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Jing Du
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| |
Collapse
|
14
|
Huang C, Zeng Q, Chen J, Wen Q, Jin W, Dai X, Ruan R, Zhong H, Xia Y, Wu Z, Huang R, Zhang J, Yao Y, Li L, Lei W, Xiong J, Deng J. TMEM160 inhibits KEAP1 to suppress ferroptosis and induce chemoresistance in gastric cancer. Cell Death Dis 2025; 16:287. [PMID: 40223081 PMCID: PMC11994801 DOI: 10.1038/s41419-025-07621-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/15/2025]
Abstract
Chemoresistance is the most significant challenge affecting the clinical efficacy of the treatment of patients with gastric cancer (GC). Here we reported that transmembrane protein 160 (TMEM160) suppressed ferroptosis and induced chemoresistance in GC cells. Mechanistically, TMEM160 recruited the E3 ligase TRIM37 to promote K48-linked ubiquitination and degradation of KEAP1, thereby activating NRF2 and transcriptionally upregulating the target genes GPX4 and SLC7A11 to inhibit ferroptosis. Further in vitro and in vivo experiments demonstrated that the combination of TMEM160 targeting and chemotherapy had a synergistic inhibitory effect on the growth of GC cells, which was partially NRF2-dependent. Moreover, TMEM160 and NRF2 protein levels were markedly overexpressed in GC tissues, and their co-overexpression was an independent factor for poor prognosis. Collectively, these findings indicate that TMEM160, as a pivotal negative regulator of ferroptosis, exerts a crucial influence on the chemoresistance of GC through the TRIM37-KEAP1/NRF2 axis, providing a potential new prognostic factor and combination therapy strategy for patients with GC.
Collapse
Affiliation(s)
- Chunye Huang
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Qinru Zeng
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Jingyi Chen
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Qin Wen
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Weilun Jin
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Xiaofeng Dai
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Ruiwen Ruan
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Hongguang Zhong
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Yang Xia
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Zhipeng Wu
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Ruixuan Huang
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Jianxi Zhang
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Yangyang Yao
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Li Li
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Wan Lei
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| | - Jun Deng
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China.
- Postdoctoral Innovation Practice Base, The First Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
15
|
Liu X, Zheng Z, Xue C, Wang X, Li J, Liu Z, Xin W, Xu X, Zhou D, Yao L, Lu G. LRRK2 Mediates α-Synuclein-Induced Neuroinflammation and Ferroptosis through the p62-Keap1-Nrf2 Pathway in Parkinson's Disease. Inflammation 2025:10.1007/s10753-025-02291-8. [PMID: 40169487 DOI: 10.1007/s10753-025-02291-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/23/2025] [Accepted: 03/06/2025] [Indexed: 04/03/2025]
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder worldwide, characterized by the progressive degeneration of dopaminergic neurons in the substantia nigra pars compacta and the abnormal aggregation of α-synuclein (α-syn). Despite extensive research, the mechanisms underlying microglial-mediated neuroinflammation and ferroptosis in PD remain inadequately understood. In particular, the role of leucine-rich repeat kinase 2 (LRRK2) in microglial cells and its modulation of the p62-Keap1-Nrf2 signaling pathway warrant further investigation.In this study, we present novel findings demonstrating that LRRK2 regulates microglial neuroinflammation and ferroptosis through the p62-Keap1-Nrf2 signaling axis in the context of PD. Using α-syn-stimulated BV2 microglial cells, we found that LRRK2 inhibition significantly reduced the production of pro-inflammatory cytokines and enhanced the activation of the p62-Keap1-Nrf2 pathway, thereby mitigating ferroptosis and oxidative stress. Furthermore, conditioned medium from LRRK2-inhibited microglia conferred neuroprotective effects on cultured neurons, highlighting the therapeutic potential of targeting LRRK2 in microglia.Importantly, these in vitro findings were corroborated in the MPTP-induced PD mouse model, where LRRK2 inhibition led to diminished microglial activation, decreased apoptosis of midbrain dopaminergic neurons, and upregulation of the p62-Keap1-Nrf2 pathway.Our study fills a critical gap in understanding the microglial mechanisms mediated by LRRK2 and provides novel insights into the pathogenesis of PD. These findings suggest that targeting LRRK2 in microglia may represent a promising therapeutic strategy for PD.
Collapse
Affiliation(s)
- Xinjie Liu
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Zijian Zheng
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Cheng Xue
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xiangrong Wang
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Jianwei Li
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Zheng Liu
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Wenqiang Xin
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xinping Xu
- Jiangxi Institute of Respiratory Disease, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Dongwei Zhou
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Longping Yao
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
- Department of Neuroanatomy, Group for Regeneration and Reprogramming, Institute for Anatomy and Cell Biology, Medical Faculty, Heidelberg University, Heidelberg, Germany.
| | - Guohui Lu
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
16
|
Zheng J, Conrad M. Ferroptosis: when metabolism meets cell death. Physiol Rev 2025; 105:651-706. [PMID: 39661331 DOI: 10.1152/physrev.00031.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/18/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024] Open
Abstract
We present here a comprehensive update on recent advancements in the field of ferroptosis, with a particular emphasis on its metabolic underpinnings and physiological impacts. After briefly introducing landmark studies that have helped to shape the concept of ferroptosis as a distinct form of cell death, we critically evaluate the key metabolic determinants involved in its regulation. These include the metabolism of essential trace elements such as selenium and iron; amino acids such as cyst(e)ine, methionine, glutamine/glutamate, and tryptophan; and carbohydrates, covering glycolysis, the citric acid cycle, the electron transport chain, and the pentose phosphate pathway. We also delve into the mevalonate pathway and subsequent cholesterol biosynthesis, including intermediate metabolites like dimethylallyl pyrophosphate, squalene, coenzyme Q (CoQ), vitamin K, and 7-dehydrocholesterol, as well as fatty acid and phospholipid metabolism, including the biosynthesis and remodeling of ester and ether phospholipids and lipid peroxidation. Next, we highlight major ferroptosis surveillance systems, specifically the cyst(e)ine/glutathione/glutathione peroxidase 4 axis, the NAD(P)H/ferroptosis suppressor protein 1/CoQ/vitamin K system, and the guanosine triphosphate cyclohydrolase 1/tetrahydrobiopterin/dihydrofolate reductase axis. We also discuss other potential anti- and proferroptotic systems, including glutathione S-transferase P1, peroxiredoxin 6, dihydroorotate dehydrogenase, glycerol-3-phosphate dehydrogenase 2, vitamin K epoxide reductase complex subunit 1 like 1, nitric oxide, and acyl-CoA synthetase long-chain family member 4. Finally, we explore ferroptosis's physiological roles in aging, tumor suppression, and infection control, its pathological implications in tissue ischemia-reperfusion injury and neurodegeneration, and its potential therapeutic applications in cancer treatment. Existing drugs and compounds that may regulate ferroptosis in vivo are enumerated.
Collapse
Affiliation(s)
- Jiashuo Zheng
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
- Translational Redox Biology, Technical University of Munich (TUM), TUM Natural School of Sciences, Garching, Germany
| |
Collapse
|
17
|
Zhang L, Li Y, Qian Y, Xie R, Peng W, Zhou W. Advances in the Development of Ferroptosis-Inducing Agents for Cancer Treatment. Arch Pharm (Weinheim) 2025; 358:e202500010. [PMID: 40178208 DOI: 10.1002/ardp.202500010] [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: 01/04/2025] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 04/05/2025]
Abstract
Cancer is the main leading cause of death worldwide and poses a great threat to human life and health. Although pharmacological treatment with chemotherapy and immunotherapy is the main therapeutic strategy for cancer patients, there are still many shortcomings during the treatment such as incomplete killing of cancer cells and development of drug resistance. Emerging evidence indicates the promise of inducing ferroptosis for cancer treatment, particularly for eliminating aggressive malignancies that are resistant to conventional therapies. This review covers recent advances in important regulatory targets in the ferroptosis metabolic pathway and ferroptosis inducers (focusing mainly on the last 3 years) to delineate their design, mechanisms of action, and anticancer applications. To date, many compounds, including inhibitors, degraders, and active molecules from traditional Chinese medicine, have been demonstrated to have ferroptosis-inducing activity by targeting the different biomolecules in the ferroptosis pathway. However, strictly defined ferroptosis inducers have not yet been approved for clinical use; therefore, the discovery of new highly active, less toxic, and selective compounds remains the goal of further research in the coming years.
Collapse
Affiliation(s)
- Li Zhang
- Maternal and Child Health Department, Shaoxing Maternity and Child Health Care Hospital, Shaoxing, Zhejiang Province, China
| | - Yulong Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yufeng Qian
- Medical Research Center, Shaoxing People's Hospital, Shaoxing, Zhejiang Province, China
| | - Ruliang Xie
- Jiangsu Institute of Marine Resources Development, Jiangsu Ocean University, Lianyungang, Jiangsu Province, China
| | - Wei Peng
- Medical Research Center, Shaoxing People's Hospital, Shaoxing, Zhejiang Province, China
| | - Wen Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| |
Collapse
|
18
|
Feng X, Zhang W, Liu X, Wang Q, Dang X, Han J, Zhang X. Ferroptosis-associated signaling pathways and therapeutic approaches in depression. Front Neurosci 2025; 19:1559597. [PMID: 40177374 PMCID: PMC11961976 DOI: 10.3389/fnins.2025.1559597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 03/05/2025] [Indexed: 04/05/2025] Open
Abstract
Ferroptosis, a newly identified form of cell death, is characterized by excessive iron accumulation and lipid peroxidation. Studies indicate a strong association between ferroptosis and depression; however, the precise signaling pathways and underlying molecular mechanisms remain unclear. This review summarizes the role of ferroptosis in depression and its associated signaling pathways. Additionally, therapeutic approaches for depression based on ferroptosis theory are reviewed, providing novel targets for the prevention and treatment of depression and laying a foundation for future research on the relationship between ferroptosis and depression.
Collapse
Affiliation(s)
- Xuyang Feng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoxi Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Qiuxuan Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiao Dang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jingxian Han
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xuezhu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
19
|
He Y, Lin Y, Song J, Song M, Nie X, Sun H, Xu C, Han Z, Cai J. From mechanisms to medicine: Ferroptosis as a Therapeutic target in liver disorders. Cell Commun Signal 2025; 23:125. [PMID: 40055721 PMCID: PMC11889974 DOI: 10.1186/s12964-025-02121-2] [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: 11/20/2024] [Accepted: 02/22/2025] [Indexed: 05/13/2025] Open
Abstract
In recent 10 years, ferroptosis has become a hot research direction in the scientific research community as a new way of cell death. Iron toxicity accumulation and lipotoxicity are unique features. Several studies have found that ferroptosis is involved in the regulation of the hepatic microenvironment and various hepatic metabolisms, thereby mediating the progression of related liver diseases. For example, NRF2 and FSP1, as important regulatory proteins of ferroptosis, are involved in the development of liver tumors and liver failure. In this manuscript, we present the mechanisms involved in ferroptosis, the concern of ferroptosis with the liver microenvironment and the progression of ferroptosis in various liver diseases. In addition, we summarize recent clinical advances in targeted ferroptosis therapy for related diseases. We expect that this manuscript can provide a new perspective for clinical treatment of related diseases.
Collapse
Affiliation(s)
- Yuqi He
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China
| | - Yumeng Lin
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jinfeng Song
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China
| | - Mingzhu Song
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China
| | - Xiaoxia Nie
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China
| | - Hong Sun
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China
| | - Changyun Xu
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China
| | - Zhongyu Han
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China.
| | - Juan Cai
- Department of Transfusion, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu'an People's Hospital, Lu'an, Anhui Province, China.
| |
Collapse
|
20
|
Elseweidy MM, Harb NG, Ali AA, El-Aziz RMA, Elrashidy RA. Sulforaphane substantially impedes testicular ferroptosis in adult rats exposed to di-2-ethylhexyl phthalate through activation of NRF-2/SLC7A11/GPX-4 trajectory. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3163-3175. [PMID: 39352535 PMCID: PMC11920001 DOI: 10.1007/s00210-024-03440-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/07/2024] [Indexed: 03/19/2025]
Abstract
Di-2-ethylhexyl phthalate (DEHP) is a common plasticizer with a deleterious impact on testicular functionality and male fertility. Growing evidence implicates ferroptosis as one of the plausible mechanisms for DEHP-induced testicular injury. Sulforaphane (SFN) is a natural isothiocyanate displaying beneficial effects on testicular injury in several animal models. Herein, we explored the potential protective effect of SFN on testicular ferroptosis and toxicity evoked by DEHP. Adult male Wistar rats were equally distributed into three groups (n = 6/group): (i) CON group; (ii) DEHP group, received DEHP (2 g/kg PO) for 4 weeks; and (iii) DEHP + SFN group, received SFN (10 mg/kg, PO) 1 week prior to DEHP then concurrently with DEHP for further 4 weeks. Compared to CON group, exposure to DEHP caused testicular atrophy, deteriorated testicular architecture, testicular fibrosis, reduced sperm count and motility, higher sperm deformity, and declined serum testosterone level. All these abnormalities were ameliorated by SFN preconditioning. Additionally, pretreatment with SFN reversed the increased aromatase level and upregulated the steroidogenic markers in testes of DEHP-exposed rats. SFN pretreatment also counteracted DEHP-induced oxidative stress and boosted the total antioxidant capacity in testicular tissue via activation of the nuclear factor erythroid 2-related factor 2 (NRF-2) and its downstream target, hemeoxygenase-1 (HO-1). Moreover, SFN preconditioning mitigated DEHP-induced ferroptosis through up-surging SLC7A11, GPX-4, and GSH, while suppressing iron overload and ACSL4-induced lipid peroxidation in testicular tissue of rats. These findings may nominate SFN as a promising protective intervention to alleviate testicular ferroptosis associated with DEHP exposure through activation of NRF-2/SLC7A11/GPX-4 trajectory.
Collapse
Affiliation(s)
- Mohammed M Elseweidy
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Nouran G Harb
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Abdelmoniem A Ali
- Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Reda M Abd El-Aziz
- Department of Physiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Rania A Elrashidy
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
21
|
Khawas S, Sharma N. Cell death crosstalk in respiratory diseases: unveiling the relationship between pyroptosis and ferroptosis in asthma and COPD. Mol Cell Biochem 2025; 480:1305-1326. [PMID: 39112808 DOI: 10.1007/s11010-024-05062-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/29/2024] [Indexed: 02/21/2025]
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) are heterogeneous obstructive diseases characterized by airflow limitations and are recognized as significant contributors to fatality all over the globe. Asthma accounts for about 4, 55,000 deaths, and COPD is the 3rd leading contributor of mortality worldwide. The pathogenesis of these two obstructive disorders is complex and involves numerous mechanistic pathways, including inflammation-mediated and non-inflammation-mediated pathways. Among all the pathological categorizations, programmed cell deaths (PCDs) play a dominating role in the progression of these obstructive diseases. The two major PCDs that are involved in structural and functional remodeling in the progression of asthma and COPD are Pyroptosis and Ferroptosis. Pyroptosis is a PCD mechanism mediated by the activation of the Nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome, leading to the maturation and release of Interleukin-1β and Interleukin-18, whereas ferroptosis is a lipid peroxidation-associated cell death. In this review, the major molecular pathways contributing to these multifaceted cell deaths have been discussed, and crosstalk among them regarding the pathogenesis of asthma and COPD has been highlighted. Further, the possible therapeutic approaches that can be utilized to mitigate both cell deaths at once have also been illustrated.
Collapse
Affiliation(s)
- Sayak Khawas
- Department of Pharmaceutical Science & Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Neelima Sharma
- Department of Pharmaceutical Science & Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India.
| |
Collapse
|
22
|
Du F, Wang G, Dai Q, Huang J, Li J, Liu C, Du K, Tian H, Deng Q, Xie L, Zhao X, Zhang Q, Yang L, Li Y, Wu Z, Zhang Z. Targeting novel regulated cell death: disulfidptosis in cancer immunotherapy with immune checkpoint inhibitors. Biomark Res 2025; 13:35. [PMID: 40012016 DOI: 10.1186/s40364-025-00748-4] [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: 01/07/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
The battle against cancer has evolved over centuries, from the early stages of surgical resection to contemporary treatments including chemotherapy, radiation, targeted therapies, and immunotherapies. Despite significant advances in cancer treatment over recent decades, these therapies remain limited by various challenges. Immune checkpoint inhibitors (ICIs), a cornerstone of tumor immunotherapy, have emerged as one of the most promising advancements in cancer treatment. Although ICIs, such as CTLA-4 and PD-1/PD-L1 inhibitors, have demonstrated clinical efficacy, their therapeutic impact remains suboptimal due to patient-specific variability and tumor immune resistance. Cell death is a fundamental process for maintaining tissue homeostasis and function. Recent research highlights that the combination of induced regulatory cell death (RCD) and ICIs can substantially enhance anti-tumor responses across multiple cancer types. In cells exhibiting high levels of recombinant solute carrier family 7 member 11 (SLC7A11) protein, glucose deprivation triggers a programmed cell death (PCD) pathway characterized by disulfide bond formation and REDOX (reduction-oxidation) reactions, termed "disulfidptosis." Studies suggest that disulfidptosis plays a critical role in the therapeutic efficacy of SLC7A11high cancers. Therefore, to investigate the potential synergy between disulfidptosis and ICIs, this study will explore the mechanisms of both processes in tumor progression, with the goal of enhancing the anti-tumor immune response of ICIs by targeting the intracellular disulfidptosis pathway.
Collapse
Affiliation(s)
- Fei Du
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Guojun Wang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qian Dai
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Jiang Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Junxin Li
- Department of pharmacy, Zigong Fourth People's Hospital, Zigong, 643000, China
| | - Congxing Liu
- Department of Pharmacy, Chengfei Hospital, Chengdu, 610000, China
| | - Ke Du
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pediatrics, Luzhou Maternal and Child Health Hospital, Luzhou Second People's Hospital, Luzhou, 646000, Sichuan, China
| | - Hua Tian
- School of Nursing, Chongqing College of Humanities, Science & Technology, Chongqing, 401520, China
| | - Qiwei Deng
- Heruida Pharmaceutical Co.,ltd, Haikou, Hainan, 570100, China
| | - Longxiang Xie
- The TCM Hospital of Longquanyi District, Chengdu, 610100, Sichuan, China
| | - Xin Zhao
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qimin Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Lan Yang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Yaling Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhigui Wu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhuo Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
23
|
Wu K, Zhao W, Hou Z, Zhang W, Qin L, Qiu J, Wang D, Zhuang L, Xue X, Sun D. Ferritinophagy: multifaceted roles and potential therapeutic strategies in liver diseases. Front Cell Dev Biol 2025; 13:1551003. [PMID: 40070880 PMCID: PMC11893559 DOI: 10.3389/fcell.2025.1551003] [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: 12/24/2024] [Accepted: 02/06/2025] [Indexed: 03/14/2025] Open
Abstract
Ferritinophagy, the selective autophagic degradation of ferritin to release iron, is emerging as a critical regulator of iron homeostasis and a key player in the pathogenesis of various liver diseases. This review comprehensively examines the mechanisms, regulation, and multifaceted roles of ferritinophagy in liver health and disease. Ferritinophagy is intricately regulated by several factors, including Nuclear Receptor Coactivator 4 (NCOA4), Iron regulatory proteins and signaling pathways such as mTOR and AMPK. These regulatory mechanisms ensure proper iron utilization and prevent iron overload, which can induce oxidative stress and ferroptosis. In liver diseases, ferritinophagy exhibits dual roles. In liver fibrosis, promoting ferritinophagy in hepatic stellate cells (HSCs) can induce cell senescence and reduce fibrosis progression. However, in non-alcoholic fatty liver disease (NAFLD), chronic ferritinophagy may exacerbate liver injury through iron overload and oxidative stress. In hepatocellular carcinoma (HCC), ferritinophagy can be harnessed as a novel therapeutic strategy by inducing ferroptosis in cancer cells. Additionally, ferritinophagy is implicated in drug-induced liver injury and sepsis-associated liver damage, highlighting its broad impact on liver pathology. This review also explores the crosstalk between ferritinophagy and other selective autophagy pathways, such as mitophagy and lipophagy, which collectively influence cellular homeostasis and disease progression. Understanding these interactions is essential for developing comprehensive therapeutic strategies targeting multiple autophagy pathways. In summary, ferritinophagy is a complex and dynamic process with significant implications for liver diseases. This review provides an in-depth analysis of ferritinophagy's regulatory mechanisms and its potential as a therapeutic target, emphasizing the need for further research to elucidate its role in liver health and disease.
Collapse
Affiliation(s)
- Kejia Wu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Zhao
- Department of Anesthesiology, Xinyi People’s Hospital, Xinyi, Jiangsu, China
| | - Zeyu Hou
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Weigang Zhang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lei Qin
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Junyi Qiu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Daobin Wang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lin Zhuang
- Department of General Surgery, Wujin Affiliated Hospital of Jiangsu University and The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Xiaofeng Xue
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ding Sun
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
24
|
Wang L, Tang Y, Hou T, Gao Y, Ci X, Peng L. Hesperetin alleviates PM2.5-induced lung injury by inhibiting ferroptosis in an Nrf2-dependent manner. Int Immunopharmacol 2025; 148:114123. [PMID: 39884081 DOI: 10.1016/j.intimp.2025.114123] [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: 09/26/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Fine particulate matter (PM2.5) is a global environmental problem that threatens public health because it can induce ferroptosis and cause lung injury. Hesperetin (Hes), a natural compound widely present in fruits and vegetables, can activate nuclear factor erythroid 2-related factor 2 (Nrf2), thereby exerting powerful antioxidant effects. PURPOSE We explored the antioxidant effects of Hes on lung injury caused by PM2.5 exposure. METHODS In vivo, a mouse model of PM2.5-induced lung injury was used to evaluate the protective effect of Hes. In vitro, the effects of Hes on PM2.5-induced ferroptosis were examined in BEAS-2B cells. RESULTS In vivo, Hes activated the Nrf2 signaling pathway and protected lung tissues from damage induced by PM2.5. In vitro, Hes activated Nrf2 by promoting PI3K/AKT phosphorylation and inhibiting PM2.5-induced ferroptosis. However, in siNrf2-treated BEAS-2B cells, the protective effects of Hes were eliminated. In addition, Nrf2-KO mice exhibited more severe lung injury than did wild-type (WT) mice after PM2.5 exposure. Besides, the protective effects of Hes on PM2.5-exposed Nrf2-KO mice were strongly compromised. CONCLUSION Hes activates Nrf2 by promoting PI3K/AKT phosphorylation to exert a protective effect on PM2.5-induced ferroptosis and lung injury.
Collapse
Affiliation(s)
- Lu Wang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun 130021 China
| | - Ying Tang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun 130021 China
| | - Tianhua Hou
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun 130021 China
| | - Yun Gao
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun 130021 China
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130001 China.
| | - Liping Peng
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun 130021 China.
| |
Collapse
|
25
|
Bao X, Chen Y, Chang J, Du J, Yang C, Wu Y, Sha Y, Li M, Chen S, Yang M, Liu SB. Machine learning-based bulk RNA analysis reveals a prognostic signature of 13 cell death patterns and potential therapeutic target of SMAD3 in acute myeloid leukemia. BMC Cancer 2025; 25:273. [PMID: 39955536 PMCID: PMC11830216 DOI: 10.1186/s12885-025-13658-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Dysregulation or abnormality of the programmed cell death (PCD) pathway is closely related to the occurrence and development of many tumors, including acute myeloid leukemia (AML). Studying the abnormal characteristics of PCD pathway-related molecular markers can provide a basis for prognosis prediction and targeted drug design in AML patients. METHODS A total of 1394 genes representing 13 different PCD pathways were examined in AML patients and healthy donors. The upregulated genes were analyzed for their ability to predict overall survival (OS) individually, and these prognostic genes were subsequently combined to construct a PCD-related prognostic signature via an integrated approach consisting of 101 models based on ten machine learning algorithms. RNA transcriptome and clinical data from multiple AML cohorts (TCGA-AML, GSE106291, GSE146173 and Beat AML) were obtained to develop and validate the AML prognostic model. RESULTS A total of 214 upregulated PCD-related genes were identified in AML patients, 39 of which were proven to be prognostic genes in the training cohort. On the basis of the average C-index and number of model genes identified from the machine learning combinations, a PCD index was developed and validated for predicting AML OS. A prognostic nomogram was then generated and validated on the basis of the PCD index, age and ELN risk stratification in the Beat AML cohort and the GSE146173 cohort, revealing satisfactory predictive power (AUC values ≥ 0.7). With different mutation patterns, a higher PCD index was associated with a worse OS. The PCD index was significantly related to higher scores for immunosuppressive cells and mature leukemia cell subtypes. As the gene most closely related to the PCD index, the expression of SMAD3 was further validated in vitro. AML cells harboring KMT2A rearrangements were more sensitive to the SMAD3 inhibitor SIS3, and the expression of the autophagy-related molecular marker LC3 was increased in KMT2A-rearranged cell lines after SIS3 monotherapy and combined treatment. CONCLUSION The PCD index and SMAD3 gene expression levels have potential prognostic value and can be used in targeted therapy for AML, and these findings can lead to the development of effective strategies for the combined treatment of high-risk AML patients.
Collapse
Affiliation(s)
- Xiebing Bao
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yao Chen
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, 28 Kehua Road, Suzhou, 215009, China
| | - Jie Chang
- School of Public Health, Medical College of Soochow University, Suzhou, 215123, China
| | - Jiahui Du
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, 28 Kehua Road, Suzhou, 215009, China
| | - Chen Yang
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China
| | - Yijie Wu
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China
| | - Yu Sha
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, 28 Kehua Road, Suzhou, 215009, China
| | - Ming Li
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, 28 Kehua Road, Suzhou, 215009, China
| | - Suning Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| | - Minfeng Yang
- School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong, 226019, China.
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China.
| | - Song-Bai Liu
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in Oncology, Suzhou Vocational Health College, 28 Kehua Road, Suzhou, 215009, China.
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China.
| |
Collapse
|
26
|
Eskander G, Abdelhamid SG, Wahdan SA, Radwan SM. Insights on the crosstalk among different cell death mechanisms. Cell Death Discov 2025; 11:56. [PMID: 39929794 PMCID: PMC11811070 DOI: 10.1038/s41420-025-02328-9] [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: 08/06/2024] [Revised: 12/23/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025] Open
Abstract
The phenomenon of cell death has garnered significant scientific attention in recent years, emerging as a pivotal area of research. Recently, novel modalities of cellular death and the intricate interplay between them have been unveiled, offering insights into the pathogenesis of various diseases. This comprehensive review delves into the intricate molecular mechanisms, inducers, and inhibitors of the underlying prevalent forms of cell death, including apoptosis, autophagy, ferroptosis, necroptosis, mitophagy, and pyroptosis. Moreover, it elucidates the crosstalk and interconnection among the key pathways or molecular entities associated with these pathways, thereby paving the way for the identification of novel therapeutic targets, disease management strategies, and drug repurposing.
Collapse
Affiliation(s)
- Georgette Eskander
- Postgraduate program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | | | - Sara A Wahdan
- Pharmacology and toxicology Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Sara M Radwan
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
27
|
Zhang Z, Wang H, Kan X, Zhang X, Xu S, Cai J, Guo J. The interplay of ferroptosis and oxidative stress in the pathogenesis of aortic dissection. Front Pharmacol 2025; 16:1519273. [PMID: 39974735 PMCID: PMC11835687 DOI: 10.3389/fphar.2025.1519273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/09/2025] [Indexed: 02/21/2025] Open
Abstract
Aortic dissection (AD) is a life-threatening vascular condition marked by the separation or tearing of the aortic media. Ferroptosis, a form of iron-dependent programmed cell death, occurs alongside lipid peroxidation and the accumulation of reactive oxygen species (ROS). The relationship between ferroptosis and AD lies in its damaging effect on vascular cells. In AD, ferroptosis worsens the damage to vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), thereby weakening the vascular wall's structural integrity and accelerating the onset and progression of the condition. However, the molecular mechanisms through which ferroptosis regulates the onset and progression of AD remain poorly understood. This article explores the relationship between ferroptosis and AD.
Collapse
Affiliation(s)
- Zhaoshan Zhang
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, China
- Department of Stomatology, The First Affiliated Hospital of Yangtze University, Yangtze University, Jingzhou, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou, China
| | - Haichao Wang
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, China
- Department of Stomatology, The First Affiliated Hospital of Yangtze University, Yangtze University, Jingzhou, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou, China
| | - Xi Kan
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, China
- Department of Stomatology, The First Affiliated Hospital of Yangtze University, Yangtze University, Jingzhou, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou, China
| | - Xiaozhao Zhang
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, China
- Department of Stomatology, The First Affiliated Hospital of Yangtze University, Yangtze University, Jingzhou, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou, China
| | - Senping Xu
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, China
- Department of Stomatology, The First Affiliated Hospital of Yangtze University, Yangtze University, Jingzhou, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou, China
| | - Jie Cai
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, China
- Department of Stomatology, The First Affiliated Hospital of Yangtze University, Yangtze University, Jingzhou, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou, China
| | - Jiawei Guo
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, China
- Department of Stomatology, The First Affiliated Hospital of Yangtze University, Yangtze University, Jingzhou, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou, China
| |
Collapse
|
28
|
Yu Y, Zhang L, Zhang D, Dai Q, Hou M, Chen M, Gao F, Liu XL. The role of ferroptosis in acute kidney injury: mechanisms and potential therapeutic targets. Mol Cell Biochem 2025; 480:759-784. [PMID: 38943027 DOI: 10.1007/s11010-024-05056-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024]
Abstract
Acute kidney injury (AKI) is one of the most common and severe clinical renal syndromes with high morbidity and mortality. Ferroptosis is a form of programmed cell death (PCD), is characterized by iron overload, reactive oxygen species accumulation, and lipid peroxidation. As ferroptosis has been increasingly studied in recent years, it is closely associated with the pathophysiological process of AKI and provides a target for the treatment of AKI. This review offers a comprehensive overview of the regulatory mechanisms of ferroptosis, summarizes its role in various AKI models, and explores its interaction with other forms of cell death, it also presents research on ferroptosis in AKI progression to other diseases. Additionally, the review highlights methods for detecting and assessing AKI through the lens of ferroptosis and describes potential inhibitors of ferroptosis for AKI treatment. Finally, the review presents a perspective on the future of clinical AKI treatment, aiming to stimulate further research on ferroptosis in AKI.
Collapse
Affiliation(s)
- Yanxin Yu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Lei Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Die Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Qiangfang Dai
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Mingzheng Hou
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Meini Chen
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Feng Gao
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Xiao-Long Liu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China.
| |
Collapse
|
29
|
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
|
30
|
Lu C, Xu C, Li S, Ni H, Yang J. Liraglutide and GLP-1(9-37) alleviated hepatic ischemia-reperfusion injury by inhibiting ferroptosis via GSK3β/Nrf2 pathway and SMAD159/Hepcidin/FTH pathway. Redox Biol 2025; 79:103468. [PMID: 39693850 PMCID: PMC11719303 DOI: 10.1016/j.redox.2024.103468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/07/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024] Open
Abstract
Ferroptosis plays a pivotal role in the pathogenesis of ischemia-reperfusion injury (IRI). Liraglutide, as a GLP-1 receptor (GLP-1R) agonist, has exhibited extensive biological effects beyond its hypoglycemic action. Recent studies have shed light on the regulatory influence of Liraglutide on ferroptosis, yet the precise underlying mechanism remains elusive. GLP-1(9-37), as a metabolite of GLP-1, has a low affinity to GLP-1R. Its effect on ferroptosis remains unknown. In this study, we investigated the effects of Liraglutide and GLP-1(9-37) on the ferroptosis during hepatic ischemia-repferfusion (I/R), as well as the underlying specific mechanisms. We found that the administration of Liraglutide alleviated I/R-induced liver injury with less iron accumulation and lower lipid peroxidation, which was not entirely dependent on the presence of GLP-1R. Similarly, GLP-1(9-37) also exhibited these effects. Besides, both of them increased GPX4 expression and decreased COX2 expression. These effects were reversed by a High-Iron Diet. In vitro study showed similar results. In mechanism study, we found that both Liraglutide and GLP-1(9-37) treatment promoted the nuclear translocation of Nrf2 by inhibiting GSK-3β, thereby reducing lipid peroxides. Furthermore, they increased FTH and FTL expression via the SMAD159/Hepcidin pathway, which contributed to the decreased iron accumulation. In conclusion, this study determined that both Liraglutide and GLP-1(9-37) alleviated hepatic ischemia-reperfusion injury (HIRI) by suppressing ferroptosis via the activation of the GSK3β/Nrf2 pathway and the SMAD159/Hepcidin/FTH pathway.
Collapse
Affiliation(s)
- Chenqi Lu
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Cong Xu
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shanglin Li
- Department of General Surgery, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haiqiang Ni
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Jun Yang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
31
|
Desterke C, Fu Y, Bonifacio-Mundaca J, Monge C, Pineau P, Mata-Garrido J, Francés R. Ferroptosis Transcriptional Regulation and Prognostic Impact in Medulloblastoma Subtypes Revealed by RNA-Seq. Antioxidants (Basel) 2025; 14:96. [PMID: 39857430 PMCID: PMC11761645 DOI: 10.3390/antiox14010096] [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: 12/11/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in children, typically arising during infancy and childhood. Despite multimodal therapies achieving a response rate of 70% in children older than 3 years, treatment remains challenging. Ferroptosis, a form of regulated cell death, can be induced in medulloblastoma cells in vitro using erastin or RSL3. Using two independent medulloblastoma RNA-sequencing cohorts (MB-PBTA and MTAB-10767), we investigated the expression of ferroptosis-related molecules through multiple approaches, including Weighted Gene Co-Expression Network Analysis (WGCNA), molecular subtype stratification, protein-protein interaction (PPI) networks, and univariable and multivariable overall survival analyses. A prognostic expression score was computed based on a cross-validated ferroptosis signature. In training and validation cohorts, the regulation of the ferroptosis transcriptional program distinguished the four molecular subtypes of medulloblastoma. WGCNA identified nine gene modules in the MB tumor transcriptome; five correlated with molecular subtypes, implicating pathways related to oxidative stress, hypoxia, and trans-synaptic signaling. One module, associated with disease recurrence, included epigenetic regulators and nucleosome organizers. Univariable survival analyses identified a 45-gene ferroptosis prognostic signature associated with nutrient sensing, cysteine and methionine metabolism, and trans-sulfuration within a one-carbon metabolism. The top ten unfavorable ferroptosis genes included CCT3, SNX5, SQOR, G3BP1, CARS1, SLC39A14, FAM98A, FXR1, TFAP2C, and ATF4. Patients with a high ferroptosis score showed a worse prognosis, particularly in the G3 and SHH subtypes. The PPI network highlighted IL6 and CBS as unfavorable hub genes. In a multivariable overall survival model, which included gender, age, and the molecular subtype classification, the ferroptosis expression score was validated as an independent adverse prognostic marker (hazard ratio: 5.8; p-value = 1.04 × 10-9). This study demonstrates that the regulation of the ferroptosis transcriptional program is linked to medulloblastoma molecular subtypes and patient prognosis. A cross-validated ferroptosis signature was identified in two independent RNA-sequencing cohorts, and the ferroptosis score was confirmed as an independent and adverse prognostic factor in medulloblastoma.
Collapse
Affiliation(s)
- Christophe Desterke
- INSERM UMRS-1310, Faculté de Médecine du Kremlin Bicêtre, Université Paris-Saclay, F-94270 Le Kremlin-Bicêtre, France;
| | - Yuanji Fu
- INSERM, CNRS, Institut Necker Enfants Malades, Université Paris Cité, F-75015 Paris, France;
| | - Jenny Bonifacio-Mundaca
- National Tumor Bank, Department of Pathology, National Institute of Neoplastic Diseases, Surquillo 15038, Peru;
| | - Claudia Monge
- Unité Organisation Nucléaire et Oncogenèse, Institut Pasteur, Université Paris Cité, INSERM U993, F-75015 Paris, France; (C.M.); (P.P.)
| | - Pascal Pineau
- Unité Organisation Nucléaire et Oncogenèse, Institut Pasteur, Université Paris Cité, INSERM U993, F-75015 Paris, France; (C.M.); (P.P.)
| | - Jorge Mata-Garrido
- Unité Organisation Nucléaire et Oncogenèse, Institut Pasteur, Université Paris Cité, INSERM U993, F-75015 Paris, France; (C.M.); (P.P.)
| | - Raquel Francés
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, F-75006 Paris, France
| |
Collapse
|
32
|
Jing C, Wu Y, Zhang Y, Zhu Z, Zhang Y, Liu Z, Sun D. Epigenetic regulation and post-translational modifications of ferroptosis-related factors in cardiovascular diseases. Clin Epigenetics 2025; 17:4. [PMID: 39799367 PMCID: PMC11724467 DOI: 10.1186/s13148-024-01809-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 12/23/2024] [Indexed: 01/15/2025] Open
Abstract
As an important element of the human body, iron participates in numerous physiological and biochemical reactions. In the past decade, ferroptosis (a form of iron-dependent regulated cell death) has been reported to contribute to the pathogenesis and progression of various diseases. The stability of iron in cardiomyocytes is crucial for the maintenance of normal physiological cardiac activity. Ferroptosis has been detected in many cardiovascular diseases (CVDs), including coronary heart disease, myocardial ischemia-reperfusion injury, heart failure, and chemotherapy-induced myocardial damage. In cardiomyocytes, epigenetic regulation and post-translational modifications regulate the expression of ferroptosis-related factors, maintain iron homeostasis, and participate in the progression of CVDs. Currently, there is no detailed mechanism to explain the relationship between epigenetic regulation and ferroptosis in CVDs. In this review, we provide an initial summary of the core mechanisms of ferroptosis in cardiomyocytes, with first focus on the epigenetic regulation and expression of ferroptosis-related factors in the context of common cardiovascular diseases. We anticipate that the new insights into the pathogenesis of CVDs provided here will inspire the development of clinical interventions to specifically target the active sites of these factors, reducing the harmfulness of ferroptosis to human health.
Collapse
Affiliation(s)
- Chunlu Jing
- Department of Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, People's Republic of China
- Shenyang Clinical Medical Research Center for Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, People's Republic of China
- Dalian Medical University, Dalian, 116044, Liaoning, People's Republic of China
| | - Yupeng Wu
- Department of Neurosurgery, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, People's Republic of China
| | - Yuzhu Zhang
- Department of Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, People's Republic of China
- Shenyang Clinical Medical Research Center for Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, People's Republic of China
| | - Zaihan Zhu
- Department of Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, People's Republic of China
- Shenyang Clinical Medical Research Center for Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, People's Republic of China
| | - Yong Zhang
- Department of Urology, The People's Hospital of Liaoning Province, The People's Hospital of China Medical University, 33 Wenyi Road, Shenhe District, Shenyang, 110016, People's Republic of China
| | - Zhen Liu
- Department of Urology, The People's Hospital of Liaoning Province, The People's Hospital of China Medical University, 33 Wenyi Road, Shenhe District, Shenyang, 110016, People's Republic of China.
| | - Dandan Sun
- Department of Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, People's Republic of China.
- Shenyang Clinical Medical Research Center for Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, People's Republic of China.
| |
Collapse
|
33
|
Revol-Cavalier J, Quaranta A, Newman JW, Brash AR, Hamberg M, Wheelock CE. The Octadecanoids: Synthesis and Bioactivity of 18-Carbon Oxygenated Fatty Acids in Mammals, Bacteria, and Fungi. Chem Rev 2025; 125:1-90. [PMID: 39680864 PMCID: PMC11719350 DOI: 10.1021/acs.chemrev.3c00520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024]
Abstract
The octadecanoids are a broad class of lipids consisting of the oxygenated products of 18-carbon fatty acids. Originally referring to production of the phytohormone jasmonic acid, the octadecanoid pathway has been expanded to include products of all 18-carbon fatty acids. Octadecanoids are formed biosynthetically in mammals via cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP) activity, as well as nonenzymatically by photo- and autoxidation mechanisms. While octadecanoids are well-known mediators in plants, their role in the regulation of mammalian biological processes has been generally neglected. However, there have been significant advancements in recognizing the importance of these compounds in mammals and their involvement in the mediation of inflammation, nociception, and cell proliferation, as well as in immuno- and tissue modulation, coagulation processes, hormone regulation, and skin barrier formation. More recently, the gut microbiome has been shown to be a significant source of octadecanoid biosynthesis, providing additional biosynthetic routes including hydratase activity (e.g., CLA-HY, FA-HY1, FA-HY2). In this review, we summarize the current field of octadecanoids, propose standardized nomenclature, provide details of octadecanoid preparation and measurement, summarize the phase-I metabolic pathway of octadecanoid formation in mammals, bacteria, and fungi, and describe their biological activity in relation to mammalian pathophysiology as well as their potential use as biomarkers of health and disease.
Collapse
Affiliation(s)
- Johanna Revol-Cavalier
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Larodan
Research Laboratory, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Alessandro Quaranta
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - John W. Newman
- Western
Human Nutrition Research Center, Agricultural
Research Service, USDA, Davis, California 95616, United States
- Department
of Nutrition, University of California, Davis, Davis, California 95616, United States
- West
Coast Metabolomics Center, Genome Center, University of California, Davis, Davis, California 95616, United States
| | - Alan R. Brash
- Department
of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Mats Hamberg
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Larodan
Research Laboratory, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Craig E. Wheelock
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Department
of Respiratory Medicine and Allergy, Karolinska
University Hospital, Stockholm SE-141-86, Sweden
| |
Collapse
|
34
|
Yang Y, Yu S, Liu W, Zhuo Y, Qu C, Zeng Y. Ferroptosis-related signaling pathways in cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:1. [PMID: 39935430 PMCID: PMC11813627 DOI: 10.20517/cdr.2024.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/16/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025]
Abstract
Ferroptosis is an iron-dependent form of programmed cell death induced by lipid peroxidation. This process is regulated by signaling pathways associated with redox balance, iron metabolism, and lipid metabolism. Cancer cells' increased iron demand makes them especially susceptible to ferroptosis, significantly influencing cancer development, therapeutic response, and metastasis. Recent findings indicate that cancer cells can evade ferroptosis by downregulating key signaling pathways related to this process, contributing to drug resistance. This underscores the possibility of modulating ferroptosis as an approach to counteract drug resistance and enhance therapeutic efficacy. This review outlines the signaling pathways involved in ferroptosis and their interactions with cancer-related signaling pathways. We also highlight the current understanding of ferroptosis in cancer drug resistance, offering insights into how targeting ferroptosis can provide novel therapeutic approaches for drug-resistant cancers. Finally, we explore the potential of ferroptosis-inducing compounds and examine the challenges and opportunities for drug development in this evolving field.
Collapse
Affiliation(s)
- Yang Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- XiangYa School of Medicine, Central South University, Changsha 410013, Hunan, China
| | - Simin Yu
- XiangYa School of Medicine, Central South University, Changsha 410013, Hunan, China
- Department of Urology, Innovation Institute for Integration of Medicine and Engineering, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wanyao Liu
- XiangYa School of Medicine, Central South University, Changsha 410013, Hunan, China
| | - Yi Zhuo
- First Clinical Department of Changsha Medical University, Changsha 410219, Hunan, China
| | - Chunrun Qu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yu Zeng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| |
Collapse
|
35
|
Wang Y, He Z, Dong X, Yao Y, Chen Q, Shi Y, Deng Y, Zhang Q, Yu L, Wang C. Regulation and therapy: the role of ferroptosis in DLBCL. Front Pharmacol 2025; 15:1458412. [PMID: 39834804 PMCID: PMC11743434 DOI: 10.3389/fphar.2024.1458412] [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: 07/02/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of B-cell non-Hodgkin's lymphoma (NHL), up to 30%-40% of patients will relapse and 10%-15% of patients have primary refractory disease, so exploring new treatment options is necessary. Ferroptosis is a non-apoptotic cell death mode discovered in recent years. Its occurrence pathway plays an essential impact on the therapeutic effect of tumors. Numerous studies have shown that modulating critical factors in the ferroptosis pathway can influence the growth of tumor cells in hematological malignancies including DLBCL. This review highlights recent advances in ferroptosis-related genes (FRGs), including STAT3, Nrf2, and ZEB1, and focuses on the clinical potential of ferroptosis inducers such as IKE, α-KG, DMF, and APR-246, which are currently being explored in clinical studies for their therapeutic effects in DLBCL. Correlational studies provide a novel idea for the research and treatment of ferroptosis in DLBCL and other hematological malignancies and lay a solid foundation for future studies.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Zhengmei He
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Xinyu Dong
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Huai’an, China
| | - Yiming Yao
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Huai’an, China
| | - Qiuni Chen
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Yuye Shi
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Yuan Deng
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Quane Zhang
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Liang Yu
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Huai’an, China
| | - Chunling Wang
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Huai’an, China
| |
Collapse
|
36
|
Liu X, Tuerxun H, Zhao Y, Li Y, Wen S, Li X, Zhao Y. Crosstalk between ferroptosis and autophagy: broaden horizons of cancer therapy. J Transl Med 2025; 23:18. [PMID: 39762980 PMCID: PMC11702107 DOI: 10.1186/s12967-024-06059-w] [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: 10/04/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
Ferroptosis and autophagy are two main forms of regulated cell death (RCD). Ferroptosis is a newly identified RCD driven by iron accumulation and lipid peroxidation. Autophagy is a self-degradation system through membrane rearrangement. Autophagy regulates the metabolic balance between synthesis, degradation and reutilization of cellular substances to maintain intracellular homeostasis. Numerous studies have demonstrated that both ferroptosis and autophagy play important roles in cancer pathogenesis and cancer therapy. We also found that there are intricate connections between ferroptosis and autophagy. In this article, we tried to clarify how different kinds of autophagy participate in the process of ferroptosis and sort out the common regulatory pathways between ferroptosis and autophagy in cancer. By exploring the complex crosstalk between ferroptosis and autophagy, we hope to broaden horizons of cancer therapy.
Collapse
Affiliation(s)
- Xingyu Liu
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Halahati Tuerxun
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yixin Zhao
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yawen Li
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Shuhui Wen
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xi Li
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yuguang Zhao
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
37
|
Yang XC, Jin YJ, Ning R, Mao QY, Zhang PY, Zhou L, Zhang CC, Peng YC, Chen N. Electroacupuncture attenuates ferroptosis by promoting Nrf2 nuclear translocation and activating Nrf2/SLC7A11/GPX4 pathway in ischemic stroke. Chin Med 2025; 20:4. [PMID: 39755657 DOI: 10.1186/s13020-024-01047-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 12/12/2024] [Indexed: 01/06/2025] Open
Abstract
OBJECTIVE Electroacupuncture has been shown to play a neuroprotective role following ischemic stroke, but the underlying mechanism remains poorly understood. Ferroptosis has been shown to play a key role in the injury process. In the present study, we wanted to explore whether electroacupuncture could inhibit ferroptosis by promoting nuclear factor erythroid-2-related factor 2 (Nrf2) nuclear translocation. METHODS The ischemic stroke model was established by middle cerebral artery occlusion/reperfusion (MCAO/R) in adult rats. These rats have been randomly divided into the EA + MCAO/R group, the MCAO/R group, the EA + MCAO/R + Brusatol group (the inhibitor of Nrf2), and the EA + MCAO/R + DMSO group, and the Sham group. The EA + MCAO/R group, EA + MCAO/R + Brusatol group, and the EA + MCAO/R + DMSO group received EA intervention 24 h after modeling for 7 consecutive days. The behavioral function was evaluated by Neurologic severity score (NSS), Garcia score, Foot-fault Test, and Rotarod Test. The infarct volume was detected by TTC staining, and the neuronal damage was observed by Nissl staining. The levels of Fe2+, reactive oxygen species (ROS), superoxide dismutase (SOD), and malondialdehyde (MDA) were measured by ELISA. The immunofluorescence and Western blotting were used to detect the expression of Total Nrf2, p-Nrf2, Nuclear Nrf2, and Cytoplasmic Nrf2, and the essential ferroptosis proteins, including glutathione peroxidase 4 (GPX4), solute carrier family 7 member 11 (SLC7A11) and ferritin heavy chain 1 (FTH1). The mitochondria were observed by transmission electron microscopy (TEM). RESULTS Electroacupuncture improved neurological deficits in rats model of MCAO/R, decreased the brain infarct volume, alleviated neuronal damage, inhibited the Fe2+, ROS, and MDA accumulation, increased SOD levels, increased the expression of GPX4, SLC7A11 and FTH1, and rescued injured mitochondria. Especially, we found that the electroacupuncture up-regulated the expression of Nrf2, and promoted phosphorylation of Nrf2 and nuclear translocation, However, Nrf2 inhibitor Brusatol reversed the neuroprotective effect of electroacupuncture. CONCLUSION Electroacupuncture can alleviate cerebral I/R injury-induced ferroptosis by promoting Nrf2 nuclear translocation. It is expected that these data will provide novel insights into the mechanisms of electroacupuncture protecting against cerebral I/R injury and potential targets underlying ferroptosis in the stroke.
Collapse
Affiliation(s)
- Xi-Chen Yang
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Ya-Ju Jin
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China.
| | - Rong Ning
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Qiu-Yue Mao
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Peng-Yue Zhang
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Li Zhou
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Cheng-Cai Zhang
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Yi-Chen Peng
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Na Chen
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| |
Collapse
|
38
|
Kim JM, Kim Y, Na HJ, Hur HJ, Lee SH, Sung MJ. Magnolia kobus DC. suppresses neointimal hyperplasia by regulating ferroptosis and VSMC phenotypic switching in a carotid artery ligation mouse model. Chin Med 2025; 20:3. [PMID: 39754271 PMCID: PMC11699803 DOI: 10.1186/s13020-024-01051-4] [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: 09/06/2024] [Accepted: 12/19/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Magnolia kobus DC (MO), as a plant medicine, has been reported to have various physiological activities, including neuroprotective, anti-inflammatory, and anti-diabetic effects. However, vascular protective effects of MO remain incompletely understood. In this study, we evaluated the vascular protective effect of MO against ferroptosis in a carotid artery ligation (CAL)-induced neointimal hyperplasia mouse model and in aortic thoracic smooth muscle A7r5 cells. METHODS This study was conducted to estimate the vascular protective effects of MO by systematically measuring histopathological analysis and western blot analysis in CAL animal model. In vitro protective effects of MO were evaluated by estimating cell viability, reactive oxygen species (ROS) content, glutathione (GSH) levels, lipid peroxidation, mitochondrial morphological change, cell proliferation, migration, western blot analysis, and qRT-PCR against erastin (Era)-induced A7r5 cells. RESULTS MO intake significantly improved neointimal formation, inhibited ferroptosis and vascular smooth muscle cell (VSMC) phenotypes, and ameliorated the antioxidant system of carotid artery tissues. In addition, MO treatment effectively ameliorated Era-induced ferroptotic cytotoxicity, including cellular death, ROS production, and cell migration status. MO treatment also suppressed proliferation and migration in Era-induced A7r5 cells. MO considerably regulated Era-induced abnormal mechanisms related to ferroptotic changes, VSMC phenotype switching, and the ROS scavenging system in A7r5 cells. CONCLUSION MO has the potential for use as a functional food supplement, nutraceutical, or medicinal food, with protective effects on vascular health by regulating ferroptosis and VSMC phenotypic switching.
Collapse
Affiliation(s)
- Jong Min Kim
- Aging and Metabolism Research Group, Korea Food Research Institute, Wanju‑gun, 55365, Republic of Korea
| | - Yiseul Kim
- Aging and Metabolism Research Group, Korea Food Research Institute, Wanju‑gun, 55365, Republic of Korea
| | - Hyun-Jin Na
- Aging and Metabolism Research Group, Korea Food Research Institute, Wanju‑gun, 55365, Republic of Korea
| | - Haeng Jeon Hur
- Aging and Metabolism Research Group, Korea Food Research Institute, Wanju‑gun, 55365, Republic of Korea
| | - Sang Hee Lee
- Aging and Metabolism Research Group, Korea Food Research Institute, Wanju‑gun, 55365, Republic of Korea
| | - Mi Jeong Sung
- Aging and Metabolism Research Group, Korea Food Research Institute, Wanju‑gun, 55365, Republic of Korea.
| |
Collapse
|
39
|
Tian R, Guo S, Chen S, Wu J, Long A, Cheng R, Wang X, Huang L, Li C, Mao W, Xu P, Yu L, Pan H, Liu L. Natural products as Nrf2 modulators for ferroptosis inhibition in renal disease therapy: Recent progress and future prospects. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156342. [PMID: 39742572 DOI: 10.1016/j.phymed.2024.156342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 01/03/2025]
Abstract
BACKGROUND The transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2, NFE2L2) is a pivotal regulator of redox balance, metabolism, protein homeostasis and inflammation. Nrf2 is critically involved in both ferroptosis and renal diseases, and may serve as a significant target for many natural products in the treatment of renal diseases. However, a comprehensive overview on this topic is still lacking. PURPOSE To review the protective or therapeutic effects of natural products regulating Nrf2-related ferroptosis against various renal diseases. METHODS We systematically searched the electronic databases involving PubMed, Web of Science, Google Scholar, China National Knowledge Internet (CNKI), Wanfang Database and VIP Database. To ensure a comprehensive exploration, keywords including Nrf2, ferroptosis, natural products, phytochemicals, renal disease, kidney disease, kidney injury and nephropathy were employed. RESULTS Ferroptosis is deeply implicated in various kinds of renal diseases, notably including cisplatin-induced acute kidney injury, sepsis-associated acute kidney injury, renal ischemia/reperfusion injury, diabetic nephropathy, kidney stones and renal fibrosis. Nrf2 plays a regulatory role on many important genes related to iron metabolism, antioxidant system and lipid metabolism, thereby modulating ferroptosis. More than twenty natural products exert renoprotective effects by inhibiting ferroptosis via the regulation of Nrf2. This review presents a comprehensive overview of recent advancements in elucidating the ferroptosis involvement in renal diseases, the role of Nrf2 in regulating ferroptosis, and summarizes the renoprotective natural products as Nrf2 modulators for ferroptosis inhibition. CONCLUSION Through the comprehensive insights, this review clarifies the protective or therapeutic effects of natural products as Nrf2 modulators for ferroptosis inhibition in renal disease therapy, in the pursuit of providing new research ideas and directions for the treatment of renal diseases. Further drug development aimed at discovering more natural products and optimizing their utilization for disease treatment is necessary.
Collapse
Affiliation(s)
- Ruimin Tian
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Shan Guo
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shudong Chen
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jiaqi Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Aoyang Long
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Ran Cheng
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiaowan Wang
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Lihua Huang
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Chuang Li
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Wei Mao
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China
| | - Peng Xu
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China
| | - Lili Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Hudan Pan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China.
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China.
| |
Collapse
|
40
|
Xie Z, Hou Q, He Y, Xie Y, Mo Q, Wang Z, Zhao Z, Chen X, Peng T, Li L, Xie W. Ferritin Hinders Ferroptosis in Non-Tumorous Diseases: Regulatory Mechanisms and Potential Consequences. Curr Protein Pept Sci 2025; 26:89-104. [PMID: 39225224 DOI: 10.2174/0113892037315874240826112422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
Ferritin, as an iron storage protein, has the potential to inhibit ferroptosis by reducing excess intracellular free iron concentrations and lipid reactive oxygen species (ROS). An insufficient amount of ferritin is one of the conditions that can lead to ferroptosis through the Fenton reaction mediated by ferrous iron. Consequently, upregulation of ferritin at the transcriptional or posttranscriptional level may inhibit ferroptosis. In this review, we have discussed the essential role of ferritin in ferroptosis and the regulatory mechanism of ferroptosis in ferritin-deficient individuals. The description of the regulatory factors governing ferritin and its properties in regulating ferroptosis as underlying mechanisms for the pathologies of diseases will allow potential therapeutic approaches to be developed.
Collapse
Affiliation(s)
- Zhongcheng Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Qin Hou
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yinling He
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yushu Xie
- Class of Clinical Medicine, University of South China, Hengyang 421001, Hunan, China
| | - Qinger Mo
- Class of Clinical Medicine, University of South China, Hengyang 421001, Hunan, China
| | - Ziyi Wang
- Class of Clinical Medicine, University of South China, Hengyang 421001, Hunan, China
| | - Ziye Zhao
- Class of Clinical Medicine, University of South China, Hengyang 421001, Hunan, China
| | - Xi Chen
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Tianhong Peng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Liang Li
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Wei Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| |
Collapse
|
41
|
Lucas O, Ward S, Zaidi R, Bunkum A, Frankell AM, Moore DA, Hill MS, Liu WK, Marinelli D, Lim EL, Hessey S, Naceur-Lombardelli C, Rowan A, Purewal-Mann SK, Zhai H, Dietzen M, Ding B, Royle G, Aparicio S, McGranahan N, Jamal-Hanjani M, Kanu N, Swanton C, Zaccaria S. Characterizing the evolutionary dynamics of cancer proliferation in single-cell clones with SPRINTER. Nat Genet 2025; 57:103-114. [PMID: 39614124 PMCID: PMC11735394 DOI: 10.1038/s41588-024-01989-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/15/2024] [Indexed: 12/01/2024]
Abstract
Proliferation is a key hallmark of cancer, but whether it differs between evolutionarily distinct clones co-existing within a tumor is unknown. We introduce the Single-cell Proliferation Rate Inference in Non-homogeneous Tumors through Evolutionary Routes (SPRINTER) algorithm that uses single-cell whole-genome DNA sequencing data to enable accurate identification and clone assignment of S- and G2-phase cells, as assessed by generating accurate ground truth data. Applied to a newly generated longitudinal, primary-metastasis-matched dataset of 14,994 non-small cell lung cancer cells, SPRINTER revealed widespread clone proliferation heterogeneity, orthogonally supported by Ki-67 staining, nuclei imaging and clinical imaging. We further demonstrated that high-proliferation clones have increased metastatic seeding potential, increased circulating tumor DNA shedding and clone-specific altered replication timing in proliferation- or metastasis-related genes associated with expression changes. Applied to previously generated datasets of 61,914 breast and ovarian cancer cells, SPRINTER revealed increased single-cell rates of different genomic variants and enrichment of proliferation-related gene amplifications in high-proliferation clones.
Collapse
Affiliation(s)
- Olivia Lucas
- Computational Cancer Genomics Research Group, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- University College London Hospitals, London, UK
| | - Sophia Ward
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Genomics Science Technology Platform, The Francis Crick Institute, London, UK
| | - Rija Zaidi
- Computational Cancer Genomics Research Group, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Abigail Bunkum
- Computational Cancer Genomics Research Group, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
| | - Alexander M Frankell
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - David A Moore
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Department of Cellular Pathology, University College London Hospitals, London, UK
| | - Mark S Hill
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Wing Kin Liu
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
| | - Daniele Marinelli
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, University College London Cancer Institute, London, UK
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Emilia L Lim
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Sonya Hessey
- Computational Cancer Genomics Research Group, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- University College London Hospitals, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
| | | | - Andrew Rowan
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | | | - Haoran Zhai
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Michelle Dietzen
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Genome Evolution Research Group, University College London Cancer Institute, London, UK
| | - Boyue Ding
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Gary Royle
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Samuel Aparicio
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, University College London Cancer Institute, London, UK
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- University College London Hospitals, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
| | - Nnennaya Kanu
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
- University College London Hospitals, London, UK.
| | - Simone Zaccaria
- Computational Cancer Genomics Research Group, University College London Cancer Institute, London, UK.
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
| |
Collapse
|
42
|
Fan L, Guo D, Zhu C, Gao C, Wang Y, Yin F, Liu M, Zhou Y, Wei T, Xiong X, Yu K, Le A. LRRC45 accelerates bladder cancer development and ferroptosis inhibition via stabilizing NRF2 by competitively KEAP1 interaction. Free Radic Biol Med 2025; 226:29-42. [PMID: 39522565 DOI: 10.1016/j.freeradbiomed.2024.11.001] [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: 09/14/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Centrosomal dysregulation is closely linked to the genesis and progression of tumors. A comprehensive analysis of single-cell RNA sequencing (scRNA-seq) data has revealed that leucine-rich repeat-containing protein 45 (LRRC45), a centrosome linker protein crucial for maintaining centrosome cohesion and a member of the leucine-rich repeat-containing proteins (LRRCs) family, is significantly upregulated in bladder cancer. Notably, the elevated expression levels of LRRC45 were strongly correlated with a poor prognosis in patients. Furthermore, the depletion of LRRC45 in bladder cancer cells markedly inhibited tumorigenic proliferation and increased intracellular iron and reactive oxygen species (ROS) levels. It ultimately triggered ferroptosis, an iron-dependent form of programmed cell death characterized by lipid peroxidation. Mechanistic studies revealed that LRRC45 exerts its oncogenic effects through competitive interaction with Kelch-like ECH-associated protein 1 (KEAP1), which inhibits the ubiquitin-proteasome-mediated degradation of nuclear factor erythroid 2-related factor 2 (NRF2). This interaction enhances the nuclear translocation of NRF2 and its subsequent anti-ferroptotic activity. In conclusion, our studies highlight the critical role of LRRC45 in enhancing the stability of NRF2, thereby promoting the tumorigenic potential of bladder cancer. These insights suggest that targeting LRRC45 could serve as a promising molecular target for developing novel therapeutic interventions for bladder cancer.
Collapse
Affiliation(s)
- Linwei Fan
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Dingfan Guo
- The First Clinical Medical School of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Chao Zhu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Chenqi Gao
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Yu Wang
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Fang Yin
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Mengwei Liu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Yanyu Zhou
- The First Clinical Medical School of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Tiancheng Wei
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Xinxin Xiong
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Kuai Yu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| | - Aiping Le
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
43
|
Zhang S, Guo L, Tao R, Liu S. Ferroptosis-targeting drugs in breast cancer. J Drug Target 2025; 33:42-59. [PMID: 39225187 DOI: 10.1080/1061186x.2024.2399181] [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: 05/19/2024] [Revised: 07/07/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
In 2020, breast cancer surpassed lung cancer as the most common cancer in the world for the first time. Due to the resistance of some breast cancer cell lines to apoptosis, the therapeutic effect of anti-breast cancer drugs is limited. According to recent report, the susceptibility of breast cancer cells to ferroptosis affects the progress, prognosis and drug resistance of breast cancer. For instance, roblitinib induces ferroptosis of trastuzumab-resistant human epidermal growth factor receptor 2 (HER2)-positive breast cancer cells by diminishing fibroblast growth factor receptor 4 (FGFR4) expression, thereby augmenting the susceptibility of these cells to HER2-targeted therapies. In tamoxifen-resistant breast cancer cells, Fascin exacerbates their resistance by repressing solute carrier family 7 member 11 (SLC7A11) expression, which in turn heightens their responsiveness to tamoxifen. In recent years, Chinese herbs extracts and therapeutic drugs have been demonstrated to elicit ferroptosis in breast cancer cells by modulating a spectrum of regulatory factors pertinent to ferroptosis, including SLC7A11, glutathione peroxidase 4 (GPX4), acyl-CoA synthetase long chain family member 4 (ACSL4), and haem oxygenase 1 (HO-1). Here, we review the roles and mechanisms of Chinese herbal extracts and therapeutic drugs in regulating ferroptosis in breast cancer, providing potential therapeutic options for anti-breast cancer.
Collapse
Affiliation(s)
- Shuxian Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, China
| | - Lijuan Guo
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, China
| | - Ran Tao
- Department of Anatomy, Medical College, Dalian University, Dalian, China
| | - Shuangping Liu
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, China
| |
Collapse
|
44
|
Shimizu K, Inuzuka H, Tokunaga F. The interplay between cell death and senescence in cancer. Semin Cancer Biol 2025; 108:1-16. [PMID: 39557316 DOI: 10.1016/j.semcancer.2024.11.001] [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: 08/15/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
Cellular senescence is a state of permanent proliferative arrest that occurs in response to DNA damage-inducing endogenous and exogenous stresses, and is often accompanied by dynamic molecular changes such as a senescence-associated secretory phenotype (SASP). Accumulating evidence indicates that age-associated increases in the upstream and downstream signals of regulated cell death, including apoptosis, necroptosis, pyroptosis, and ferroptosis, are closely related to the induction of cellular senescence and its phenotype. Furthermore, elevated levels of pro-inflammatory SASP factors with aging can be both a cause and consequence of several cell death modes, suggesting the reciprocal effects of cellular senescence and cells undergoing regulated cell death. Here, we review the critical molecular pathways of the regulated cell death forms and describe the crosstalk between aging-related signals and cancer. In addition, we discuss how targeting regulated cell death could be harnessed in therapeutic interventions for cancer. ABBREVIATIONS: Abbreviations that are not standard in this field are defined at their first occurrence in the article and are used consistently throughout the article.
Collapse
Affiliation(s)
- Kouhei Shimizu
- Department of Medical Biochemistry, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan.
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA02215, USA
| | - Fuminori Tokunaga
- Department of Medical Biochemistry, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan
| |
Collapse
|
45
|
Wei Z, Zhou Z, Zhang Y, Wang J, Huang K, Ding Y, Sun Y, Gu M, Kong X, Xi E, Zeng S. PRKAA2 Promotes Tumor Growth and Inhibits Ferroptosis through SLC7A11/GSH/GPX4 Pathway in Non-Small Cell Lung Cancer. Biotechnol Appl Biochem 2024. [PMID: 39722125 DOI: 10.1002/bab.2710] [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: 07/08/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024]
Abstract
Non-small cell lung cancer (NSCLC) is the most pervasive sort of lung cancer with deadly outcome. According to recent studies, a number of neoplastic disorders and ferroptosis are intimately connected. This study aims to identify the role of key ferroptosis-related gene (protein kinase AMP-activated catalytic subunit alpha 2, PRKAA2) and explore new directions for the diagnosis and treatment of NSCLC. The PRKAA2 expression and its influence on survival were analyzed in multiple public databases (TCGA, TIMER2.0, and GEPIA). And PRKAA2 mRNA level in NSCLC cells were examined by qRT-PCR. Silencing of PRKAA2 (sh-PRKAA2) were used to cell transfection. CCK-8, EdU, and flow cytometry assays were used to measure cell proliferation and apoptosis. The protein levels of ferroptosis markers (SLC7A11, GPX4, and NRF2) were determined by western blotting. Meanwhile, the related ferroptosis analysis, such as malondialdehyde (MDA) and glutathione (GSH), reactive oxygen species (ROS), iron, and Fe2+ levels were also detected in the transfected cells. Moreover, the relationship between PRKAA2 expression and SLC7A11 was analyzed. NSCLC xenograft mouse models were used for in vivo verification of the PRKAA2 function. Here, our data revealed that PRKAA2 was upregulated in NSCLC cells. Additionally, PRKAA2 strengthened cell proliferation and attenuated apoptosis and ferroptosis of NSCLC cells. The depletion of PRKAA2 enhanced the erastin-induced inhibition effect on cell growth, and notably increased the levels of MDA, ROS, iron, and Fe2+, while decreased GSH level in NSCLC cells. In the mechanism exploration, we discovered that PRKAA2 could activate the SLC7A11/GSH/GPx4 antioxidant pathway. The rescue experiments showed that SLC7A11 abrogated the inhibitive impacts of PRKAA2 repression on cellular proliferation, cell apoptosis, and ferroptosis in NSCLC. Besides, animal experiments proved that PRKAA2 enhanced NSCLC tumor growth in vivo. The results discovered that PRKAA2 accelerated the malignant progression, diminished apoptosis and ferroptosis in NSCLC through SLC7A11/GSH/GPX4 pathway. This study provide a novel target in the application of PRKAA2 for NSCLC treatment.
Collapse
Affiliation(s)
- Zhiqiang Wei
- Department of Radiation and Medical Oncology, Affiliated Sanming First Hospital of Fujian Medical University, Sanming, Fujian, China
| | - Zhilian Zhou
- Department of Radiation and Medical Oncology, Affiliated Sanming First Hospital of Fujian Medical University, Sanming, Fujian, China
| | - Yu Zhang
- Department of Thoracic Cardiovascular Surgery, Central Theater General Hospital of PLA, Wuhan, Hubei, China
| | - Jie Wang
- Department of Thoracic Cardiovascular Surgery, Central Theater General Hospital of PLA, Wuhan, Hubei, China
| | - Ke Huang
- Department of Thoracic Cardiovascular Surgery, Central Theater General Hospital of PLA, Wuhan, Hubei, China
| | - Yuanyu Ding
- Department of Thoracic Cardiovascular Surgery, Central Theater General Hospital of PLA, Wuhan, Hubei, China
| | - Yingming Sun
- Department of Radiation and Medical Oncology, Affiliated Sanming First Hospital of Fujian Medical University, Sanming, Fujian, China
| | - Mingming Gu
- Department of Thoracic Cardiovascular Surgery, Central Theater General Hospital of PLA, Wuhan, Hubei, China
| | - Xiangang Kong
- Department of Thoracic Cardiovascular Surgery, Central Theater General Hospital of PLA, Wuhan, Hubei, China
| | - Erping Xi
- Department of Thoracic Cardiovascular Surgery, Central Theater General Hospital of PLA, Wuhan, Hubei, China
| | - Shaoshan Zeng
- Department of Thoracic Surgery, Affiliated Sanming First Hospital of Fujian Medical University Sanming, Fujian, China
| |
Collapse
|
46
|
Tan R, Ge C, Yan Y, Guo H, Han X, Zhu Q, Du Q. Deciphering ferroptosis in critical care: mechanisms, consequences, and therapeutic opportunities. Front Immunol 2024; 15:1511015. [PMID: 39737174 PMCID: PMC11682965 DOI: 10.3389/fimmu.2024.1511015] [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: 10/14/2024] [Accepted: 12/03/2024] [Indexed: 01/01/2025] Open
Abstract
Ischemia-reperfusion injuries (IRI) across various organs and tissues, along with sepsis, significantly contribute to the progression of critical illnesses. These conditions disrupt the balance of inflammatory mediators and signaling pathways, resulting in impaired physiological functions in human tissues and organs. Ferroptosis, a distinct form of programmed cell death, plays a pivotal role in regulating tissue damage and modulating inflammatory responses, thereby influencing the onset and progression of severe illnesses. Recent studies highlight that pharmacological agents targeting ferroptosis-related proteins can effectively mitigate oxidative stress caused by IRI in multiple organs, alleviating associated symptoms. This manuscript delves into the mechanisms and signaling pathways underlying ferroptosis, its role in critical illnesses, and its therapeutic potential in mitigating disease progression. We aim to offer a novel perspective for advancing clinical treatments for critical illnesses.
Collapse
Affiliation(s)
- Ruimin Tan
- School of Clinical Medical, North China University of Science and Technology, Tangshan, Hebei, China
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Chen Ge
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Yating Yan
- School of Clinical Medical, North China University of Science and Technology, Tangshan, Hebei, China
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - He Guo
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
- School of Graduate, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xumin Han
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
- School of Graduate, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qiong Zhu
- Department of Orthopaedics, The People’s Hospital Of Shizhu, Chongqing, China
| | - Quansheng Du
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
47
|
Carlos A, Mendes M, Cruz MT, Pais A, Vitorino C. Ferroptosis driven by nanoparticles for tackling glioblastoma. Cancer Lett 2024; 611:217392. [PMID: 39681210 DOI: 10.1016/j.canlet.2024.217392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
Glioblastoma (GBM) is the most aggressive, malignant, and drug-resistant brain tumor. There are no effective treatment options for GBM, which usually leads to relapses that cause patients to die a few months later. Ferroptosis, a newly discovered mechanism of regulated cell death, has been identified as a tumor suppressor in solid tumors and represents an alternative to apoptosis resistance. This mechanism of cell death is characterized by iron overload, which is responsible for generating reactive oxygen species (ROS) in the cell. Understanding the ferroptosis pathway and its key regulators can be used to develop rational delivery systems that specifically target these regulators in GBM cells and promote cell death. This review conducted a systematic literature search to better understand the potential of ferroptosis as a target for developing nanoparticles to tackle GBM. The mechanisms of action, design parameters, efficacy, and safety concerns of 16 nanoparticles were evaluated, demonstrating the potential of combining ferroptosis inducers with nanocarriers to promote a selective delivery to the tumor microenvironment.
Collapse
Affiliation(s)
- Ana Carlos
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Maria Mendes
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences-IMS, Departmente of Chemistry, University of Coimbra, 3004-535, Coimbra, Portugal
| | - Maria T Cruz
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal; Center for Neurosciences and Cell Biology (CNC) and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548, Coimbra, Portugal
| | - Alberto Pais
- Coimbra Chemistry Centre, Institute of Molecular Sciences-IMS, Departmente of Chemistry, University of Coimbra, 3004-535, Coimbra, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences-IMS, Departmente of Chemistry, University of Coimbra, 3004-535, Coimbra, Portugal.
| |
Collapse
|
48
|
Schiavoni V, Emanuelli M, Milanese G, Galosi AB, Pompei V, Salvolini E, Campagna R. Nrf2 Signaling in Renal Cell Carcinoma: A Potential Candidate for the Development of Novel Therapeutic Strategies. Int J Mol Sci 2024; 25:13239. [PMID: 39769005 PMCID: PMC11675435 DOI: 10.3390/ijms252413239] [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: 11/06/2024] [Revised: 12/05/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Renal cell carcinoma (RCC) is the most common type of kidney cancer arising from renal tubular epithelial cells and is characterized by a high aggressive behavior and invasiveness that lead to poor prognosis and high mortality rate. Diagnosis of RCC is generally incidental and occurs when the stage is advanced and the disease is already metastatic. The management of RCC is further complicated by an intrinsic resistance of this malignancy to chemotherapy and radiotherapy, which aggravates the prognosis. For these reasons, there is intense research focused on identifying novel biomarkers which may be useful for a better prognostic assessment, as well as molecular markers which could be utilized for targeted therapy. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcriptional factor that has been identified as a key modulator of oxidative stress response, and its overexpression is considered a negative prognostic feature in several types of cancers including RCC, since it is involved in various key cancer-promoting functions such as proliferation, anabolic metabolism and resistance to chemotherapy. Given the key role of Nrf2 in promoting tumor progression, this enzyme could be a promising biomarker for a more accurate prediction of RCC course and it can also represent a valuable therapeutic target. In this review, we provide a comprehensive literature analysis of studies that have explored the role of Nrf2 in RCC, underlining the possible implications for targeted therapy.
Collapse
Affiliation(s)
- Valentina Schiavoni
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy; (V.S.); (M.E.); (G.M.); (A.B.G.); (V.P.)
| | - Monica Emanuelli
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy; (V.S.); (M.E.); (G.M.); (A.B.G.); (V.P.)
- New York-Marche Structural Biology Center (NY-MaSBiC), Polytechnic University of Marche, 60131 Ancona, Italy
| | - Giulio Milanese
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy; (V.S.); (M.E.); (G.M.); (A.B.G.); (V.P.)
| | - Andrea Benedetto Galosi
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy; (V.S.); (M.E.); (G.M.); (A.B.G.); (V.P.)
| | - Veronica Pompei
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy; (V.S.); (M.E.); (G.M.); (A.B.G.); (V.P.)
| | - Eleonora Salvolini
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy; (V.S.); (M.E.); (G.M.); (A.B.G.); (V.P.)
| | - Roberto Campagna
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy; (V.S.); (M.E.); (G.M.); (A.B.G.); (V.P.)
| |
Collapse
|
49
|
Xie Y, Liang B, Meng Z, Guo R, Liu C, Yuan Y, Mu W, Wang Y, Cao J. Down-regulation of HSPB1 and MGST1 promote ferroptosis and impact immune infiltration in diabetic cardiomyopathy. RESEARCH SQUARE 2024:rs.3.rs-5153598. [PMID: 39711549 PMCID: PMC11661379 DOI: 10.21203/rs.3.rs-5153598/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Diabetic cardiomyopathy (DCM) is a leading cause of death in diabetic patients. Current therapies do not adequately resolve this problem and focus only on the optimal level of blood glucose for patients. Ferroptosis plays an important role in diabetes mellitus and cardiovascular diseases. However, the role of ferroptosis in DCM remains unclear. Differentially expressed ferroptosis-related genes (DE-FRGs) were identified by intersecting GSE26887 dataset and the Ferroptosis Database (FerrDb). The associations between the DE-FRGs and immune cells in DCM, estimated by CIBERSORTx algorithm, were analyzed. Using ow cytometry (FCM) to evaluated the infiltration of immune cells of myocardial tissues. The expression of DE-FRGs, Glutathione peroxidase 4 (GPX4) and Solute carrier family 7 member 11 (SLC7A11) were examined by real-time quantitative PCR and western blotting. 3 DE-FRGs were identified, which are Heat shock protein family B (small) member 1 (HSPB1), Microsomal glutathione S-transferase 1 (MGST1) and solute carrier family 40 member 1 (SLC40A1) respectively, and they were closely linked to immune cells in DCM. In vivo, the levels of CD8 + T cells, B cells and Treg cells were significantly decreased in the DCM group, while the levels of CD4 + T cells, M1 cells, M2 cells and monocytes were increased. Diabetes significantly decreased HSPB1 and MGST1 levels and increased ferroptosis compared to normal group. Furthermore, ferroptosis inhibitor ferrostatin-1 (Fer-1) alleviated high-fat diet (HFD)-induced cadiomyocyte injury and rescued the ferroptosis. This study suggests that ferroptosis related gene HSPB1 and MGST1 are closely related to immune cell infiltration, which may become therapeutic targets for DCM.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Mu
- The Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital)
| | | | | |
Collapse
|
50
|
Tang L, He D, Su B. Nrf2: A critical participant in regulation of apoptosis, ferroptosis, and autophagy in gastric cancer. Acta Histochem 2024; 126:152203. [PMID: 39342913 DOI: 10.1016/j.acthis.2024.152203] [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: 05/05/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024]
Abstract
Nuclear factor erythroid 2-related factor-2 (Nrf2) is a specific transcription factor that maintains redox homeostasis by regulating the expression of anti-oxidative stress-related genes. Hyperactivation of Nrf2 is involved in tumor progression and is associated with chemoresistance in a large number of solid tumors. Programmatic cell death (PCD), such as apoptosis, ferroptosis, and autophagy, plays a crucial role in tumor development and chemotherapy sensitivity. Accumulating evidence suggests that some anti-tumor compounds and genes can induce massive production of reactive oxygen species (ROS) via inhibiting Nrf2 expression, which exacerbates oxidative stress and promotes Gastric cancer (GC) cell death, thereby enhancing the sensitivity of GC cells to chemotherapy-induced PCD. In this review, we summarize the role of antitumor drugs in interfering in three different types of PCD (apoptosis, ferroptosis, and autophagy) in GC cells by modulating Nrf2 expression, as well as the molecular mechanisms through which targeting Nrf2 brings about PCD and chemosensitivity. It is reasonable to believe that Nrf2 serves as a potential therapeutic target, and targeting Nrf2 by drug or gene regulation could provide a new strategy for the treatment of GC.
Collapse
Affiliation(s)
- LiJie Tang
- Institute of Pharmacy and Pharmacology, School of Pharmacy, Hengyang Medical School, University of South China, Hengyang, China
| | - DongXiu He
- Institute of Pharmacy and Pharmacology, School of Pharmacy, Hengyang Medical School, University of South China, Hengyang, China
| | - Bo Su
- Institute of Pharmacy and Pharmacology, School of Pharmacy, Hengyang Medical School, University of South China, Hengyang, China.
| |
Collapse
|