1
|
Lv X, Yang C, Li X, Liu Y, Yang Y, Jin T, Chen Z, Jia J, Wang M, Li L. Ferroptosis and hearing loss: from molecular mechanisms to therapeutic interventions. J Enzyme Inhib Med Chem 2025; 40:2468853. [PMID: 39992186 PMCID: PMC11852237 DOI: 10.1080/14756366.2025.2468853] [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: 08/27/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025] Open
Abstract
Hearing loss profoundly affects social engagement, mental health, cognition, and brain development, with sensorineural hearing loss (SNHL) being a major concern. Linked to ototoxic medications, ageing, and noise exposure, SNHL presents significant treatment challenges, highlighting the need for effective prevention and regeneration strategies. Ferroptosis, a distinct form of cell death featuring iron-dependent lipid peroxidation, has garnered interest due to its potential role in cancer, ageing, and neuronal degeneration, especially hearing loss. The emerging role of ferroptosis as a crucial mediator in SNHL suggests that it may offer a novel therapeutic target for otoprotection. This review aims to summarise the intricate connection between ferroptosis and SNHL, offering a fresh perspective for exploring targeted therapeutic strategies that could potentially mitigate cochlear cells damage and enhance the quality of life for individuals with hearing impairments.
Collapse
Affiliation(s)
- Xingyi Lv
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Chenyi Yang
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Xianying Li
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Yun Liu
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Yu Yang
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Tongyan Jin
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhijian Chen
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Jinjing Jia
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Min Wang
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Li Li
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
2
|
Shao N, Cai K, Hong Y, Wu L, Luo Q. USP9X suppresses ferroptosis in diabetic kidney disease by deubiquitinating Nrf2 in vitro. Ren Fail 2025; 47:2458761. [PMID: 39967230 PMCID: PMC11841168 DOI: 10.1080/0886022x.2025.2458761] [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: 08/21/2024] [Revised: 12/22/2024] [Accepted: 01/21/2025] [Indexed: 02/20/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) regulates many critical genes associated with iron storage and transportation, the activity of which is influenced by E3 ligase-mediated ubiquitination. We wondered whether there is a deubiquitinase that mediates the deubiquitination of Nrf2 to stabilize Nrf2 expression and further prevent diabetic kidney disease (DKD). High glucose (HG) was applied to induce an in vitro model of DKD. The effects of HG on HK-2 cell viability, apoptosis, Fe2+ level, Nrf2, and ubiquitin-specific protease 9X (USP9X) were assessed by cell counting kit-8 (CCK-8) assay, flow cytometry, iron assay, and Western blot. The direct interaction between Nrf2 and USP9X was analyzed using co-immunoprecipitation and ubiquitination assay. After transfection and ferrostatin-1 (Fer-1) intervention, Nrf2 and USP9X levels, cell viability, apoptosis, and Fe2+ level were tested again. Reactive oxygen species (ROS), malondialdehyde (MDA), glutathione (GSH) contents, and ferroptosis-related markers were assessed by ROS assay kit, ELISA, and Western blot. HG reduced cell viability and levels of USP9X and Nrf2, while elevating apoptosis and Fe2+ level. An interaction between USP9X and Nrf2 has been verified and USP9X deubiquitinated Nrf2. Nrf2 up-regulation augmented the viability, GSH content, and ferroptosis-related protein expressions, while suppressing the apoptosis, Fe2+ level, MDA, and ROS content in HG-mediated HK-2 cells, which was reversed by USP9X silencing. Fer-1 offset the combined modulation of Nrf2 and siUSP9X on HG-induced HK-2 cells. USP9X mediates Nrf2 deubiquitinase to hamper the ferroptosis in DKD in vitro.
Collapse
Affiliation(s)
- Ningjun Shao
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, China
| | - Kedan Cai
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, China
| | - Yue Hong
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, China
| | - Lingping Wu
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, China
| | - Qun Luo
- Department of Nephrology, Ningbo No.2 Hospital, Ningbo, China
| |
Collapse
|
3
|
Yang H, Xia Y, Ma Y, Gao M, Hou S, Xu S, Wang Y. Inhibition of the cGAS-STING pathway: contributing to the treatment of cerebral ischemia-reperfusion injury. Neural Regen Res 2025; 20:1900-1918. [PMID: 38993125 PMCID: PMC11691458 DOI: 10.4103/nrr.nrr-d-24-00015] [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: 01/05/2024] [Revised: 03/05/2024] [Accepted: 05/02/2024] [Indexed: 07/13/2024] Open
Abstract
The cGAS-STING pathway plays an important role in ischemia-reperfusion injury in the heart, liver, brain, and kidney, but its role and mechanisms in cerebral ischemia-reperfusion injury have not been systematically reviewed. Here, we outline the components of the cGAS-STING pathway and then analyze its role in autophagy, ferroptosis, cellular pyroptosis, disequilibrium of calcium homeostasis, inflammatory responses, disruption of the blood-brain barrier, microglia transformation, and complement system activation following cerebral ischemia-reperfusion injury. We further analyze the value of cGAS-STING pathway inhibitors in the treatment of cerebral ischemia-reperfusion injury and conclude that the pathway can regulate cerebral ischemia-reperfusion injury through multiple mechanisms. Inhibition of the cGAS-STING pathway may be helpful in the treatment of cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Hang Yang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yulei Xia
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yue Ma
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Mingtong Gao
- Department of Emergency, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| | - Shuai Hou
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Shanshan Xu
- Department of Emergency, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| | - Yanqiang Wang
- Department of Neurology II, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| |
Collapse
|
4
|
Zhang H, Tian L, Wang P, Li L, Wang K, Li Y, Zhang Y, Feng L, Yao S, Guan H, Ren W. Ferrostatin-1 mitigates acute lung injury by reducing ferroptosis levels in gas explosions. Tissue Cell 2025; 94:102773. [PMID: 39954560 DOI: 10.1016/j.tice.2025.102773] [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: 11/26/2024] [Revised: 01/15/2025] [Accepted: 02/01/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Gas explosion injuries are a severe form of trauma with high incidence and mortality rates, both in daily life and industrial settings. Acute lung injury (ALI) is one of the most serious complications of gas explosion injuries and is a leading cause of mortality in such cases. However, the mechanisms underlying gas explosion-induced ALI have not been fully elucidated, and the treatment process consumes a significant amount of medical resources. Therefore, it is crucial to conduct research on the injury mechanisms of gas explosion injuries, especially the mechanisms of gas explosion-induced ALI, which can effectively improve the treatment rate of this condition. In this study, we analyzed the relationship between a novel form of cell death, ferroptosis, and gas explosion-induced ALI, and explored its specific mechanisms. METHODS We established ALI rat model by Shock tube biological injury system, and detected lung injury-related indexes as well as ferroptosis related indexes, such as glutathione peroxidase 4(GPX4), 4-hydroxynonenal(4HNE), Malondialdehyde(MDA), Fe2 + . We also investigated the therapeutic effects of the ferroptosis inhibitor ferrostatin-1 (Fer-1) in ALI induced by gas explosion, as well as its specific mechanisms of action. RESULTS A rat ALI model by gas explosion was successfully established. After the gas explosion treatment, we observed that the systemic inflammatory reaction of rats was increased, and lung function, liver function, kidney function and cardiac function were damaged to different degrees. The inflammatory infiltration in the lung tissue was more severe, and the degree of lung injury and pulmonary edema increased. The ferroptosis markers GPX4 was decreased, while the levels of 4HNE, MDA and Fe2 + were increased. Treatment with Fer-1 significantly ameliorated gas explosion ALI damage and down-regulated the expression level of ferroptosis. CONCLUSIONS Gas explosion-induced ALI in rats is characterized by enhanced inflammatory responses and reduced antioxidant capacity in lung tissues. The specific mechanism of injury involves ferroptosis. Fer-1 has been shown to mitigate the severity of ALI caused by gas explosion by suppressing ferroptosis expression levels in lung tissues via the Nrf2/GPX4 axis.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; Department of Orthopedics of the General Hospital of Western Theater Command, Chengdu 610083, China
| | - Linqiang Tian
- Institutes of Health Central Plain, Xinxiang Medical University, Jinhui Road #601, Xinxiang, Henan 453003, China; Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China; Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, China
| | - Peng Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Long Li
- Institutes of Health Central Plain, Xinxiang Medical University, Jinhui Road #601, Xinxiang, Henan 453003, China; Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China; Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, China
| | - Kunxi Wang
- Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Yanyan Li
- Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Yue Zhang
- Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Lili Feng
- Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Sanqiao Yao
- Institutes of Health Central Plain, Xinxiang Medical University, Jinhui Road #601, Xinxiang, Henan 453003, China; School of Public Health, Xinxiang Medical University, Xinxiang 453003, China.
| | - Hao Guan
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Wenjie Ren
- Institutes of Health Central Plain, Xinxiang Medical University, Jinhui Road #601, Xinxiang, Henan 453003, China; Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China; Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, China.
| |
Collapse
|
5
|
de Morais Gomes V, Santos DM, Macedo-da-Silva J, Lazari LC, Machado RRG, Dos Santos AF, Araujo DB, Coutinho JVP, Arini GS, Angeli CB, de Souza EE, Marques RF, Boscardin SB, Wrenger C, Marinho CRF, Oliveira DBL, Durigon EL, Labriola L, Rosa-Fernandes L, Palmisano G. P.1 and P.2 SARS-CoV-2 Brazilian variants activate the unfolded protein response with a time and pathway specificity. J Proteomics 2025; 315:105397. [PMID: 39909104 DOI: 10.1016/j.jprot.2025.105397] [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: 01/07/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
COVID-19 is a human respiratory syndrome caused by the infection of the SARS-CoV-2 virus that has a high rate of infection and mortality. Viruses modulate the host machinery by altering cellular mechanisms that favor their replication. One of the mechanisms that viruses exploit is the protein folding and processing of post-translational modifications that occur in the endoplasmic reticulum (ER). When ER function is impaired, there is an accumulation of misfolded proteins leading to endoplasmic reticulum stress (ER stress). To maintain homeostasis, cells trigger an adaptive signaling mechanism called the Unfolded Protein Response (UPR) which helps cells deal with stress, but under severe conditions, can activate the apoptotic cell death mechanism. This study elucidated an activation of a diversity of molecular mechanisms by Brazilian variants of SARS-CoV-2 by a time-resolved and large-scale characterization of SARS-CoV-2-infected cells proteomics and immunoblotting. Furthermore, it was shown that pharmacological UPR modulation could reduce viral release by counteracting the different viral activations of its cellular response. Analysis of human clinical specimens and disease outcomes focusing on ER stress reinforces the importance of UPR modulation as a host regulatory mechanism during viral infection and could point to novel therapeutic targets. SIGNIFICANCE: Since the emergence of SARS-CoV-2 and the consequent COVID-19 pandemic, the rapid emergence of variants of this new coronavirus has been a cause for concern since many of them have significantly higher rates of transmissibility and virulence, being called Variants of Concern (VOC). In this work, we studied the VOCs Gamma (P.1) and Zeta (P.2), also known as Brazilian variants. Constant evidence has reported that there are particularities related to each variant of SARS-CoV-2, with different rates of transmissibility, replication and modulation of host biological processes being observed, in addition to the mutations present in the variants. For this reason, this work focused on infections caused by the Brazilian variants of SARS-CoV-2 in different cell lines, in which we were able to observe that the infections caused by the variants induced endoplasmic reticulum stress in the infected cells and activated the UPR pathways, presenting specific modulations of each variant in this pathway. Furthermore, transcriptome analysis of patients revealed a correlation between ER-related genes and COVID-19 progression. Finally, we observed that the use of UPR modulators in host cells decreased viral release of all variants without affecting cell viability. The data presented in this work complement the observations of other studies that aim to understand the pathogenicity of SARS-CoV-2 VOCs and possible new therapeutic strategies, mainly targeting biological processes related to the endoplasmic reticulum.
Collapse
Affiliation(s)
| | - Deivid Martins Santos
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Janaina Macedo-da-Silva
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Lucas C Lazari
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | | | | | - Danielle Bastos Araujo
- Laboratory of Clinical and Molecular Virology, Department of Microbiology, ICB, University of São Paulo, Brazil
| | | | - Gabriel Santos Arini
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Brazil
| | - Claudia B Angeli
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Edmarcia E de Souza
- Unit for Drug Discovery, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Rodolfo F Marques
- Laboratory of Antigen Targeting for Dendritic Cells, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Silvia Beatriz Boscardin
- Laboratory of Antigen Targeting for Dendritic Cells, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Carsten Wrenger
- Unit for Drug Discovery, Department of Parasitology, ICB, University of São Paulo, Brazil
| | | | - Danielle B L Oliveira
- Laboratory of Clinical and Molecular Virology, Department of Microbiology, ICB, University of São Paulo, Brazil
| | - Edison L Durigon
- Laboratory of Clinical and Molecular Virology, Department of Microbiology, ICB, University of São Paulo, Brazil; Scientific Platform Pasteur USP, Sao Paulo, Brazil
| | - Leticia Labriola
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Brazil
| | - Livia Rosa-Fernandes
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil; Laboratory of Experimental Immunoparasitology, Department of Parasitology, ICB, University of São Paulo, Brazil; Centre for Motor Neuron Disease Research, Faculty of Medicine, Health & Human Sciences, Macquarie Medical School, Sydney, Australia
| | - Giuseppe Palmisano
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil; School of Natural Sciences, Macquarie University, Sydney, Australia.
| |
Collapse
|
6
|
Wang D, Qu X, Zhang Z, Zhou G. New developments in the role of ferroptosis in sepsis‑induced cardiomyopathy (Review). Mol Med Rep 2025; 31:118. [PMID: 40052561 PMCID: PMC11904766 DOI: 10.3892/mmr.2025.13483] [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: 09/25/2024] [Accepted: 01/15/2025] [Indexed: 03/15/2025] Open
Abstract
Sepsis is a life‑threatening organ dysfunction disorder caused by dysfunctional host response to infection. Sepsis‑induced cardiomyopathy (SIC) is a common and serious complication of sepsis, and it is associated with increased mortality rates; however, its specific pathogenesis is still unclear. Ferroptosis, which is an iron‑dependent form of programmed cell death, is involved in the pathophysiology of SIC. Further study on the mechanism and therapeutic targets of ferroptosis in SIC may provide new strategies for clinical diagnosis and treatment of this condition. The present article reviews the mechanisms between SIC and ferroptosis, summarizes the progress in research of the involvement of ferroptosis in SIC and provides new potential strategies for further research and treatment in the future.
Collapse
Affiliation(s)
- Dingdeng Wang
- Department of Critical Care Medicine, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei 443003, P.R. China
| | - Xinguang Qu
- Department of Critical Care Medicine, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei 443003, P.R. China
| | - Zhaohui Zhang
- Department of Critical Care Medicine, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei 443003, P.R. China
| | - Gaosheng Zhou
- Department of Critical Care Medicine, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei 443003, P.R. China
| |
Collapse
|
7
|
Lu Q, Shao N, Fang Z, Ouyang Z, Shen Y, Yang R, Liu H, Cai B, Wei T. The anti-Alzheimer's disease effects of ganoderic acid A by inhibiting ferroptosis-lipid peroxidation via activation of the NRF2/SLC7A11/GPX4 signaling pathway. Chem Biol Interact 2025; 412:111459. [PMID: 40054827 DOI: 10.1016/j.cbi.2025.111459] [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: 01/03/2025] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025]
Abstract
Alzheimer's disease (AD) is a degenerative disease of the central nervous system, characterized by a gradual decline in cognitive and memory abilities, social disorders, and behavioral abnormalities. Ferroptosis, an iron-dependent type of programmed cell death, is closely associated with the pathogenesis of AD. Ferroptosis is characterized by the accumulation of iron within cells, leading to increased oxidative stress, and ultimately lipid peroxidation and cell death. Ganoderic acid A (GAA), one of the major pharmacologically active components in Ganoderma lucidum, exhibits an excellent neuroprotective effect against AD. However, it is unclear whether GAA improves the symptoms of AD by inhibiting ferroptosis. This study investigated the anti-AD effects of GAA through both in vivo and in vitro experiments, and determined its molecular mechanism from the perspective of ferroptosis modulation. The results showed that GAA administration attenuated hippocampal neuronal loss, improved mitochondrial ultrastructure, and enhanced the memory and learning ability of the AD mice. In vitro assays suggested that GAA effectively protected HT22 AD cells against ferroptosis-related morphological damage, enhanced their antioxidant capacity, maintained their iron metabolism, and reduced mitochondrial dysfunction. Moreover, the immunofluorescence and western blotting results showed that the levels of NFE2 like bZIP transcription factor 2 (NRF2), glutathione peroxidase 4 (GPX4), and solute carrier family 7 member 11 (SLC7A11) both in the hippocampus of APP/PS1 mice and amyloid beta (Aβ)25-35-induced HT22 AD cells were markedly enhanced after GAA administration. In summary, these results revealed that GAA improves AD by activating on the NRF2/SLC7A11/GPX4 axis to inhibit ferroptosis-lipid peroxidation.
Collapse
Affiliation(s)
- Qingyang Lu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Nan Shao
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Ziyi Fang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Zhaorong Ouyang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Yiran Shen
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Ruiling Yang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China; Medical Basic Research Innovation Center for Integrated Chinese and Western Medicine in the Prevention and Treatment of Neurodegenerative Diseases, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Houli Liu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China; Medical Basic Research Innovation Center for Integrated Chinese and Western Medicine in the Prevention and Treatment of Neurodegenerative Diseases, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Biao Cai
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China; Medical Basic Research Innovation Center for Integrated Chinese and Western Medicine in the Prevention and Treatment of Neurodegenerative Diseases, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Tao Wei
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China; Medical Basic Research Innovation Center for Integrated Chinese and Western Medicine in the Prevention and Treatment of Neurodegenerative Diseases, Anhui University of Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
8
|
Liu C, Liu X, Wang Y, Yu H, Li Q, Zheng Y, Fu Y, Yao G, Sun L. Mesenchymal stromal cells reduce ferroptosis of podocytes by activating the Nrf2/HO-1/GPX4 pathway in lupus nephritis. Int Immunopharmacol 2025; 153:114537. [PMID: 40147265 DOI: 10.1016/j.intimp.2025.114537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Ferroptosis has been reported to be involved in the occurrence and development of various kidney diseases. Emerging evidence suggests that ferroptosis also plays a critical role in systemic lupus erythematosus (SLE) and lupus nephritis (LN), contributing to podocyte injury and renal dysfunction. Mesenchymal stromal cells (MSCs) have become an attractive option for podocyte injury repairing in LN. The aim of this research was to determine whether MSCs regulate ferroptosis of podocytes in LN. METHODS MSCs were injected into female MRL/lpr mice via tail vein. The symptoms of LN and the detection of ferroptosis-related biomarkers in podocytes were detected. In vitro validation was conducted by mouse podocyte cell line MPC-5. RESULTS The occurrence of ferroptosis and involvement of Nrf2/heme oxygenase-1 (HO-1) signaling pathway in podocytes were observed. We found increased expression of the podocyte marker, Wilm's tumor 1 (WT-1) and synaptopodin, following the improvement of lupus-like symptoms after MSC transplantation in MRL/lpr mice. The expression of ferroptosis-related protein glutathione peroxidase 4 (GPX4) and long chain acyl-CoA synthetase 4 (ACSL4) were elevated in renal, along with the Nrf2 and HO-1 activity enhancement. In vitro, MSC treatment maintain a stabilization of podocyte actin stress fibers, leading to an improvement of cell viability. Furthermore, our results showed that puromycin aminonucleoside (PAN) induce accumulation of cellular lipid reactive oxygen species (ROS) and glutathione depletion, and the expression of Nrf2, HO-1 and GPX4 were all downregulated whereas the expression of ACSL4 was upregulated. However, these effects were reversed by MSCs and ferroptosis inhibitor ferrastatin-1 (Fer-1). The promotion of Nrf2 nuclear translocation was observed after the treatment with MSCs. CONCLUSION Ferroptosis activation is involved in the development of LN. MSCs could ameliorate podocyte injury in LN by inhibiting ferroptosis through the Nrf2/HO-1/GPX4 pathway, which will provide novel potential therapeutic targets for LN.
Collapse
Affiliation(s)
- Chang Liu
- Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China
| | - Xuanqi Liu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yujiao Wang
- Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China
| | - Honghong Yu
- Department of Rheumatology and Immunology, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Qi Li
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuanyuan Zheng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yao Fu
- Department of Pathology, Affiliated Drum Tower Hospital, Medical School of Nanjing University.
| | - Genhong Yao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Department of Rheumatology and Immunology, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China.
| | - Lingyun Sun
- Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China; Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Department of Rheumatology and Immunology, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
9
|
Yang Y, Chen Z, Song D, Wu J, Wang J, YouyouYan. Inhibition of ferroptosis alleviates atherosclerosis and foam cell formation by regulating lipid metabolism via AMPK activation. Int Immunopharmacol 2025; 153:114553. [PMID: 40147262 DOI: 10.1016/j.intimp.2025.114553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/14/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Atherosclerosis (AS) is a lipid disorder characterised by lipid accumulation in the aortic wall and foam cell formation. Recent studies have shown that excess iron accelerates AS progression and foam cell formation by inducing ferroptosis. GPx4, an anti-erroptotic protein, promotes SCARB1 expression, which inhibits macrophage foam cell formation by interacting with HDL. Thus, a complex association exists between ferroptosis and lipid metabolism. However, the underlying mechanisms remain unclear. AMPK signalling is a key regulator of metabolism and is involved in the regulation of ferroptosis. In this study, we used the ferroptosis inhibitor ferrostatin-1 (Fer-1) to assay the effect of ferroptosis inhibition on AS and foam cell formation and to investigate the underlying mechanism. Our results showed that Fer-1 alleviated AS lesions and foam cell formation both in vivo and in vitro. Additionally, Fer-1 reduced iron content and lipid accumulation in oxidized low-density lipoprotein (ox-LDL)-treated macrophages by upregulating the levels of FTH, GPx4, and SCARB1 via AMPK activation. The inhibition of AMPK reduces the effect of Fer-1 on iron and lipid accumulation in macrophages, which may contribute to a deeper understanding of the pathological process of AS and provide a therapeutic target for AS.
Collapse
Affiliation(s)
- Yunfan Yang
- Department of Cardiology, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Zhenzhen Chen
- Department of Cardiology, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Dandan Song
- Department of Clinical Laboratory, Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Junduo Wu
- Department of Cardiology, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Junnan Wang
- Department of Cardiology, Second Hospital of Jilin University, Changchun, Jilin 130041, China.
| | - YouyouYan
- Department of Cardiology, Second Hospital of Jilin University, Changchun, Jilin 130041, China.
| |
Collapse
|
10
|
Shu H, Liao Q, Chen Z, Liang M, Zhang S, Liu J, Wu Y, Hu P, Luo M, Zhu W, Zhu X, Yang L, Yan T. Flavonoids serve as a promising therapeutic agent for ischemic stroke. Brain Res 2025; 1853:149528. [PMID: 39999903 DOI: 10.1016/j.brainres.2025.149528] [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: 01/08/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 02/27/2025]
Abstract
Ischemic stroke (IS) continues to be a major public health concern and is characterized by significantly high mortality and disabling rates. Inhibiting nerve cells death and enhancing the repair of ischemic tissue are important treatment concepts for IS. Currently, the mainstream treatment strategies mainly focus on short-term care, which underscores the urgent need for novel therapeutic strategies for long-term care. Emerging data reveal that flavonoids have surfaced as promising candidates for IS patients' long-term care. Flavonoids can alleviate neuroinflammation and anti-apoptosis due to their characteristic pharmacological mechanisms. Clinical evidence suggests that long-term flavonoids intake improves IS patients' long-term outcomes. Though the effect of flavonoids in IS treatment has been explored for decades, the neuroprotective pharmacodynamics have not been well established. Thereby, the aim of current review is to summarize the pathways involved in neuroprotective effect of flavonoids. This review will also advance the potential of flavonoids as a viable clinical candidate for the treatment of IS.
Collapse
Affiliation(s)
- Hongxin Shu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qiuye Liao
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Zhihao Chen
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Mingyu Liang
- School of life sciences, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Si Zhang
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Junzhe Liu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Yanze Wu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ping Hu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ming Luo
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Wenping Zhu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xingen Zhu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Li Yang
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Tengfeng Yan
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
11
|
Wang X, Xue Y, Chang L, Zhu X, Liu W, Liang T. The Regulation of Trace Metal Elements in Cancer Ferroptosis. Adv Biol (Weinh) 2025:e2400821. [PMID: 40200790 DOI: 10.1002/adbi.202400821] [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: 12/19/2024] [Revised: 03/11/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis, as novel type of regulated cell death that has garnered widespread attention over the past decade, has witnessed the continuous discovery of an increasing number of regulatory mechanisms. Trace metal elements play a multifaceted and crucial role in oncology. Interestingly, it has been increasingly evident that these elements, such as copper, are involved in the regulation of iron accumulation, lipid peroxidation and antiferroptotic systems, suggesting the existence of "nonferrous" mechanisms in ferroptosis. In this review, a comprehensive overview of the composition and mechanism of ferroptosis is provided. The interaction between copper metabolism (including cuproptosis) and ferroptosis in cancer, as well as the roles of other trace metal elements (such as zinc, manganese, cobalt, and molybdenum) in ferroptosis are specifically focused. Furthermore, the applications of nanomaterials based on these metals in cancer therapy are also reviewed and potential strategies for co-targeting ferroptosis and cuproptosis are explored. Nevertheless, in light of the intricate and ambiguous nature of these interactions, ongoing research is essential to further elucidate the "nonferrous" mechanisms of ferroptosis, thereby facilitating the development of novel therapeutic targets and approaches for cancer treatment.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuanyuan Xue
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Lei Chang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xuena Zhu
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Wenjun Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
- Zhejiang University Cancer Center, Hangzhou, 310003, China
| |
Collapse
|
12
|
Liu S, Liao S, He J, Zhou Y, He Q. IGF2BP2: an m 6A reader that affects cellular function and disease progression. Cell Mol Biol Lett 2025; 30:43. [PMID: 40205577 PMCID: PMC11983839 DOI: 10.1186/s11658-025-00723-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 03/27/2025] [Indexed: 04/11/2025] Open
Abstract
Insulin-like growth factor 2 messenger RNA (mRNA)-binding protein 2 (IGF2BP2) is a widely studied N6-methyladenosine (m6A) modification reader, primarily functioning to recognize and bind to m6A modification sites on the mRNA of downstream target genes, thereby enhancing their stability. Previous studies have suggested that the IGF2BP2-m6A modification plays an essential role in cellular functions and the progression of various diseases. In this review, we focus on summarizing the molecular mechanisms by which IGF2BP2 enhances the mRNA stability of downstream target genes through m6A modification, thereby regulating cell ferroptosis, epithelial-mesenchymal transition (EMT), stemness, angiogenesis, inflammatory responses, and lipid metabolism, ultimately affecting disease progression. Additionally, we update the related research progress on IGF2BP2. This article aims to elucidate the effects of IGF2BP2 on cell ferroptosis, EMT, stemness, angiogenesis, inflammatory responses, and lipid metabolism, providing a new perspective for a comprehensive understanding of the relationship between IGF2BP2 and cell functions such as ferroptosis and EMT, as well as the potential for targeted IGF2BP2 therapy for tumors and other diseases.
Collapse
Affiliation(s)
- Siyi Liu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University/Hunan Cancer Hospital, Changsha, 410013, Hunan, China
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410011, Hunan, China
| | - Shan Liao
- Department of Pathology, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Junyu He
- Department of Clinical Laboratory, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, 410007, Hunan, People's Republic of China
| | - Yanhong Zhou
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410011, Hunan, China.
| | - Qian He
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University/Hunan Cancer Hospital, Changsha, 410013, Hunan, China.
| |
Collapse
|
13
|
Gao S, Wang X, Xu Q, Li R, Yao L, Zhang A, Zhou Q, Xiao Z, Li S, Meng X, Wu J, Qin L. Total Sanghuangporus vaninii extract inhibits hepatocyte ferroptosis and intestinal microbiota disturbance to attenuate liver fibrosis in mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119571. [PMID: 40023344 DOI: 10.1016/j.jep.2025.119571] [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: 11/17/2024] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sanghuangporus, the dried fruiting body of Sanghuangporus vaninii (Ljub) L.W.Zhou et Y.C.Dai. As the main species of Sanghuang, it has been well-known and used commonly as a traditional medicinal and edible macrofungi for thousands of years in many countries, including China, Korea and Japan. Although it has good hepatoprotective activity, its potential efficacy and mechanism on liver fibrosis remain elusive. AIM OF THE STUDY Total Sanghuangporus vaninii extract (TSH) was prepared by ethanol extraction to investigate its chemical components and to conduct an initial assessment of its efficacy and underlying mechanism in a murine model of liver fibrosis. MATERIALS AND METHODS The chemical components of TSH were initially analyzed by UHPLC-Q-Orbitrap HRMS. To elucidate the effects of TSH, an in vivo model of fibrosis was established in mice using carbon tetrachloride (CCl4), followed by assessments of serum liver function and histopathological analysis. Besides, indicators related to liver fibrosis, hepatic stellate cells (HSCs) activation, inflammation response and ferroptosis related indicators were detected by western blotting, immunohistochemistry and real-time quantitative PCR (RT-qPCR) analysis. Additionally, the 16S rDNA sequencing and untargeted metabolomics analysis of intestinal microbiota were employed to investigating the role of TSH in gut microbiome. In vitro, the human hepatocyte line L02 was stimulated with erastin and treated with or without TSH to elucidate its underlying mechanism. RESULTS The administration of TSH significantly improved serum indicators of liver injury in CCl4-induced fibrosis mice, reduced HSCs activation and collagen deposition, while inhibiting the expressions of transforming growth factor-β1(TGF-β1)/Smad signaling pathway. Notably, TSH treatment attenuated hepatocyte ferroptosis and lipid peroxidation both in vivo and in vitro, as evidenced by a marked decrease in liver iron and malondialdehyde (MDA) contents. In particular, TSH was demonstrated to activate the nuclear factor erythroid 2-related factor 2 (Nrf2)-glutathione peroxidase 4 (GPX4) signaling pathway, thereby protecting hepatocytes from ferroptosis with a particular enhancement of Nrf2 nuclear transcription. Furthermore, TSH influenced gut microbiota composition and ameliorated intestinal metabolic disorders. The increased abundance of Parasutterella and Olsenellas due to TSH treatment was significantly positively correlated with elevated phosphatidylcholines involved in linoleic acid metabolism, and negatively correlated with the reduction of fatty acyls. And the enrichment of intestinal linoleic acid metabolism presented a negative correlation in liver fibrosis biomarkers. CONCLUSIONS Our findings indicate that the TSH treatment exerts a significantly protective effect on CCl4-induced mice by ameliorating hepatic injury and ferroptosis damage, inhibiting HSCs activation and collagen deposition, and remodeling gut microbiota homeostasis and metabolic imbalance. Notably, TSH attenuated hepatocyte ferroptosis in liver fibrosis and exhibited upregulation of the Nrf2-GPX4 signaling pathway. Furthermore, TSH could enrich the abundance of Parasutterella and Olsenellas, which may contribute to intestinal linoleic acid metabolism, thereby contributing to the reduction of liver fibrosis damage. Our study provides more effective and unreported evidence of TSH in anti-fibrosis activity.
Collapse
Affiliation(s)
- Siqi Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Xingxing Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Qiuying Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Rongsheng Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Lumeng Yao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Anna Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Qun Zhou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Zhun Xiao
- Department of Digestive Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Shengsheng Li
- Department of Digestive Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiongyu Meng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Jianjun Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Luping Qin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| |
Collapse
|
14
|
Yang Q, Shen X, Luo Y, Li R, Meng X, Xu P, Liu X, Bian D, Wang J, Shi J, Chen J. ELANE enhances KEAP1 protein stability and reduces NRF2-mediated ferroptosis inhibition in metabolic dysfunction-associated fatty liver disease. Cell Death Dis 2025; 16:266. [PMID: 40204709 PMCID: PMC11982220 DOI: 10.1038/s41419-025-07603-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/11/2025]
Abstract
Neutrophil elastase (Elane) is upregulated in metabolic-associated fatty liver disease (MAFLD) and has the capacity to promote disease progression. However, the mechanism by which Elane promotes MAFLD development remains unclear. Ferroptosis, which is an iron-dependent nonapoptotic form of cell death characterized by the iron-induced accumulation of lipid reactive oxygen species (ROS), has been recently considered as an important mechanism for the development of MAFLD. In this study, we used mice of Elane-knockout (Elane-KO) and wild-type (WT), and their primary mouse hepatocytes to establish MAFLD models in vivo and vitro for elucidating the role of Elane in ferroptosis of hepatocytes and MAFLD development. Elane-KO in vivo reduced high-fat diet (HFD) induced hepatic lipid peroxidation levels and the proportion of hepatocyte death, upregulated the expression of Nrf2 and Gpx4, and downregulated Keap1 expression. Treatment with recombinant Elane increased the lipid peroxidation level of hepatocytes, increased the ferroptosis rate of hepatocytes, upregulated the expression of Keap1, enhanced the ubiquitination of Nrf2, and downregulated the expression of Nrf2 and Gpx4 in an FFA-induced MAFLD in vitro model. However, primary hepatocytes from Elane-KO mice presented opposite changes. Furthermore, an in vitro experiment revealed that Elane enhanced the protein stability of Keap1 and thus increased Keap1 expression in hepatocytes by inhibiting the lysosomal degradation of the Keap1 protein. Finally, in vitro Co-IP experiments revealed that Elane increased the protein stability of Keap1 by weakening the binding between P62 and Keap1 and ultimately promoted hepatocyte Nrf2 ubiquitination and ferroptosis in MAFLD. In conclusion, our results suggested that Elane promoted hepatocyte ferroptosis in MAFLD through the P62-Keap1-Nrf2-Gpx4 axis. Elane promotes ferroptosis in hepatocytes from fatty livers. Elane reduces the binding of P62 to Keap1, thereby increasing Keap1 protein stability and subsequently inhibiting the Nrf2/Gpx4 pathway, ultimately leading to ferroptosis in hepatocytes.
Collapse
Affiliation(s)
- Qingqing Yang
- Department of Gastroenterology, The First People's Hospital of Yancheng, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, China
| | - Xuan Shen
- College of Basic Medicine, Jiangsu Medical college, Yancheng, Jiangsu, China
| | - Yan Luo
- Department of Liver Diseases, Hangzhou Normal University Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Rongqing Li
- College of Clinical Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiangrui Meng
- Department of Nuclear Medicine, Xinxiang Central Hospital, The Fourth Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Ping Xu
- Department of Gastroenterology, The First People's Hospital of Yancheng, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, China
| | - Xuan Liu
- Department of Gastroenterology, The First People's Hospital of Yancheng, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, China
| | - Dongxue Bian
- Department of Gastroenterology, Yancheng TCM Hospital Affiliated with Nanjing University of Chinese Medicine, Yancheng, Jiangsu, China
| | - Jianhua Wang
- Department of Gastroenterology, The First People's Hospital of Yancheng, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, China.
| | - Junping Shi
- Department of Liver Diseases, Hangzhou Normal University Affiliated Hospital, Hangzhou, Zhejiang, China.
| | - Jin Chen
- Department of Gastroenterology, The First People's Hospital of Yancheng, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, China.
| |
Collapse
|
15
|
Hou Q, Cheng S, Li Z, Lei C, Chen Y, Ma M, Liu J, Chen X, Wang L, Xue Q, Qi X. Peste des petits ruminants virus (PPRV) induces ferroptosis via LONP1-mediated mitochondrial GPX4 degradation in cell culture. J Virol 2025:e0231024. [PMID: 40197059 DOI: 10.1128/jvi.02310-24] [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: 01/07/2025] [Accepted: 03/12/2025] [Indexed: 04/09/2025] Open
Abstract
Peste des petits ruminants virus (PPRV) is an important pathogen that seriously affects the productivity of small ruminants worldwide. Ferroptosis is a programmed cell death characterized by iron-dependent lipid peroxidation and the accumulation of reactive oxygen species (ROS). Emerging evidence has demonstrated that mitochondria play diverse roles in the process of ferroptosis, but the interaction between mitochondria and ferroptosis during virus infection remains largely unknown. Here, we demonstrate that PPRV induces ferroptosis, including Fe2+ overload, accumulation of lipid peroxidation, and shrinkage of mitochondria. Importantly, mitochondria play a crucial role in the process of PPRV-induced ferroptosis characterized by decreased mitochondrial GPX4 and lipid peroxidation in mitochondria. Mechanistically, PPRV infection downregulates mitochondrial Lon protease-1 (LONP1) expression, an important multifaceted enzyme that is essential for maintaining mitochondrial homeostasis and function, which leads to mitochondrial GPX4 degradation through the Nrf2/Keap pathway and accumulation of ROS in mitochondria. More importantly, PPRV-induced ferroptosis is tightly associated with inflammatory responses and enhanced virus replication. Overall, this study is the first to show that LONP1-mediated ferroptosis is involved in the inflammatory responses during PPRV infection. IMPORTANCE Peste des petits ruminants virus (PPRV) infection induces a transient but severe immunosuppression in the host, which threatens both small livestock and endangered susceptible wildlife populations in many countries. Despite extensive research, it is unknown whether PPRV causes ferroptosis and what the mechanism of regulation is. Our data provide the first direct evidence that the relationship between Lon protease-1 (LONP1)-mediated dysfunctional mitochondria and the consequent induction of ferroptosis is involved in PPRV-induced pathogenesis. Importantly, we demonstrate that PPRV infection induces ferroptosis via the LONP1-mediated GPX4 degradation and ROS accumulation in mitochondria, and PPRV-induced ferroptosis is tightly associated with inflammatory responses and enhanced virus replication levels. Taken together, our research has provided new insight into understanding the effect of ferroptosis on PPRV replication and pathogenesis and revealed a potential therapeutic target for antiviral intervention.
Collapse
Affiliation(s)
- Qiaodi Hou
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs of the People's Republic of China, Yangling, Shaanxi, China
| | - Shuijin Cheng
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs of the People's Republic of China, Yangling, Shaanxi, China
| | - Zhijun Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs of the People's Republic of China, Yangling, Shaanxi, China
| | - Congshang Lei
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs of the People's Republic of China, Yangling, Shaanxi, China
| | - Yan Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs of the People's Republic of China, Yangling, Shaanxi, China
| | - Mingzhuo Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs of the People's Republic of China, Yangling, Shaanxi, China
| | - Jinming Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs of the People's Republic of China, Yangling, Shaanxi, China
| | - Xiwen Chen
- Animal Disease Prevention and Control, Mianyang Normal University, Mianyang, Sichuan, China
- Healthy Breeding Engineering Technology Research Center, Mianyang Normal University, Mianyang, Sichuan, China
| | - Lizhen Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs of the People's Republic of China, Yangling, Shaanxi, China
| | - Qinghong Xue
- China Institute of Veterinary Drug Control, Beijing, China
| | - Xuefeng Qi
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs of the People's Republic of China, Yangling, Shaanxi, China
| |
Collapse
|
16
|
Zhou J, Lu P, He H, Zhang R, Yang D, Liu Q, Liu Q, Liu M, Zhang G. The metabolites of gut microbiota: their role in ferroptosis in inflammatory bowel disease. Eur J Med Res 2025; 30:248. [PMID: 40189555 PMCID: PMC11974165 DOI: 10.1186/s40001-025-02524-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/27/2025] [Indexed: 04/09/2025] Open
Abstract
Inflammatory bowel disease (IBD) includes chronic inflammatory conditions, such as Crohn's disease and ulcerative colitis, characterized by impaired function of the intestinal mucosal epithelial barrier. In recent years, ferroptosis, a novel form of cell death, has been confirmed to be involved in the pathological process of IBD and is related to various pathological changes, such as oxidative stress and inflammation. Recent studies have further revealed the complex interactions between the microbiome and ferroptosis, indicating that ferroptosis is an important target for the regulation of IBD by the gut microbiota and its metabolites. This article reviews the significant roles of gut microbial metabolites, such as short-chain fatty acids, tryptophan, and bile acids, in ferroptosis in IBD. These metabolites participate in the regulation of ferroptosis by influencing the intestinal microenvironment, modulating immune responses, and altering oxidative stress levels, thereby exerting an impact on the pathological development of IBD. Treatments based on the gut microbiota for IBD are gradually becoming a research hotspot. Finally, we discuss the potential of current therapeutic approaches, including antibiotics, probiotics, prebiotics, and fecal microbiota transplantation, in modulating the gut microbiota, affecting ferroptosis, and improving IBD symptoms. With a deeper understanding of the interaction mechanisms between the gut microbiota and ferroptosis, it is expected that more precise and effective treatment strategies for IBD will be developed in the future.
Collapse
Affiliation(s)
- Jingying Zhou
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Penghui Lu
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Haolong He
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ruhan Zhang
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Dican Yang
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Qiong Liu
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Qianyan Liu
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Mi Liu
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Guoshan Zhang
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
17
|
Soni P, Sharma SM, Pieper AA, Paul BD, Thomas B. Nrf2/Bach1 signaling axis: A promising multifaceted therapeutic strategy for Alzheimer's disease. Neurotherapeutics 2025:e00586. [PMID: 40199685 DOI: 10.1016/j.neurot.2025.e00586] [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: 10/31/2024] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia, which continues to elude effective treatment despite decades of research and numerous clinical trials. While existing therapeutic strategies have primarily targeted neuropathological hallmarks such as amyloid plaques and tau tangles, they have failed to halt disease progression, leaving patients with limited options. This persistent failure reveals a critical gap in our understanding of AD and calls for a fresh perspective - one that goes beyond the traditional targets and dives deeper into the fundamental cellular processes that drive neurodegeneration. Recent advances in molecular biology underscore the significance of nuclear factor E2-related factor 2 (Nrf2), often termed the "guardian of redox homeostasis," in the pathophysiology of AD. Nrf2 orchestrates cellular responses to oxidative stress and neuroinflammation - two interlinked pathological features of AD. In the brains of AD patients, Nrf2 activity is diminished, weakening the brain's ability to counteract oxidative damage. Additionally, the BTB and CNC homology 1 (Bach1) protein, a transcriptional repressor of Nrf2, has emerged as a potential therapeutic target. Here, we review the current landscape of clinical trials in AD and identify the limitations of the conventional approaches. We then explore the prospects of a novel approach that combines Nrf2 activation with Bach1 inhibition to achieve a multipronged defense against oxidative stress, neuroinflammation, and other molecular culprits driving AD. This innovative strategy holds promise for synergistically modulating multiple neuroprotective pathways to advance AD treatment.
Collapse
Affiliation(s)
- Priyanka Soni
- Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC, USA; Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Sudarshana M Sharma
- Department of Biochemistry and Molecular Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Andrew A Pieper
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Bindu D Paul
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Bobby Thomas
- Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC, USA; Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA; Department of Drug Discovery, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
18
|
Yang Y, Deng X, Li W, Leng Y, Xiong Y, Wang B, Gong S, Wang Y, Yang B, Li W. Targeting the epigenetic regulation of ferroptosis: a potential therapeutic approach for sepsis-associated acute kidney injury. Clin Epigenetics 2025; 17:57. [PMID: 40189571 PMCID: PMC11974148 DOI: 10.1186/s13148-025-01861-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/13/2025] [Indexed: 04/09/2025] Open
Abstract
Sepsis is a syndrome of organ dysfunction caused by the invasion of pathogenic microorganisms. In clinical practice, patients with sepsis are prone to concurrent acute kidney injury, which has high morbidity and mortality rates. Thus, understanding the pathogenesis of sepsis-associated acute kidney injury is of significant clinical importance. Ferroptosis is an iron-dependent programmed cell death pathway, which is proved to play a critical role in the process of sepsis-associated acute kidney injury through various mechanisms. Epigenetic regulation modulates the content and function of nucleic acids and proteins within cells through various modifications. Its impact on ferroptosis has garnered increasing attention; however, the role of epigenetic regulation targeting ferroptosis in sepsis-associated acute kidney injury has not been fully elucidated. Growing evidence suggests that epigenetic regulation can modulate ferroptosis through complex pathway networks, thereby affecting the development and prognosis of sepsis-associated acute kidney injury. This paper summarizes the impact of ferroptosis on sepsis-associated acute kidney injury and the regulatory mechanisms of epigenetic regulation on ferroptosis, providing new insights for the targeted therapy of sepsis-associated acute kidney injury.
Collapse
Affiliation(s)
- Yuhang Yang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Xinqi Deng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wenyuan Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yan Leng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yonghong Xiong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Bihan Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Siyuan Gong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yunhao Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Baichuan Yang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wei Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
19
|
Li M, Wang Y, Li X, Xu J, Yan L, Tang S, Liu C, Shi M, Liu R, Zhao Y, Zhang Y, Yang L, Zhang Y, Wang G, Li Z, Guo Y, Feng Y, Liu P. Pharmacological targeting of the mitochondrial phosphatase PTPMT1 sensitizes hepatocellular carcinoma to ferroptosis. Cell Death Dis 2025; 16:257. [PMID: 40189563 PMCID: PMC11973169 DOI: 10.1038/s41419-025-07581-5] [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: 07/31/2024] [Revised: 03/07/2025] [Accepted: 03/21/2025] [Indexed: 04/09/2025]
Abstract
Protein tyrosine phosphatase mitochondrial 1 (PTPMT1), is a member of the protein tyrosine phosphatase superfamily localized on the mitochondrial inner membrane, and regulates the biosynthesis of cardiolipin. Given the important position of PTPMT1 in mitochondrial function and metabolism, pharmacological targeting of PTPMT1 is considered a promising manner in disease treatments. In this study, we mainly investigated the role of PTPMT1 in hepatocellular carcinoma (HCC) ferroptosis, a new type of cell death accompanied by significant iron accumulation and lipid peroxidation. Herein, the pharmacological inhibition of PTPMT1 was induced by alexidine dihydrochloride (AD, a dibiguanide compound). Human HCC cell lines with PTPMT1 knockout and PTPMT1 overexpression were established using CRISPR/Cas9 and lentiviral transduction methods, respectively. The position of PTPMT1 in regulating HCC ferroptosis was evaluated in vitro and in vivo. Our results indicated that pharmacological inhibition of PTPMT1, facilitated by AD treatment, heightens the susceptibility of HCC to cystine deprivation-ferroptosis, and AD treatment promoted the conversion from ferritin-bound Fe3+ to free Fe2+, which contributed to the labile iron pool in cytoplasm. Meanwhile, pharmacological inhibition of PTPMT1 also induced the formation of both swollen mitochondria and donut mitochondria, and enhanced the metabolism process form succinate to fumarate in mitochondrial tricarboxylic acid (TCA) cycle, which increased the sensitivity of HCC cells to cystine deprivation-induced ferroptosis. In total, our work reveals the close association of PTPMT1 with cysteine deprivation-induced ferroptosis, providing a novel insight into chemotherapy strategies against human HCC.
Collapse
Affiliation(s)
- Miaomiao Li
- Department of Critical Care Medicine, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Regenerative Medicine, School of Pharmaceutical Science, Jilin University, Changchun, China
| | - Yi Wang
- Department of Regenerative Medicine, School of Pharmaceutical Science, Jilin University, Changchun, China
| | - Xinyan Li
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiayi Xu
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liangwen Yan
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shenkang Tang
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Oncology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Chenyue Liu
- Department of Medical Image, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mengjiao Shi
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rongrong Liu
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yaping Zhao
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yi Zhang
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lan Yang
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yinggang Zhang
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Gang Wang
- Department of Critical Care Medicine, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Surgical Critical Care and Life Support, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, China
| | - Zongfang Li
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ying Guo
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Yetong Feng
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Core Research Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Pengfei Liu
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Key Laboratory of Environment and Genes Related To Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, China.
| |
Collapse
|
20
|
Huang M, Wu Y, Wei X, Cheng L, Fu L, Yan H, Wei W, Li B, Ru H, Mo X, Tang W, Su Z, Yan L. Trifluridine/tipiracil induces ferroptosis by targeting p53 via the p53-SLC7A11 axis in colorectal cancer 3D organoids. Cell Death Dis 2025; 16:255. [PMID: 40188162 PMCID: PMC11972347 DOI: 10.1038/s41419-025-07541-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: 09/19/2024] [Revised: 02/15/2025] [Accepted: 03/14/2025] [Indexed: 04/07/2025]
Abstract
Trifluridine/Tipiracil (FTD/TPI, TAS102) has been approved for the treatment of patients with colorectal cancer (CRC) for its promising anticancer activity enabled by its incorporation into double strands during DNA synthesis. However, the mechanisms underlying the anticancer targets of FTD/TPI remain not fully understood. Here we report our observation of the activation of ferroptosis in CRC by FTD/TPI. Mechanistically, FTD/TPI directly promotes the ubiquitination and degradation of MDM2, thereby stabilizing the p53. Nuclear accumulation of p53 subsequently downregulates SLC7A11 expression, leading to ferroptosis. Furthermore, we observed that FTD/TPI combined with sulfasalazine (SAS), a system Xc- inhibitor, works in a synergistic manner to induce ferroptosis and further inhibit the proliferation of CRC cells. Finally, we confirmed the synergistic effect of SAS and FTD/TPI on patient-derived organoids in vitro and patient-derived xenograft mouse models in vivo. Our findings are the first to reveal that FTD/TPI induces ferroptosis via the p53-SLC7A11 axis and that SAS enhances the sensitivity and therapeutic effect of FTD/TPI. These findings suggest that the synergistic effect of FTD/TPI and SAS may represent a new therapeutic strategy for patients with CRC.
Collapse
Affiliation(s)
- Maosen Huang
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Yancen Wu
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Xiaoxia Wei
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Linyao Cheng
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Lihua Fu
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Haochao Yan
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Wene Wei
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Bo Li
- Liaoning Provincial Engineering Laboratory of Anti-tumor Immunity and Molecular Theranostics Technology, Collaborative Innovation Center for Age-related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Haiming Ru
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Xianwei Mo
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Weizhong Tang
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Zijie Su
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
| | - Linhai Yan
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
21
|
Zhao Y, Lu L, Chen X, Yin Q. Natural compounds targeting ferroptosis in ovarian cancer: Research progress and application potential. Pharmacol Res 2025; 215:107729. [PMID: 40194611 DOI: 10.1016/j.phrs.2025.107729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/12/2025] [Accepted: 04/01/2025] [Indexed: 04/09/2025]
Abstract
Ovarian cancer (OC) is among the most common malignancies in the female reproductive system, marked by high rates of recurrence and mortality. Conventional chemotherapy, however, faces limitations due to the development of drug resistance, which hinders its effectiveness. Ferroptosis, an atypical form of programmed cell death distinct from autophagy, apoptosis, and necrosis, the relationship with tumors has become a hot research area in cancer studies in recent years. Anticancer therapies that target ferroptosis show strong potential in improving prognosis and counteracting chemotherapy resistance. Natural compounds, as a valuable source of novel targeted anticancer agents, its significant role in inhibiting tumor cell proliferation and metastasis and improving therapeutic sensitivity has been demonstrated in numerous existing studies. This review summarizes a range of natural compounds that target ferroptosis in OC cells, discussing their active components, mechanisms of action, and therapeutic potential, thereby providing useful insights for future targeted therapy and research in OC.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China.
| | - Lichao Lu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China.
| | - Xingying Chen
- Yuebei People's Hospital, Shaoguan, Guangdong 512000, China.
| | - Qiaozhi Yin
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China.
| |
Collapse
|
22
|
Lu Y, Mi L, Zhang P, Chen Y, Bai X, Li K, Zhang Y, Li J. Induction of Cuproptosis by Dichloromethane Extract From Patrinia scabiosaefolia Fisch on K562 Cells. ENVIRONMENTAL TOXICOLOGY 2025. [PMID: 40177825 DOI: 10.1002/tox.24516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 03/06/2025] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
Cuproptosis is a newly identified form of cell death that relies on copper (Cu) ionophores to transport Cu into cancer cells. As a perennial herb, Patrinia scabiosaefolia Fisch (PS) has garnered significant attention owing to its analgesic, anti-inflammatory, antibacterial, and antitumor properties. Previous research has shown that the extract from PS (DEPS) can inhibit the growth of leukemia cell lines. However, the specific mechanism of its anti-leukemic effect has not been fully clarified. Therefore, this study was conducted to investigate the molecular mechanism of cuproptosis in the treatment of leukemia with DEPS. Our results demonstrated that DEPS up-regulated SLC31A1 and down-regulated ATP7B expression, which increased intracellular copper concentration, down-regulated FDX1, influenced the lipoylation of DLAT and DLD, and subsequently increased the expression of the stress protein HSP70 and the expression of PDHA1, inducing copper death in K562 cells. In addition, we investigated the toxicity of DEPS in vivo and demonstrated its low in vivo toxicity and adequate in vivo safety. In conclusion, our results suggest that DEPS may induce cuproptosis in cells, offering valuable insights for the future application of PS in leukemia treatment.
Collapse
Affiliation(s)
- Yuan Lu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Leyuan Mi
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Clinical Laboratory Center, Gansu Provincial Maternity and Child-Care Hospital/Central Hospital of Gansu Province, Lanzhou, China
| | - Peipei Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yang Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xinyi Bai
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Kejing Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Ying Zhang
- Clinical Laboratory Division II, the First Hospital of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Juan Li
- Clinical Laboratory Division II, the First Hospital of Lanzhou University, Lanzhou University, Lanzhou, China
| |
Collapse
|
23
|
Mu F, Luo P, Zhu Y, Nie P, Li B, Bai X. Iron Metabolism and Ferroptosis in Diabetic Kidney Disease. Cell Biochem Funct 2025; 43:e70067. [PMID: 40166850 DOI: 10.1002/cbf.70067] [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: 09/15/2024] [Revised: 02/20/2025] [Accepted: 03/04/2025] [Indexed: 04/02/2025]
Abstract
Diabetic kidney disease (DKD) is a major diabetic microvascular complication that still lacks effective therapeutic drugs. Ferroptosis is a recently identified form of programmed cell death that is triggered by iron overload. It is characterized by unrestricted lipid peroxidation and subsequent membrane damage and is found in various diseases. Accumulating evidence has highlighted the crucial roles of iron overload and ferroptosis in DKD. Here, we review iron metabolism and the biology of ferroptosis. The role of aberrant ferroptosis in inducing diverse renal intrinsic cell death, oxidative stress, and renal fibrosis in DKD is summarized, and we elaborate on critical regulatory factors related to ferroptosis in DKD. Finally, we focused on the significance of ferroptosis in the treatment of DKD and highlight recent data regarding the novel activities of some drugs as ferroptosis inhibitors in DKD, aiming to provide new research targets and treatment strategies on DKD.
Collapse
Affiliation(s)
- Fangxin Mu
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Ping Luo
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Yuexin Zhu
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Ping Nie
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Bing Li
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Xue Bai
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
24
|
Luo L, Wang J, Zhao J, Yang B, Ma W, Lin J. Dental pulp stem cells derived exosomes inhibit ferroptosis via regulating the Nrf2-keap1/GPX4 signaling pathway to ameliorate chronic kidney disease injury. Tissue Cell 2025; 93:102670. [PMID: 39667244 DOI: 10.1016/j.tice.2024.102670] [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: 11/09/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION Chronic kidney disease (CKD) has long represented a substantial global health challenge. Regrettably, current therapeutic interventions exhibit limited efficacy in halting the progression of CKD. Ferroptosis may play a crucial role in CKD, as indicated by substantial evidence. Dental pulp stem cell-derived exosomes (DPSC-Exos) possess advantages such as abundant sources and low immunogenicity, holding promising prospects in CKD treatment. METHODS This study constructed a mouse CKD model to investigate the therapeutic effects of DPSC-Exos. First, we successfully extracted and identified DPSC-Exos. Then, mice were randomly divided into sham, PBS, CKD, and CKD+Exos groups. Our study determined the expression of ferroptosis-related pathway molecules Nrf2, GPX4, Keap1, and HO-1 in each group. Finally, we detected the expression levels of inflammatory factors, TNF-α, IL-1β, and IL-6, at the injury site. RESULTS Mice treated with DPSC-Exos showed increased expression of the ferroptosis inhibitory factor Nrf2 and its downstream regulatory factors GPX4 and HO-1, while the expression of Keap1 decreased. The expression of TNF-α, IL-1β, and IL-6 also decreased. CONCLUSION DPSC-Exos may help inhibit ferroptosis through the Keap1-Nrf2/GPX4 pathway and reduce the inflammatory response at the injury site, revealing their potential therapeutic effects on CKD.
Collapse
Affiliation(s)
- Lin Luo
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, Macao; Department of spine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jing Wang
- Department of spine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jie Zhao
- Department of Magnetic Resonance Imaging, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Bin Yang
- Department of Magnetic Resonance Imaging, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Wenzhe Ma
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, Macao.
| | - Jiaru Lin
- Department of nephropathy, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
25
|
Feng C, Zhang L, Zhou X, Lu S, Guo R, Song C, Zhang X. Redox imbalance drives magnetic property and function changes in mice. Redox Biol 2025; 81:103561. [PMID: 40020452 PMCID: PMC11910372 DOI: 10.1016/j.redox.2025.103561] [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: 01/06/2025] [Revised: 02/05/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025] Open
Abstract
The magnetic properties of substances directly determine their response to an externally applied magnetic field, which are closely associated with magnetoreception, magnetic resonance imaging (MRI), and magnetic bioeffects. However, people's understanding of the magnetic properties of living organisms remains limited. In this study, we utilized NRF2 (nuclear factor erythroid 2-related factor 2) deficient mice to investigate the contribution of redox (oxidation-reduction) homeostasis, in which the key process is the transfer of electron, a direct target of magnetic field and origin of paramagnetism. Our results show that the NRF2-/- mice exhibit significantly altered systemic redox state, accompanied by increased magnetic susceptibility, particularly in the liver and spleen. Further analyses reveal that the levels of paramagnetic reactive oxygen species (ROS) in these tissues are markedly elevated compared to wild-type mice. Moreover, the concentrations of Fe2+ and Fe3+ are significantly elevated in NRF2-/- mice, which are directly correlated with the increased magnetic susceptibility. The disrupted redox balance in NRF2-/- mice not only exacerbates oxidative stress and iron deposition, but also induces impairment to the liver and spleen. The findings highlight the combined effects of ROS and iron metabolism in driving magnetic susceptibility changes, providing valuable theoretical insights for further research into magnetic bioeffects and organ-specific sensitivity to magnetic fields.
Collapse
Affiliation(s)
- Chuanlin Feng
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, 230026, China
| | - Lei Zhang
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Xiaoyuan Zhou
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230039, China
| | - Shiyu Lu
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, 230026, China
| | - Ruowen Guo
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, 230026, China
| | - Chao Song
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
| | - Xin Zhang
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, 230026, China; Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230039, China.
| |
Collapse
|
26
|
Ye Y, Xie X, Bi Y, Liu Q, Qiu L, Zhao H, Wang C, Zhu W, Zeng T. Nrf2 alleviates acute ischemic stroke induced ferroptosis via regulating xCT/GPX4 pathway. Free Radic Biol Med 2025; 231:153-162. [PMID: 40020881 DOI: 10.1016/j.freeradbiomed.2025.02.040] [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: 01/13/2025] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/03/2025]
Abstract
Ferroptosis is a form of regulating cell death, and iron accumulation in the brain after acute ischemic stroke (AIS) is associated with the triggering of iron metabolism. Nuclear factor erythroid 2-related factor 2 (Nrf2), one of the most critical antioxidant transcription factors in cells, is closely associated with ferroptosis and oxidative stress.In the present study, we explore the intrinsic mechanisms by which Nrf2 exerts neuroprotective effects against AIS-induced ferroptosis.In vivo experiments, we explored the protective effects of AIS induced by middle cerebral artery occlusion (MCAO) and its mechanisms by using intraperitoneal injections of ferrostatin-1 (Fer-1, an inhibitor of ferroptosis), Oltipraz (an agonist of Nrf2) and ML385 (an inhibitor of Nrf2) in wild-type (WT) mice, as well as using Nrf2-/- mice. In vitro experiments, we investigated the mechanism of action of Nrf2 on the establishment of a ferroptosis cell model induced by Erastin by overexpressing or silencing Nrf2 expression using shRNA in SH-SY5Y cells.Ferroptosis played an important role in AIS, and Fer-1 inhibited iron accumulation and alleviated neuronal damage caused by AIS.Oltipraz attenuated AIS-induced neuronal damage and cerebral infarction by increasing cortical blood flow (CBF). Additionally, Oltipraz protected against AIS-induced ferroptosis by reducing oxidative stress and iron overload. Meanwhile, in Oltipraz-treated AIS mice, Nrf2, solute carrier family 7 member 11 (SLC7A11/xCT), and glutathione peroxidase 4 (GPX4) were upregulated. Conversely, ML385 decreased CBF and exacerbated IS-induced neuronal damage. Furthermore, both ML385 treatment and Nrf2 knockout mice exacerbated oxidative stress injury and iron overload and downregulated the expression of both xCT and GPX4. Consistent with the in vivo results, Nrf2 conferred ferroptosis resistance in vitro upon exposure to compounds that induce ferroptosis, by modulating the xCT/GPX4 pathway.The present study confirmed that Nrf2 could attenuate AIS-induced neuronal ferroptosis and oxidative stress by regulating xCT/GPX4.
Collapse
Affiliation(s)
- Yujun Ye
- Department of Brain Diseases, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangdong, China; School of Combine Traditional Chinese and Western Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China; Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xuexin Xie
- Department of Brain Diseases, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangdong, China; School of Combine Traditional Chinese and Western Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China; Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yiming Bi
- Department of Brain Diseases, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangdong, China; Sleep Research Institute of Traditional Chinese Medicine, Guangzhou Medical University, Guangdong, China
| | - Qing Liu
- Department of Brain Diseases, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangdong, China; Sleep Research Institute of Traditional Chinese Medicine, Guangzhou Medical University, Guangdong, China
| | - Lingling Qiu
- Department of Brain Diseases, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangdong, China; Sleep Research Institute of Traditional Chinese Medicine, Guangzhou Medical University, Guangdong, China
| | - He Zhao
- Department of Brain Diseases, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangdong, China; Sleep Research Institute of Traditional Chinese Medicine, Guangzhou Medical University, Guangdong, China
| | - Chengyin Wang
- Department of Brain Diseases, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangdong, China; Sleep Research Institute of Traditional Chinese Medicine, Guangzhou Medical University, Guangdong, China.
| | - Weifeng Zhu
- Department of Brain Diseases, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangdong, China; Sleep Research Institute of Traditional Chinese Medicine, Guangzhou Medical University, Guangdong, China.
| | - Ting Zeng
- Department of Brain Diseases, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangdong, China; Sleep Research Institute of Traditional Chinese Medicine, Guangzhou Medical University, Guangdong, China.
| |
Collapse
|
27
|
Zhang H, Shi H, Li X, Zhou S, Song X, Ma N, Meng M, Chang G, Shen X. Quercetin alleviates LPS/iE-DAP-induced liver injury by suppressing ferroptosis via regulating ferritinophagy and intracellular iron efflux. Redox Biol 2025; 81:103557. [PMID: 39986118 PMCID: PMC11904602 DOI: 10.1016/j.redox.2025.103557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/06/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025] Open
Abstract
Ruminal dysbiosis-induced liver injury is prevalent in dairy cows, yet its underlying mechanisms remain incompletely understood. Ferroptosis, a newly identified form of programmed cell death distinct from apoptosis and necrosis, has been implicated in various liver diseases by emerging studies. In the present study, lipopolysaccharide (LPS) and γ-D-glutamyl-meso-diaminopimelic acid (iE-DAP) were employed to establish in vitro and in vivo models of liver injury using bovine hepatocytes and mice, respectively. It was observed that noncytotoxic iE-DAP alone did not influence lipid peroxidation or GPX4, but exacerbated LPS-induced ferroptosis and hepatocyte injury. Notably, co-treatment with LPS and iE-DAP (LPS/iE-DAP)-induced hepatocyte injury was mitigated by intervention with the ferroptosis inhibitor ferrostatin-1 (Fer-1). Mechanistically, the activated IL-6/STAT3 signaling pathway was found to mediate LPS/iE-DAP-induced ferroptosis. Suppression of IL-6/STAT3, either through IL6 and STAT3 knockdown or pharmacological intervention, reduced Fe2+ accumulation and alleviated ferroptotic cell death. Further investigations identified that IL-6/STAT3 signaling enhanced ferritinophagy and impaired iron export. Either disrupting ferritinophagy by knocking down NCOA4 or restoring iron export via HAMP knockdown relieved intracellular iron overload and inhibited ferroptosis. Specifically, LPS/iE-DAP treatment increased the interaction between hepcidin and ferroportin, promoting ferroportin ubiquitination and degradation, thereby blocking iron efflux. Furthermore, we provided several evidence to prove that quercetin pretreatment alleviated LPS/iE-DAP-induced ferroptosis and liver injury by decreasing hepatic iron accumulation via targeting the IL-6/STAT3 signaling in vitro and in vivo, effects reversed by the addition of recombinant bovine IL-6. Based on these findings, we concluded that LPS/iE-DAP-induced liver injury by triggering ferroptosis through regulating IL-6/STAT3/ferritinophagy-dependent iron release and IL-6/STAT3/hepcidin/ferroportin-dependent iron export, while quercetin could alleviate this liver injury by inhibiting ferroptosis via IL-6/STAT3 signaling pathway. This study provides novel insights into the mechanisms whereby ruminal dysbiosis induces liver injury and presents a prospective solution for ruminal dysbiosis-induced liver injury.
Collapse
Affiliation(s)
- Hongzhu Zhang
- Ministry of Education Joint International Research Laboratory of Animal Health and Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Huimin Shi
- Ministry of Education Joint International Research Laboratory of Animal Health and Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Xuerui Li
- Ministry of Education Joint International Research Laboratory of Animal Health and Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Shendong Zhou
- Ministry of Education Joint International Research Laboratory of Animal Health and Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Xiaokun Song
- Ministry of Education Joint International Research Laboratory of Animal Health and Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Nana Ma
- Ministry of Education Joint International Research Laboratory of Animal Health and Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Meijuan Meng
- Ministry of Education Joint International Research Laboratory of Animal Health and Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Guangjun Chang
- Ministry of Education Joint International Research Laboratory of Animal Health and Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Xiangzhen Shen
- Ministry of Education Joint International Research Laboratory of Animal Health and Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China.
| |
Collapse
|
28
|
Li P, Chu D, Ding G, Qin D, Bu Y, Tian B. IGF2BP3 suppresses ferroptosis in lung adenocarcinoma by m6A-dependent regulation of TFAP2A to transcriptionally activate SLC7A11/GPX4. Mol Cell Biochem 2025; 480:2361-2375. [PMID: 39026029 DOI: 10.1007/s11010-024-05068-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: 05/27/2024] [Accepted: 06/30/2024] [Indexed: 07/20/2024]
Abstract
Ferroptosis is recently discovered as an important player in the initiation, proliferation, and progression of human tumors. Insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3) has been reported as an oncogene in multiple types of cancers, including lung adenocarcinoma (LUAD). However, little research has been designed to investigate the regulation of IGF2BP3 on ferroptosis in LUAD. qRT-PCR and western blot were used to measure the mRNA and protein expression of IGF2BP3 and transcription factor AP-2 alpha (TFAP2A). CCK-8 assay was performed to determine cell viability. DCFH-DA and C11-BODIPY staining were used to detect the levels of intracellular reactive oxygen species (ROS) and lipid ROS. The corresponding assay kits were used to analyze the levels of malondialdehyde (MDA) and glutathione (GSH). SRAMP website and m6A RNA immunoprecipitation (Me-RIP) were used to predict and confirm the m6A modification of TFAP2A. RIP experiments were conducted to confirm the binding of IGF2BP3 and TFAP2A. RNA stability assay was performed using actinomycin D. Chromatin immunoprecipitation (ChIP) and dual-luciferase reporter experiments were performed to confirm the interaction between TFAP2A and cystine/glutamate antiporter solute carrier family 7 member 11 (SLC7A11) or glutathione peroxidase 4 (GPX4). Mice xenotransplant model was also constructed to explore the effect of IGF2BP3 on LUAD tumor growth and ferroptosis. IGF2BP3 and TFAP2A were both highly expressed in LUAD. IGF2BP3 or TFAP2A knockdown induced ferroptosis by aggravating erastin-induced cell viability suppression, increasing the production of intracellular ROS, lipid ROS, and MDA, and decreasing GSH synthesis, GSH/GSSG ratio, and cystine uptake. Mechanistically, IGF2BP3 stabilized TFAP2A expression via m6A modification. Moreover, sh-IGF2BP3-mediated ferroptosis was significantly abated by TFAP2A overexpression. Furthermore, TFAP2A binds to the promoters of SLC7A11 and GPX4 to promote their transcription. Also, IGF2BP3 depletion suppressed LUAD tumor growth by inducing ferroptosis in mice. IGF2BP3 suppresses ferroptosis in LUAD by m6A-dependent regulation of TFAP2A to promote the transcription of SLC7A11 and GPX4. Our findings suggest that targeting IGF2BP3/TFAP2A/SLC7A11/GPX4 axis might be a potential therapeutic choice to increase ferroptosis sensitivity in LUAD.
Collapse
Affiliation(s)
- Pengpeng Li
- Tumor Treatment Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, 450052, China
| | - Dan Chu
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, China.
| | - Guangcheng Ding
- Tumor Treatment Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, 450052, China
| | - Dehua Qin
- Tumor Treatment Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, 450052, China
| | - Yajing Bu
- Tumor Treatment Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, 450052, China
| | - Bi Tian
- Tumor Treatment Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, 450052, China.
| |
Collapse
|
29
|
Wu H, Lan Q, He YX, Xue JY, Liu H, Zou Y, Liu P, Luo G, Chen MT, Liu MN. Programmed cardiomyocyte death in myocardial infarction. Apoptosis 2025; 30:597-615. [PMID: 39833636 DOI: 10.1007/s10495-025-02075-3] [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: 01/02/2025] [Indexed: 01/22/2025]
Abstract
Cardiovascular disease (CVD) is a leading cause of human mortality worldwide, with patients often at high risk of heart failure (HF) in myocardial infarction (MI), a common form of CVD that results in cardiomyocyte death and myocardial necrosis due to inadequate myocardial perfusion. As terminally differentiated cells, cardiomyocytes possess a severely limited capacity for regeneration, and an excess of dead cardiomyocytes will further stress surviving cells, potentially exacerbating to more extensive heart disease. The article focuses on the relationship between programmed cell death (PCD) of cardiomyocytes, including different forms of apoptosis, necrosis, and autophagy, and MI, as well as the potential application of these mechanisms in the treatment of MI. By gaining a deeper understanding of the mechanisms of cardiomyocyte death, it aims to provide new insights into the prevention and treatment of MI.
Collapse
Affiliation(s)
- Hao Wu
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Qi Lan
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Yi-Xiang He
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Jin-Yi Xue
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Hao Liu
- Department of Pediatrics, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Yuan Zou
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Ping Liu
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Gang Luo
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
| | - Ming-Tai Chen
- Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, 518033, Shenzhen, People's Republic of China.
| | - Meng-Nan Liu
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
| |
Collapse
|
30
|
You YL, Choi HS. Nootkatone (NK), a grapefruit-derived sesquiterpenoid, suppresses UVB-induced damage by regulating NRF2-HO-1 and AhR-CYP1A1 signaling pathways in HaCaT cells. Food Sci Biotechnol 2025; 34:1751-1761. [PMID: 40151610 PMCID: PMC11937452 DOI: 10.1007/s10068-024-01791-x] [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: 09/09/2024] [Revised: 11/07/2024] [Accepted: 12/05/2024] [Indexed: 03/29/2025] Open
Abstract
Nootkatone (NK), a sesquiterpene naturally derived from citrus species, was investigated for its protective effect against UVB-induced damage in HaCaT cells and its underlying mechanisms. NK effectively suppressed UVB-mediated cell death and significantly modulated expression of skin hydration genes; NK (100 μM) increased mRNA levels of collagen-1 and HAS by 44.6 and 34.7%, respectively, while downregulating HYAL by 46.8%. NK also reduced MMP1/2 expression, key matrix metalloproteinases, but enhanced mRNA levels of skin barrier factors, Filaggrin, Loricrin, and Involucrin by up to 45%. Additionally, NK lowered UVB-induced ROS production and elevated antioxidant levels (NRF2, HO-1, catalase, SOD1, and Gpx), and decrease the protein levels of xenobiotic factors, AhR and CYP1A1. These findings suggest that NK protects skin integrity against UVB-induced photoaging through the modulation of NRF2-HO-1 and AhR-CYP1A1 signaling pathways. NK shows promise as a functional agent, either edible or topical, for protecting against UVB-induced skin damage. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-024-01791-x.
Collapse
Affiliation(s)
- Ye-Lim You
- Department of Food Nutrition, Sangmyung University, Hongjimun 2-Gil 20, Jongno-Gu, Seoul, 03016 Republic of Korea
| | - Hyeon-Son Choi
- Department of Food Nutrition, Sangmyung University, Hongjimun 2-Gil 20, Jongno-Gu, Seoul, 03016 Republic of Korea
| |
Collapse
|
31
|
Wu M, Zhang S, Wu X, Zhou Y, Zhou M, Du A, Zhang Y, Wei T, Wang B, Wang S, Jiang C, Hu S, Xiao J, Wu Y. Acute hyperglycemia impedes spinal cord injury recovery via triggering excessive ferroptosis of endothelial cells. Int J Biol Macromol 2025; 301:140453. [PMID: 39884601 DOI: 10.1016/j.ijbiomac.2025.140453] [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: 10/09/2023] [Revised: 01/03/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Spinal cord injury (SCI) is a serious central nervous system injury that often causes sensory and motor dysfunction in patients. Diabetes seriously destroys the blood spinal cord barrier (BSCB) and aggravates SCI. Ferroptosis is a new type of programmed cell death. The role of ferroptosis in diabetes-medicated BSCB destruction has not been clearly elucidated. Here, we built a type 1 diabetes (T1D) combined with SCI rat model and confirmed that hyperglycemia exacerbates SCI-mediated BSCB destruction. Pathological mechanism demonstrated that except for apoptosis, the excessive ferroptosis is another caused factor for endothelial cells (ECs) loss under hyperglycemic condition. More importantly, ferrostatin-1(a ferroptosis inhibitor) treatment significantly inhibited the ferroptosis of ECs, and promoted the BSCB repair in T1D combined with SCI rat. The mechanism study further revealed that hyperglycemia not only induces the elevated reactive oxygen species (ROS) via activating RAGE, but also suppresses the xCT expression in system Xc- in ECs, which decreases GPX4 expression and induces ferroptosis. Additionally, hyperglycemia also accelerated the transfer of iron ions from serum to spinal cord after SCI. In summary, our results suggest that the excessive ferroptosis of ECs is essential for the severe BSCB destruction in T1D combined with spinal cord injury rat.
Collapse
Affiliation(s)
- Man Wu
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Wenzhou University, Wenzhou 325035, China; The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China; National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Susu Zhang
- The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China
| | - Xuejuan Wu
- The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China
| | - Yongxiu Zhou
- The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China
| | - Mei Zhou
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Wenzhou University, Wenzhou 325035, China
| | - Anyu Du
- The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China
| | - Yanren Zhang
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Wenzhou University, Wenzhou 325035, China
| | - Tao Wei
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Wenzhou University, Wenzhou 325035, China
| | - Beini Wang
- The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China
| | - Shuangshuang Wang
- The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China
| | - Chang Jiang
- The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China
| | - Siwang Hu
- The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China.
| | - Jian Xiao
- The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China.
| | - Yanqing Wu
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Wenzhou University, Wenzhou 325035, China; The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China.
| |
Collapse
|
32
|
Li S, Xiao X, Chang Y, Xu Z, Zheng X, Zhou H, Ding H, Lu W, Li T, Tao Y. Berberine Inhibits Abdominal Aortic Aneurysm Formation and Vascular Smooth Muscle Cell Phenotypic Switching by Regulating the Nrf2 Pathway. J Cell Mol Med 2025; 29:e70509. [PMID: 40193135 PMCID: PMC11974455 DOI: 10.1111/jcmm.70509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/12/2025] [Accepted: 03/10/2025] [Indexed: 04/10/2025] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening disease featuring extensive membrane destruction in the vascular wall, which is closely associated with the phenotypic switching of vascular smooth muscle cells (VSMC). A thorough understanding of the changes in regulatory factors during the pathogenesis of VSMC phenotypic switching is essential for medical treatments in AAA. NRF2 was deemed to hold a pivotal position in developing AAA, especially as it can regulate VSMC phenotypic switching. In this study, we found that berberine prevents the formation of AAA by regulating the phenotypic switching of VSMC, which was well validated in both in vitro and in vivo functional experiments. Mechanically, we found that berberine regulates VSMC phenotypic switching by promoting the expression of downstream VSMC contraction genes through the deubiquitination of Keap1, in which the deubiquitinating enzyme USP15 plays an important mediating role in this process.
Collapse
MESH Headings
- Berberine/pharmacology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/drug therapy
- Aortic Aneurysm, Abdominal/prevention & control
- NF-E2-Related Factor 2/metabolism
- NF-E2-Related Factor 2/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Animals
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Signal Transduction/drug effects
- Phenotype
- Mice
- Humans
- Male
- Kelch-Like ECH-Associated Protein 1/metabolism
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Sanjun Li
- Department of CardiologyJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangChina
| | - Xiaoyong Xiao
- Department of Emergency MedicineThe First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's HospitalShenzhenGuangdongChina
| | - Yuechen Chang
- Experimental Center of Medical School of Shihezi UniversityShiheziChina
| | - Ziyao Xu
- Senior Department of General SurgeryThe First Medical Center of Chinese PLA General HospitalBeijingChina
| | | | - Haiwen Zhou
- Department of CardiologyJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangChina
| | - Haiqiang Ding
- Department of CardiologyJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangChina
| | - Weiling Lu
- Department of CardiologyGuangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Prevention and Treatment of Coronary Heart Disease,Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
| | - Tian Li
- Tianjin Key Laboratory of Acute Abdomen Disease‐Associated Organ Injury and ITCWM RepairInstitute of Integrative Medicine of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical UniversityTianjinChina
| | - Yu Tao
- Department of CardiologyJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangChina
| |
Collapse
|
33
|
Qi L, Qian L, Yu X, Qiu K. SIRT6 mitigates oxidative stress and RSL3-induced ferroptosis in HTR-8/SVneo cells. Tissue Cell 2025; 93:102639. [PMID: 39642638 DOI: 10.1016/j.tice.2024.102639] [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: 08/25/2024] [Revised: 11/06/2024] [Accepted: 11/24/2024] [Indexed: 12/09/2024]
Abstract
Dysregulation in placental trophoblast cells frequently results in oxidative stress, culminating in pregnancy-related complications. While iron is essential for fetal development, cellular ferroptosis due to elevated iron levels might mediate the emergence of preeclampsia (PE), presenting significant risks during gestation. We found abnormally activated oxidative stress and increased iron concentration in the placental tissues of PE patients. Subsequently, we treated placental trophoblasts with hydrogen peroxide and RSL3 to induce oxidative stress and ferroptosis models. The results revealed that SIRT6 overexpression activates the Nrf2/HO-1 pathway, restores the oxidative imbalance of the cells, and protects the cells from ferroptosis. Meanwhile, activation of the Nrf2/HO-1 pathway alone showed similar results. Thus, we posit that SIRT6, via the Nrf2/HO-1 pathway, alleviates cellular oxidative stress and diminishes ferroptosis, offering a novel therapeutic avenue for PE.
Collapse
Affiliation(s)
- Lifang Qi
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China
| | - Liyan Qian
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China
| | - Xiaoting Yu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China
| | - Kan Qiu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, China.
| |
Collapse
|
34
|
Kim YK, Song J. Metabolic imbalance and brain tumors: The interlinking metabolic pathways and therapeutic actions of antidiabetic drugs. Pharmacol Res 2025; 215:107719. [PMID: 40174814 DOI: 10.1016/j.phrs.2025.107719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
Brain tumors are complex, heterogeneous malignancies, often associated with significant morbidity and mortality. Emerging evidence suggests the important role of metabolic syndrome, such as that observed in diabetes mellitus, in the progression of brain tumors. Several studies indicated that hyperglycemia, insulin resistance, oxidative stress, and altered adipokine profiles influence tumor growth, proliferation, and treatment resistance. Intriguingly, antidiabetic drugs (e.g., metformin, sulfonylureas, dipeptidyl peptidase-4 (DPP-4) inhibitors, glucagon-like peptide-1 (GLP-1) receptor agonists, and thiazolidinediones) have shown promise as adjunctive or repurposed agents in managing brain tumors. Metformin can impair tumor cell proliferation, enhance treatment sensitivity, and modify the tumor microenvironment by activating AMP-activated protein kinase (AMPK) and inhibiting mammalian target of rapamycin (mTOR) signaling pathways. DPP-4 inhibitors and GLP-1 receptor agonists can target both metabolic and inflammatory aspects of brain tumors, while thiazolidinediones may induce apoptosis in tumor cells and synergize with other therapeutics. Consequently, further studies and clinical trials are needed to confirm the efficacy, safety, and utility of metabolic interventions in treating brain tumors. Here, we review the evidence for the metabolic interconnections between metabolic diseases and brain tumors and multiple actions of anti-diabetes drugs in brain tumors.
Collapse
Affiliation(s)
- Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, 58128, Korea.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun, 58128, Korea.
| |
Collapse
|
35
|
Zhan J, Chen Y, Liu Y, Chen Y, Li Z, Li X, He Z, Meng F, Qian X, Yang L, Yang Q. IDO1-mediated AhR activation up-regulates pentose phosphate pathway via NRF2 to inhibit ferroptosis in lung cancer. Biochem Pharmacol 2025; 236:116913. [PMID: 40164341 DOI: 10.1016/j.bcp.2025.116913] [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: 01/08/2025] [Revised: 02/28/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Ferroptosis is a type of cell death marked by iron-dependent lipid peroxide accumulation. Indoleamine 2,3-dioxygenase 1 (IDO1), a key enzyme in the catabolism of tryptophan through kynurenine pathway, participates in the development of multiple tumor types. However, the role of IDO1 in tumor ferroptosis is unclear. In this study, we identified IDO1 as a key regulator of ferroptosis in lung cancer. With Erastin-treated lung cancer cells, we found that IDO1 inhibited ferroptosis, reduced the generation of lipid peroxide and ROS. Mechanistically, IDO1 promoted the expression of nuclear factor erythroid 2-related factor 2 (NRF2) through activating aryl hydrocarbon receptor (AhR) pathway. IDO1 up-regulated the expression of solute carrier family 7 member 11 (SLC7A11) and the activity of pentose phosphate pathway (PPP) via AhR-NRF2 axis, promoted the production of reduced nicotinamide adenine dinucleotide phosphate (NADPH) and glutathione (GSH), thereby inhibiting ferroptosis. Moreover, combined treatment with IDO1 inhibitor and Erastin inhibited tumor growth, down-regulated SLC7A11 expression and PPP activity, promoted tumor ferroptosis in lung cancer-bearing mice. In conclusion, this study revealed the function of IDO1 in lung cancer ferroptosis and provided a new strategy for lung cancer therapy.
Collapse
Affiliation(s)
- Jiani Zhan
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, MOE Engineering Research Center of Gene Technology, Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai, China
| | - Yijia Chen
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, MOE Engineering Research Center of Gene Technology, Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai, China
| | - Yuying Liu
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, MOE Engineering Research Center of Gene Technology, Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai, China
| | - Yunqiu Chen
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, MOE Engineering Research Center of Gene Technology, Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai, China
| | - Zhiyao Li
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, MOE Engineering Research Center of Gene Technology, Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai, China
| | - Xuewen Li
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, MOE Engineering Research Center of Gene Technology, Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai, China
| | - Zhenning He
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, MOE Engineering Research Center of Gene Technology, Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai, China
| | - Fangzhou Meng
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, MOE Engineering Research Center of Gene Technology, Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai, China
| | - Xiaoyang Qian
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, MOE Engineering Research Center of Gene Technology, Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai, China
| | - Lili Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.
| | - Qing Yang
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, MOE Engineering Research Center of Gene Technology, Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai, China.
| |
Collapse
|
36
|
Huang H, Liu S, Wu J, Zhu J, Xu J, Yu S, Bei L, Zhang B, Luo Y. Buyang Huanwu Decoction Alleviates Ischemic Stroke Injury by Inhibiting Ferroptosis via the Nrf2/GPX4 Pathway. Drug Des Devel Ther 2025; 19:2285-2305. [PMID: 40170790 PMCID: PMC11960815 DOI: 10.2147/dddt.s503424] [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: 11/26/2024] [Accepted: 03/20/2025] [Indexed: 04/03/2025] Open
Abstract
Purpose Acute ischemic stroke poses major challenges due to high disability and mortality rates. Ferroptosis, a form of regulated cell death triggered by iron-induced oxidative stress, plays a key role in stroke injury. Despite its long history in stroke treatment, the mechanism of Buyang Huanwu Decoction (BHD) in ferroptosis remains unclear. Methods Network pharmacology predicted BHD's active components and pathways, while Ultra Performance Liquid Chromatography-Tandem Mass Spectrometry (UPLC-MS/MS) confirmed its main ingredients. Middle Cerebral Artery Occlusion (MCAO) was induced in C57 mice, with neurological deficits and infarct size assessed by Longa scoring and TTC staining. Histopathological and ultrastructural changes were assessed by staining and electron microscopy, and biochemical markers (MDA, GSH, SOD, Fe²+) measured by kits. Western blotting and qPCR analyzed ferroptosis-related proteins, Nrf2 localization, and gene expression. In vitro, HT22 cells viability and ROS levels were assessed under Oxygen-Glucose Deprivation/Reoxygenation (OGD/R) conditions. Protein expression, Nrf2 interactions, and nuclear translocation were also investigated. Results Network pharmacology showed BHD targets key pathways in cerebral infarction, including ferroptosis and antioxidant pathways. BHD improved neurological function and reduced the infarct size in MCAO mice by 10% - 50%, and also significantly decreased the levels of oxidative stress markers (MDA, Fe2+) while increasing the activities of antioxidants (GSH, SOD). Histopathological and ultrastructural analyses demonstrated reduced neuronal damage and improved mitochondrial structure. Western blot and qPCR indicated upregulation of GPX4 and Nrf2, downregulation of Keap1, and Nrf2 nuclear translocation. In vitro, BHD enhanced HT22 cell viability and reduced ROS under stress. Protein analysis confirmed increased Nrf2, GPX4, and HO-1, with decreased Keap1 and enhanced Nrf2 nuclear translocation. Nrf2 inhibitor experiments confirmed BHD's effects are Nrf2-mediated. Conclusion In pre-clinical studies, BHD exerts neuroprotective effects in ischemic stroke by inhibiting ferroptosis through the Nrf2/GPX4 pathway.
Collapse
Affiliation(s)
- Hao Huang
- Department of Neurology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, People’s Republic of China
| | - Sijie Liu
- Department of Neurology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, People’s Republic of China
| | - Jing Wu
- Department of Traditional Chinese Medicine, Community Health Service Center of the Urban Area, Suzhou, People’s Republic of China
| | - Jiayi Zhu
- Department of Neurology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, People’s Republic of China
| | - Jiaxiang Xu
- Department of Neurology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, People’s Republic of China
| | - Shuhong Yu
- Department of Neurology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, People’s Republic of China
| | - Lingna Bei
- Department of Neurology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, People’s Republic of China
| | - Biao Zhang
- Department of Neurology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, People’s Republic of China
- Central laboratory, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, People’s Republic of China
| | - Yi Luo
- Department of Neurology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, People’s Republic of China
| |
Collapse
|
37
|
Jiang C, Yan Y, Long T, Xu J, Chang C, Kang M, Wang X, Chen Y, Qiu J. Ferroptosis: a potential therapeutic target in cardio-cerebrovascular diseases. Mol Cell Biochem 2025:10.1007/s11010-025-05262-7. [PMID: 40148662 DOI: 10.1007/s11010-025-05262-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
Cardio-cerebrovascular diseases (CCVDs) are the leading cause of global mortality, yet effective treatment options remain limited. Ferroptosis, a novel form of regulated cell death, has emerged as a critical player in various CCVDs, including atherosclerosis, myocardial infarction, ischemia-reperfusion injury, cardiomyopathy, and ischemic/hemorrhagic strokes. This review highlights the core mechanisms of ferroptosis, its pathological implications in CCVDs, and the therapeutic potential of targeting this process. Additionally, it explores the role of Chinese herbal medicines (CHMs) in mitigating ferroptosis, offering novel therapeutic strategies for CCVDs management. Ferroptosis is regulated by several key pathways. The GPX4-GSH-System Xc- axis is central to ferroptosis execution, involving GPX4 using GSH to neutralize lipid peroxides, with system Xc- being crucial for GSH synthesis. The NAD(P)H/FSP1/CoQ10 axis involves FSP1 regenerating CoQ10 via NAD(P)H, inhibiting lipid peroxidation independently of GPX4. Lipid peroxidation, driven by PUFAs and enzymes like ACSL4 and LPCAT3, and iron metabolism, regulated by proteins like TfR1 and ferritin, are also crucial for ferroptosis. Inhibiting ferroptosis shows promise in managing CCVDs. In atherosclerosis, ferroptosis inhibitors reduce iron accumulation and lipid peroxidation. In myocardial infarction, inhibitors protect cardiomyocytes by preserving GPX4 and SLC7A11 levels. In ischemia-reperfusion injury, targeting ferroptosis reduces myocardial and cerebral damage. In diabetic cardiomyopathy, Nrf2 activators alleviate oxidative stress and iron metabolism irregularities. CHMs offer natural compounds that mitigate ferroptosis. They possess antioxidant properties, chelate iron, and modulate signaling pathways like Nrf2 and AMPK. For example, Salvia miltiorrhiza and Astragalus membranaceus reduce oxidative stress, while some CHMs chelate iron, reducing its availability for ferroptosis. In conclusion, ferroptosis plays a pivotal role in CCVDs, and targeting it offers novel therapeutic avenues. CHMs show promise in reducing ferroptosis and improving patient outcomes. Future research should explore combination therapies and further elucidate the molecular interactions in ferroptosis.
Collapse
Affiliation(s)
- Chenlong Jiang
- School of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
| | - Yang Yan
- Department of Cardiology, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China
| | - Tianlin Long
- Department of Neurosurgery, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China
| | - Jiawei Xu
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
| | - Cuicui Chang
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
- Department of Cardiology, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China
| | - Meili Kang
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
| | - Xuanqi Wang
- Department of Cardiology, First Hospital of Northwestern University, Northwest University, No. 512 Xianning East Road, Xi'an, 710043, Shaanxi, China.
| | - Yuhua Chen
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China.
- Department of Neurosurgery, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China.
- School of Life and Health Science, Hainan University, No. 58 People's Avenue, Haikou, 570100, Hainan, China.
| | - Junlin Qiu
- Department of Cardiology, First Hospital of Northwestern University, Northwest University, No. 512 Xianning East Road, Xi'an, 710043, Shaanxi, China.
| |
Collapse
|
38
|
Lan D, Huang S, Li J, Zhou S, Deng J, Qin S, Zhou T, Meng F, Li W. Ferroptosis in Endometriosis: Traditional Chinese Medicine Interventions and Mechanistic Insights. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2025; 53:385-408. [PMID: 40145281 DOI: 10.1142/s0192415x25500156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Endometriosis (EMS) is a chronic, estrogen-dependent inflammatory disease affecting 5-10% of women of reproductive age, characterized by the growth of endometrial tissue on the outside of the uterus. The dysregulation of iron metabolism leads to the accumulation of iron ions at the lesion sites, resulting in oxidative stress and pro-inflammatory responses that promote the progression of EMS. The mechanisms underlying ferroptosis in EMS primarily involve iron accumulation, lipid peroxidation, and loss of glutathione peroxidase 4 activity. These mechanisms confer resistance to ferroptosis within the ectopic tissues and facilitate cell survival and proliferation. Traditional Chinese medicine (TCM) has demonstrated therapeutic potential for modulating ferroptosis. Studies have shown that TCM monomers may regulate ferroptosis by modulating iron transport proteins and anti-oxidant defense mechanisms. TCM formulas employ distinct treatment strategies depending on the stage of EMS: in the early stages, they promote ferroptosis to control lesion growth, whereas in the later stages, they inhibit ferroptosis to reduce oxidative stress and inflammation in order to improve reproductive health and slow disease progression. This study provides a new perspective on potential therapeutic strategies for the management of EMS by summarizing the role of ferroptosis in its pathological mechanisms and reviewing findings on the use of TCM in regulating ferroptosis.
Collapse
Affiliation(s)
- Dingli Lan
- Graduate School Guangxi University of Chinese Medicine Nanning, P. R. China
| | - Shuping Huang
- Graduate School Guangxi University of Chinese Medicine Nanning, P. R. China
| | - Jing Li
- Graduate School Guangxi University of Chinese Medicine Nanning, P. R. China
| | - Shilang Zhou
- Graduate School Guangxi University of Chinese Medicine Nanning, P. R. China
| | - Jianli Deng
- Graduate School Guangxi University of Chinese Medicine Nanning, P. R. China
| | - Shuiyun Qin
- Graduate School Guangxi University of Chinese Medicine Nanning, P. R. China
| | - Ting Zhou
- Graduate School Guangxi University of Chinese Medicine Nanning, P. R. China
| | - Fengyun Meng
- Yao College of Medicine Guangxi University of Chinese Medicine Nanning, P. R. China
| | - Weihong Li
- Department of Nursing Guangxi University of Chinese Medicine Nanning, P. R. China
| |
Collapse
|
39
|
Yin Y, Mu F, Zhang L, Zhao J, Gong R, Yin Y, Zheng L, Du Y, Jin F, Wang J. Wedelolactone activates the PI3K/AKT/NRF2 and SLC7A11/GPX4 signalling pathways to alleviate oxidative stress and ferroptosis and improve sepsis-induced liver injury. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119557. [PMID: 40010556 DOI: 10.1016/j.jep.2025.119557] [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: 12/20/2024] [Revised: 02/11/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025]
Abstract
INTRODUCTION Sepsis-induced liver injury (SILI) is a severe complication of sepsis. Wedelolactone (WEL) can be used to treat liver diseases. However, its therapeutic mechanisms and efficacy in SILI remain unclear. To investigate the therapeutic effects of WEL on SILI and its potential mechanisms of action through in vitro and in vivo experiments. METHODS A SILI model based on lipopolysaccharide (LPS), and AML12 cells were treated with different concentrations of WEL, LY294002 and ML385. The SILI model was established by caecal ligation and puncture (CLP). C57BL/6 mice were administered WEL and biphenyl diester for seven consecutive days, and CLP was then performed 1 h later. Blood and liver tissue were collected 24 h later for subsequent analysis. HE staining, liver function index, oxidative stress index, JC-1 staining, transmission electron microscopy, immunofluorescence staining, Western blot, and inflammatory cytokines were used to detect oxidative stress and ferroptosis-related markers. RESULTS The in vivo experiments showed that WEL treatment reduced the pathological damage of the liver and decreased ALT and AST, MMP and ROS (the product of iron and lipid peroxidation) and inflammatory factors. WEL also decreased hepatocyte viability in vitro. Inhibition of NRF2 can lead to exacerbation of SILI. The expressions of P-PI3K and P-AKT were up-regulated while HO-1, GPX4, NRF2, and SLC7A11 were down-regulated in vitro and in vivo. CONCLUSIONS Ferroptosis and oxidative stress are pivotal in SILI. WEL mitigates SILI by inhibiting ferroptosis and oxidative stress, primarily through the PI3K/AKT/NRF2 and SLC7A11/GPX4 signalling pathways, thus suggesting a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yanping Yin
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China; College of Life Science, Northwest University, Xi'an, 710069, China
| | - Fei Mu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lulu Zhang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jinyi Zhao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Rui Gong
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yanli Yin
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lingling Zheng
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yang Du
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Fuxing Jin
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jingwen Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
40
|
Gu M, Sun L, Yang J, Wang K, Wu F, Zheng L, Shen X, Lai X, Gong L, Peng Y, Xu S, Yang J, Yang C. Ditan Decoction ameliorates vascular dementia-induced cognitive dysfunction through anti-ferroptosis via the HIF1α pathway: Integrating network pharmacology and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119459. [PMID: 39978449 DOI: 10.1016/j.jep.2025.119459] [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: 11/08/2024] [Revised: 01/06/2025] [Accepted: 02/06/2025] [Indexed: 02/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Vascular dementia (VaD) represents a frequently seen cognitive dysfunction syndrome and has ranks second among dementia subtypes following Alzheimer's disease. At present, Ditan Decoction (DTD), the traditional Chinese herbal prescription, is clinically applied in treating VaD. However, the material basis of its efficacy and therapeutic mechanism still remain unknown. AIM OF THE STUDY This experiment investigated the protection induced by DTD against VaD and the associated mechanism through network pharmacology, mass spectrometry analysis, and in vivo validation. MATERIALS AND METHODS We induced VaD in a rat model using bilateral common carotid artery ligation method (2-VO) and administered DTD at doses of 2.14, 4.28 and 8.55 g/kg, with Memantine (0.9 mg/kg) being the positive control. Following oral administration with DTD or Memantine for 4 weeks, behavioral tests were used for assessing cognitive function. H&E and Nissl staining was used for evaluating hippocampal pathology. TEM was used to visualize the ultrastructure of the hippocampal tissue. ELISA was carried out for measuring inflammatory factor levels in rat serum, and biochemical assays were employed to assess oxidative stress levels. Ferroptosis in the hippocampus was examined through analyzing corresponding biomarkers and protein expression. Additionally, HPLC-Q-Exactive-MS technology was employed for identifying DTD components, whereas network pharmacology was conducted for predicting DTD's targets for treating VaD. HIF1α expression levels were assessed by Western blotting and immunofluorescence. We also further validated whether the protective effects of DTD on VaD were mediated through the HIF1α-regulated ferroptosis signaling pathway by using an HIF1α inhibitor in rats. RESULTS DTD demonstrated protective effects against 2VO-induced hippocampal injury through alleviating oxidative stress, lowering systemic inflammation, while preventing ferroptosis of hippocampal tissue. As revealed by network pharmacology, DTD probably executes its function in VaD by activating HIF1α pathway. According to immunofluorescence and Western blotting, DTD activated HIF1α within hippocampal tissue. Furthermore, DTD's protection against VaD and ferroptosis was reversed when an HIF1α inhibitor was applied. CONCLUSION These findings suggested that DTD rescued cognitive dysfunction in VaD by inhibiting ferroptosis via activating HIF1α pathway.
Collapse
Affiliation(s)
- Mengyu Gu
- Hubei University of Chinese Medicine, Wuhan, 430065, China; Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Lieqian Sun
- Hubei University of Chinese Medicine, Wuhan, 430065, China; Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Jie Yang
- Hubei Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China; Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Kaiyi Wang
- Hubei University of Chinese Medicine, Wuhan, 430065, China; Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Fan Wu
- Hubei University of Chinese Medicine, Wuhan, 430065, China; Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Li Zheng
- Hubei University of Chinese Medicine, Wuhan, 430065, China; Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Xiangzhong Shen
- Hubei University of Chinese Medicine, Wuhan, 430065, China; Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Xing Lai
- Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Lili Gong
- Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Ying Peng
- Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Shujie Xu
- Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China
| | - Jia Yang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Chao Yang
- Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430022, China.
| |
Collapse
|
41
|
Ma J, Zhong X, Li Z, Jiang Y, Jiang Y, Liu X, Hu Y, Yang Z, Zhai G. Di-Dang-Tang suppresses ferroptosis in the hippocampal CA1 region by targeting PGK1/NRF2/GPX4 signaling pathway to exert neuroprotection in vascular dementia. Int Immunopharmacol 2025; 150:114233. [PMID: 39946772 DOI: 10.1016/j.intimp.2025.114233] [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: 10/29/2024] [Revised: 01/27/2025] [Accepted: 02/03/2025] [Indexed: 03/03/2025]
Abstract
Increasing evidence has emphasized the crucial role of ferroptosis in the pathogenesis of Vascular dementia (VaD). Di-Dang-Tang (DDT) has the effects of removing blood stasis according to the theory of Traditional Chinese medicine (TCM), while its effects on ferroptosis and mechanisms remain unclear. To elucidate whether the neuroprotective effect of DDT treatment is associated with ferroptosis mediated by the Phosphoglycerate kinase 1 (PGK1)/ Nuclear Factor Erythroid 2-related factor (NRF2)/ Glutathione Peroxidase 4 (GPX4) signaling pathway in the hippocampal CA1 region of rats with the 2-vessel occlusion (2VO) model, we conducted a series of experiments. Nissl staining, HE staining and FJB staining were used to assess the effects of DDT on the degeneration and apoptosis of neurons in the CA1 region of the hippocampus. DDT's suppression on ferroptosis and its protective effects were also evaluated by ELISA and DHE fluorescence. Immunofluorescence assay, immunohistochemistry examination, and western blot analysis further validated DDT's regulatory effects on ferroptosis via PGK1/NRF2/GPX4 pathway. Additionally, we explored the key mediating role of PGK1 in the DDT treatment of VaD by overexpressing PGK1 using AAV-OE-PGK1 plasmid injection. DDT significantly attenuated neuronal apoptosis and degeneration in CA1 region and ameliorated cognitive dysfunctions in VaD rats. DDT inhibited ferroptosis in this brain region, as evidenced by an up-regulation of GPX4 and SLC7A11, and a decline in ferroptosis-related indices. Further, DDT activated protein expression of the PGK1/NRF2/GPX4 pathway, alleviating the lipid peroxidation. Notably, the inhibition of ferroptosis by DDT was achieved by suppression of the PGK1 axis signaling pathway.
Collapse
MESH Headings
- Animals
- Ferroptosis/drug effects
- NF-E2-Related Factor 2/metabolism
- Male
- Signal Transduction/drug effects
- Phosphoglycerate Kinase/metabolism
- Phosphoglycerate Kinase/genetics
- Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism
- Phospholipid Hydroperoxide Glutathione Peroxidase/genetics
- Dementia, Vascular/drug therapy
- Dementia, Vascular/metabolism
- Dementia, Vascular/pathology
- Rats
- Drugs, Chinese Herbal/therapeutic use
- Drugs, Chinese Herbal/pharmacology
- Neuroprotective Agents/pharmacology
- Neuroprotective Agents/therapeutic use
- Rats, Sprague-Dawley
- CA1 Region, Hippocampal/pathology
- CA1 Region, Hippocampal/drug effects
- CA1 Region, Hippocampal/metabolism
- Disease Models, Animal
- Neurons/drug effects
- Neurons/metabolism
- Neurons/pathology
Collapse
Affiliation(s)
- Junjie Ma
- School of Intergrative Medicine, Nanjing University of Chinese Medicine, No. 138, Xianlin Road, Qixia District, Nanjing City, Jiangsu 210000, China
| | - Xinxin Zhong
- School of Intergrative Medicine, Nanjing University of Chinese Medicine, No. 138, Xianlin Road, Qixia District, Nanjing City, Jiangsu 210000, China
| | - Zhiyuan Li
- School of Intergrative Medicine, Nanjing University of Chinese Medicine, No. 138, Xianlin Road, Qixia District, Nanjing City, Jiangsu 210000, China
| | - Yongxia Jiang
- Lianyungang Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, No. 160, Chaoyang Middle Road, Haizhou District, Lianyungang City, Jiangsu 222004, China
| | - Yongqu Jiang
- Lianyungang Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, No. 160, Chaoyang Middle Road, Haizhou District, Lianyungang City, Jiangsu 222004, China
| | - Xiaoli Liu
- Lianyungang Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, No. 160, Chaoyang Middle Road, Haizhou District, Lianyungang City, Jiangsu 222004, China
| | - Yue Hu
- School of Intergrative Medicine, Nanjing University of Chinese Medicine, No. 138, Xianlin Road, Qixia District, Nanjing City, Jiangsu 210000, China
| | - Zhou Yang
- Lianyungang Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, No. 160, Chaoyang Middle Road, Haizhou District, Lianyungang City, Jiangsu 222004, China
| | - Guojie Zhai
- Department of Neurology, Suzhou Ninth People's Hospital, Suzhou Ninth Hospital Affiliated to Soochow University, No. 2666, Ludang Road, Wujiang District, Suzhou City, Jiangsu 215200, China.
| |
Collapse
|
42
|
Cai Z, Zhang Y, He L, Cui M, Zhang W, E L, Yang H, Ling Q, Hoffmann PR, He J, Gou S, Liu F, Huang Z. Methylseleninic Acid Elevating the Nrf2-GPX4 Axis Relieves Endothelial Dysfunction and Ferroptosis Induced by Arsenic Exposure. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:7445-7455. [PMID: 40071728 DOI: 10.1021/acs.jafc.4c12515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Chronic exposure to arsenic (As), a ubiquitous contaminant, poses deleterious health risks to humans, including cardiovascular disease. Recent studies have implicated ferroptosis, in which the essential micronutrient selenium (Se) plays a crucial role, in several As-induced pathological processes. However, whether Se can counteract As-induced endothelial dysfunction through ferroptosis remains unclear. Herein, methylseleninic acid (MSA), a methylselenium metabolite, was used as a Se supplement to investigate the underlying effect and mechanism of Se in As-induced endothelial dysfunction involving ferroptosis in vivo and in vitro. As exposure induced endothelial dysfunction in mice, as indicated by increased aortic permeability, increased number of circulating endothelial cells, and endothelial mitochondria exhibiting ferroptosis-related alterations. Additionally, As induced ferroptosis-related cell death in mouse aortic endothelial cells, accompanied by impaired redox homeostasis, relatively low Se status, and decreased expressions of selenoproteome, including GPX4. Notably, these were attenuated by MSA via activation of Nrf2 and upregulation of three GPX4 isoforms, which were further abrogated by the Nrf2 antagonist ML385. Finally, MSA exhibited ameliorative effects on endothelial ferroptosis and dysfunction in the aortas of As-exposed mice. These results demonstrate that As causes endothelial ferroptosis and dysfunction by affecting the Se-Nrf2/GPX4 axis, which can be relieved by MSA. This study provides novel evidence implicating Se in As-induced endothelial dysfunction by mitigating ferroptosis.
Collapse
Affiliation(s)
- Zhihui Cai
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Yutian Zhang
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Leting He
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Miao Cui
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Weijie Zhang
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Lingling E
- Institute of Stomatology & Oral Maxilla Facial Key Laboratory, First Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Hui Yang
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Qinjie Ling
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Peter R Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii 96813, United States
| | - Jingjun He
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Shan Gou
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Fei Liu
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
| | - Zhi Huang
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine; Department of Bioscience and Biotechnology of the College of Life Science and Technology, Jinan University, Guangzhou 510317, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong 510632, China
| |
Collapse
|
43
|
Yu D, Hu Y, Ma M, Li W, Zhao X. The landscape of research on ferroptosis under hypoxic conditions: a bibliometric analysis. Front Pharmacol 2025; 16:1519000. [PMID: 40206079 PMCID: PMC11979267 DOI: 10.3389/fphar.2025.1519000] [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: 10/29/2024] [Accepted: 03/04/2025] [Indexed: 04/11/2025] Open
Abstract
Background Ferroptosis is a newly identified type of iron-dependent cell death that characterized by an increase in intracellular iron ions, which disrupt the balance of the cellular lipid peroxidation system, causing lipid peroxidation and ultimately resulting in cell death. Interestingly, ferroptosis is modulated by hypoxia and plays a role in hypoxia-related diseases. Therefore, we performed a bibliometric review of the Web of Science Core Collection (WoSCC) database to investigate the link between ferroptosis and hypoxia from January 2013 to December 2023. Method The core collection within the Web of Science bibliographic index was consulted to extract relevant articles and reviews. Data on publications, countries, institutions, authors, journals, citations, and keywords in the included studies were systematically analyzed using Microsoft Excel 2019 and CiteSpace 6.3.R1 software. Result A comprehensive analysis and visualization of 472 research papers on ferroptosis under hypoxic conditions published between 2013 and 2023 revealed emerging research hotspots and trends. Initially, a scarcity of studies existed in this field. However, this was succeeded by a significant increase in research interest in subsequent years, culminating in a peak of 204 publications in 2023. Research in this field focused primarily on the Asian region. Notably, research hotspots include diseases related to hypoxia, treatment therapy and pathogenesis. Among the researchers in this field, Supuran emerged as the most prolific author. Wuhan University was the leading institution in terms of research output, and China was the most prolific country in this area of study. Among the top ten journals ranked by the number of publications, nine were classified as Q1, indicating the high level of credibility of these studies. The research conducted by Stockwell et al., featured in the journal "Cell," currently has the most citations. Present scholarly pursuits are primarily focused on comprehending the mechanisms through which interventions affect hypoxia-related diseases through the ferroptosis pathway, as well as on probing and pinpointing prospective treatment targets. Conclusion This study highlights key areas of interest and emerging trends in ferroptosis research in the presence of hypoxic conditions, thus providing valuable insights for future directions of exploration for the diagnosis and treatment of hypoxia-related diseases.
Collapse
Affiliation(s)
- Di Yu
- Department of Basic Medical Sciences, Qinghai Unversity Medical College, Xining, Qinghai, China
| | - Yibo Hu
- Department of Orthopaedic Trauma, The Affiliated Hospital of Qinghai University, Xining, Qinghai, China
| | - Meijuan Ma
- Department of Basic Medical Sciences, Qinghai Unversity Medical College, Xining, Qinghai, China
| | - Wenjia Li
- Department of Basic Medical Sciences, Qinghai Unversity Medical College, Xining, Qinghai, China
| | - Xiaohui Zhao
- Department of Basic Medical Sciences, Qinghai Unversity Medical College, Xining, Qinghai, China
| |
Collapse
|
44
|
Liu Y, Fu Z, Wang X, Yang Q, Liu S, Zhu D. Metformin attenuates diabetic osteoporosis by suppressing ferroptosis via the AMPK/Nrf2 pathway. Front Pharmacol 2025; 16:1527316. [PMID: 40206070 PMCID: PMC11979264 DOI: 10.3389/fphar.2025.1527316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/17/2025] [Indexed: 04/11/2025] Open
Abstract
Background Ferroptosis is a critical factor in the impairment of osteoblast function in osteoporosis. Metformin (Met), a biguanide antidiabetic drug, has demonstrated anti-osteoporotic effects and has been confirmed to exert therapeutic benefits in diabetic osteoporosis (DOP). Nevertheless, the underlying mechanisms through which Met affects bone metabolism remain ambiguous. Objective This study seeks to elucidate the function of Met in DOP and to explore the potential mechanisms through which it mediates treatment effects. Methods In vitro, we utilized osteoblasts to explore the impact of Met on osteoblast differentiation and anti-ferroptosis in a high glucose and palmitic acid (HGHF) environment. In vivo, we developed a DOP model utilizing a high-fat diet along with streptozocin injections and evaluated the bone-protective effects of Met through micro-CT and histomorphological analyses. Results Met inhibits HGHF-induced ferroptosis in osteoblasts, as indicated by the elevation of ferroptosis-protective proteins (GPX4, FTH1, and SLAC7A11), along with decreased lipid peroxidation and ferrous ion levels. Furthermore, Met augmented the levels of osteogenic markers (RUNX2 and COL1A1) and enhanced alkaline phosphatase activity in osteoblasts under HGHF conditions. Mechanistic investigations revealed that Met activates the AMPK/Nrf2 pathway, effectively preventing ferroptosis progression. Additionally, in vivo results demonstrated Met alleviates bone loss and microstructural deterioration in DOP rats. Conclusion Met can activate the AMPK/Nrf2 pathway to prevent ferroptosis, thereby protecting against DOP.
Collapse
Affiliation(s)
- Yanwei Liu
- Department of Orthopaedics, The First Hospital of Jilin University, Changchun, China
| | - Zhaoyu Fu
- Department of Emergency Surgery, The First Affiliated Hospital of Zhengzhou University, Henan Medical Key Laboratory of Emergency and Trauma Research, Zhengzhou, Henan, China
| | - Xinyu Wang
- Department of Orthopaedics, The First Hospital of Jilin University, Changchun, China
| | - Qifan Yang
- Department of Orthopaedics, The First Hospital of Jilin University, Changchun, China
| | - Shun Liu
- Department of Orthopaedics, The First Hospital of Jilin University, Changchun, China
| | - Dong Zhu
- Department of Orthopaedics, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
45
|
Li H, Li JX, Zeng YD, Zheng CX, Dai SS, Yi J, Song XD, Liu T, Liu WH. Luteolin ameliorates ischemic/reperfusion injury by inhibiting ferroptosis. Metab Brain Dis 2025; 40:159. [PMID: 40138029 DOI: 10.1007/s11011-025-01588-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 03/13/2025] [Indexed: 03/29/2025]
Abstract
Ischaemic stroke is a large disease burden worldwide. Thrombolysis and thrombectomy are the main treatment methods for cerebral ischemia-reperfusion (I/R) injury. Luteolin, as a flavonoid compound, has an antagonistic effect on inflammation, oxidative stress, and tumorigenesis in disease. Therefore, the primary objective of this study is to determine the role of luteolin in cerebral I/R injury. Oxygen glucose deprivation/reoxygenation (OGD/R)-treated BV2 cells and a cerebral I/R rat model were established. Cell viability and death were verified using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and propidium iodide staining. The glutathione/oxidized glutathione (GSH/GSSG) ratio, superoxide dismutase (SOD) activity, and reactive oxygen species (ROS) and malondialdehyde (MDA) levels were determined using corresponding kits. Solute Carrier Family 7 Member 11 (SLC7A11), nuclear transcription factor erythroid 2-related factor 2 (NRF2), and glutathione peroxidase 4 (GPX4) levels were analyzed by western blotting. In addition, the infarct volume of brain tissues was examined by tetrazolium chloride (TTC) staining. Luteolin treatment significantly enhanced cell viability, decreased LDH release and intracellular ROS and MDA levels, and increased the GSH/GSSG ratio and SOD activity in OGD/R-treated BV2 cells. PI staining demonstrated that cell death was inhibited after luteolin treatment. Additionally, luteolin treatment significantly increased the SLC7A11, NRF2, and GPX4 protein levels. After treatment with ML385, an NRF2 inhibitor, the influence of luteolin on OGD/R-treated BV2 cells was reversed. Moreover, luteolin treatment significantly decreased the neurological score and infarct area in the brain tissues of cerebral I/R rats. Our research demonstrated that luteolin treatment inhibited ferroptosis by enhancing antioxidant functions through the NRF2 pathway, which provides a promising method for treating cerebral I/R injury.
Collapse
Affiliation(s)
- Hua Li
- Hunan University of Traditional Chinese Medicine Cultivates World First-class Disciplines of Traditional Chinese Medicine, Changsha, Hunan, 410208, China
- The Key Laboratory of Diagnostics of TCM in Hunan Province, Changsha, Hunan, 410208, China
- Key Laboratory of TCM Heart and Lung Syndrome Differentiation & Medicated Diet and Dietotherapy, Changsha, Hunan, 410208, China
| | - Jin-Xia Li
- Hunan University of Traditional Chinese Medicine Cultivates World First-class Disciplines of Traditional Chinese Medicine, Changsha, Hunan, 410208, China
- The Key Laboratory of Diagnostics of TCM in Hunan Province, Changsha, Hunan, 410208, China
| | - Yi-di Zeng
- Hunan University of Traditional Chinese Medicine Cultivates World First-class Disciplines of Traditional Chinese Medicine, Changsha, Hunan, 410208, China
- The Key Laboratory of Diagnostics of TCM in Hunan Province, Changsha, Hunan, 410208, China
| | - Cai-Xing Zheng
- Hunan University of Traditional Chinese Medicine Cultivates World First-class Disciplines of Traditional Chinese Medicine, Changsha, Hunan, 410208, China
- The Key Laboratory of Diagnostics of TCM in Hunan Province, Changsha, Hunan, 410208, China
| | - Si-Si Dai
- Hunan University of Traditional Chinese Medicine Cultivates World First-class Disciplines of Traditional Chinese Medicine, Changsha, Hunan, 410208, China
- The Key Laboratory of Diagnostics of TCM in Hunan Province, Changsha, Hunan, 410208, China
| | - Jian Yi
- Hunan University of Traditional Chinese Medicine Cultivates World First-class Disciplines of Traditional Chinese Medicine, Changsha, Hunan, 410208, China
| | - Xu-Dong Song
- Hunan University of Traditional Chinese Medicine Cultivates World First-class Disciplines of Traditional Chinese Medicine, Changsha, Hunan, 410208, China
- The Key Laboratory of Diagnostics of TCM in Hunan Province, Changsha, Hunan, 410208, China
| | - Ting Liu
- Hunan University of Traditional Chinese Medicine Cultivates World First-class Disciplines of Traditional Chinese Medicine, Changsha, Hunan, 410208, China
- The Key Laboratory of Diagnostics of TCM in Hunan Province, Changsha, Hunan, 410208, China
| | - Wang-Hua Liu
- Hunan University of Traditional Chinese Medicine Cultivates World First-class Disciplines of Traditional Chinese Medicine, Changsha, Hunan, 410208, China.
- The Key Laboratory of Diagnostics of TCM in Hunan Province, Changsha, Hunan, 410208, China.
- Key Laboratory of TCM Heart and Lung Syndrome Differentiation & Medicated Diet and Dietotherapy, Changsha, Hunan, 410208, China.
- Hunan Engineering Technology Research Center For Medicinal and Functional Food, Hunan University of Traditional Chinese Medicine, XueShi Road, Changsha, Hunan, 410208, China.
| |
Collapse
|
46
|
Li H, Li G, Gao Y, Ma Y, Yu Z, Zhang A, Yang G, Hou Z, Zhang Y, Yu Y, Zhang Z. Oligo-peptide I-C-F-6 mitigates polymicrobial sepsis-induced cardiac dysfunction in mice. Eur J Pharmacol 2025; 996:177545. [PMID: 40139420 DOI: 10.1016/j.ejphar.2025.177545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/04/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Cardiomyopathy accounts for worse clinical outcome and higher mortality rate during sepsis globally. Here we assessed whether post-operative administration of I-C-F-6, a small molecule oligo-peptide (Gly-Ala-Gly-Pro-His-Gly-Gly) derived from Carapax trionycis, protected against septic cardiomyopathy in mice. Male adult mice were exposed to cecal ligation and puncture (CLP) and I-C-F-6 was administered intravenously (0.4 mg/kg or 4.0 mg/kg) 30 min following surgery. Administration of I-C-F-6 extended survival period and decreased sepsis severity score in septic mice. Furthermore, administration of I-C-F-6 mitigated cardiac atrophy and preserved cardiac function in septic mice. Mechanistically, I-C-F-6 inhibited inflammation and promoted M2 polarization in myocardium of septic mice. In addition, I-C-F-6 activated nuclear factor erythroid 2-related factor 2 (Nrf2)/haem oxygenase-1 (HO-1)/glutathione peroxidase 4 (GPX4) pathway, mitigated oxidative damage and inhibited ferroptosis in myocardium of septic mice. In conclusion, post-operative administration of I-C-F-6 in mice exposed to CLP improved survival and mitigated myocardial impairment. Our work established a clear therapeutic potential of I-C-F-6 for sepsis-induced cardiomyopathy.
Collapse
Affiliation(s)
- Hongxiao Li
- Department of Cardiology, Shanghai East Hospital, Tongji University, Jimo Road 150, Shanghai, 200120, China
| | - Guang Li
- Department of Emergency Trauma Surgery, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Yuan Gao
- School of Medicine, Shanghai University, Shangda Road 99, Shanghai, 200444, China
| | - Yulin Ma
- School of Medicine, Shanghai University, Shangda Road 99, Shanghai, 200444, China
| | - Zixuan Yu
- School of Medicine, Shanghai University, Shangda Road 99, Shanghai, 200444, China
| | - Anna Zhang
- School of Medicine, Shanghai University, Shangda Road 99, Shanghai, 200444, China
| | - Guoling Yang
- School of Medicine, Shanghai University, Shangda Road 99, Shanghai, 200444, China
| | - Zhiqi Hou
- School of Medicine, Shanghai University, Shangda Road 99, Shanghai, 200444, China
| | - Yuefan Zhang
- School of Medicine, Shanghai University, Shangda Road 99, Shanghai, 200444, China
| | - Yongsheng Yu
- School of Medicine, Shanghai University, Shangda Road 99, Shanghai, 200444, China.
| | - Zhigang Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University, Jimo Road 150, Shanghai, 200120, China.
| |
Collapse
|
47
|
Li Q, Yang X, Li T. Natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in central nervous system diseases: current preclinical evidence and future perspectives. Front Pharmacol 2025; 16:1570069. [PMID: 40196367 PMCID: PMC11973303 DOI: 10.3389/fphar.2025.1570069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 02/24/2025] [Indexed: 04/09/2025] Open
Abstract
Flavonoids are a class of important polyphenolic compounds, renowned for their antioxidant properties. However, recent studies have uncovered an additional function of these natural flavonoids: their ability to inhibit ferroptosis. Ferroptosis is a key mechanism driving cell death in central nervous system (CNS) diseases, including both acute injuries and chronic neurodegenerative disorders, characterized by iron overload-induced lipid peroxidation and dysfunction of the antioxidant defense system. This review discusses the therapeutic potential of natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in CNS diseases, focusing on their molecular mechanisms, summarizing findings from preclinical animal models, and providing insights for clinical translation. We specifically highlight natural flavonoids such as Baicalin, Baicalein, Chrysin, Vitexin, Galangin, Quercetin, Isoquercetin, Eriodictyol, Proanthocyanidin, (-)-epigallocatechin-3-gallate, Dihydromyricetin, Soybean Isoflavones, Calycosin, Icariside II, and Safflower Yellow, which have shown promising results in animal models of acute CNS injuries, including ischemic stroke, cerebral ischemia-reperfusion injury, intracerebral hemorrhage, subarachnoid hemorrhage, traumatic brain injury, and spinal cord injury. Among these, Baicalin and its precursor Baicalein stand out due to extensive research and favorable outcomes in acute injury models. Mechanistically, these flavonoids not only regulate the Nrf2/ARE pathway and activate GPX4/GSH-related antioxidant pathways but also modulate iron metabolism proteins, thereby alleviating iron overload and inhibiting ferroptosis. While flavonoids show promise as ferroptosis inhibitors for CNS diseases, especially in acute injury settings, further studies are needed to evaluate their efficacy, safety, pharmacokinetics, and blood-brain barrier penetration for clinical application.
Collapse
Affiliation(s)
- Qiuhe Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaohang Yang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Tiegang Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
48
|
Shi J, Cheng Y, Wang L, Xing W, Li Y, Sun X, Lv Y, Zhang Y, Li Y, Zhao W. SR-B1 deficiency suppresses progression in acute myeloid leukemia via ferroptosis and reverses resistance to venetoclax. Free Radic Biol Med 2025; 233:24-38. [PMID: 40122151 DOI: 10.1016/j.freeradbiomed.2025.03.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Increase of immature myeloid cells in the bone marrow drives the development of acute myeloid leukemia (AML). The study aimed to clarify the biological function and regulatory mechanism of scavenger receptor class B type 1 (SR-B1) in AML, mainly its effect on ferroptosis and the influences on leukemogenesis and resistance to venetoclax. In this study, we found that the SR-B1 deficiency directly reduced the invasion and promoted death of malignant cells in AML. Strikingly, SR-B1 deficiency could up-regulated the expression of ferroptosis-related proteins to facilitate the occurrence of ferroptosis in vivo, and could also down-regulated the expression of apoptosis related protein B-cell lymphoma-2 (BCL-2). And then, we confirmed SR-B1 inhibitor block lipid transport-1 (BLT-1) had a superior efficacy in AML cells and AML model mice. The study found that whether SR-B1 deficiency or BLT-1 treatment could cause iron deposition and the accumulation of lipid peroxides in vivo, thereby suppressing leukemogenesis through ferroptosis. Critically, we found that SR-B1 inhibitor BLT-1 could reverse drug-resistance of venetoclax to promote AML cells death via ferroptosis. Our finding identified that SR-B1 as a critical regulator of the proliferation in AML which could provide a promising therapeutic strategy against malignant myeloid leukemia cells and drug-resistance.
Collapse
Affiliation(s)
- Junfeng Shi
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yifeng Cheng
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Lixue Wang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China
| | - Wen Xing
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yudi Li
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xiulin Sun
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yunpeng Lv
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yichuan Zhang
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yanming Li
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Wenhua Zhao
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
49
|
Wang Z, Zhu Y, Yao Y, Zhang W, Wang B, Wang J, Yang Y, Liu L. Natural products targeting regulated cell deaths for adriamycin-induced cardiotoxicity. Cell Death Discov 2025; 11:112. [PMID: 40118839 PMCID: PMC11928682 DOI: 10.1038/s41420-025-02389-w] [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: 10/04/2024] [Revised: 02/01/2025] [Accepted: 03/06/2025] [Indexed: 03/24/2025] Open
Abstract
Adriamycin (ADR), as an anti-cancer drug in routine clinical application, is utilized to treat various cancers such as ovarian cancer, hematological malignant tumor, and endometrial carcinoma. However, its serious dose-dependent cardiotoxicity extremely limits its clinical application. Currently, there remains a dearth of therapeutic agents to mitigate ADR-induced cardiotoxicity. Extensive research has demonstrated that ADR can simultaneously trigger various regulated cell death (RCD) pathways, such as apoptosis, autophagy, ferroptosis, necroptosis, and pyroptosis. Therefore, drugs targeting these RCD pathways may represent effective strategies for treating ADR-induced cardiotoxicity. Natural products, with their wide availability, low cost, and diverse pharmacological activities, have increasingly gained attention. Various natural products, including polyphenols, flavonoids, terpenoids, and alkaloids, can target the RCD pathways involved in ADR-induced cardiotoxicity. Furthermore, these natural products have exhibited excellent properties in preclinical studies or in vitro experiments. This review summarizes the mechanisms of RCD in ADR-induced cardiotoxicity and systematically reviews the natural products targeting these RCD pathways. Finally, we propose future research directions of natural products in this field.
Collapse
Affiliation(s)
- Zheng Wang
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, 627 Wuluo Road, Wuhan, 430070, China
| | - Yanli Zhu
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yu Yao
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Wenyu Zhang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Bo Wang
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Jing Wang
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Yang Yang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Liwen Liu
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
50
|
Liu C, Deng Y, Huang L, Nie X, Jiang Y, Zhang X, Zhang H. USP5 Suppresses Ferroptosis in Bladder Cancer Through Stabilization of GPX4. Curr Issues Mol Biol 2025; 47:211. [PMID: 40136465 PMCID: PMC11941033 DOI: 10.3390/cimb47030211] [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: 02/28/2025] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
USP5 has been proven to play an important role in the proliferation of bladder cancer (BC). In this study, we focused on investigating the molecular mechanism of ferroptosis induced by USP5 in bladder cancer. The role of USP5 in bladder cancer was evaluated using T24 wild-type cells (WT) and USP5 knockout (USP5-/-) by CCK8 and colony formation assays. The contents of ferrobivalent ions (Fe2+), reactive oxygen species (ROS), and malondialdehyde (MDA) were detected using a determination kit to observe the relationship between USP5 and ferroptosis. Furthermore, the molecular mechanism study was evaluated by employing Western blotting, co-immunoprecipitation, RT-qPCR, ubiquitination assays, etc. This study showed genetic ablation of USP5 significantly inhibited the viability and proliferation of bladder cancer cells. Genetic ablation of USP5 promoted increases in Fe2+ content, ROS, and MDA levels. The addition of erastin significantly increased the viability and proliferation of T24 USP5-/- cells and significantly increased their ROS and MDA contents. We verified that USP5 deficiency led to a significant reduction in GPX4 protein levels and that the overexpression of USP5 could stabilize the GPX4 protein. Further studies showed that USP5 interacts with GPX4 and stabilizes GPX4 by inhibiting its ubiquitination These findings revealed USP5 inhibits ferroptosis in bladder cancer cells by stabilizing GPX4. The relationship between USP5 and ferroptosis could be a potential therapeutic target for bladder cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Xia Zhang
- School of Medical Technology and Translational Medicine, Hunan Normal University, 371 Tongzipo Road, Yuelu District, Changsha 410013, China; (C.L.); (Y.D.); (L.H.); (X.N.); (Y.J.)
| | - Huihui Zhang
- School of Medical Technology and Translational Medicine, Hunan Normal University, 371 Tongzipo Road, Yuelu District, Changsha 410013, China; (C.L.); (Y.D.); (L.H.); (X.N.); (Y.J.)
| |
Collapse
|