1
|
He Y, Lu J, Luo Y, Pang R, Hu X, Ding L, Xiao H, Wang Y, Wang W. Exploring the therapeutic mechanism of curcumin in spinal cord injury treatment based on network pharmacology, molecular dynamics simulation, and experimental validation. Front Chem 2025; 13:1568551. [PMID: 40224222 PMCID: PMC11985754 DOI: 10.3389/fchem.2025.1568551] [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: 01/30/2025] [Accepted: 03/07/2025] [Indexed: 04/15/2025] Open
Abstract
Introduction Curcumin, a natural active compound derived from plants, is widely used as a pigment across the globe. Research has demonstrated that curcumin possesses neuroprotective properties in spinal cord injuries (SCIs); however, its specific mechanisms of action remain unclear. This study aimed to elucidate the potential mechanisms underlying curcumin's therapeutic effects in SCI. Methods We screened the targets of curcumin in the treatment of spinal cord injury using network pharmacology across a variety of public databases. The interaction between the compound and the target was analyzed through bioinformatics analysis, molecular docking, and molecular dynamics simulation. Finally, the prediction results were verified by simulating spinal cord injury through oxygen-glucose deprivation (OGD) injury in PC12 cells. Results Initial screening indicated 13 core targets involved in mitigating SCI. Curcumin may regulate the HIF pathway, immune cells, inflammation, oxidative stress, and other processes. Matrix metalloproteinase-9 (MMP9), tumor necrosis factor (TNF), interleukin-1β (IL-1β), signal transducer and activator of transcription 3 (STAT3), and caspase 3 (CASP3) were identified as key targets of curcumin in SCI regulation. Molecular docking results demonstrated that curcumin exhibited favorable affinity with the core targets, with MMP9 showing the highest binding affinity (-8.76 kcal/mol). Further studies confirmed that curcumin stably binds with MMP9, and the binding site was located at residues 220-225. Cell counting kit-8 (CCK8) assay results showed that curcumin exerted a good therapeutic effect. Western blot results showed that curcumin inhibited the expression of MMP9 protein but had no significant effect on the expression of TNF-α. Conclusion Curcumin exerts its effects on SCI through multiple targets and pathways. Its specific mechanisms involve the inhibition of inflammation, prevention of apoptosis and ferroptosis, and promotion of neuronal repair. MMP9 may be a key target mediating curcumin's protective effects against SCI. These findings provide scientific evidence for further research and development of drugs.
Collapse
Affiliation(s)
- Yongzhi He
- School of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, Sichuan, China
- Department of Rehabilitation Medicine, Xichong County People’s Hospital, Nanchong, Sichuan, China
| | - Jiachun Lu
- Chengdu Eighth People’s Hospital (Geriatric Hospital of Chengdu Medical College), Chengdu, Sichuan, China
| | - Yushan Luo
- Department of Rehabilitation Medicine, Xichong County People’s Hospital, Nanchong, Sichuan, China
| | - Rizhao Pang
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Xiaoming Hu
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Lijuan Ding
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Hua Xiao
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Yunyun Wang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine(Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, , China
| | - Wenchun Wang
- School of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Zhou L, Li L, Yang J, Mansuer M, Deng X, Wang Y, Ren H, Cui D, Jiang Y, Gao L. TNFAIP3 affects ferroptosis after traumatic brain injury by affecting the deubiquitination and ubiquitination pathways of the HMOX1 protein and ACSL3. Free Radic Biol Med 2025; 228:221-239. [PMID: 39743027 DOI: 10.1016/j.freeradbiomed.2024.12.048] [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: 02/06/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/04/2025]
Abstract
The occurrence and progression of traumatic brain injury involve a complex process. The pathophysiological mechanisms triggered by neuronal damage include various forms of programmed cell death, including ferroptosis. We observed upregulation of TNFAIP3 in mice after traumatic brain injury. Overexpression of TNFAIP3 inhibits HT-22 proliferation and cell viability through ferroptosis. Mechanistically, TNFAIP3 interacts with the HMOX1 protein and promotes its stability through the deubiquitination pathway. Additionally, TNFAIP3 can enhance lipoperoxidation, mitochondrial damage, and neuronal cell death by promoting ACSL3 degradation via NEDD4-mediated ubiquitination. Mice injected with AAV-shTNFAIP3 exhibited reduced neuronal degeneration and improved motor and cognitive function following cortical impact injury. In conclusion, our findings demonstrate that TNFAIP3 deficiency inhibits neuronal cell ferroptosis and ameliorates cognitive impairment caused by traumatic brain injury and demonstrate its potential applicability in the treatment of traumatic brain injury.
Collapse
Affiliation(s)
- Lin Zhou
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Lei Li
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Jinghao Yang
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Maierdan Mansuer
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Xianyu Deng
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yida Wang
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Hui Ren
- Department of Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200435, China
| | - Daming Cui
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yang Jiang
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Liang Gao
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| |
Collapse
|
3
|
Shi C, Wang B, Zhai T, Zhang C, Ma J, Guo Y, Yang Y, Chen C, Gao J, Zhao L. Exploring Ubiquitination in Spinal Cord Injury Therapy: Multifaceted Targets and Promising Strategies. Neurochem Res 2025; 50:82. [PMID: 39833619 DOI: 10.1007/s11064-025-04332-y] [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: 10/18/2024] [Revised: 01/02/2025] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Spinal cord injury (SCI) is a severely debilitating neurological condition that often results in significant functional impairment and is associated with poor long-term prognosis. Edema, oxidative stress, inflammatory responses, and cell death are the primary factors contributing to secondary injury following spinal cord damage. Ubiquitination is a crucial intracellular mechanism for protein regulation that has garnered significant attention as a therapeutic target in a variety of diseases. Numerous studies have shown that ubiquitination plays a key role in modulating processes such as inflammatory responses, apoptosis, and nerve regeneration following SCI, thereby influencing injury repair. Accordingly, targeting ubiquitination has the potential for mitigating harmful inflammatory responses, inhibiting dysregulated programmed cell death, and protecting the integrity of the blood-spinal cord barrier, thereby providing a novel therapeutic strategy for SCI. In this review, we discuss the role of ubiquitination and its potential as a therapeutic target in SCI, aiming to offer a foundation for developing ubiquitination-targeted therapies for this condition.
Collapse
Affiliation(s)
- Caizhen Shi
- Medical School of Yan'an University, Yan'an, Shaanxi, China
| | - Bingbing Wang
- Medical School of Yan'an University, Yan'an, Shaanxi, China
| | - Tianyu Zhai
- Medical School of Yan'an University, Yan'an, Shaanxi, China
| | - Can Zhang
- Medical School of Yan'an University, Yan'an, Shaanxi, China
| | - Jiarui Ma
- Medical School of Yan'an University, Yan'an, Shaanxi, China
| | - Yanjie Guo
- Medical School of Yan'an University, Yan'an, Shaanxi, China
| | - Yanling Yang
- Medical School of Yan'an University, Yan'an, Shaanxi, China
| | - Chen Chen
- Yulin First People's Hospital, Yulin, Shaanxi, China
| | - Jianzhong Gao
- Yulin First People's Hospital, Yulin, Shaanxi, China.
| | - Lin Zhao
- Medical School of Yan'an University, Yan'an, Shaanxi, China.
| |
Collapse
|
4
|
Yang D, Zhang T, Qu H, Li S, Lu J, Cao W, Chen Z, Zhang H, Yang J, Wang J. Inhibition of ubiquitin-specific protease 7 ameliorates ferroptosis-mediated myocardial infarction by contrasting oxidative stress: An in vitro and in vivo analysis. Cell Signal 2024; 124:111423. [PMID: 39304097 DOI: 10.1016/j.cellsig.2024.111423] [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: 08/20/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Our prior research determined that USP7 exacerbates myocardial injury. Additionally, existing studies indicate a strong connection between USP7 and ferroptosis. However, the influence of USP7 on ferroptosis-mediated myocardial infarction (MI) remains unclear. Given these findings, we are particularly interested in USP7's regulatory role in ferroptosis-mediated MI and its underlying mechanisms. METHODS In this study, we established MI models and lentivirus-transfected groups to inhibit USP7 expression both in vivo and in vitro. Cardiac function was detected with Echocardiography. TTC and HE staining were employed to assess myocardial alterations. The expression of ferroptosis markers (PTGS2, ACSL4, GPX4) were analyzed by RT-qPCR and Western blotting. Flow cytometry and ELISA were used for measuring Fe2+, lipid ROS, GSH, and GSSG levels. TEM and Prussian blue staining were used to observe mitochondrial alterations and iron deposition. RT-qPCR, Western blotting, and immunofluorescence were conducted to analyze Keap1, Nrf2, and nuclear Nrf2 expression in vitro and in vivo. RESULTS In the MI model group, USP7 expression significantly increased, worsening ferroptosis-mediated MI. Conversely, in the USP7-inhibited group, activation of the Keap1-Nrf2 signaling pathway improved ferroptosis-mediated MI outcomes. In vitro, the MI model exhibited a marked decline in cardiomyocyte viability and notable mitochondrial damage. However, these issues improved in the USP7-inhibited groups. In vivo, USP7 intensified MI and iron deposition within the MI model group, with decreased values of LVEF, LVFS, SV, LVAWd, and LVPWs, all of which showed improvement in the USP7-inhibited group, except for LVPWd and LVPWs, which showed no significant variation. Importantly, both the in vitro and in vivo experiments revealed analogous results: a reduction in Keap1 expression and an increase in both Nrf2 and nuclear Nrf2 post USP7 inhibition. Additionally, GPX4 expression decreased while PTGS2 and ACSL4 expressions increased. Notably, concentrations of Fe2+, lipid ROS, GSH, and GSSG significantly decreased. CONCLUSION In vitro and in vivo studies have found that inhibition of USP7 attenuates iron deposition and suppresses oxidative stress, resulting in amelioration of ferroptosis-induced MI.
Collapse
Affiliation(s)
- Dong Yang
- Yan'an Hospital Affiliated To Kunming Medical University, Kunming, China.
| | - Tiling Zhang
- Yan'an Hospital Affiliated To Kunming Medical University, Kunming, China
| | - Hai Qu
- Yan'an Hospital Affiliated To Kunming Medical University, Kunming, China
| | - Shaolong Li
- Yan'an Hospital Affiliated To Kunming Medical University, Kunming, China
| | - Jing Lu
- Yan'an Hospital Affiliated To Kunming Medical University, Kunming, China
| | - Wanyan Cao
- Yan'an Hospital Affiliated To Kunming Medical University, Kunming, China
| | - Zhipeng Chen
- Yan'an Hospital Affiliated To Kunming Medical University, Kunming, China
| | - Han Zhang
- Yan'an Hospital Affiliated To Kunming Medical University, Kunming, China
| | - Jing Yang
- Yan'an Hospital Affiliated To Kunming Medical University, Kunming, China
| | - Jin Wang
- Yan'an Hospital Affiliated To Kunming Medical University, Kunming, China.
| |
Collapse
|
5
|
Wang L, Zhang Y, Yu T, Wu H. The Role and Mechanism of Deubiquitinase USP7 in Tumor-Associated Inflammation. Biomedicines 2024; 12:2734. [PMID: 39767641 PMCID: PMC11726842 DOI: 10.3390/biomedicines12122734] [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: 10/18/2024] [Revised: 11/19/2024] [Accepted: 11/27/2024] [Indexed: 01/16/2025] Open
Abstract
Deubiquitinating enzymes are a class of proteases that remove ubiquitin tags from proteins, thereby controlling protein stability and function. Tumor inflammation arises from interactions between tumor cells and their microenvironment, which trigger an inflammatory response. The deubiquitinating enzyme USP7 plays a central role in this process. Research suggests that USP7 may modulate various signaling pathways related to inflammatory responses through its deubiquitinating activity, thereby influencing tumor development and progression, including regulating T cell immune activity, improving macrophage anti-tumor activity, and regulating NF-κB signal pathways. Overall, describing the role and mechanism of USP7 in the tumor inflammatory response is of great importance for elucidating the regulatory mechanism of tumor inflammation and developing new therapeutic strategies. This article mainly reviews the structure, function, role, and mechanism of USP7 in the tumor inflammation response.
Collapse
Affiliation(s)
- Luhong Wang
- Cancer Hospital Affiliated to Dalian University of Technology, Shenyang 110042, China; (L.W.); (Y.Z.)
- Dalian Key Laboratory of Protein Modification and Disease, Faculty of Medicine, School of Biological Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yong Zhang
- Cancer Hospital Affiliated to Dalian University of Technology, Shenyang 110042, China; (L.W.); (Y.Z.)
| | - Tao Yu
- Cancer Hospital Affiliated to Dalian University of Technology, Shenyang 110042, China; (L.W.); (Y.Z.)
| | - Huijian Wu
- Dalian Key Laboratory of Protein Modification and Disease, Faculty of Medicine, School of Biological Engineering, Dalian University of Technology, Dalian 116024, China
| |
Collapse
|
6
|
Zhu L, Zhang J, Fan W, Su C, Jin Z. Identification of iron metabolism-related genes in coronary heart disease and construction of a diagnostic model. Front Cardiovasc Med 2024; 11:1409605. [PMID: 39610972 PMCID: PMC11602506 DOI: 10.3389/fcvm.2024.1409605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 10/31/2024] [Indexed: 11/30/2024] Open
Abstract
Background Coronary heart disease is a common cardiovascular disease, yferroptosiset its relationship with iron metabolism remains unclear. Methods Gene expression data from peripheral blood samples of patients with coronary heart disease and a healthy control group were utilized for a comprehensive analysis that included differential expression analysis, weighted gene co-expression network analysis, gene enrichment analysis, and the development of a logistic regression model to investigate the associations and differences between the groups. Additionally, the CIBERSORT algorithm was employed to examine the composition of immune cell types within the samples. Results Eight central genes were identified as being both differentially expressed and related to iron metabolism. These central genes are mainly involved in the cellular stress response. A logistic regression model based on the central genes achieved an AUC of 0.64-0.65 in the diagnosis of coronary heart disease. A higher proportion of M0 macrophages was found in patients with coronary heart disease, while a higher proportion of CD8T cells was observed in the normal control group. Conclusion The study identified important genes related to iron metabolism in the pathogenesis of coronary heart disease and constructed a robust diagnostic model. The results suggest that iron metabolism and immune cells may play a significant role in the development of coronary heart disease, providing a basis for further research.
Collapse
Affiliation(s)
| | | | | | | | - Zhi Jin
- Department of Traditional Chinese Medicine, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
7
|
Xu Q, Kong F, Zhao G, Jin J, Feng S, Li M. USP7 alleviates neuronal inflammation and apoptosis in spinal cord injury via deubiquitinating NRF1/KLF7 axis. Neurol Res 2024; 46:1008-1017. [PMID: 39007840 DOI: 10.1080/01616412.2024.2376999] [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: 12/28/2023] [Accepted: 07/02/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Ubiquitin-specific protease 7 (USP7) has been found to be associated with motor function recovery after spinal cord injury (SCI). Therefore, its role and mechanism in SCI process need further exploration. METHODS SCI rat models were established via performing laminectomy at the T9-T11 spinal vertebrae and cutting spinal cord tissues. SCI cell models were constructed by inducing PC12 cells with lipopolysaccharide (LPS). The protein levels of USP7, nuclear respiratory factor 1 (NRF1), Krüppel-like factor 7 (KLF7) and apoptosis-related markers were detected by western blot. Cell viability and apoptosis were tested by cell counting kit-8 assay and flow cytometry. The contents of inflammatory factors were examined using ELISA. The interaction between NRF1 and USP7 or KLF7 was analyzed by co-immunoprecipitation assay, chromatin immunoprecipitation assay and dual-luciferase reporter assay, respectively. RESULTS USP7 was downregulated in SCI rat models and LPS-induced PC12 cells. Overexpressed USP7 promoted viability, while repressed apoptosis and inflammation in LPS-induced PC12 cells. USP7 could stabilize NRF1 protein expression via deubiquitination, and NRF1 knockdown reversed the protective effect of USP7 against LPS-induced PC12 cell injury. NRF1 is bound to KLF7 promoter to enhance its transcription. NRF1 overexpression inhibited LPS-induced PC12 cell inflammation and apoptosis via increasing KLF7 expression. CONCLUSION USP7 alleviated inflammation and apoptosis in LPS-induced PC12 cells via NRF1/KLF7 axis, indicating that targeting of USP7/NRF1/KLF7 axis might be a promising treatment strategy for SCI.
Collapse
Affiliation(s)
- Qifei Xu
- Department of Orthopedics, The First People's Hospital of Pingdingshan, Pingdingshan, China
| | - Fanguo Kong
- Department of Orthopedics, Henan Provincial Orthopedic Hospital, Zhengzhou, China
| | - Guanghui Zhao
- Department of Orthopedics, The First People's Hospital of Pingdingshan, Pingdingshan, China
| | - Junwei Jin
- Department of Orthopedics, The First People's Hospital of Pingdingshan, Pingdingshan, China
| | - Shengkai Feng
- Department of Orthopedics, The First People's Hospital of Pingdingshan, Pingdingshan, China
| | - Ming Li
- Department of Orthopedics, The First People's Hospital of Pingdingshan, Pingdingshan, China
| |
Collapse
|
8
|
Liao X, Huang Z, Ling H, Li W, Liu J, Lao Y, Su W. Mechanism of minocycline activating Nrf2/Hmox1 pathway to prevent ferroptosis and alleviate acute compartment syndrome. J Orthop Surg Res 2024; 19:686. [PMID: 39443986 PMCID: PMC11515506 DOI: 10.1186/s13018-024-05183-z] [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: 08/19/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Acute compartment syndrome(ACS) is a perilous consequence of trauma. Acute compartment syndrome's precise cause is yet unknown. We performed studies to confirm that acute compartment syndrome can be relieved by suppressing ferroptosis and activating the Nrf2/Hmox1 pathway. METHODS We generated an ACS rat model and we conducted next-generation sequencing(NGS) of skeletal muscle tissue and identified differentially expressed target genes. Ultimately, we performed in vivo experiments to validate the presence of ferroptosis and the Nrf2/Hmox1 pathway in ACS rats. After the minocycline intervention, the drug was evaluated for its effects on ACS by examining changes associated with ferroptosis. RESULTS The bioinformatics analysis identified that the genetic changes in the disease were mostly focused on ferroptosis, with noticeable modifications in Nrf2/Hmox1. Based on the in vivo results, it was observed that ACS rats exhibited significantly elevated levels of ferroptosis compared to the control rats. The suppression of the Nrf2/Hmox1 pathway mediated by minocycline improves outcomes in ACS and reduces tissue damage after intervention. CONCLUSION Minocycline hinders ferroptosis via stimulating the Nrf2/Hmox1 pathway, which slows down the advancement of acute compartment syndrome.
Collapse
Affiliation(s)
- Xiong Liao
- Department of Orthopedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, NO.6 ShuangYong Road, Nanning, Guangxi, 530022, China
- Department of Orthopedic Trauma, The Affiliated Changsha Central Hospital (Changsha Central Hospital), Hengyang Medical School, University of South China, Changsha, Hunan, 410004, China
| | - Zhao Huang
- Department of Orthopedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, NO.6 ShuangYong Road, Nanning, Guangxi, 530022, China
| | - He Ling
- Department of Orthopedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, NO.6 ShuangYong Road, Nanning, Guangxi, 530022, China
| | - Wencai Li
- Department of Orthopedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, NO.6 ShuangYong Road, Nanning, Guangxi, 530022, China
| | - Junjie Liu
- Department of Orthopedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, NO.6 ShuangYong Road, Nanning, Guangxi, 530022, China
| | - Yonghui Lao
- Department of Orthopedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, NO.6 ShuangYong Road, Nanning, Guangxi, 530022, China
| | - Wei Su
- Department of Orthopedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, NO.6 ShuangYong Road, Nanning, Guangxi, 530022, China.
| |
Collapse
|
9
|
Ru Q, Li Y, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in human diseases: mechanisms and therapeutic prospects. Signal Transduct Target Ther 2024; 9:271. [PMID: 39396974 PMCID: PMC11486532 DOI: 10.1038/s41392-024-01969-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/08/2024] [Accepted: 09/02/2024] [Indexed: 10/15/2024] Open
Abstract
Iron, an essential mineral in the body, is involved in numerous physiological processes, making the maintenance of iron homeostasis crucial for overall health. Both iron overload and deficiency can cause various disorders and human diseases. Ferroptosis, a form of cell death dependent on iron, is characterized by the extensive peroxidation of lipids. Unlike other kinds of classical unprogrammed cell death, ferroptosis is primarily linked to disruptions in iron metabolism, lipid peroxidation, and antioxidant system imbalance. Ferroptosis is regulated through transcription, translation, and post-translational modifications, which affect cellular sensitivity to ferroptosis. Over the past decade or so, numerous diseases have been linked to ferroptosis as part of their etiology, including cancers, metabolic disorders, autoimmune diseases, central nervous system diseases, cardiovascular diseases, and musculoskeletal diseases. Ferroptosis-related proteins have become attractive targets for many major human diseases that are currently incurable, and some ferroptosis regulators have shown therapeutic effects in clinical trials although further validation of their clinical potential is needed. Therefore, in-depth analysis of ferroptosis and its potential molecular mechanisms in human diseases may offer additional strategies for clinical prevention and treatment. In this review, we discuss the physiological significance of iron homeostasis in the body, the potential contribution of ferroptosis to the etiology and development of human diseases, along with the evidence supporting targeting ferroptosis as a therapeutic approach. Importantly, we evaluate recent potential therapeutic targets and promising interventions, providing guidance for future targeted treatment therapies against human diseases.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
10
|
Stojkovic L, Djordjevic A, Stefanovic M, Stankovic A, Dincic E, Djuric T, Zivkovic M. Circulatory Indicators of Lipid Peroxidation, the Driver of Ferroptosis, Reflect Differences between Relapsing-Remitting and Progressive Multiple Sclerosis. Int J Mol Sci 2024; 25:11024. [PMID: 39456806 PMCID: PMC11507982 DOI: 10.3390/ijms252011024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Ferroptosis, a lipid peroxidation- and iron-mediated type of regulated cell death, relates to both neuroinflammation, which is common in relapsing-remitting multiple sclerosis (RRMS), and neurodegeneration, which is prevalent in progressive (P)MS. Currently, findings related to the molecular markers proposed in this paper in patients are scarce. We analyzed circulatory molecular indicators of the main ferroptosis-related processes, comprising lipid peroxidation (malondialdehyde (MDA), 4-hydroxynonenal (4-HNE), and hexanoyl-lysine adduct (HEL)), glutathione-related antioxidant defense (total glutathione (reduced (GSH) and oxidized (GSSG)) and glutathione peroxidase 4 (GPX4)), and iron metabolism (iron, transferrin and ferritin) to estimate their contributions to the clinical manifestation of MS and differences between RRMS and PMS disease course. In 153 patients with RRMS and 69 with PMS, plasma/serum lipid peroxidation indicators and glutathione were quantified using ELISA and colorimetric reactions, respectively. Iron serum concentrations were determined using spectrophotometry, and transferrin and ferritin were determined using immunoturbidimetry. Compared to those with RRMS, patients with PMS had decreased 4-HNE (median, 1368.42 vs. 1580.17 pg/mL; p = 0.03). Interactive effects of MS course (RRMS/PMS) and disease-modifying therapy status on MDA (p = 0.009) and HEL (p = 0.02) levels were detected. In addition, the interaction of disease course and self-reported fatigue revealed significant impacts on 4-HNE levels (p = 0.01) and the GSH/GSSG ratio (p = 0.04). The results also show an association of MS course (p = 0.03) and EDSS (p = 0.04) with GSH levels. No significant changes were observed in the serum concentrations of iron metabolism indicators between the two patient groups (p > 0.05). We suggest circulatory 4-HNE as an important parameter related to differences between RRMS and PMS. Significant interactions of MS course and other clinically relevant parameters with changes in redox processes associated with ferroptosis support the further investigation of MS with a larger sample while taking into account both circulatory and central nervous system estimation.
Collapse
Affiliation(s)
- Ljiljana Stojkovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia; (A.D.); (A.S.); (T.D.)
| | - Ana Djordjevic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia; (A.D.); (A.S.); (T.D.)
| | - Milan Stefanovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia; (A.D.); (A.S.); (T.D.)
| | - Aleksandra Stankovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia; (A.D.); (A.S.); (T.D.)
| | - Evica Dincic
- Clinic for Neurology, Military Medical Academy, 11000 Belgrade, Serbia;
- Medical Faculty of the Military Medical Academy, University of Defence, 11000 Belgrade, Serbia
| | - Tamara Djuric
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia; (A.D.); (A.S.); (T.D.)
| | - Maja Zivkovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia; (A.D.); (A.S.); (T.D.)
| |
Collapse
|
11
|
Feng S, Liu L, Cheng Y, Zhou M, Zhu H, Zhao X, Chen Z, Kan S, Fu X, Hu W, Zhu R. Icariin promotes functional recovery in rats after spinal cord injury by inhibiting YAP and regulating PPM1B ubiquitination to inhibiting the activation of reactive astrocytes. Front Pharmacol 2024; 15:1434652. [PMID: 39439899 PMCID: PMC11493691 DOI: 10.3389/fphar.2024.1434652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Objective The limited ability to regenerate axons after spinal cord injury (SCI) is influenced by factors such as astrocyte activation, reactive proliferation, and glial scar formation. The TGF-β/Smad (transforming growth factor-β/mothers against decapentaplegic homolog) pathway, associated with astrocytic scarring, plays a crucial role in recovery post-injury. This study aims to investigate how icariin (ICA) interacts with reactive astrocytes in the treatment of spinal cord injury. Methods A rat SCI model was constructed, and the recovery of motor function was observed after treatment with ICA.HE staining, LFB staining, immunofluorescence staining, and Western blotting were employed to assess ICA's ability to inhibit astrocyte proliferation in rats following spinal cord injury by modulating YAP, as well as to evaluate the reparative effects of ICA on the injured spinal cord tissue. Primary astrocytes were isolated and cultured. Immunoprecipitation-Western Blot (IP-WB) ubiquitination and cytoplasm-nuclear separation were employed to assess PPM1B ubiquitination and nuclear translocation. Results The CatWalk XT gait analysis, BBB (Basso, Beattie, and Bresnahan) score, electrophysiological measurements, HE staining, and LFB staining collectively demonstrated that ICA promotes motor function and tissue recovery following spinal cord injury in rats. Immunofluorescence staining and Western Blot analyses revealed that ICA inhibits astrocyte proliferation in rats post-spinal cord injury by suppressing YAP activity. Furthermore, the activation of YAP by XMU-MP-1 was shown to compromise the efficacy of ICA in these rats after spinal cord injury. Additional immunofluorescence staining and Western Blot experiments confirmed that ICA inhibits TGFβ1-induced astrocyte activation through the regulation of YAP. The knockdown of PPM1B (protein phosphatase, Mg2+/Mn2+-dependent 1B) in astrocytes was found to inhibit TGFβ signaling. Additionally, YAP was shown to regulate PPM1B ubiquitination and nuclear translocation through immunoprecipitation-Western blot analysis, along with the segregation of cytoplasm and nucleus. Conclusion Icariin promotes functional recovery in rats after spinal cord injury by inhibiting YAP and regulating PPM1B ubiquitination to inhibiting the activation of reactive astrocytes.
Collapse
Affiliation(s)
- Sa Feng
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Linyan Liu
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Yuelin Cheng
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Mengmeng Zhou
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Haoqiang Zhu
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Xinyan Zhao
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Ziyu Chen
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Shunli Kan
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Xuanhao Fu
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Wei Hu
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Rusen Zhu
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Tianjin Institute of Spinal Surgery, Tianjin Union Medical Center, Tianjin, China
| |
Collapse
|
12
|
Ma Z, Liu T, Liu L, Pei Y, Wang T, Wang Z, Guan Y, Zhang X, Zhang Y, Chen X. Epidermal Neural Crest Stem Cell Conditioned Medium Enhances Spinal Cord Injury Recovery via PI3K/AKT-Mediated Neuronal Apoptosis Suppression. Neurochem Res 2024; 49:2854-2870. [PMID: 39023805 PMCID: PMC11365850 DOI: 10.1007/s11064-024-04207-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 04/19/2024] [Accepted: 07/05/2024] [Indexed: 07/20/2024]
Abstract
This study aimed to assess the impact of conditioned medium from epidermal neural crest stem cells (EPI-NCSCs-CM) on functional recovery following spinal cord injury (SCI), while also exploring the involvement of the PI3K-AKT signaling pathway in regulating neuronal apoptosis. EPI-NCSCs were isolated from 10-day-old Sprague-Dawley rats and cultured for 48 h to obtain EPI-NCSC-CM. SHSY-5Y cells were subjected with H2O2 treatment to induce apoptosis. Cell viability and survival rates were evaluated using the CCK-8 assay and calcein-AM/PI staining. SCI contusion model was established in adult Sprague-Dawley rats to assess functional recovery, utilizing the Basso, Beattie and Bresnahan (BBB) scoring system, inclined test, and footprint observation. Neurological restoration after SCI was analyzed through electrophysiological recordings. Histological analysis included hematoxylin and eosin (H&E) staining and Nissl staining to evaluate tissue organization. Apoptosis and oxidative stress levels were assessed using TUNEL staining and ROS detection methods. Additionally, western blotting was performed to examine the expression of apoptotic markers and proteins related to the PI3K/AKT signaling pathway. EPI-NCSC-CM significantly facilitated functional and histological recovery in SCI rats by inhibiting neuronal apoptosis through modulation of the PI3K/AKT pathway. Administration of EPI-NCSCs-CM alleviated H2O2-induced neurotoxicity in SHSY-5Y cells in vitro. The use of LY294002, a PI3K inhibitor, underscored the crucial role of the PI3K/AKT signaling pathway in regulating neuronal apoptosis. This study contributes to the ongoing exploration of molecular pathways involved in spinal cord injury (SCI) repair, focusing on the therapeutic potential of EPI-NCSC-CM. The research findings indicate that EPI-NCSC-CM exerts a neuroprotective effect by suppressing neuronal apoptosis through activation of the PI3K/AKT pathway in SCI rats. These results highlight the promising role of EPI-NCSC-CM as a potential treatment strategy for SCI, emphasizing the significance of the PI3K/AKT pathway in mediating its beneficial effects.
Collapse
Affiliation(s)
- Ziqian Ma
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, 8 Workers Stadium South Road, Chaoyang District, Beijing, China
| | - Tao Liu
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Liang Liu
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yilun Pei
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Tianyi Wang
- Department of Orthopedics, 981st Hospital of the Chinese People's Liberation Army Joint Logistics Support Force, Chengde, 067000, Hebei Province, P.R. China
| | - Zhijie Wang
- Department of Pediatric Internal Medicine, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurological Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Xinwei Zhang
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yan Zhang
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China.
| | - Xueming Chen
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
13
|
Guan X, Wang Y, Yu W, Wei Y, Lu Y, Dai E, Dong X, Zhao B, Hu C, Yuan L, Luan X, Miao K, Chen B, Cheng X, Zhang W, Qin J. Blocking Ubiquitin-Specific Protease 7 Induces Ferroptosis in Gastric Cancer via Targeting Stearoyl-CoA Desaturase. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307899. [PMID: 38460164 PMCID: PMC11095140 DOI: 10.1002/advs.202307899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/21/2024] [Indexed: 03/11/2024]
Abstract
Gastric cancer (GC) presents a formidable global health challenge, and conventional therapies face efficacy limitations. Ubiquitin-specific protease 7 (USP7) plays pivotal roles in GC development, immune response, and chemo-resistance, making it a promising target. Various USP7 inhibitors have shown selectivity and efficacy in preclinical studies. However, the mechanistic role of USP7 has not been fully elucidated, and currently, no USP7 inhibitors have been approved for clinical use. In this study, DHPO is identified as a potent USP7 inhibitor for GC treatment through in silico screening. DHPO demonstrates significant anti-tumor activity in vitro, inhibiting cell viability and clonogenic ability, and preventing tumor migration and invasion. In vivo studies using orthotopic gastric tumor mouse models validate DHPO's efficacy in suppressing tumor growth and metastasis without significant toxicity. Mechanistically, DHPO inhibition triggers ferroptosis, evidenced by mitochondrial alterations, lipid Reactive Oxygen Species (ROS), Malondialdehyde (MDA) accumulation, and iron overload. Further investigations unveil USP7's regulation of Stearoyl-CoA Desaturase (SCD) through deubiquitination, linking USP7 inhibition to SCD degradation and ferroptosis induction. Overall, this study identifies USP7 as a key player in ferroptosis of GC, elucidates DHPO's inhibitory mechanisms, and highlights its potential for GC treatment by inducing ferroptosis through SCD regulation.
Collapse
Affiliation(s)
- Xiaoqing Guan
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
- Key Laboratory of PreventionDiagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang ProvinceHangzhouZhejiang310022China
| | - Yichao Wang
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
- College of Pharmaceutical SciencesZhejiang University of TechnologyHangzhouZhejiang310014China
| | - Wenkai Yu
- School of PharmacyZhejiang Chinese Medical UniversityHangzhouZhejiang310053China
| | - Yong Wei
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
| | - Yang Lu
- Hangzhou Institute of Innovative MedicineInstitute of Drug Discovery and DesignCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Enyu Dai
- Department of Genomic MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexas77030USA
| | - Xiaowu Dong
- Hangzhou Institute of Innovative MedicineInstitute of Drug Discovery and DesignCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Bing Zhao
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
| | - Can Hu
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
- Key Laboratory of PreventionDiagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang ProvinceHangzhouZhejiang310022China
| | - Li Yuan
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
- Key Laboratory of PreventionDiagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang ProvinceHangzhouZhejiang310022China
| | - Xin Luan
- Institute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Kai Miao
- MOE Frontier Science Centre for Precision OncologyUniversity of MacauMacau SAR999078China
| | - Bonan Chen
- Department of Anatomical and Cellular PathologyPrince of Wales HospitalThe Chinese University of Hong KongHong Kong999077China
| | - Xiang‐Dong Cheng
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
- Key Laboratory of PreventionDiagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang ProvinceHangzhouZhejiang310022China
| | - Weidong Zhang
- Institute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
- School of PharmacyNaval Medical UniversityShanghai200433China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di HerbsInstitute of Medicinal Plant DevelopmentChinese Academy of Medical Science and Peking Union Medical CollegeBeijing100193China
| | - Jiang‐Jiang Qin
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
- Key Laboratory of PreventionDiagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang ProvinceHangzhouZhejiang310022China
| |
Collapse
|
14
|
Wang Y, Hu J, Wu S, Fleishman JS, Li Y, Xu Y, Zou W, Wang J, Feng Y, Chen J, Wang H. Targeting epigenetic and posttranslational modifications regulating ferroptosis for the treatment of diseases. Signal Transduct Target Ther 2023; 8:449. [PMID: 38072908 PMCID: PMC10711040 DOI: 10.1038/s41392-023-01720-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/16/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
Ferroptosis, a unique modality of cell death with mechanistic and morphological differences from other cell death modes, plays a pivotal role in regulating tumorigenesis and offers a new opportunity for modulating anticancer drug resistance. Aberrant epigenetic modifications and posttranslational modifications (PTMs) promote anticancer drug resistance, cancer progression, and metastasis. Accumulating studies indicate that epigenetic modifications can transcriptionally and translationally determine cancer cell vulnerability to ferroptosis and that ferroptosis functions as a driver in nervous system diseases (NSDs), cardiovascular diseases (CVDs), liver diseases, lung diseases, and kidney diseases. In this review, we first summarize the core molecular mechanisms of ferroptosis. Then, the roles of epigenetic processes, including histone PTMs, DNA methylation, and noncoding RNA regulation and PTMs, such as phosphorylation, ubiquitination, SUMOylation, acetylation, methylation, and ADP-ribosylation, are concisely discussed. The roles of epigenetic modifications and PTMs in ferroptosis regulation in the genesis of diseases, including cancers, NSD, CVDs, liver diseases, lung diseases, and kidney diseases, as well as the application of epigenetic and PTM modulators in the therapy of these diseases, are then discussed in detail. Elucidating the mechanisms of ferroptosis regulation mediated by epigenetic modifications and PTMs in cancer and other diseases will facilitate the development of promising combination therapeutic regimens containing epigenetic or PTM-targeting agents and ferroptosis inducers that can be used to overcome chemotherapeutic resistance in cancer and could be used to prevent other diseases. In addition, these mechanisms highlight potential therapeutic approaches to overcome chemoresistance in cancer or halt the genesis of other diseases.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Jing Hu
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300060, PR China
| | - Shuang Wu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, 430000, PR China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Yulin Li
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Yinshi Xu
- Department of Outpatient, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Wailong Zou
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Jinhua Wang
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| | - Yukuan Feng
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, PR China.
| | - Jichao Chen
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China.
| | - Hongquan Wang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, PR China.
| |
Collapse
|
15
|
Bejarano E, Whitcomb EA, Pfeiffer RL, Rose KL, Asensio MJ, Rodríguez-Navarro JA, Ponce-Mora A, Canto A, Almansa I, Schey KL, Jones BW, Taylor A, Rowan S. Unbalanced redox status network as an early pathological event in congenital cataracts. Redox Biol 2023; 66:102869. [PMID: 37677999 PMCID: PMC10495660 DOI: 10.1016/j.redox.2023.102869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/08/2023] [Accepted: 08/31/2023] [Indexed: 09/09/2023] Open
Abstract
The lens proteome undergoes dramatic composition changes during development and maturation. A defective developmental process leads to congenital cataracts that account for about 30% of cases of childhood blindness. Gene mutations are associated with approximately 50% of early-onset forms of lens opacity, with the remainder being of unknown etiology. To gain a better understanding of cataractogenesis, we utilized a transgenic mouse model expressing a mutant ubiquitin protein in the lens (K6W-Ub) that recapitulates most of the early pathological changes seen in human congenital cataracts. We performed mass spectrometry-based tandem-mass-tag quantitative proteomics in E15, P1, and P30 control or K6W-Ub lenses. Our analysis identified targets that are required for early normal differentiation steps and altered in cataractous lenses, particularly metabolic pathways involving glutathione and amino acids. Computational molecular phenotyping revealed that glutathione and taurine were spatially altered in the K6W-Ub cataractous lens. High-performance liquid chromatography revealed that both taurine and the ratio of reduced glutathione to oxidized glutathione, two indicators of redox status, were differentially compromised in lens biology. In sum, our research documents that dynamic proteome changes in a mouse model of congenital cataracts impact redox biology in lens. Our findings shed light on the molecular mechanisms associated with congenital cataracts and point out that unbalanced redox status due to reduced levels of taurine and glutathione, metabolites already linked to age-related cataract, could be a major underlying mechanism behind lens opacities that appear early in life.
Collapse
Affiliation(s)
- Eloy Bejarano
- JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA; School of Health Sciences and Veterinary School, Universidad Cardenal Herrera-CEU, CEU Universities, Moncada, Valencia, Spain
| | - Elizabeth A Whitcomb
- JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Rebecca L Pfeiffer
- Moran Eye Center, The University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Kristie L Rose
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Maria José Asensio
- Servicio de Neurobiología, Departamento de Investigación, Hospital Ramón y Cajal, IRYCIS, Madrid, Spain
| | - José Antonio Rodríguez-Navarro
- Servicio de Neurobiología, Departamento de Investigación, Hospital Ramón y Cajal, IRYCIS, Madrid, Spain; Department of Cell Biology, Complutense University of Madrid, Madrid, Spain
| | - Alejandro Ponce-Mora
- School of Health Sciences and Veterinary School, Universidad Cardenal Herrera-CEU, CEU Universities, Moncada, Valencia, Spain
| | - Antolín Canto
- School of Health Sciences and Veterinary School, Universidad Cardenal Herrera-CEU, CEU Universities, Moncada, Valencia, Spain
| | - Inma Almansa
- School of Health Sciences and Veterinary School, Universidad Cardenal Herrera-CEU, CEU Universities, Moncada, Valencia, Spain
| | - Kevin L Schey
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Bryan W Jones
- Moran Eye Center, The University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Allen Taylor
- JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA; Friedman School of Nutrition and Science Policy, Tufts University, Boston, MA, USA.
| | - Sheldon Rowan
- JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA; Friedman School of Nutrition and Science Policy, Tufts University, Boston, MA, USA.
| |
Collapse
|