1
|
Xu Z, Zhang M, Zhang X, Han H, Ye W, Chen Z, Lv Z, Liu Y, Liu Z, Gong J, Zhu B, Zhou S, Zhu R, Tao C, Zhang G, Yan X. Dihydromyricetin protects against cisplatin-induced renal injury and mitochondria-mediated apoptosis via the EGFR/HSP27/STAT3 signaling pathway. Ren Fail 2025; 47:2490202. [PMID: 40230054 PMCID: PMC12001862 DOI: 10.1080/0886022x.2025.2490202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 03/06/2025] [Accepted: 03/22/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Cisplatin (CP) has been used as an effective chemotherapy drug for different types of cancers. Despite its therapeutic benefits, the clinical utility of CP is often hindered by adverse effects, notably acute kidney injury (AKI), which restricts its widespread application. Dihydromyricetin (DHM) is a flavonoid acquired from Ampelopsis grossedentata, exhibiting a range of pharmacological activities. The major objective of this research was to examine the possible molecular mechanism involved in CP-induced AKI and the protective function of DHM. METHODS In this study, the protective function of DHM against CP-induced AKI was assessed in both mice and HK-2 cells. Kidney dysfunction parameters and renal morphology were evaluated to ascertain the extent of protection. Additionally, proteomics techniques were employed to investigate the protective effect of DHM and elucidate the underlying molecular mechanisms involved in mitigating CP-induced AKI. In addition, protein levels of epidermal growth factor receptor (EGFR), p-EGFR, heat shock protein 27 (HSP27), p-HSP27, STAT3, and p-STAT3 in renal tissues were investigated. Furthermore, an EGFR-blocking agent (gefitinib) or si-RNA of HSP27 was used to study the effects of inhibiting EGFR or HSP27 on CP-induced renal injury. RESULTS DHM decreased blood urea nitrogen (BUN) and creatinine in serum, alleviated renal morphological injury and downregulated the expression of CP-induced kidney injury molecule-1 and neutrophil gelatinase-related lipocalin. Proteomic data revealed HSP27 as a potential therapeutic target for AKI. DHM treatment resulted in the downregulation of EGFR, HSP27, and STAT3 phosphorylation, ultimately mitigating CP-induced AKI. In addition, the inhibition of EGFR or HSP27 reduced mitochondria-mediated apoptosis and CP-induced cell damage in HK-2 cells. CONCLUSIONS DHM effectively inhibited CP-induced oxidative stress, inflammation, and mitochondria-mediated apoptosis through the EGFR/HSP27/STAT3 pathway.
Collapse
Affiliation(s)
- Zheming Xu
- Department of Urology, Pediatric Urolith Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Minjing Zhang
- Department of Urology, Pediatric Urolith Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Xue Zhang
- Department of Urology, Pediatric Urolith Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Huirong Han
- School of Anesthesiology, Shandong Second Medical University, Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, Weifang, China
| | - Weifeng Ye
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Zhenjie Chen
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Zhisu Lv
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Yang Liu
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Zhengye Liu
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jianguang Gong
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Bin Zhu
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Suhan Zhou
- Department of Physiology, School of Basic Medical Sciences, and Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Runzhi Zhu
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Chang Tao
- Department of Urology, Pediatric Urolith Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Gensheng Zhang
- Department of Urology, Pediatric Urolith Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Xiang Yan
- Department of Urology, Pediatric Urolith Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| |
Collapse
|
2
|
Luo Y, He Y, Xu S, Chen Y, Qin F, Hu W. Ferroptosis: a potential target for non-surgical treatment of laryngeal cancer. Eur Arch Otorhinolaryngol 2025:10.1007/s00405-025-09279-y. [PMID: 40087171 DOI: 10.1007/s00405-025-09279-y] [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: 10/24/2024] [Accepted: 02/17/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Laryngeal cancer (LC) is among the most prevalent tumors of the respiratory tract. In recent years, the implementation of non-surgical treatments like radiotherapy and chemotherapy has significantly enhanced the therapeutic outcomes for LC. Nevertheless, the underlying therapeutic mechanisms remain unclear, posing a hindrance to the progression of subsequent treatment strategies. OBJECTIVES To explore the potential mechanisms from existing effective treatments for LC and identify relevant targets, thereby providing guidance for subsequent therapeutic research on LC. METHODS This study focuses on ferroptosis, a common type of non-apoptotic cell death that is closely linked to various malignancies. It examines the relationship between ferroptosis and LC by analyzing how regulating ferroptosis-related targets in LC cells can influence the development of the cancer. RESULTS There is a strong association between ferroptosis and LC. Regulating the targets related to ferroptosis in LC cells can effectively counteract the progression of LC. CONCLUSIONS Taking ferroptosis as an entry point, analyzing its potential mechanism in inhibiting LC can provide a direction for the treatment of laryngeal cancer, which may contribute to the improvement of therapeutic strategies for this disease.
Collapse
Affiliation(s)
- Yang Luo
- Department of Otolaryngology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yuzhu He
- Department of Otolaryngology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Shuang Xu
- Department of Otolaryngology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yuxiang Chen
- Department of Otolaryngology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Fengfeng Qin
- Department of Otolaryngology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Wenjian Hu
- Department of Otolaryngology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University of Luzhou, NO. 182, Chunhui Road, Longmatan District, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
3
|
Hong H, Guo D, Xia T, Zhang Y. Dihydromyricetin attenuates intervertebral disc degeneration by inhibiting NLRP3 inflammasome activation via the Keap1/Nrf2/HO-1 pathway. Eur J Pharmacol 2025; 998:177501. [PMID: 40058758 DOI: 10.1016/j.ejphar.2025.177501] [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: 01/16/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/20/2025]
Abstract
Intervertebral disc degeneration (IVDD) is a highly prevalent chronic degenerative condition that significantly compromises patients' quality of life. Currently employed clinical treatments include surgical intervention and symptom management strategies; however, effective pharmacological strategies are lacking. Dihydromyricetin (DHM) has remarkable anti-inflammatory and antioxidative properties. On the basis of these biological characteristics, we hypothesized that DHM might have therapeutic potential in IVDD through its anti-inflammatory effects. Network pharmacology analysis revealed 130 overlapping targets between DHM and IVDD, with the Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway emerging as a crucial regulatory axis. Experimental validation demonstrated that DHM treatment significantly ameliorated LSI-induced disc degeneration, as evidenced by reduced histopathological scores, upregulated expression of extracellular matrix (ECM) proteins. In vitro studies revealed that DHM effectively inhibited IL-1β-induced NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome activation and pyroptosis by decreasing Keap1 expression and activating the Nrf2/HO-1 signaling pathway. Specific silencing of Nrf2 significantly attenuated the protective effects of DHM, further confirming the crucial role of the Keap1/Nrf2/HO-1 pathway in the therapeutic action of DHM. Through integrated network pharmacology analysis and experimental validation, this study demonstrated for the first time that DHM alleviates IVDD by inhibiting Keap1-mediated Nrf2 degradation and activating the Nrf2/HO-1 pathway to suppress NLRP3 inflammasome-mediated pyroptosis. Furthermore, these findings validate the therapeutic potential of natural bioactive compounds in IVDD, providing experimental evidence and a theoretical foundation for the development of novel therapeutic strategies against IVDD.
Collapse
Affiliation(s)
- Hainan Hong
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Di Guo
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Tao Xia
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China.
| | - Yuhang Zhang
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China; Enze Hospital, Taizhou Enze Medical Center (Group), Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China.
| |
Collapse
|
4
|
Hu J, Zhang Y, Zhang Y, Shi N, Miu Y, Huang J, Miao M, Ci X. Bergenin inhibits ferritinophagy and ferroptosis in cisplatin-induced acute kidney injury by activating the p-GSK3β/Nrf2/PPARγ pathway. Int Immunopharmacol 2025; 147:114004. [PMID: 39793228 DOI: 10.1016/j.intimp.2024.114004] [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: 10/28/2024] [Revised: 12/20/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025]
Abstract
Ferroptosis plays a key role in cisplatin-induced acute kidney injury (AKI). Bergenin, which is extracted from Ardisiae Japonicae Herba and has long been used in folk tea and herbal tea drinks, is known to activate Nrf2 and has anti-inflammatory and antioxidant properties, however, its protective influence on CI-AKI has not been elucidated. We used models of cisplatin-induced nephrotoxicity in vitro and CI-AKI models in vivo. In vitro, we found that ferroptosis and ferritinophagy biomarkers were strongly regulated by bergenin treatment. Mechanistic experiments demonstrated that bergenin bound to and phosphorylated GSK3β, which inhibited its activity, to promote the nuclear translocation of Nrf2 and its subsequent binding to the PPARγ promoter sequence to activate PPARγ. However, the protective effects of bergenin on ferroptosis and ferritinophagy in cisplatin-exposed HK-2 cells were diminished when Nrf2 or PPARγ was inhibited. In vivo, bergenin effectively inhibited renal damage induced by cisplatin. Furthermore, bergenin attenuated ferritinophagy-mediated ferroptosis caused by cisplatin; these effects were abolished in Nrf2 knockout mice. Our findings revealed that bergenin effectively protected against ferritinophagy and ferroptosis in CI-AKI, which was largely dependent on the activation of the p-GSK3β/Nrf2/PPARγ pathway.
Collapse
Affiliation(s)
- Jianqiang Hu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Yan Zhang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Yanmin Zhang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Ningmohan Shi
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Yufan Miu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Jing Huang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Mochi Miao
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China.
| |
Collapse
|
5
|
Yu Y, Zhang L, Zhang D, Dai Q, Hou M, Chen M, Gao F, Liu XL. The role of ferroptosis in acute kidney injury: mechanisms and potential therapeutic targets. Mol Cell Biochem 2025; 480:759-784. [PMID: 38943027 DOI: 10.1007/s11010-024-05056-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024]
Abstract
Acute kidney injury (AKI) is one of the most common and severe clinical renal syndromes with high morbidity and mortality. Ferroptosis is a form of programmed cell death (PCD), is characterized by iron overload, reactive oxygen species accumulation, and lipid peroxidation. As ferroptosis has been increasingly studied in recent years, it is closely associated with the pathophysiological process of AKI and provides a target for the treatment of AKI. This review offers a comprehensive overview of the regulatory mechanisms of ferroptosis, summarizes its role in various AKI models, and explores its interaction with other forms of cell death, it also presents research on ferroptosis in AKI progression to other diseases. Additionally, the review highlights methods for detecting and assessing AKI through the lens of ferroptosis and describes potential inhibitors of ferroptosis for AKI treatment. Finally, the review presents a perspective on the future of clinical AKI treatment, aiming to stimulate further research on ferroptosis in AKI.
Collapse
Affiliation(s)
- Yanxin Yu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Lei Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Die Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Qiangfang Dai
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Mingzheng Hou
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Meini Chen
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Feng Gao
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Xiao-Long Liu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China.
| |
Collapse
|
6
|
Shapiro O, Woods C, Gleixner AM, Sannino S, Ngo M, McDaniels MD, Wipf P, Hukriede NA, Donnelly CJ, Brodsky JL. Assays to measure small molecule Hsp70 agonist activity in vitro and in vivo. Anal Biochem 2025; 697:115712. [PMID: 39522672 PMCID: PMC11952136 DOI: 10.1016/j.ab.2024.115712] [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: 09/03/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Hsp70 prevents protein aggregation and is cytoprotective, but sustained Hsp70 overexpression is problematic. Therefore, we characterized small molecule agonists that augment Hsp70 activity. Because cumbersome assays were required to assay agonists, we developed cell-based and in vivo assays in which disease-associated consequences of Hsp70 activation can be quantified. One assay uses an optogenetic system in which the formation of TDP-43 inclusions can be controlled, and the second assay employs a zebrafish model for acute kidney injury (AKI). These complementary assays will facilitate future work to identify new Hsp70 agonists as well as optimized agonist derivatives.
Collapse
Affiliation(s)
- Olivia Shapiro
- Department of Neurobiology, LiveLikeLou Center for ALS Research, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Clara Woods
- Department of Cell Biology, Center for Integrative Organ Systems, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Amanda M Gleixner
- Department of Neurobiology, LiveLikeLou Center for ALS Research, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Sara Sannino
- Department of Biological Science, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Marilyn Ngo
- Department of Neurobiology, LiveLikeLou Center for ALS Research, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, PA, 15213, USA
| | - Michael D McDaniels
- Department of Cell Biology, Center for Integrative Organ Systems, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Neil A Hukriede
- Department of Cell Biology, Center for Integrative Organ Systems, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Christopher J Donnelly
- Department of Neurobiology, LiveLikeLou Center for ALS Research, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Jeffrey L Brodsky
- Department of Biological Science, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
7
|
Xie J, Yang Q, Zeng X, Zeng Q, Xiao H. Dihydromyricetin inhibits injury caused by ischemic stroke through the lncRNA SNHG17/miR-452-3p/CXCR4 axis. PeerJ 2025; 13:e18876. [PMID: 39897488 PMCID: PMC11786715 DOI: 10.7717/peerj.18876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/27/2024] [Indexed: 02/04/2025] Open
Abstract
Ischemic stroke (IS) is an important cause of death worldwide. Dihydromyricetin (DHM) has been reported to have neuroprotective potential, but its role and mechanism in IS have not been fully elucidated. The MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay was used to determine the safe dose of DHM in BV2 microglia and its applicability in OGD/R-treated cells. The mechanism of action of DHM was explored by RT-qPCR, ELISA, luciferase reporter gene assay and western blotting. DHM dose-dependently enhanced BV2 cell viability post-OGD/R and attenuated inflammation and oxidative stress. The protective effects of DHM were found to be mediated through the downregulation of SNHG17, which in turn modulated miR-452-3p expression. miR-452-3p was identified as a negative regulator of pro-inflammatory CXCR4, a direct target whose expression was inversely affected by SNHG17. The interaction between SNHG17 and miR-452-3p was further confirmed by RNA pull-down assays. Furthermore, manipulation of the SNHG17/miR-452-3p/CXCR4 axis was shown to modulate the NF-κB signaling pathway as evidenced by changes in phosphorylation levels. In conclusion, our findings elucidate a novel DHM-mediated neuroprotective mechanism in microglial cells involving the SNHG17/miR-452-3p/CXCR4 regulatory axis. This axis attenuates OGD/R-induced inflammatory and oxidative stress, suggesting a therapeutic potential for DHM in conditions characterized by such pathological processes.
Collapse
Affiliation(s)
- Jiacheng Xie
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Qiuyue Yang
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xueliang Zeng
- Department of Pharmacology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Qi Zeng
- Department of Ultrasound, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Hai Xiao
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
8
|
He C, Chen Y, Xie J, Luo M, Fisher D, Hien NTT, Musabaev E, Dang Y, Zhao L, Xia Y. Dihydromyricetin: an emerging compound with comprehensive effects on multiple systems. Front Pharmacol 2025; 15:1488003. [PMID: 39830336 PMCID: PMC11739078 DOI: 10.3389/fphar.2024.1488003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Dihydromyricetin (DHM or DMY) is a flavonoid derived from natural sources with a range of confirmed biological benefits. It exhibits anti-inflammatory, antioxidant, anti-tumor, and anti-viral activities. DHM is recognized for its high biosafety, making it a promising subject for further research. This article offers a comprehensive overview of DHM's pharmacological properties, mechanisms, and recent research developments in the cardiovascular, urinary, digestive, nervous, and respiratory systems. The review summarizes DHM's biological effects and associated signaling pathways, providing novel insights for its clinical application.
Collapse
Affiliation(s)
- Chengyi He
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yunfei Chen
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiao Xie
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Miao Luo
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - David Fisher
- Department of Medical Biosciences, Faculty of Natural Sciences, University of The Western Cape, Cape Town, South Africa
| | | | - Erkin Musabaev
- The Research Institute of Virology, Ministry of Health, Tashkent, Uzbekistan
| | - Yiping Dang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yin Xia
- Department of Vascular Surgery, The Affiliated People’s Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
9
|
Tian R, Guo S, Chen S, Wu J, Long A, Cheng R, Wang X, Huang L, Li C, Mao W, Xu P, Yu L, Pan H, Liu L. Natural products as Nrf2 modulators for ferroptosis inhibition in renal disease therapy: Recent progress and future prospects. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156342. [PMID: 39742572 DOI: 10.1016/j.phymed.2024.156342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 01/03/2025]
Abstract
BACKGROUND The transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2, NFE2L2) is a pivotal regulator of redox balance, metabolism, protein homeostasis and inflammation. Nrf2 is critically involved in both ferroptosis and renal diseases, and may serve as a significant target for many natural products in the treatment of renal diseases. However, a comprehensive overview on this topic is still lacking. PURPOSE To review the protective or therapeutic effects of natural products regulating Nrf2-related ferroptosis against various renal diseases. METHODS We systematically searched the electronic databases involving PubMed, Web of Science, Google Scholar, China National Knowledge Internet (CNKI), Wanfang Database and VIP Database. To ensure a comprehensive exploration, keywords including Nrf2, ferroptosis, natural products, phytochemicals, renal disease, kidney disease, kidney injury and nephropathy were employed. RESULTS Ferroptosis is deeply implicated in various kinds of renal diseases, notably including cisplatin-induced acute kidney injury, sepsis-associated acute kidney injury, renal ischemia/reperfusion injury, diabetic nephropathy, kidney stones and renal fibrosis. Nrf2 plays a regulatory role on many important genes related to iron metabolism, antioxidant system and lipid metabolism, thereby modulating ferroptosis. More than twenty natural products exert renoprotective effects by inhibiting ferroptosis via the regulation of Nrf2. This review presents a comprehensive overview of recent advancements in elucidating the ferroptosis involvement in renal diseases, the role of Nrf2 in regulating ferroptosis, and summarizes the renoprotective natural products as Nrf2 modulators for ferroptosis inhibition. CONCLUSION Through the comprehensive insights, this review clarifies the protective or therapeutic effects of natural products as Nrf2 modulators for ferroptosis inhibition in renal disease therapy, in the pursuit of providing new research ideas and directions for the treatment of renal diseases. Further drug development aimed at discovering more natural products and optimizing their utilization for disease treatment is necessary.
Collapse
Affiliation(s)
- Ruimin Tian
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Shan Guo
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shudong Chen
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jiaqi Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Aoyang Long
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Ran Cheng
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiaowan Wang
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Lihua Huang
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Chuang Li
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Wei Mao
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China
| | - Peng Xu
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China
| | - Lili Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Hudan Pan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China.
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao In-Depth Cooperation Zone in Hengqin, 519000, China.
| |
Collapse
|
10
|
Wang Y, Xue P, Gao L, Wang X, Zhou S, Wu X, Guo C. Improved bioavailability of polydatin and its protective effect against cisplatin induced nephrotoxicity through self-assembled fucoidan and carboxymethyl chitosan delivery system. Int J Biol Macromol 2025; 287:138577. [PMID: 39657878 DOI: 10.1016/j.ijbiomac.2024.138577] [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: 09/24/2024] [Revised: 11/26/2024] [Accepted: 12/07/2024] [Indexed: 12/12/2024]
Abstract
Cisplatin induced acute kidney injury (AKI) is clinically prevalent, with a complex pathogenesis and a lack of effective therapeutic drugs. Polydatin (Po) has excellent biological activity, but its low solubility and bioavailability limit its application. In this study, fucoidan (Fu) and carboxymethyl chitosan (Cs) self-assembled into nanoparticles through electrostatic interactions/hydrogen bonding and loaded Po (Fu/Cs Po NPs). In vitro studies found that Fu/Cs Po NPs protected human renal tubular epithelial (HK-2) cells from cisplatin induced damage and accumulation of reactive oxygen species (ROS). Mechanistic studies showed that Fu/Cs Po NPs inhibited cisplatin induced DNA damage and activation of cyclic guanosine monophosphate synthase (cGAS) and intron gene stimulator (STING) pathways. In vivo studies showed that Fu/Cs Po NPs treatment alleviated cisplatin induced AKI symptoms, including elevated blood urea nitrogen (BUN) and serum creatinine (SCr), as well as pathological damage to kidney tissues. In vivo mechanism studies also showed that Fu/Cs Po NPs treatment inhibited cisplatin induced DNA damage and activation of the cGAS-STING pathway. The pharmacokinetic and tissue distribution results demonstrated that the Fu/Cs delivery system enhanced the bioavailability and kidney accumulation of Po in vivo. In summary, our study provided potential drugs for the treatment of cisplatin induced AKI.
Collapse
Affiliation(s)
- Yinghan Wang
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Pengyu Xue
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Liang Gao
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xuefei Wang
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Shilin Zhou
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xiaochen Wu
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Chuanlong Guo
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China.
| |
Collapse
|
11
|
Zhang R, Zhang X, Zhu X, Li T, Li Y, Zhang P, Chen Y, Li G, Han X. Nanoparticles transfected with plasmid-encoded lncRNA-OIP5-AS1 inhibit renal ischemia-reperfusion injury in mice via the miR-410-3p/Nrf2 axis. Ren Fail 2024; 46:2319327. [PMID: 38419565 PMCID: PMC10906121 DOI: 10.1080/0886022x.2024.2319327] [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: 08/17/2023] [Accepted: 02/09/2024] [Indexed: 03/02/2024] Open
Abstract
Nanostructures composed of liposomes and polydopamine (PDA) have demonstrated efficacy as carriers for delivering plasmids, effectively alleviating renal cell carcinoma. However, their role in acute kidney injury (AKI) remains unclear. This study aimed to investigate the effects of the plasmid-encoded lncRNA-OIP5-AS1@PDA nanoparticles (POP-NPs) on renal ischemia/reperfusion (RI/R) injury and explore the underlying mechanisms. RI/R or OGD/R models were established in mice and HK-2 cells, respectively. In vivo, vector or POP-NPs were administered (10 nmol, IV) 48 h after RI/R treatment. In the RI/R mouse model, the OIP5-AS1 and Nrf2/HO-1 expressions were down-regulated, while miR-410-3p expression was upregulated. POP-NPs treatment effectively reversed RI/R-induced renal tissue injury, restoring altered levels of blood urea nitrogen, creatinine, malondialdehyde, inflammatory factors (IL-8, IL-6, TNF-α), ROS, apoptosis, miR-410-3p, as well as the suppressed expression of SOD and Nrf2/HO-1 in the model mice. Similar results were obtained in cell models treated with POP-NPs. Additionally, miR-410-3p mimics could reverse the effects of POP-NPs on cellular models, partially counteracted by Nrf2 agonists. The binding relationship between OIP5-AS1 and miR-410-3p, alongside miR-410-3p and Nrf2, has been substantiated by dual-luciferase reporter and RNA pull-down assays. The study revealed that POP-NPs can attenuate RI/R-induced injury through miR-410-3p/Nrf2 axis. These findings lay the groundwork for future targeted therapeutic approaches utilizing nanoparticles for RI/R-induced AKI.
Collapse
Affiliation(s)
- Rongjie Zhang
- Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Xin Zhang
- Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Xuhui Zhu
- Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Tao Li
- Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Yansheng Li
- Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Peng Zhang
- Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Yuanhao Chen
- Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Gao Li
- Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Xiuwu Han
- Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| |
Collapse
|
12
|
Sun KY, Bai XY, Zhang L, Zhang X, Hu QQ, Song YX, Qiang RR, Zhang N, Zou JL, Yang YL, Xiang Y. A new strategy for the treatment of intracerebral hemorrhage: Ferroptosis. Exp Neurol 2024; 382:114961. [PMID: 39288829 DOI: 10.1016/j.expneurol.2024.114961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/28/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
Intracerebral hemorrhage, is a cerebrovascular disease with high morbidity, mortality, and disability. Due to the lack of effective clinical treatments, the development of new drugs to treat intracerebral hemorrhage is necessary. In recent years, ferroptosis has been found to play an important role in the pathophysiological process of intracerebral hemorrhage, which can be treated by inhibiting ferroptosis and thus intracerebral hemorrhage. This article aims to explain the mechanism of ferroptosis and its relationship to intracerebral hemorrhage. In the meantime, it briefly discusses the molecules identified to alleviate intracerebral hemorrhage by inhibiting ferroptosis, along with other clinical agents that are expected to treat intracerebral hemorrhage through this mechanism. In addition, a brief overview of the morphological alterations of different forms of cell death and their role in ICH is provided. Finally, the challenges that may arise in translating ferroptosis inhibitors from basic research to clinical use are presented. This article serves as a reference and provides insights to aid in the treatment of intracerebral hemorrhage in the clinic.
Collapse
Affiliation(s)
- Ke Yao Sun
- School of Medicine, Yan'an University, Yan'an, China
| | - Xin Yue Bai
- School of Medicine, Yan'an University, Yan'an, China
| | - Lei Zhang
- School of Medicine, Yan'an University, Yan'an, China
| | - Xin Zhang
- School of Medicine, Yan'an University, Yan'an, China
| | - Qian Qian Hu
- School of Medicine, Yan'an University, Yan'an, China
| | - Yu Xuan Song
- School of Medicine, Yan'an University, Yan'an, China
| | | | - Ning Zhang
- School of Medicine, Yan'an University, Yan'an, China
| | - Jia Lun Zou
- School of Medicine, Yan'an University, Yan'an, China
| | - Yan Ling Yang
- School of Medicine, Yan'an University, Yan'an, China
| | - Yang Xiang
- School of Medicine, Yan'an University, Yan'an, China; College of Physical Education, Yan'an University, Yan'an, China.
| |
Collapse
|
13
|
Miao J, Zhu H, Wang J, Chen J, Han F, Lin W. Experimental models for preclinical research in kidney disease. Zool Res 2024; 45:1161-1174. [PMID: 39257378 PMCID: PMC11491777 DOI: 10.24272/j.issn.2095-8137.2024.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 06/04/2024] [Indexed: 09/12/2024] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are significant public health issues associated with a long-term increase in mortality risk, resulting from various etiologies including renal ischemia, sepsis, drug toxicity, and diabetes mellitus. Numerous preclinical models have been developed to deepen our understanding of the pathophysiological mechanisms and therapeutic approaches for kidney diseases. Among these, rodent models have proven to be powerful tools in the discovery of novel therapeutics, while the development of kidney organoids has emerged as a promising advancement in the field. This review provides a comprehensive analysis of the construction methodologies, underlying biological mechanisms, and recent therapeutic developments across different AKI and CKD models. Additionally, this review summarizes the advantages, limitations, and challenges inherent in these preclinical models, thereby contributing robust evidence to support the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Jin Miao
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine
- Institute of Nephrology, Zhejiang University
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang 310003, China
| | - Huanhuan Zhu
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine
- Institute of Nephrology, Zhejiang University
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang 310003, China
| | - Junni Wang
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine
- Institute of Nephrology, Zhejiang University
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang 310003, China
| | - Jianghua Chen
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine
- Institute of Nephrology, Zhejiang University
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang 310003, China
| | - Fei Han
- Kidney Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine
- Institute of Nephrology, Zhejiang University
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang 310003, China. E-mail:
| | - Weiqiang Lin
- Department of Nephrology, Center for Regeneration and Aging Medicine, Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang 322000, China. E-mail:
| |
Collapse
|
14
|
Zhao X, Meng Y, Dang C, Xue L, Zhang J, Ma S, Li H. VALD-2 mitigates cisplatin-induced acute kidney injury: Mechanistic insights into oxidative stress modulation and inflammation suppression. J Biochem Mol Toxicol 2024; 38:e23786. [PMID: 39072927 DOI: 10.1002/jbt.23786] [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: 01/18/2024] [Revised: 07/02/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
This study explores the compelling antitumor properties of VALD-2, a synthetic Schiff base ligand known for its low toxicity. The focus is on investigating VALD-2's protective role against cisplatin-induced acute kidney injury (AKI) in mice, with a specific emphasis on mitigating oxidative stress and inflammation. The study involves daily intraperitoneal injections of amifostine or VALD-2 over 7 days to establish an AKI model. Subsequently, mice were assigned to normal control, cisplatin group, cisplatin + amifostine group, and cisplatin + VALD-2 10 mg/kg group, cisplatin + VALD-2 20 mg/kg, and cisplatin + VALD-2 40 mg/kg. Kidney injury is assessed through serum blood urea nitrogen (BUN) and creatinine (Cr) activity assays. Levels of inflammatory factors, tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), in kidney tissue of mice were assessed through enzyme-linked immunosorbent assay (ELISA). The protective effect of VALD-2 is further examined through HE staining to observe pathological changes in kidney injury. The ultrastructural changes of renal cells and tubular epithelial cells were observed by electron microscopy under experimental conditions, indicating the effect of VALD-2 on reversing cisplatin-induced renal injury. The study delves into VALD-2's protective mechanisms against cisplatin-induced kidney injury by using western blot analysis to assess the expression levels of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and malondialdehyde (MDA) in kidney tissues. VALD-2 demonstrates significant improvement in cisplatin-induced AKI, as evidenced by increased BUN and Cr levels. It effectively protects kidney tissue from oxidative damage, enhancing SOD and GSH-Px activities while reducing MDA levels. The study also reveals a decrease in TNF-α and IL-6 levels, supported by ELISA results, and histological findings confirm anti-nephrotoxic effects. Western blot analysis shows an upregulation of antioxidant enzymes (SOD, GSH-Px) and a reduction in MDA production. VALD-2 emerges as a promising mitigator of cisplatin-induced AKI, showcasing its ability to enhance oxidative stress-related protein expression. The findings suggest VALD-2 as a potential therapeutic agent for protecting against cisplatin-induced kidney injury.
Collapse
Affiliation(s)
- Xuhui Zhao
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Yuna Meng
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Chunyan Dang
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Li Xue
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Jing Zhang
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Shuping Ma
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Hongling Li
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| |
Collapse
|
15
|
Emad D, Bayoumi AMA, Gebril SM, Ali DME, Waz S. Modulation of keap-1/Nrf2/HO-1 and NF-ĸb/caspase-3 signaling pathways by dihydromyricetin ameliorates sodium valproate-induced liver injury. Arch Biochem Biophys 2024; 758:110084. [PMID: 38971420 DOI: 10.1016/j.abb.2024.110084] [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: 05/30/2024] [Revised: 06/30/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Nuclear factor erythroid factor 2 (Nrf2) is the key regulatory of the antioxidant response elements. Also, Nrf2 interacts with nuclear factor kappa B (NF-ĸB) to inhibit subsequent inflammatory cascade. Activation of Nrf2 signaling ameliorates drug-induced liver injury. Sodium valproate (SVP) is an anti-epilepsy drug with a hepatotoxic adverse effect that restricts its clinical use. In this study, coadministration of Dihydromyricetin (DHM), a natural flavonoid, with SVP to rats upregulated gene expression of Nrf2 and its downstream gene, heme oxygenase 1 (HO-1), while suppressed the Nrf2 repressor, Keap-1. Additionally, DHM led to downregulation of proinflammatory factors in liver tissues, including NF-ĸB, interleukin 1 beta (IL-1β), and tumor necrosis factor alpha (TNF-α). This was accompanied by a decrease in the proapoptotic protein (cleaved caspase-3) expression level. Furthermore, biochemical and histopathological studies showed that DHM treatment improved liver function and lipid profile while decreased inflammatory cell infiltration, congestion, and hepatocellular damage. According to our knowledge, prior research has not examined the protective effect of DHM on the liver injury induced by SVP. Consequently, this study provides DHM as a promising herbal medication that, when used with SVP, can prevent its induced hepatotoxicity owing to its potential anti-oxidative, anti-inflammatory, and anti-apoptotic properties.
Collapse
Affiliation(s)
- Doaa Emad
- Department of Biochemistry, Faculty of Pharmacy, Sohag University, Sohag, Egypt.
| | - Asmaa M A Bayoumi
- Department of Biochemistry, Faculty of Pharmacy, Minia University, El-Minia, 61511, Egypt.
| | - Sahar M Gebril
- Department of Histology and Cell biology, Faculty of Medicine, Sohag University, Sohag, Egypt.
| | | | - Shaimaa Waz
- Department of Biochemistry, Faculty of Pharmacy, Minia University, El-Minia, 61511, Egypt.
| |
Collapse
|
16
|
You L. Dihydromyricetin Inhibits Ferroptosis to Attenuate Cisplatin-Induced Muscle Atrophy. Physiol Res 2024; 73:405-413. [PMID: 39027957 PMCID: PMC11299785 DOI: 10.33549/physiolres.935317] [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: 01/03/2024] [Accepted: 05/14/2024] [Indexed: 07/27/2024] Open
Abstract
Cisplatin is a widely used chemotherapy drug for the treatment of various cancers. However, although cisplatin is effective in targeting cancer cells, it has severe side effects including skeletal muscle atrophy. In this study, we aimed to characterize the role of Dihydromyricetin in cisplatin-induced muscle atrophy in mice. 5-week-old male C57BL/6 mice were treated with Dihydromyricetin for 14 days orally followed by in intraperitoneally cisplatin administration for 6 days. Gastrocnemius muscles were isolated for the following experiments. Antioxidative stress were determined by peroxidative product malondialdehyde (MDA) and antioxidants superoxide dismutase (SOD) and glutathione peroxidase (GPx) activities. Quadriceps muscle mass and grip strength were significantly restored by Dihydromyricetin in a dose-dependent manner. Moreover, muscle fibers were improved in Dihydromyricetin treated group. Excessive skeletal muscle E3 ubiquitin-protein ligases in cisplatin group were significantly repressed by Dihydromyricetin treatment. Dihydromyricetin significantly reduced oxidative stress induced by cisplatin by decreasing MDA level and restored SOD and GPx activities. In addition, ferroptosis was significantly reduced by Dihydromyricetin characterized by reduced iron level and ferritin heavy chain 1 and improved Gpx4 level. The present study demonstrated that Dihydromyricetin attenuated cisplatin-induced muscle atrophy by reducing skeletal muscle E3 ubiquitin-protein ligases, oxidative stress, and ferroptosis.
Collapse
Affiliation(s)
- L You
- The Second Affiliated Hospital, Department of Clinical Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
17
|
Pan Y, Peng Z, Fang Z, Iddrisu L, Sun L, Deng Q, Gooneratne R. A Tripeptide (Ser-Arg-Pro, SRP) from Sipunculus nudus L. Improves Cadmium-Induced Acute Kidney Injury by Targeting the MAPK, Inflammatory, and Apoptosis Pathways in Mice. Mar Drugs 2024; 22:286. [PMID: 38921597 PMCID: PMC11204732 DOI: 10.3390/md22060286] [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: 05/10/2024] [Revised: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
Cadmium (Cd) is a toxic heavy metal that causes nephrosis, including acute kidney injury. To prevent and treat acute kidney injury (AKI) following Cd exposure, a tripeptide, Ser-Arg-Pro (SRP), from Sipunculus nudus L. was employed, and its potential efficacy in AKI was assessed. Oral administration of SRP significantly alleviated Cd-induced kidney damage, leading to improved renal function and the attenuation of structural abnormalities. A network pharmacology analysis revealed the potential of SRP in renal protection by targeting various pathways, including mitogen-activated protein kinase (MAPK) signaling, inflammatory response, and apoptosis pathways. Mechanistic studies indicated that SRP achieves renal protection by inhibiting the activation of MAPK pathways (phosphorylation of p38, p56, ERK, and JNK) in the oxidative stress cascade, suppressing inflammatory responses (iNOS, Arg1, Cox2, TNF-α, IL-1β, and IL-6), and restoring altered apoptosis factors (caspase-9, caspase-3, Bax, and Bcl-2). Hence, SRP has the potential to be used as a therapeutic agent for the treatment of Cd-induced nephrotoxicity.
Collapse
Affiliation(s)
- Yanmei Pan
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Marine Food, Key Laboratory of Advanced Processing of Aquatic Products of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.P.); (L.I.); (L.S.); (Q.D.)
| | - Zhilan Peng
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang 524023, China;
| | - Zhijia Fang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Marine Food, Key Laboratory of Advanced Processing of Aquatic Products of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.P.); (L.I.); (L.S.); (Q.D.)
| | - Lukman Iddrisu
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Marine Food, Key Laboratory of Advanced Processing of Aquatic Products of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.P.); (L.I.); (L.S.); (Q.D.)
| | - Lijun Sun
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Marine Food, Key Laboratory of Advanced Processing of Aquatic Products of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.P.); (L.I.); (L.S.); (Q.D.)
| | - Qi Deng
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Marine Food, Key Laboratory of Advanced Processing of Aquatic Products of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.P.); (L.I.); (L.S.); (Q.D.)
| | - Ravi Gooneratne
- Department of Wine, Food and Molecular Biosciences, Lincoln University, Lincoln 7647, New Zealand;
| |
Collapse
|
18
|
Tao X, Pan X, Zhao G, Xue M, Rui Y. Dihydromyricetin regulates KEAP1-Nrf2 pathways to enhance the survival of ischemic flap. Food Sci Nutr 2024; 12:3893-3909. [PMID: 38873488 PMCID: PMC11167164 DOI: 10.1002/fsn3.4049] [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: 10/31/2023] [Revised: 02/02/2024] [Accepted: 02/07/2024] [Indexed: 06/15/2024] Open
Abstract
In clinical flap practice, there are a lot of studies being done on how to promote the survival of distal random flap necrosis in the hypoxic and ischemic state. As a traditional Chinese medicine, dihydromyricetin (DHM) is crucial in preventing oxidative stress and apoptosis in a number of disorders. In this work, we examined the impact of DHM on the ability to survive of ischemia flaps and looked into its fundamental mechanism. Our results showed that DHM significantly increased the ischemic flaps' survival area, encouraged angiogenesis and blood flow, reduced oxidative stress and apoptosis, and stimulated KEAP1-Nrf2 (Kelch-like ECH-associated protein 1-nuclear factor erythroid 2-related factor) signaling pathways. Adeno-associated virus (AAV) upregulation of KEAP1 expression also negated the favorable effects of DHM on flap survival. By activating KEAP1-Nrf2 signaling pathways, DHM therapy promotes angiogenesis while reducing oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Xianyao Tao
- Suzhou Medical College of Soochow UniversitySuzhouJiangsuChina
- Department of Hand SurgeryWuxi Ninth People's Hospital Affiliated to Soochow UniversityWuxiJiangsuChina
| | - Xiaoyun Pan
- Department of Hand SurgeryWuxi Ninth People's Hospital Affiliated to Soochow UniversityWuxiJiangsuChina
| | - Gang Zhao
- Department of Hand SurgeryWuxi Ninth People's Hospital Affiliated to Soochow UniversityWuxiJiangsuChina
| | - Mingyu Xue
- Department of Hand SurgeryWuxi Ninth People's Hospital Affiliated to Soochow UniversityWuxiJiangsuChina
| | - Yongjun Rui
- Department of Hand SurgeryWuxi Ninth People's Hospital Affiliated to Soochow UniversityWuxiJiangsuChina
| |
Collapse
|
19
|
Jia X, Zhu L, Zhu Q, Zhang J. The role of mitochondrial dysfunction in kidney injury and disease. Autoimmun Rev 2024; 23:103576. [PMID: 38909720 DOI: 10.1016/j.autrev.2024.103576] [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: 01/22/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024]
Abstract
Mitochondria are the main sites of aerobic respiration in the cell and mainly provide energy for the organism, and play key roles in adenosine triphosphate (ATP) synthesis, metabolic regulation, and cell differentiation and death. Mitochondrial dysfunction has been identified as a contributing factor to a variety of diseases. The kidney is rich in mitochondria to meet energy needs, and stable mitochondrial structure and function are essential for normal kidney function. Recently, many studies have shown a link between mitochondrial dysfunction and kidney disease, maintaining mitochondrial homeostasis has become an important target for kidney therapy. In this review, we integrate the role of mitochondrial dysfunction in different kidney diseases, and specifically elaborate the mechanism of mitochondrial reactive oxygen species (mtROS), autophagy and ferroptosis involved in the occurrence and development of kidney diseases, providing insights for improved treatment of kidney diseases.
Collapse
Affiliation(s)
- Xueqian Jia
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, PR China
| | - Lifu Zhu
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, PR China
| | - Qixing Zhu
- Institute of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; Key Laboratory of Dermatology, Ministry of Education, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China.
| | - Jiaxiang Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, PR China; Key Laboratory of Dermatology, Ministry of Education, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China; The Center for Scientific Research, Anhui Medical University, Hefei, PR China.
| |
Collapse
|
20
|
Zhang L, Luo YL, Xiang Y, Bai XY, Qiang RR, Zhang X, Yang YL, Liu XL. Ferroptosis inhibitors: past, present and future. Front Pharmacol 2024; 15:1407335. [PMID: 38846099 PMCID: PMC11153831 DOI: 10.3389/fphar.2024.1407335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/06/2024] [Indexed: 06/09/2024] Open
Abstract
Ferroptosis is a non-apoptotic mode of programmed cell death characterized by iron dependence and lipid peroxidation. Since the ferroptosis was proposed, researchers have revealed the mechanisms of its formation and continue to explore effective inhibitors of ferroptosis in disease. Recent studies have shown a correlation between ferroptosis and the pathological mechanisms of neurodegenerative diseases, as well as diseases involving tissue or organ damage. Acting on ferroptosis-related targets may provide new strategies for the treatment of ferroptosis-mediated diseases. This article specifically describes the metabolic pathways of ferroptosis and summarizes the reported mechanisms of action of natural and synthetic small molecule inhibitors of ferroptosis and their efficacy in disease. The paper also describes ferroptosis treatments such as gene therapy, cell therapy, and nanotechnology, and summarises the challenges encountered in the clinical translation of ferroptosis inhibitors. Finally, the relationship between ferroptosis and other modes of cell death is discussed, hopefully paving the way for future drug design and discovery.
Collapse
Affiliation(s)
- Lei Zhang
- School of Medicine, Yan’an University, Yan’an, China
| | - Yi Lin Luo
- School of Medicine, Yan’an University, Yan’an, China
| | - Yang Xiang
- College of Physical Education, Yan’an University, Yan’an, China
| | - Xin Yue Bai
- School of Medicine, Yan’an University, Yan’an, China
| | | | - Xin Zhang
- School of Medicine, Yan’an University, Yan’an, China
| | - Yan Ling Yang
- School of Medicine, Yan’an University, Yan’an, China
| | - Xiao Long Liu
- School of Medicine, Yan’an University, Yan’an, China
| |
Collapse
|
21
|
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: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [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
|
22
|
Mapuskar KA, Pulliam CF, Zepeda-Orozco D, Griffin BR, Furqan M, Spitz DR, Allen BG. Redox Regulation of Nrf2 in Cisplatin-Induced Kidney Injury. Antioxidants (Basel) 2023; 12:1728. [PMID: 37760031 PMCID: PMC10525889 DOI: 10.3390/antiox12091728] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/30/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Cisplatin, a potent chemotherapeutic agent, is marred by severe nephrotoxicity that is governed by mechanisms involving oxidative stress, inflammation, and apoptosis pathways. The transcription factor Nrf2, pivotal in cellular defense against oxidative stress and inflammation, is the master regulator of the antioxidant response, upregulating antioxidants and cytoprotective genes under oxidative stress. This review discusses the mechanisms underlying chemotherapy-induced kidney injury, focusing on the role of Nrf2 in cancer therapy and its redox regulation in cisplatin-induced kidney injury. We also explore Nrf2's signaling pathways, post-translational modifications, and its involvement in autophagy, as well as examine redox-based strategies for modulating Nrf2 in cisplatin-induced kidney injury while considering the limitations and potential off-target effects of Nrf2 modulation. Understanding the redox regulation of Nrf2 in cisplatin-induced kidney injury holds significant promise for developing novel therapeutic interventions. This knowledge could provide valuable insights into potential strategies for mitigating the nephrotoxicity associated with cisplatin, ultimately enhancing the safety and efficacy of cancer treatment.
Collapse
Affiliation(s)
- Kranti A. Mapuskar
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| | - Casey F. Pulliam
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| | - Diana Zepeda-Orozco
- Pediatric Nephrology and Hypertension at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
| | - Benjamin R. Griffin
- Division of Nephrology, The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
- Department of Internal Medicine, The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Muhammad Furqan
- Department of Internal Medicine, The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| | - Bryan G. Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| |
Collapse
|