1
|
Chen C, Wang X, Zhao Y, Duan X, Hu Y, Lv Z, He Q, Yangyang Z, Wu G, Luo H, Zuo Q, Hao X, Zhao Y, Ding X, Zhang F. Exosomes inhibit ferroptosis to alleviate intervertebral disc degeneration via the p62-KEAP1-NRF2 pathway. Free Radic Biol Med 2025; 232:171-184. [PMID: 39986487 DOI: 10.1016/j.freeradbiomed.2025.02.027] [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: 11/28/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
Ferroptosis, an iron-dependent form of regulated cell death, has been reported to affect the activity of nucleus pulposus (NP) cells in the intervertebral disc (IVD), thereby contributing to intervertebral disc degeneration (IVDD). Exosomes (EXOs), extracellular nanovesicles that participate in intercellular communication, are potential therapeutic options for IVDD. Interestingly, while EXOs play an important role in inhibiting ferroptosis, whether EXOs from mesenchymal stem cells (MSCs) modulate the progression of IVDD through regulating ferroptosis is unclear. To reveal the role of ferroptosis in IVDD, IVD tissues with varying degrees of degeneration were collected and abnormal expression of ferroptosis markers was detected. Ferroptotic death was observed in TBHP-induced NP cell death in vitro, which can be specifically inhibited by the ferroptosis inhibitors DFO and Fer-1. Interestingly, MSC-derived EXOs alleviated TBHP-induced or RSL3-induced ferroptosis and rescued NP cell degeneration. Mechanistically, either an NRF2 inhibitor or p62 knockdown dampened the inhibitory effects of EXOs on ferroptosis, suggesting that EXOs attenuated oxidative stress-induced ferroptosis in NP cells by regulating the p62/KEAP1/NRF2 axis. Moreover, EXOs effectively alleviated IVDD in an in vivo rat model. The current study revealed that ferroptosis is associated with the development of IVDD. MSC-derived EXOs slowed IVDD progression by inhibiting NP cell ferroptosis through the p62/KEAP1/NRF2 signaling pathway, suggesting that EXOs are a potential therapeutic option for IVDD.
Collapse
Affiliation(s)
- Chao Chen
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, PR China; State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, PR China
| | - Xuenan Wang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, PR China; State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, PR China
| | - Yueqin Zhao
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Xianle Duan
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Yaoquan Hu
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, PR China
| | - Zhengpin Lv
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, PR China
| | - Qicong He
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, PR China
| | - Zijiu Yangyang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, PR China; State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, PR China
| | - Guishuai Wu
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, PR China
| | - Haoyan Luo
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, PR China
| | - Qianlin Zuo
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, PR China
| | - Xiaojiang Hao
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, PR China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing, 100730, PR China
| | - Yuhan Zhao
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, PR China.
| | - Xiao Ding
- State Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, PR China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing, 100730, PR China.
| | - Fan Zhang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, PR China.
| |
Collapse
|
2
|
Kong X, Liu T, Wei J. Parkinson's Disease: The Neurodegenerative Enigma Under the "Undercurrent" of Endoplasmic Reticulum Stress. Int J Mol Sci 2025; 26:3367. [PMID: 40244210 PMCID: PMC11989508 DOI: 10.3390/ijms26073367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Parkinson's disease (PD), a prevalent neurodegenerative disorder, demonstrates the critical involvement of endoplasmic reticulum stress (ERS) in its pathogenesis. This review comprehensively examines the role and molecular mechanisms of ERS in PD. ERS represents a cellular stress response triggered by imbalances in endoplasmic reticulum (ER) homeostasis, induced by factors such as hypoxia and misfolded protein aggregation, which activate the unfolded protein response (UPR) through the inositol-requiring enzyme 1 (IRE1), protein kinase R-like endoplasmic reticulum kinase (PERK), and activating transcription factor 6 (ATF6) pathways. Clinical, animal model, and cellular studies have consistently demonstrated a strong association between PD and ERS. Abnormal expression of ERS-related molecules in PD patients' brains and cerebrospinal fluid (CSF) correlates with disease progression. In animal models (e.g., Drosophila and mice), ERS inhibition alleviates dopaminergic neuronal damage. Cellular experiments reveal that PD-mimicking pathological conditions induce ERS, while interactions between ERS and mitochondrial dysfunction promote neuronal apoptosis. Mechanistically, (1) pathological aggregation of α-synuclein (α-syn) and ERS mutually reinforce dopaminergic neuron damage; (2) leucine-rich repeat kinase 2 (LRRK2) gene mutations induce ERS through thrombospondin-1 (THBS1)/transforming growth factor beta 1 (TGF-β1) interactions; (3) molecules such as Parkin and PTEN-induced kinase 1 (PINK1) regulate ERS in PD. Furthermore, ERS interacts with mitochondrial dysfunction, oxidative stress, and neuroinflammation to exacerbate neuronal injury. Emerging therapeutic strategies show significant potential, including artificial intelligence (AI)-assisted drug design targeting ERS pathways and precision medicine approaches exploring non-pharmacological interventions such as personalized electroacupuncture. Future research should focus on elucidating ERS-related mechanisms and identifying novel therapeutic targets to develop more effective treatments for PD patients, ultimately improving their quality of life.
Collapse
Affiliation(s)
- Xiangrui Kong
- Wushu College, Henan University, Kaifeng 475004, China;
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China;
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China;
| | - Jianshe Wei
- Wushu College, Henan University, Kaifeng 475004, China;
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China;
| |
Collapse
|
3
|
Li X, Li W, Xie X, Fang T, Yang J, Shen Y, Wang Y, Wang H, Tao L, Zhang H. ROS Regulate Rotenone-induced SH-SY5Y Dopamine Neuron Death Through Ferroptosis-mediated Autophagy and Apoptosis. Mol Neurobiol 2025:10.1007/s12035-025-04824-6. [PMID: 40097764 DOI: 10.1007/s12035-025-04824-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
Rotenone, a plant-derived natural insecticide, is widely used to induce Parkinson's disease (PD) models. However, the mechanisms of rotenone-induced cell death remain unclear. Here, we found that rotenone (0.01, 0.1, or 1 μmol/L) suppressed SH-SY5Y dopamine neuron viability and led to PD-like pathological changes, such as reduced tyrosine hydroxylase (TH) but increased α-synuclein. Rotenone increased the levels of intracellular reactive oxygen species (ROS) and mitochondrial ROS, as well as the levels of the antioxidants nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), ultimately resulting in oxidative stress. Moreover, rotenone significantly downregulated the expression of GPX4 and xCT but upregulated the expression of COX2 and NCOA4, which are markers of ferroptosis. Furthermore, rotenone decreased phosphorylated mTOR level but increased Beclin-1, ATG5, LC3 and p62 expression, suggesting that rotenone enhances autophagy and reduces autophagy flux. Additionally, rotenone reduced Bcl-2 levels and the mitochondrial membrane potential (MMP) while promoting BAX and Caspase-3 expression, thus initiating cell apoptosis. N-acetylcysteine (NAC), a ROS scavenger, and ferrostatin-1 (Fer-1) and deferoxamine (DFO), two ferroptosis inhibitors, significantly eliminated rotenone-induced autophagy and apoptosis. Moreover, ML385, a specific inhibitor of Nrf2, suppressed rotenone-induced ferroptosis. Our results demonstrated that ROS might mediate rotenone-induced PD-like pathological changes by regulating iron death, autophagy, and apoptosis. Inhibiting ferroptosis blocked the rotenone-induced increase in autophagy and apoptosis. Thus, the ability of ROS to regulate rotenone-induced death through autophagy and apoptosis is dependent on ferroptosis. The findings require validation in multiple neuronal cell lines and in vivo.
Collapse
Affiliation(s)
- Xinying Li
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Weiran Li
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
- Department of Clinical Medicine, School of Medicine, Qinghai University, Xining, China
| | - Xinying Xie
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Ting Fang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Jingwen Yang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Yue Shen
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Yicheng Wang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Hongyan Wang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Liqing Tao
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Heng Zhang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China.
| |
Collapse
|
4
|
Naderi S, Khodagholi F, Janahmadi M, Motamedi F, Torabi A, Batool Z, Heydarabadi MF, Pourbadie HG. Ferroptosis and cognitive impairment: Unraveling the link and potential therapeutic targets. Neuropharmacology 2025; 263:110210. [PMID: 39521042 DOI: 10.1016/j.neuropharm.2024.110210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/24/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Neurodegenerative disorders, such as Alzheimer's and Parkinson's diseases, share key characteristics, notably cognitive impairment and significant cell death in specific brain regions. Cognition, a complex mental process allowing individuals to perceive time and place, is disrupted in these conditions. This consistent disruption suggests the possibility of a shared underlying mechanism across all neurodegenerative diseases. One potential common factor is the activation of pathways leading to cell death. Despite significant progress in understanding cell death pathways, no definitive treatments have emerged. This has shifted focus towards less-explored mechanisms like ferroptosis, which holds potential due to its involvement in oxidative stress and iron metabolism. Unlike apoptosis or necrosis, ferroptosis offers a novel therapeutic avenue due to its distinct biochemical and genetic underpinnings, making it a promising target in neurodegenerative disease treatment. Ferroptosis is distinguished from other cellular death mechanisms, by distinctive characteristics such as an imbalance of iron hemostasis, peroxidation of lipids in the plasma membrane, and dysregulated glutathione metabolism. In this review, we discuss the potential role of ferroptosis in cognitive impairment. We then summarize the evidence linking ferroptosis biomarkers to cognitive impairment brought on by neurodegeneration while highlighting recent advancements in our understanding of the molecular and genetic mechanisms behind the condition. Finally, we discuss the prospective therapeutic implications of targeting ferroptosis for the treatment of cognitive abnormalities associated with neurodegeneration, including natural and synthetic substances that suppress ferroptosis via a variety of mechanisms. Promising therapeutic candidates, including antioxidants and iron chelators, are being explored to inhibit ferroptosis and mitigate cognitive decline.
Collapse
Affiliation(s)
- Soudabeh Naderi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahyar Janahmadi
- Neuroscience Research Center, Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereshteh Motamedi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolfazl Torabi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zehra Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | | | - Hamid Gholami Pourbadie
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
5
|
Jiao D, Yang Y, Wang K, Wang Y. Ferroptosis: a novel pathogenesis and therapeutic strategies for Parkinson disease: A review. Medicine (Baltimore) 2025; 104:e41218. [PMID: 39833092 PMCID: PMC11749581 DOI: 10.1097/md.0000000000041218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
Parkinson disease (PD) is the second most common neurodegenerative disease, and its incidence is climbing every year, but there is still a lack of effective clinical treatments. In recent years, many studies have shown that ferroptosis plays a key role in the progression of PD. Most importantly, many cellular and animal studies and clinical trials have shown that episodes of PD can be alleviated by inhibiting the ferroptosis process, such as utilizing inhibitors, chelating agents, and others. Here, we review the role of ferroptosis, a new form of cell death, in the pathogenesis of PD, and summarize the therapeutic strategies for targeting ferroptosis in PD, hoping to provide new thinking for the study of PD pathogenesis and the development of therapeutic strategies.
Collapse
Affiliation(s)
- Di Jiao
- School of Medicine, Zhengzhou University of Industrial Technology, Zhengzhou, China
| | - Yang Yang
- School of Medicine, Zhengzhou University of Industrial Technology, Zhengzhou, China
| | - Kejing Wang
- School of Medicine, Zhengzhou University of Industrial Technology, Zhengzhou, China
| | - Yaomei Wang
- Department of Hematology, Henan Cancer Hospital, the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
6
|
Chen J, Luo R, Li S, Shao J, Wang T, Xie S, Xu L, You Q, Feng S, Feng G. A novel NIR fluorescent probe for copper(ii) imaging in Parkinson's disease mouse brain. Chem Sci 2024; 15:13082-13089. [PMID: 39148792 PMCID: PMC11323298 DOI: 10.1039/d4sc03445g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/12/2024] [Indexed: 08/17/2024] Open
Abstract
Abnormal copper ion (Cu2+) levels are considered to be one of the pathological factors of Parkinson's disease (PD), but the internal relationship between Cu2+ and PD progression remains elusive. Visualizing Cu2+ in the brain will be pivotal for comprehending the underlying pathophysiological processes of PD. In this work, a near-infrared (NIR) fluorescent probe, DDAO-Cu, capable of detecting Cu2+ with exceptional sensitivity (about 1.8 nM of detection limit) and selectivity, rapid response (<3 min), and deep tissue penetration, was designed for quantification and visualization of the Cu2+ level. It could detect not only Cu2+ in cells but also the changes in the Cu2+ level in the rotenone-induced cell and zebrafish PD models. Moreover, DDAO-Cu can cross the blood-brain barrier to image Cu2+ in the brain of PD model mice. The imaging result showed a significant increase in Cu2+ levels in brain regions of PD model mice, including the cerebral cortex, hippocampus, and striatum. Meanwhile, Cu2+ levels in the substantia nigra region were significantly reduced in PD model mice. It revealed the nuanced relationship of Cu2+ levels in different brain regions in the disease and indicated the pathological complexity of PD. Overall, DDAO-Cu represents a novel and practical tool for investigating Cu2+-related physiological and pathological processes underlying Parkinson's disease.
Collapse
Affiliation(s)
- Jianmei Chen
- School of Pharmacy, Hubei University of Chinese Medicine Wuhan 430065 China
| | - Rongqing Luo
- School of Pharmacy, Hubei University of Chinese Medicine Wuhan 430065 China
| | - Shuang Li
- School of Pharmacy, Hubei University of Chinese Medicine Wuhan 430065 China
| | - Jinping Shao
- School of Pharmacy, Hubei University of Chinese Medicine Wuhan 430065 China
| | - Ting Wang
- School of Pharmacy, Hubei University of Chinese Medicine Wuhan 430065 China
| | - Shumei Xie
- School of Pharmacy, Hubei University of Chinese Medicine Wuhan 430065 China
| | - Li Xu
- School of Pharmacy, Hubei University of Chinese Medicine Wuhan 430065 China
- Hubei Shizhen Laboratory, Hubei University of Chinese Medicine Wuhan 430065 China
| | - Qiuyun You
- School of Pharmacy, Hubei University of Chinese Medicine Wuhan 430065 China
- Hubei Shizhen Laboratory, Hubei University of Chinese Medicine Wuhan 430065 China
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education Wuhan 430065 China
| | - Shumin Feng
- School of Pharmacy, Hubei University of Chinese Medicine Wuhan 430065 China
- Hubei Shizhen Laboratory, Hubei University of Chinese Medicine Wuhan 430065 China
| | - Guoqiang Feng
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, College of Chemistry, Central China Normal University 152 Luoyu Road Wuhan 430079 PR China
| |
Collapse
|
7
|
Zhang X, Li G, Chen H, Nie XW, Bian JS. Targeting NKAα1 to treat Parkinson's disease through inhibition of mitophagy-dependent ferroptosis. Free Radic Biol Med 2024; 218:190-204. [PMID: 38574977 DOI: 10.1016/j.freeradbiomed.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/04/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
Dysfunction of the Na+/K+-ATPase (NKA) has been documented in various neurodegenerative diseases, yet the specific role of NKAα1 in Parkinson's disease (PD) remains incompletely understood. In this investigation, we utilized NKAα1 haploinsufficiency (NKAα1+/-) mice to probe the influence of NKAα1 on dopaminergic (DA) neurodegeneration induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Our findings reveal that NKAα1+/- mice displayed a heightened loss of DA neurons and more pronounced motor dysfunction compared to the control group when exposed to MPTP. Intriguingly, this phenomenon coincided with the activation of ferroptosis and impaired mitophagy both in vivo and in vitro. To scrutinize the role and underlying mechanism of NKAα1 in PD, we employed DR-Ab, an antibody targeting the DR-region of the NKA α subunit. Our study demonstrates that the administration of DR-Ab effectively reinstated the membrane abundance of NKAα1, thereby mitigating MPTP-induced DA neuron loss and subsequent improvement in behavioral deficit. Mechanistically, DR-Ab heightened the formation of the surface NKAα1/SLC7A11 complex, inhibiting SLC7A11-dependent ferroptosis. Moreover, DR-Ab disrupted the cytosolic interaction between NKAα1 and Parkin, facilitating the translocation of Parkin to mitochondria and enhancing the process of mitophagy. In conclusion, this study establishes NKAα1 as a key regulator of ferroptosis and mitophagy, identifying its DR-region as a promising therapeutic target for PD.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Guanghong Li
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Hanbin Chen
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Xiao-Wei Nie
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (the First Affiliated Hospital, Southern University of Science and Technology; the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518055, China.
| | - Jin-Song Bian
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
8
|
Gu Y, Lv L, Jin J, Hua X, Xu Q, Wu R, Zhu S, Liu X, Lv T, Song Y, Zhang F. STING mediates LPS-induced acute lung injury by regulating ferroptosis. Exp Cell Res 2024; 438:114039. [PMID: 38641125 DOI: 10.1016/j.yexcr.2024.114039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/21/2024]
Abstract
The pathogenesis of acute lung injury is not fully understood. Stimulator of interferon genes (STING) and ferroptosis have been implicated in various pathological and physiological processes, including acute lung injury (ALI). However, the relationship between STING and ferroptosis in lipopolysaccharide (LPS)-induced ALI is unclear. We found that LPS stimulation activated STING and ferroptosis. Furthermore, STING knockout and ferroptosis inhibitor alleviated lung inflammation and epithelial cell damage. Also, STING knockout reduced inflammation injury and ferroptosis. Notably, the ferroptosis inducer reversed the alleviation of inflammation caused by STING knockout. These results show that STING participates in the inflammation injury of ALI by regulating ferroptosis. Results also showed that p-STAT3 levels increased after STING knockout, suggesting that STING negatively regulates STAT3 activation. Besides, STAT3 inhibitor aggravated ferroptosis after STING knockout, indicating that STING regulates ferroptosis through STAT3 signaling. In conclusion, STING mediates LPS-induced ALI by regulating ferroptosis, indicating that STING and ferroptosis may be new targets for ALI treatment.
Collapse
Affiliation(s)
- Yanli Gu
- Department of Respiratory and Critical Care Medicine, The Affiliated Huaian No.1 People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liting Lv
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiajia Jin
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xin Hua
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Southeast University Medical College, Nanjing, Jiangsu, China
| | - Qiuli Xu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Southeast University Medical College, Nanjing, Jiangsu, China
| | - RanPu Wu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Southeast University Medical College, Nanjing, Jiangsu, China
| | - Suhua Zhu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Xin Liu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Tangfeng Lv
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Yong Song
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China.
| | - Fang Zhang
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China.
| |
Collapse
|
9
|
Chen Y, Wu Z, Li S, Chen Q, Wang L, Qi X, Tian C, Yang M. Mapping the Research of Ferroptosis in Parkinson's Disease from 2013 to 2023: A Scientometric Review. Drug Des Devel Ther 2024; 18:1053-1081. [PMID: 38585257 PMCID: PMC10999190 DOI: 10.2147/dddt.s458026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024] Open
Abstract
Methods Related studies on PD and ferroptosis were searched in Web of Science Core Collection (WOSCC) from inception to 2023. VOSviewer, CiteSpace, RStudio, and Scimago Graphica were employed as bibliometric analysis tools to generate network maps about the collaborations between authors, countries, and institutions and to visualize the co-occurrence and trends of co-cited references and keywords. Results A total of 160 original articles and reviews related to PD and ferroptosis were retrieved, produced by from 958 authors from 162 institutions. Devos David was the most prolific author, with 9 articles. China and the University of Melbourne had leading positions in publication volume with 84 and 12 publications, respectively. Current hot topics focus on excavating potential new targets for treating PD based on ferroptosis by gaining insight into specific molecular mechanisms, including iron metabolism disorders, lipid peroxidation, and imbalanced antioxidant regulation. Clinical studies aimed at treating PD by targeting ferroptosis remain in their preliminary stages. Conclusion A continued increase was shown in the literature within the related field over the past decade. The current study suggested active collaborations among authors, countries, and institutions. Research into the pathogenesis and treatment of PD based on ferroptosis has remained a prominent topic in the field in recent years, indicating that ferroptosis-targeted therapy is a potential approach to halting the progression of PD.
Collapse
Affiliation(s)
- Yingfan Chen
- Medical School of Chinese People’s Liberation Army, Beijing, People’s Republic of China
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Zhenhui Wu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, People’s Republic of China
| | - Shaodan Li
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Qi Chen
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Liang Wang
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Xiaorong Qi
- Medical School of Chinese People’s Liberation Army, Beijing, People’s Republic of China
| | - Chujiao Tian
- Medical School of Chinese People’s Liberation Army, Beijing, People’s Republic of China
| | - Minghui Yang
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
10
|
He G, Zhang Y, Feng Y, Chen T, Liu M, Zeng Y, Yin X, Qu S, Huang L, Ke Y, Liang L, Yan J, Liu W. SBFI26 induces triple-negative breast cancer cells ferroptosis via lipid peroxidation. J Cell Mol Med 2024; 28:e18212. [PMID: 38516826 PMCID: PMC10958404 DOI: 10.1111/jcmm.18212] [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: 08/18/2023] [Revised: 01/13/2024] [Accepted: 02/09/2024] [Indexed: 03/23/2024] Open
Abstract
SBFI26, an inhibitor of FABP5, has been shown to suppress the proliferation and metastasis of tumour cells. However, the underlying mechanism by which SBFI26 induces ferroptosis in breast cancer cells remains largely unknown. Three breast cancer cell lines were treated with SBFI26 and CCK-8 assessed cytotoxicity. Transcriptome was performed on the Illumina platform and verified by qPCR. Western blot evaluated protein levels. Malondialdehyde (MDA), total superoxide dismutase (T-SOD), Fe, glutathione (GSH) and oxidized glutathione (GSSG) were measured. SBFI26 induced cell death time- and dose-dependent, with a more significant inhibitory effect on MDA-MB-231 cells. Fer-1, GSH and Vitamin C attenuated the effects but not erastin. RNA-Seq analysis revealed that SBFI26 treatment significantly enriched differentially expressed genes related to ferroptosis. Furthermore, SBFI26 increased intracellular MDA, iron ion, and GSSG levels while decreasing T-SOD, total glutathione (T-GSH), and GSH levels.SBFI26 dose-dependently up-regulates the expression of HMOX1 and ALOX12 at both gene and protein levels, promoting ferroptosis. Similarly, it significantly increases the expression of SAT1, ALOX5, ALOX15, ALOXE3 and CHAC1 that, promoting ferroptosis while downregulating the NFE2L2 gene and protein that inhibit ferroptosis. SBFI26 leads to cellular accumulation of fatty acids, which triggers excess ferrous ions and subsequent lipid peroxidation for inducing ferroptosis.
Collapse
Affiliation(s)
- Gang He
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Yiyuan Zhang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Yanjiao Feng
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Tangcong Chen
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Mei Liu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Yue Zeng
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Xiaojing Yin
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Shaokui Qu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Lifen Huang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Youqiang Ke
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Li Liang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Jun Yan
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| | - Wei Liu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education DepartmentSichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu UniversityChengduChina
| |
Collapse
|
11
|
Jia F, Han J. COVID-19 related neurological manifestations in Parkinson's disease: has ferroptosis been a suspect? Cell Death Discov 2024; 10:146. [PMID: 38503730 PMCID: PMC10951317 DOI: 10.1038/s41420-024-01915-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 03/21/2024] Open
Abstract
A rising number of patient cases point to a probable link between SARS-CoV-2 infection and Parkinson's disease (PD), yet the mechanisms by which SARS-CoV-2 affects the brain and generates neuropsychiatric symptoms in COVID-19 patients remain unknown. Ferroptosis, a distinct iron-dependent non-apoptotic type of cell death characterized by lipid peroxidation and glutathione depletion, a key factor in neurological disorders. Ferroptosis may have a pathogenic role in COVID-19, according to recent findings, however its potential contributions to COVID-19-related PD have not yet been investigated. This review covers potential paths for SARS-CoV-2 infection of the brain. Among these putative processes, ferroptosis may contribute to the etiology of COVID-19-associated PD, potentially providing therapeutic methods.
Collapse
Affiliation(s)
- Fengju Jia
- School of Nursing, Qingdao University, No. 308 Ningxia Road, Qingdao, 266071, China.
| | - Jing Han
- School of Nursing, Qingdao University, No. 308 Ningxia Road, Qingdao, 266071, China
| |
Collapse
|
12
|
Tan Y, Dong X, Zhuang D, Cao B, Jiang H, He Q, Zhao M. Emerging roles and therapeutic potentials of ferroptosis: from the perspective of 11 human body organ systems. Mol Cell Biochem 2023; 478:2695-2719. [PMID: 36913150 DOI: 10.1007/s11010-023-04694-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 02/26/2023] [Indexed: 03/14/2023]
Abstract
Since ferroptosis was first described as an iron-dependent cell death pattern in 2012, there has been increasing interest in ferroptosis research. In view of the immense potential of ferroptosis in treatment efficacy and its rapid development in recent years, it is essential to track and summarize the latest research in this field. However, few writers have been able to draw on any systematic investigation into this field based on human body organ systems. Hence, in this review, we provide a comprehensive description of the latest progress in unveiling the roles and functions, as well as the therapeutic potential of ferroptosis, in treating diseases from the aspects of 11 human body organ systems (including the nervous system, respiratory system, digestive system, urinary system, reproductive system, integumentary system, skeletal system, immune system, cardiovascular system, muscular system, and endocrine system) in the hope of providing references for further understanding the pathogenesis of related diseases and bringing an innovative train of thought for reformative clinical treatment.
Collapse
Affiliation(s)
- Yaochong Tan
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
- Medical School of Xiangya, Central South University, Changsha, 410013, Hunan, China
| | - Xueting Dong
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
- Medical School of Xiangya, Central South University, Changsha, 410013, Hunan, China
| | - Donglin Zhuang
- Department of Structural Heart Disease, National Center for Cardiovascular Disease, China & Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Buzi Cao
- Hunan Normal University School of Medicine, Changsha, 410081, Hunan, China
| | - Hua Jiang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China.
| | - Qingnan He
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| | - Mingyi Zhao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
13
|
Nasoni MG, Crinelli R, Iuliano L, Luchetti F. When nitrosative stress hits the endoplasmic reticulum: Possible implications in oxLDL/oxysterols-induced endothelial dysfunction. Free Radic Biol Med 2023; 208:178-185. [PMID: 37544487 DOI: 10.1016/j.freeradbiomed.2023.08.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/14/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
Oxidized LDL (oxLDL) and oxysterols are known to play a crucial role in endothelial dysfunction (ED) by inducing endoplasmic reticulum stress (ERS), inflammation, and apoptosis. However, the precise molecular mechanisms underlying these pathophysiological processes remain incompletely understood. Emerging evidence strongly implicates excessive nitric oxide (NO) production in the progression of various pathological conditions. The accumulation of reactive nitrogen species (RNS) leading to nitrosative stress (NSS) and aberrant protein S-nitrosylation contribute to NO toxicity. Studies have highlighted the involvement of NSS and S-nitrosylation in perturbing ER signaling through the modification of ER sensors and resident isomerases in neurons. This review focuses on the existing evidence that strongly associates NO with ERS and the possible implications in the context of ED induced by oxLDL and oxysterols. The potential effects of perturbed NO synthesis on signaling effectors linking NSS with ERS in endothelial cells are discussed to provide a conceptual framework for further investigations and the development of novel therapeutic strategies targeting ED.
Collapse
Affiliation(s)
- M G Nasoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| | - R Crinelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| | - L Iuliano
- Department of Medico-Surgical Sciences and Biotechnology, Sapienza University of Rome, Latina, Italy.
| | - F Luchetti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| |
Collapse
|
14
|
Ding XS, Gao L, Han Z, Eleuteri S, Shi W, Shen Y, Song ZY, Su M, Yang Q, Qu Y, Simon DK, Wang XL, Wang B. Ferroptosis in Parkinson's disease: Molecular mechanisms and therapeutic potential. Ageing Res Rev 2023; 91:102077. [PMID: 37742785 DOI: 10.1016/j.arr.2023.102077] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/26/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
Parkinson's Disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra (SN), leading to motor and non-motor symptoms. While the exact mechanisms remain complex and multifaceted, several molecular pathways have been implicated in PD pathology, including accumulation of misfolded proteins, impaired mitochondrial function, oxidative stress, inflammation, elevated iron levels, etc. Overall, PD's molecular mechanisms involve a complex interplay between genetic, environmental, and cellular factors that disrupt cellular homeostasis, and ultimately lead to the degeneration of dopaminergic neurons. Recently, emerging evidence highlights ferroptosis, an iron-dependent non-apoptotic cell death process, as a pivotal player in the advancement of PD. Notably, oligomeric α-synuclein (α-syn) generates reactive oxygen species (ROS) and lipid peroxides within cellular membranes, potentially triggering ferroptosis. The loss of dopamine, a hallmark of PD, could predispose neurons to ferroptotic vulnerability. This unique form of cell demise unveils fresh insights into PD pathogenesis, necessitating an exploration of the molecular intricacies connecting ferroptosis and PD progression. In this review, the molecular and regulatory mechanisms of ferroptosis and their connection with the pathological processes of PD have been systematically summarized. Furthermore, the features of ferroptosis in PD animal models and clinical trials targeting ferroptosis as a therapeutic approach in PD patients' management are scrutinized.
Collapse
Affiliation(s)
- Xv-Shen Ding
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China; Basic Medicine School, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Li Gao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Zheng Han
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Simona Eleuteri
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle 628H, Boston, MA 02215, USA
| | - Wei Shi
- Department of Neurosurgery, PLA 960th hospital, JiNan, Shandong Province, 250031, China
| | - Yun Shen
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Zi-Yao Song
- Basic Medicine School, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Mingming Su
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Qian Yang
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China.
| | - David K Simon
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle 628H, Boston, MA 02215, USA.
| | - Xue-Lian Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China.
| | - Bao Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China.
| |
Collapse
|
15
|
Li L, Guo L, Gao R, Yao M, Qu X, Sun G, Fu Q, Hu C, Han G. Ferroptosis: a new regulatory mechanism in neuropathic pain. Front Aging Neurosci 2023; 15:1206851. [PMID: 37810619 PMCID: PMC10556472 DOI: 10.3389/fnagi.2023.1206851] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 09/11/2023] [Indexed: 10/10/2023] Open
Abstract
Neuropathic pain (NP) is pain caused by damage to the somatosensory system. It is a common progressive neurodegenerative disease that usually presents with clinical features such as spontaneous pain, touch-evoked pain, nociceptive hyperalgesia, and sensory abnormalities. Due to the complexity of the mechanism, NP often persists. In addition to the traditionally recognized mechanisms of peripheral nerve damage and central sensitization, excessive iron accumulation, oxidative stress, neuronal inflammation, and lipid peroxidation damage are distinctive features of NP in pathophysiology. However, the mechanisms linking these pathological features to NP are not fully understood. The complexity of the pathogenesis of NP greatly limits the development of therapeutic approaches for NP. Ferroptosis is a novel form of cell death discovered in recent years, in which cell death is usually accompanied by massive iron accumulation and lipid peroxidation. Ferroptosis-inducing factors can affect glutathione peroxidase directly or indirectly through different pathways, leading to decreased antioxidant capacity and accumulation of lipid reactive oxygen species (ROS) in cells, ultimately leading to oxidative cell death. It has been shown that ferroptosis is closely related to the pathophysiological process of many neurological disorders such as NP. Possible mechanisms involved are changes in intracellular iron ion levels, alteration of glutamate excitability, and the onset of oxidative stress. However, the functional changes and specific molecular mechanisms of ferroptosis during this process still need to be further explored. How to intervene in the development of NP by regulating cellular ferroptosis has become a hot issue in etiological research and treatment. In this review, we systematically summarize the recent progress of ferroptosis research in NP, to provide a reference for further understanding of its pathogenesis and propose new targets for treatment.
Collapse
Affiliation(s)
- Lu Li
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lingling Guo
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Rui Gao
- Department of Anesthesiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Mengwen Yao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xinyu Qu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Guangwei Sun
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qi Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Cuntao Hu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Guang Han
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
16
|
Mansour HM, F Mohamed A, Khattab MM, El-Khatib AS. Lapatinib ditosylate rescues motor deficits in rotenone-intoxicated rats: Potential repurposing of anti-cancer drug as a disease-modifying agent in Parkinson's disease. Eur J Pharmacol 2023; 954:175875. [PMID: 37385578 DOI: 10.1016/j.ejphar.2023.175875] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/13/2023] [Accepted: 06/20/2023] [Indexed: 07/01/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by motor deficits induced by dopaminergic neuronal death in the substantia nigra (SN). Finding a successful neuroprotective therapy is still challenging despite improved knowledge of the etiology of PD and a variety of medications intended to reduce symptoms. Lapatinib (LAP), an FDA-approved anti-cancer medication, has been stated to exert its effect through the modulation of oxidative stress. Furthermore, recent studies display the neuroprotective effects of LAP in epilepsy, encephalomyelitis, and Alzheimer's disease in rodent models through the modulation of oxidative stress and ferroptosis. Nevertheless, it is questionable whether LAP exerts neuroprotective effects in PD. In the current study, administration of 100 mg/kg LAP in rotenone-treated rats for 21 days ameliorates motor impairment, debilitated histopathological alterations, and revived dopaminergic neurons by increasing tyrosine hydroxylase (TH) expression in SN, along with increased dopamine level. LAP remarkably restored the antioxidant defense mechanism system, GPX4/GSH/NRF2 axis, inhibiting oxidative markers, including iron, TfR1, PTGS2, and 4-HNE, along with suppression of p-EGFR/c-SRC/PKCβII/PLC-γ/ACSL-4 pathway. Moreover, LAP modulates HSP90/CDC37 chaperone complex, regulating many key pathological markers of PD, including LRRK2, c-ABL, and α-syn. It is concluded that LAP has neuroprotective effects in PD via modulation of many key parameters implicated in PD pathogenesis. Taken together, the current study offers insights into the potential repositioning of LAP as a disease-modifying drug in PD.
Collapse
Affiliation(s)
- Heba M Mansour
- Central Administration of Biological, Innovative Products, and Clinical Studies, Egyptian Drug Authority, EDA, Giza, Egypt
| | - Ahmed F Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Mahmoud M Khattab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Aiman S El-Khatib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
17
|
Krishnan K, Rao M, Chang N, Casazza M, Rasmussen LK. Novel Serum Biomarkers Associated With Pediatric Hepatic Encephalopathy: A Systematic Review. J Pediatr Gastroenterol Nutr 2023; 77:16-23. [PMID: 37084331 DOI: 10.1097/mpg.0000000000003801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/23/2023]
Abstract
BACKGROUND The pathophysiology of pediatric hepatic encephalopathy (HE) is not well understood. Various serum biomarkers associated with HE may provide insight into its pathology, but their use and interpretation in clinical practice for diagnosis and prognostication remain undetermined. We sought to investigate reported correlations of serum biomarkers with presence and degree of HE in children. METHODS We conducted a systematic review of studies examining novel serum biomarkers and cytokines in association with HE that included children on PubMed, Embase, Lilacs, and Scopus. We utilized Covidence for abstract and text review by 2 independent reviewers for each study. RESULTS We reviewed 2824 unique publications; 15 met criteria for inclusion. Categories of biomarkers reported were inflammatory cytokines, products of amino acid metabolism, trace elements and vitamins, and hepatic and neuro biomarkers. Of 19 individual biomarkers, only 5 were measured in more than 1 study. Elevations in interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha) were most commonly reported as associated with HE. Notably, we observed lower average IL-6 and TNF-alpha levels in pediatric-only studies compared to mixed age studies. Overall, high bias and poor applicability to our review question was observed. We encountered low numbers of studies with pediatric focus, and few conducted with low bias study designs. CONCLUSION Investigated biomarkers span a large range of categories and suggest potentially useful correlations with HE. Further well-designed prospective biomarker research is necessary to better elucidate the pathogenesis of HE in children and improve early detection and clinical care.
Collapse
Affiliation(s)
- Kavita Krishnan
- From Loyola University Stritch School of Medicine, Maywood, IL
| | - Mahil Rao
- the Department of Pediatrics, Division of Pediatric Critical Care, Stanford University, Palo Alto, CA
| | - Nathan Chang
- the Department of Pediatrics, Division of Pediatric Critical Care, Stanford University, Palo Alto, CA
| | - May Casazza
- the Department of Neurosurgery, Division of Pediatric Neurosurgery, Stanford University, Palo Alto, CA
| | - Lindsey K Rasmussen
- the Department of Pediatrics, Division of Pediatric Critical Care, Stanford University, Palo Alto, CA
| |
Collapse
|
18
|
Zhirong Z, Li H, Yi L, Lichen Z, Ruiwu D. Ferroptosis in pancreatic diseases: potential opportunities and challenges that require attention. Hum Cell 2023; 36:1233-1243. [PMID: 36929283 DOI: 10.1007/s13577-023-00894-7] [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: 12/19/2022] [Accepted: 03/07/2023] [Indexed: 03/18/2023]
Abstract
The pancreas is an abdominal organ with both endocrine and exocrine functions, and patients with pancreatic diseases suffer tremendously. The regulated cell death of various cells in the pancreas is thought to play a key role in disease development. As one of the newly discovered regulated cell death modalities, ferroptosis has the potential for therapeutic applications in the study of multiple diseases. Ferroptosis has been observed in several pancreatic diseases, but its role in pancreatic diseases has not been systematically elucidated or reviewed. Understanding the occurrence of ferroptosis in various pancreatic diseases after damage to the different cell types is crucial in determining disease progression, evaluating targeted therapies, and predicting disease prognosis. Herein, we summarize the research progress associated with ferroptosis in four common pancreatic diseases, namely acute pancreatitis, chronic pancreatitis, pancreatic ductal adenocarcinoma, and diabetes mellitus. Furthermore, the elucidation of ferroptosis in rare pancreatic diseases may provide sociological benefits in the future.
Collapse
Affiliation(s)
- Zhao Zhirong
- General Surgery Center, General Hospital of Western Theater Command, No. 270, Rongdu Rd, Jinniu District, Chengdu, 610083, Sichuan, China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Han Li
- Ultrasound Medical Center, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Liu Yi
- School of Medicine, Jianghan University, Wuhan, 430056, Hubei, China
| | - Zhou Lichen
- General Surgery Center, General Hospital of Western Theater Command, No. 270, Rongdu Rd, Jinniu District, Chengdu, 610083, Sichuan, China
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Dai Ruiwu
- General Surgery Center, General Hospital of Western Theater Command, No. 270, Rongdu Rd, Jinniu District, Chengdu, 610083, Sichuan, China.
- College of Medicine, Southwest Jiaotong University, Chengdu, China.
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, General Hospital of Western Theater Command, Chengdu, Sichuan, China.
| |
Collapse
|
19
|
FERROPTOSIS IN THE PATHOGENESIS OF BRAIN TUMORS. КЛИНИЧЕСКАЯ ПРАКТИКА 2022. [DOI: 10.17816/clinpract114787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The field of research on ferroptosis has seen explosive growth in the past few years since the term was coined in 2012. This review highlights the current state of knowledge on the developmental mechanism of this unique mode of cell death, induced by iron-dependent phospholipid peroxidation and which is regulated by a variety of cellular metabolic events, including redox homeostasis. Among these factors, the xCT. system, an amino acid antiporter that supports the synthesis of glutathione (GSH) and oxidation protection. The risk of iron accumulation in neurons, astrocytes, oligodendrocytes, microglia, and Schwann cells and the development of oxidative stress is discussed. Ferroptosis triggers a cascade of events including activation of inflammation, oxidation of neurotransmitters, impaired neuronal communication, myelin sheath degeneration, astrocyte dysregulation, dementia, and cell death. On the other hand, the exceptional vulnerability to ferroptosis of cancer cells originating from the nervous tissue is estimated. Evidence is given for the initiation of ferroptosis in tumor cells as a factor in inhibiting the growth and death of these cells. Particular attention is paid to the pharmacological modulation of ferroptosis through its induction and inhibition for the treatment of drug-resistant cancers. The choice of targets for the induction of ferroptosis in cancer cells is discussed. Glutathione peroxidase 4 and xCT amino acid antiporter are recognized as the most preferred targets and the antitumor potential of their inhibition and side effects are being evaluated.
Collapse
|
20
|
Prasad M K, Mohandas S, Kunka Mohanram R. Role of ferroptosis inhibitors in the management of diabetes. Biofactors 2022; 49:270-296. [PMID: 36468443 DOI: 10.1002/biof.1920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022]
Abstract
Ferroptosis, the iron-dependent, lipid peroxide-mediated cell death, has garnered attention due to its critical involvement in crucial physiological and pathological cellular processes. Indeed, several studies have attributed its role in developing a range of disorders, including diabetes. As accumulating evidence further the understanding of ferroptotic mechanisms, the impact this specialized mode of cell death has on diabetic pathogenesis is still unclear. Several in vivo and in vitro studies have highlighted the association of ferroptosis with beta-cell death and insulin resistance, supported by observations of marked alterations in ferroptotic markers in experimental diabetes models. The constant improvement in understanding ferroptosis in diabetes has demonstrated it as a potential therapeutic target in diabetic management. In this regard, ferroptosis inhibitors promise to rescue pancreatic beta-cell function and alleviate diabetes and its complications. This review article elucidates the key ferroptotic pathways that mediate beta-cell death in diabetes, and its complications. In particular, we share our insight into the cross talk between ferroptosis and other hallmark pathogenic mediators such as oxidative and endoplasmic reticulum stress regulators relevant to diabetes progression. Further, we extensively summarize the recent developments on the role of ferroptosis inhibitors and their therapeutic action in alleviating diabetes and its complications.
Collapse
Affiliation(s)
- Krishna Prasad M
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Sundhar Mohandas
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Ramkumar Kunka Mohanram
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| |
Collapse
|
21
|
Liu C, Liu Y, Ma B, Zhou M, Zhao X, Fu X, Kan S, Hu W, Zhu R. Mitochondrial regulatory mechanisms in spinal cord injury: A narrative review. Medicine (Baltimore) 2022; 101:e31930. [PMID: 36401438 PMCID: PMC9678589 DOI: 10.1097/md.0000000000031930] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Spinal cord injury is a severe central nervous system injury that results in the permanent loss of motor, sensory, and autonomic functions below the level of injury with limited recovery. The pathological process of spinal cord injury includes primary and secondary injuries, characterized by a progressive cascade. Secondary injury impairs the ability of the mitochondria to maintain homeostasis and leads to calcium overload, excitotoxicity, and oxidative stress, further exacerbating the injury. The defective mitochondrial function observed in these pathologies accelerates neuronal cell death and inhibits regeneration. Treatment of spinal cord injury by preserving mitochondrial biological function is a promising, although still underexplored, therapeutic strategy. This review aimed to explore mitochondrial-based therapeutic advances after spinal cord injury. Specifically, it briefly describes the characteristics of spinal cord injury. It then broadly discusses the drugs used to protect the mitochondria (e.g., cyclosporine A, acetyl-L-carnitine, and alpha-tocopherol), phenomena associated with mitochondrial damage processes (e.g., mitophagy, ferroptosis, and cuproptosis), mitochondrial transplantation for nerve cell regeneration, and innovative mitochondrial combined protection therapy.
Collapse
Affiliation(s)
- Chengjiang Liu
- Department of Spine Surgery, Tianjin Union Medical Center Tianjin, Tianjin, China
| | - Yidong Liu
- Department of Spine Surgery, Tianjin Union Medical Center Tianjin, Tianjin, China
| | - Boyuan Ma
- Department of Spine Surgery, Tianjin Union Medical Center Tianjin, Tianjin, China
| | - Mengmeng Zhou
- Department of Spine Surgery, Tianjin Union Medical Center Tianjin, Tianjin, China
| | - Xinyan Zhao
- Department of Spine Surgery, Tianjin Union Medical Center Tianjin, Tianjin, China
| | - Xuanhao Fu
- Department of Spine Surgery, Tianjin Union Medical Center Tianjin, Tianjin, China
| | - Shunli Kan
- Department of Spine Surgery, Tianjin Union Medical Center Tianjin, Tianjin, China
| | - Wei Hu
- Department of Spine Surgery, Tianjin Union Medical Center Tianjin, Tianjin, China
| | - Rusen Zhu
- Department of Spine Surgery, Tianjin Union Medical Center Tianjin, Tianjin, China
- *Correspondence: Rusen Zhu, Department of Spine Surgery, Tianjin Union Medical Center190jieyuan Road, Honggiao District, Tianjin 300121, China (e-mail: )
| |
Collapse
|
22
|
Li C, Wu Z, Xue H, Gao Q, Zhang Y, Wang C, Zhao P. Ferroptosis contributes to hypoxic-ischemic brain injury in neonatal rats: Role of the SIRT1/Nrf2/GPx4 signaling pathway. CNS Neurosci Ther 2022; 28:2268-2280. [PMID: 36184790 PMCID: PMC9627393 DOI: 10.1111/cns.13973] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/04/2022] [Accepted: 09/08/2022] [Indexed: 02/06/2023] Open
Abstract
AIMS Hypoxic-ischemic brain injury (HIBI) often results in cognitive impairments. Herein, we investigated the roles of ferroptosis in HIBI and the underlying signaling pathways. METHODS Ferrostatin-1 (Fer-1) or resveratrol (Res) treatments were administered intracerebroventricularly 30 min before HIBI in 7-day-old rats. Glutathione peroxidase 4 (GPx4) expression, malondialdehyde (MDA) concentration, iron content, mitochondrial morphology, and the expression of silent information regulator factor 2-related enzyme 1 (SIRT1) and nuclear factor erythroid-2-related factor 2 (Nrf2) were measured after HIBI. Additionally, the weight ratio of left/right hemisphere, brain morphology, Nissl staining, and the Morris water maze test were conducted to estimate brain damage. RESULTS At 24-h post-HIBI, GPx4 expression was decreased, and MDA concentration and iron content were increased in the hippocampus. HIBI led to mitochondrial atrophy, brain atrophy/damage, and resultant learning and memory impairments, which were alleviated by Fer-1-mediated inhibition of ferroptosis. Furthermore, Res-mediated SIRT1 upregulation increased Nrf2 and GPx4 expression, thereby attenuating ferroptosis, reducing brain atrophy/damage, and improving learning and memory abilities. CONCLUSION The results demonstrated that during HIBI, ferroptosis occurs via the SIRT1/Nrf2/GPx4 signaling pathway, suggesting it as a potential therapeutic target for inhibiting ferroptosis and ameliorating HIBI-induced cognitive impairments.
Collapse
Affiliation(s)
- Chang Li
- Department of AnesthesiologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Ziyi Wu
- Department of AnesthesiologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Hang Xue
- Department of AnesthesiologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Qiushi Gao
- Department of AnesthesiologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Yahan Zhang
- Department of AnesthesiologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Changming Wang
- Department of AnesthesiologyPeople's Hospital of China Medical University (Liaoning Provincial People's Hospital)ShenyangLiaoningChina
| | - Ping Zhao
- Department of AnesthesiologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
23
|
Chen Y, Li Z, Cao Q, Guan H, Mao L, Zhao M. Ferroptosis-related gene signatures in neuroblastoma associated with prognosis. Front Cell Dev Biol 2022; 10:871512. [PMID: 36147739 PMCID: PMC9486025 DOI: 10.3389/fcell.2022.871512] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 08/12/2022] [Indexed: 11/28/2022] Open
Abstract
Background: Ferroptosis, a form of regulatory cell death, has been linked to the development of various tumors. Peripheral neuroblastoma (NB) is one of the most common extracranial solid tumors in children, and it has been proposed that regulating tumor cell ferroptosis may be a future treatment for NB. However, it is unclear how ferroptosis contributes to NB development. Methods: Expression data were collected from two independent cohorts (GEO and Arrayexpress databases). Univariate Cox analysis, multivariate Cox analysis, and the least absolute shrinkage and selection operator (Lasso) algorithm were applied to create a prognostic signature, whose performance was quantified using the area under the receiver operating characteristic curve (AUC) and Kaplan–Meier curves. A prognostic meta-analysis was used to test the suitability and stability of the FRG signature. Drug sensitivity analyses were performed using the data collected from Cell Miner™. Results:PROM2, AURKA, STEAP3, CD44, ULK2, MAP1LC3A, ATP6V1G2, and STAT3 are among the eight genes in the FRG prognostic signature, all of which were highly expressed in stage 1 NB, except AURKA. Furthermore, the high-risk group, which was stratified by signature, had a lower overall survival rate than the low-risk group. GSEA revealed that high-risk groups have more biological processes related to ferroptosis. Conclusion: Ferroptosis-related genes are expressed differently between stages 1 and 4 NB. The FRG signature successfully stratified NB patients into two risk groups and can accurately predict the overall survival in NB. In addition, we found that the gene AURKA might have the potential to be a prognostic marker in NB.
Collapse
Affiliation(s)
- Yiru Chen
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zihao Li
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Pharmacy, College of Biology, Hunan University, Changsha, China
| | - Qingtai Cao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Haoyu Guan
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Longfei Mao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Pharmacy, College of Biology, Hunan University, Changsha, China
- *Correspondence: Longfei Mao, ; Mingyi Zhao,
| | - Mingyi Zhao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Longfei Mao, ; Mingyi Zhao,
| |
Collapse
|
24
|
Li X, Chen J, Yuan S, Zhuang X, Qiao T. Activation of the P62-Keap1-NRF2 Pathway Protects against Ferroptosis in Radiation-Induced Lung Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8973509. [PMID: 35847598 PMCID: PMC9277166 DOI: 10.1155/2022/8973509] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 06/22/2022] [Indexed: 12/14/2022]
Abstract
Radiation-induced lung injury (RILI) is one of the most common, serious, and dose-limiting toxicities of thoracic radiotherapy. A primary cause for this is the radiation-induced cell death. Ferroptosis is a recently recognized form of regulated cell death, characterized by the accumulation of lipid peroxidation products and lethal reactive oxygen species (ROS). The ROS generated by irradiation might be the original trigger of ferroptosis in RILI. In addition, activation of the P62-Kelch-like ECH-associated protein 1 (Keap1)-nuclear factor erythroid 2-related factor 2 (NRF2) pathway has been shown to blunt ferroptosis and thus acts as a protective factor. Therefore, this study aimed to explore the protective effect of the P62-Keap1-NRF2 pathway against radiation-induced ferroptosis in alveolar epithelial cells. First, we found that radiation induced ferroptosis in vitro using a RILI cell model, which could be significantly reduced by ferrostatin-1 (Fer-1), a specific ferroptosis inhibitor. Additionally, overexpression of P62 interacted with Keap1 to facilitate the translocation of NRF2 into the nucleus and promote the expression of its target proteins, including quinone oxidoreductase 1 (NQO1), heme oxygenase 1 (HO1), and ferritin heavy chain 1 (FTH1). In summary, our results demonstrated that the activation of the P62-Keap1-NRF2 pathway prevents radiation-induced ferroptosis in RILI cells, providing a theoretical basis of finding a potential therapeutic approach for RILI.
Collapse
Affiliation(s)
- Xuan Li
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University Shanghai Medical School, Shanghai, China
| | - Jingyao Chen
- Fudan University Shanghai Medical School, Shanghai, China
| | - Sujuan Yuan
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University Shanghai Medical School, Shanghai, China
| | - Xibing Zhuang
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University Shanghai Medical School, Shanghai, China
| | - Tiankui Qiao
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University Shanghai Medical School, Shanghai, China
| |
Collapse
|
25
|
Hirata Y, Okazaki R, Sato M, Oh-Hashi K, Takemori H, Furuta K. Effect of ferroptosis inhibitors oxindole-curcumin hybrid compound and N,N-dimethylaniline derivatives on rotenone-induced oxidative stress. Eur J Pharmacol 2022; 928:175119. [PMID: 35753403 DOI: 10.1016/j.ejphar.2022.175119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/13/2022] [Accepted: 06/21/2022] [Indexed: 11/03/2022]
Abstract
Oxidative stress is common to multiple cell death pathways, including apoptosis. We recently identified several compounds that protect against ferroptosis, another cell death pathway associated with oxidative stress, suggesting potential efficacy against apoptosis. The present study assessed the protective efficacies of the ferroptosis inhibitors oxindole-curcumin hybrid compound GIF-2165X-G1, N,N-dimethylaniline derivatives GIF-2014 and GIF-2115, and ferrostatin-1 against rotenone-induced apoptosis. Treatment of mouse hippocampal HT22 cells with the mitochondrial transport chain inhibitor rotenone for 24 h reduced mitochondrial membrane potential, increased reactive oxygen species production, promoted nuclear fragmentation, and ultimately impaired cell viability, consistent with apoptosis. Ferroptosis inhibitor cotreatment did not prevent any of these rotenone-induced apoptotic processes but did suppress delayed cell death associated with loss of plasma membrane integrity. These results suggest that GIF-2165X-G1, GIF-2014, GIF-2115, and ferrostatin-1 are selective for ferroptosis and do not affect apoptosis. Thus, erastin-induced ferroptosis and rotenone-induced apoptosis are distinct cell death pathways despite the common involvement of mitochondrial oxidative stress. Further, the cytoprotective efficacies of chemical antioxidants may depend on the specific source of oxidative stress.
Collapse
Affiliation(s)
- Yoko Hirata
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, Gifu, Japan; Graduate School of Natural Science and Technology, Gifu University, Gifu, Japan; United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan.
| | - Ruidai Okazaki
- Graduate School of Natural Science and Technology, Gifu University, Gifu, Japan
| | - Mina Sato
- Graduate School of Natural Science and Technology, Gifu University, Gifu, Japan
| | - Kentaro Oh-Hashi
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, Gifu, Japan; Graduate School of Natural Science and Technology, Gifu University, Gifu, Japan; United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan
| | - Hiroshi Takemori
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, Gifu, Japan; Graduate School of Natural Science and Technology, Gifu University, Gifu, Japan; United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan
| | - Kyoji Furuta
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, Gifu, Japan; Graduate School of Natural Science and Technology, Gifu University, Gifu, Japan; United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan.
| |
Collapse
|
26
|
Sorting Nexin 5 Plays an Important Role in Promoting Ferroptosis in Parkinson’s Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5463134. [PMID: 35571244 PMCID: PMC9098326 DOI: 10.1155/2022/5463134] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/04/2022] [Indexed: 11/30/2022]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease in the elderly, which is related to brain iron metabolism disorders. Ferroptosis is a newly discovered iron-dependent programmed cell death mode, which has been considered an essential mechanism of PD pathogenesis in recent years. However, its underlying mechanisms have not been fully understood. In the present study, the PD rat model and PD cell model were induced by 6-hydroyxdopamine (6-OHDA). The results showed that the expression of Sorting Nexin 5 (SNX5) and the level of ferroptosis will increase after treatment with 6-OHDA. Consistent with these results, ferroptosis inducer erastin synergistically reduced the expression of glutathione peroxidase 4 (GPX4) and increased the expression of SNX5 in the PD cell model, while ferroptosis inhibitor ferrostatin-1 (Fer-1) inhibited the decrease of GPX4 and the increase of SNX5 in the PD cell model. Knockdown of SNX5 in PC-12 cells could reduce intracellular lipid peroxidation and accumulation of Fe2+ and significantly inhibit the occurrence of ferroptosis. In conclusion, the present study suggested that SNX5 promotes ferroptosis in the PD model, thus providing new insights and potential for research on the pharmacological targets of PD.
Collapse
|
27
|
Zhao J, Wang Y, Tao L, Chen L. Iron Transporters and Ferroptosis in Malignant Brain Tumors. Front Oncol 2022; 12:861834. [PMID: 35530363 PMCID: PMC9071296 DOI: 10.3389/fonc.2022.861834] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Malignant brain tumors represent approximately 1.5% of all malignant tumors. The survival rate among patients is relatively low and the mortality rate of pediatric brain tumors ranks first among all childhood malignant tumors. At present malignant brain tumors remain incurable. Although some tumors can be treated with surgery and chemotherapy, new treatment strategies are urgent owing to the poor clinical prognosis. Iron is an essential trace element in many biological processes of the human body. Iron transporters play a crucial role in iron absorption and transport. Ferroptosis, an iron-dependent form of nonapoptotic cell death, is characterized by the accumulation of lipid peroxidation products and lethal reactive oxygen species (ROS) derived from iron metabolism. Recently, compelling evidence has shown that inducing ferroptosis of tumor cells is a potential therapeutic strategy. In this review, we will briefly describe the significant regulatory factors of ferroptosis, iron, its absorption and transport under physiological conditions, especially the function of iron transporters. Then we will summarize the relevant mechanisms of ferroptosis and its role in malignant brain tumors, wherein the role of transporters is not to be ignored. Finally, we will introduce the current research progress in the treatment of malignant brain tumors by inducing ferroptosis in order to explain the current biological principles of potential treatment targets and treatment strategies for malignant brain tumors.
Collapse
Affiliation(s)
- Jingyu Zhao
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, China
| | - Yaqi Wang
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, China
| | - Lei Tao
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, China
- Collaborative Innovation Center for Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Ligong Chen
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, China
- Collaborative Innovation Center for Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
- Advanced Innovation Center for Human Brain Protection, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Ligong Chen,
| |
Collapse
|
28
|
Shi Q, Liu R, Chen L. Ferroptosis inhibitor ferrostatin‑1 alleviates homocysteine‑induced ovarian granulosa cell injury by regulating TET activity and DNA methylation. Mol Med Rep 2022; 25:130. [PMID: 35169856 PMCID: PMC8867468 DOI: 10.3892/mmr.2022.12645] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 10/28/2021] [Indexed: 12/14/2022] Open
Abstract
Polycystic ovary syndrome is one of the most common endocrine and metabolic gynecological disorders, of which dysfunction of ovarian granulosa cells is a key contributing factor. The aim of the present study was to explore the role of ferrostatin-1 (Fer-1), a ferroptosis inhibitor, in a cell injury model established by homocysteine (Hcy)-induced ovarian granulosa KGN cell line and the potential underlying mechanism. Cell viability was measured using Cell Counting Kit-8 assay in the presence or absence of Hcy and Fer-1. Cell apoptosis was assessed using TUNEL staining and the expression levels of apoptosis-related proteins were measured using western blotting. To explore the effects of Fer-1 on oxidative stress in Hcy-treated ovarian granulosa cells, the levels of reactive oxygen species (ROS), malondialdehyde (MDA), lactate dehydrogenase (LDH) and glutathione (GSH) were measured using their corresponding kits. Furthermore, Fe2+ levels were assessed using Phen Green™ SK labeling and western blotting was performed to measure the protein expression levels of ferroptosis-associated proteins GPX4, SLC7A11, ASCL4 and DMT1. Subsequently, DNA methylation and ten-eleven translocation (TET) 1/2 demethylase levels were also detected to evaluate the extent of overall DNA methylation in ovarian granulosa cells after Hcy treatment. The TET1/2 inhibitor Bobcat339 hydrochloride was applied to treat ovarian granulosa cells before evaluating the possible effects of Fer-1 on TET1/2 and DNA methylation. Fer-1 was found to markedly elevate ovarian granulosa cell viability following Hcy treatment. The apoptosis rate in Fer-1-treated groups was also markedly decreased, which was accompanied by downregulated Bax and cleaved caspase-3 expression and upregulated Bcl-2 protein expression. In addition, Fer-1 treatment reduced the levels of ROS, MDA and LDH whilst enhancing the levels of GSH. Fe2+ levels were significantly decreased following Fer-1 treatment, which also elevated glutathione peroxidase 4 expression whilst reducing solute carrier family 7 member 11, achaete-scute family BHLH transcription factor 4 and divalent metal transporter 1 protein expression. Fer-1 significantly inhibited DNA methylation and enhanced TET1/2 levels, which were reversed by treatment with Bobcat339 hydrochloride. Subsequent experiments on cell viability, oxidative stress, Fe2+ content, ferroptosis- and apoptosis-related proteins levels revealed that Bobcat339 hydrochloride reversed the effects of Fer-1 on ovarian granulosa Hcy-induced cell injury. These results suggest that Fer-1 may potentially protect ovarian granulosa cells against Hcy-induced injury by increasing TET levels and reducing DNA methylation.
Collapse
Affiliation(s)
- Qing Shi
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750001, P.R. China
| | - Rui Liu
- Department of Gynecology, Hospital of Cardiovascular and Cerebrovascular Diseases, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750002, P.R. China
| | - Li Chen
- Reproductive Medicine Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| |
Collapse
|
29
|
Avcı B, Günaydın C, Külbay M, Kuruca N, Güvenç T, Bilge SS. Neuroprotective effects of sinapic acid involve the iron regulatory role on the rotenone-induced Parkinson’s disease model. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e20942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
30
|
Li Y, Song Y, Deng G, Tan Q, Xu S, Yang M, Shi H, Hong M, Ye H, Wu C, Ma S, Huang H, Zhang Y, Zeng Z, Wang M, Chen Y, Wang Y, Ma J, Li J, Gao L. Indoleamine 2, 3-dioxygenase 1 aggravates acetaminophen-induced acute liver failure by triggering excess nitroxidative stress and iron accumulation. Free Radic Biol Med 2021; 172:578-589. [PMID: 34242792 DOI: 10.1016/j.freeradbiomed.2021.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 11/17/2022]
Abstract
Acetaminophen (APAP) is the leading cause of acute liver failure (ALF), which is characterized by GSH depletion, oxidative stress and mitochondrial dysfunction. However, the specific mechanism of APAP-induced ALF remains to be clarified. In this study, we demonstrated that indoleamine 2,3-dioxygenase 1 (IDO1) aggravated APAP-induced ALF associated with excess lipid peroxidation, which was reversed by lipid peroxidation inhibitor (ferrostatin-1). Meanwhile, IDO1 deficiency effectively decreased the accumulation of reactive nitrogen species. Additionally, IDO1 deficiency prevented against APAP-induced liver injury through suppressing the activation of macrophages, thereby reduced their iron uptake and export, eventually reduced iron accumulation in hepatocytes through transferrin and transferrin receptor axis. In summary, our study confirmed that APAP-induced IDO1 aggravated ALF by triggering excess oxidative and nitrative stress and iron accumulation in liver. These results offer new insights for the clinical treatment of ALF or iron-dysregulated liver diseases in the future.
Collapse
Affiliation(s)
- Yunjia Li
- Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Yuhong Song
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, 518116, Guangdong, China
| | - Guanghui Deng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Qinxiang Tan
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, 518116, Guangdong, China
| | - Shu Xu
- Department of Oncology, Shenzhen Hospital, University of Chinese Academy of Sciences, Shenzhen, 518107, Guangdong, China
| | - Menghan Yang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Hao Shi
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Mukeng Hong
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Haixin Ye
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Chaofeng Wu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Shuoyi Ma
- Department of Traditional Chinese Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510000, Guangdong, China
| | - Huacong Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Yanhong Zhang
- Department of Traditional Chinese Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510000, Guangdong, China
| | - Zhiyun Zeng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Ming Wang
- Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Yuyao Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Yunqing Wang
- Fifth People's Hospital, Yuhang District, Hangzhou, 311100, Zhejiang, China
| | - Jun Ma
- Department of Traditional Chinese Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510000, Guangdong, China.
| | - Juan Li
- Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China.
| | - Lei Gao
- Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, Guangdong, China.
| |
Collapse
|
31
|
Skouta R. What Did We Accomplish in Fighting Radical Species in Human Health? Antioxidants (Basel) 2021; 10:antiox10030466. [PMID: 33809613 PMCID: PMC8002303 DOI: 10.3390/antiox10030466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/08/2021] [Accepted: 03/15/2021] [Indexed: 12/02/2022] Open
Affiliation(s)
- Rachid Skouta
- Department of Biology, University of Massachusetts, Amherst, MA 01003-9297, USA
| |
Collapse
|
32
|
Yao MY, Liu T, Zhang L, Wang MJ, Yang Y, Gao J. Role of ferroptosis in neurological diseases. Neurosci Lett 2021; 747:135614. [PMID: 33485988 DOI: 10.1016/j.neulet.2020.135614] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/14/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023]
Abstract
Ferroptosis is a newly identified form of nonapoptotic regulated cell death (RCD) characterized by iron-dependent accumulation of lipid peroxides which leads to oxidative stress and cell death. Recent studies have indicated that ferroptosis plays an essential role in the pathology of neurological diseases, such as intracerebral hemorrhage, ischemic stroke, epilepsy, neurodegenerative diseases, traumatic brain injury and brain cancer. This review focuses on the latest researches on the relationship of ferroptosis with nervous system diseases, highlighting the ferroptosis-based mechanisms, and elaborating the new perspective therapeutic targets of neurological disorders.
Collapse
Affiliation(s)
- Min-Yi Yao
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Tao Liu
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Liang Zhang
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Ming-Jian Wang
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yong Yang
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| | - Jing Gao
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
33
|
Shi H, Almutairi M, Moskovitz J, Xu YG. Recent advances in iron homeostasis and regulation - a focus on epigenetic regulation and stroke. Free Radic Res 2021; 55:375-383. [PMID: 33345646 DOI: 10.1080/10715762.2020.1867314] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Iron is an element with redox properties. It is active sites of many enzymes and plays an important role in various cellular and biological functions including ATP production and DNA synthesis. However, as a redox element, iron promotes free radical generation and lipid peroxidation, causing oxidative damage and cell death. Iron-mediated oxidation is a central player in ferroptosis, a type of cell death process that is different from apoptosis and necrosis. Thus, iron metabolism and homeostasis are sophisticatedly regulated. There has been exciting progress in understanding iron metabolism and regulation since hepcidin was recognized as the central regulator of iron homeostasis. Hepcidin mainly regulates the iron export function of the ferrous iron permease, ferroportin, which is the only known iron exporter expressed by mammalian cells. Particularly, epigenetic regulation has been a recent focus on iron homeostasis. Epigenetic phenomena have been demonstrated to modulate key proteins including hepcidin in iron metabolism. Here, we review the rapid progress in recent years in understanding molecular mechanisms of iron homeostasis with a focus on epigenetic regulation of hepcidin, ferritin, and ferroptosis. Interactions between methionine oxidation and iron is also discussed. Furthermore, many studies have suggested that the severity of neuronal damage after stroke is proportional to the magnitude of brain iron accumulation. Recent discoveries regarding iron metabolism in stroke is briefly discussed. Understanding the underlying mechanism in iron regulation could provide insight into the treatment of various intractable diseases including stroke.
Collapse
Affiliation(s)
- Honglian Shi
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Mohammed Almutairi
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Jackob Moskovitz
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Yuexian G Xu
- Department of Anesthesiology, School of Medicine, University of Kansas, Kansas City, KS, USA
| |
Collapse
|
34
|
Wen S, Aki T, Unuma K, Uemura K. Chemically Induced Models of Parkinson's Disease: History and Perspectives for the Involvement of Ferroptosis. Front Cell Neurosci 2020; 14:581191. [PMID: 33424553 PMCID: PMC7786020 DOI: 10.3389/fncel.2020.581191] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/30/2020] [Indexed: 12/21/2022] Open
Abstract
Ferroptosis is a newly discovered form of necrotic cell death characterized by its dependency on iron and lipid peroxidation. Ferroptosis has attracted much attention recently in the area of neurodegeneration since the involvement of ferroptosis in Parkinson’s disease (PD), a major neurodegenerative disease, has been indicated using animal models. Although PD is associated with both genetic and environmental factors, sporadic forms of PD account for more than 90% of total PD. Following the importance of environmental factors, various neurotoxins are used as chemical inducers of PD both in vivo and in vitro. In contrast to other neurodegenerative diseases such as Alzheimer’s and Huntington’s diseases (AD and HD), many of the characteristics of PD can be reproduced in vivo by the use of specific neurotoxins. Given the indication of ferroptosis in PD pathology, several studies have been conducted to examine whether ferroptosis plays role in the loss of dopaminergic neurons in PD. However, there are still few reports showing an authentic form of ferroptosis in neuronal cells during exposure to the neurotoxins used as PD inducers. In this review article, we summarize the history of the uses of chemicals to create PD models in vivo and in vitro. Besides, we also survey recent reports examining the possible involvement of ferroptosis in chemical models of PD.
Collapse
Affiliation(s)
- Shuheng Wen
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshihiko Aki
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kana Unuma
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Koichi Uemura
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
35
|
Liu Z, Lv X, Song E, Song Y. Fostered Nrf2 expression antagonizes iron overload and glutathione depletion to promote resistance of neuron-like cells to ferroptosis. Toxicol Appl Pharmacol 2020; 407:115241. [DOI: 10.1016/j.taap.2020.115241] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/29/2020] [Accepted: 09/10/2020] [Indexed: 12/24/2022]
|
36
|
Zhou RP, Chen Y, Wei X, Yu B, Xiong ZG, Lu C, Hu W. Novel insights into ferroptosis: Implications for age-related diseases. Theranostics 2020; 10:11976-11997. [PMID: 33204324 PMCID: PMC7667696 DOI: 10.7150/thno.50663] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/29/2020] [Indexed: 12/20/2022] Open
Abstract
Rapid increase in aging populations is an urgent problem because older adults are more likely to suffer from disabilities and age-related diseases (ARDs), burdening healthcare systems and society in general. ARDs are characterized by the progressive deterioration of tissues and organs over time, eventually leading to tissue and organ failure. To date, there are no effective interventions to prevent the progression of ARDs. Hence, there is an urgent need for new treatment strategies. Ferroptosis, an iron-dependent cell death, is linked to normal development and homeostasis. Accumulating evidence, however, has highlighted crucial roles for ferroptosis in ARDs, including neurodegenerative and cardiovascular diseases. In this review, we a) summarize initiation, regulatory mechanisms, and molecular signaling pathways involved in ferroptosis, b) discuss the direct and indirect involvement of the activation and/or inhibition of ferroptosis in the pathogenesis of some important diseases, and c) highlight therapeutic targets relevant for ARDs.
Collapse
Affiliation(s)
- Ren-Peng Zhou
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Yong Chen
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Xin Wei
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Bin Yu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Zhi-Gang Xiong
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Chao Lu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| |
Collapse
|
37
|
Hu B, Liu Y, Chen X, Zhao J, Han J, Dong H, Zheng Q, Nie G. Ferrostatin-1 protects auditory hair cells from cisplatin-induced ototoxicity in vitro and in vivo. Biochem Biophys Res Commun 2020; 533:1442-1448. [PMID: 33109343 DOI: 10.1016/j.bbrc.2020.10.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 10/08/2020] [Indexed: 12/23/2022]
Abstract
Cisplatin is used in a wide variety of malignancies, but cisplatin-induced ototoxicity remains a major issue in clinical practice. Experimental evidence indicates that ferroptosis plays a key role in mediating the unwanted cytotoxicity effect caused by cisplatin. However, the role of ferroptosis in cisplatin-induced ototoxicity requires elucidation. Ferrostatin-1 (Fer-1) was identified as a potent inhibitor of ferroptosis and radical-trapping antioxidant with its ability to reduce the accumulation of lipid peroxides and chain-carrying peroxyl radicals. In the current study, we investigated the effects of Fer-1 in cisplatin-induced ototoxicity in in vitro, ex vivo, and in vivo models. We found, for the first time that Fer-1 efficiently alleviated cisplatin-induced cytotoxicity in HEI-OC1 cells via a concentration-dependent manner. Furthermore, Fer-1 mitigated cisplatin cytotoxicity in transgenic zebrafish sensory hair cells. In HEI-OC1 cells, Fer-1 pretreatment not only drastically reduced the generation of intracellular reactive oxygen species but also remarkably decreased lipid peroxidation levels induced by cisplatin. This was not only ascribed to the inhibition of 4-hydroxynonenal, the final product of lipid peroxides, but also to the promotion of glutathione peroxidase 4, the protein marker of ferroptosis. MitoTracker staining and transmission electron microscopy of mitochondrial morphology suggested that in HEI-OC1 cells, Fer-1 can effectively abrogate mitochondrial damage resulting from the interaction with cisplatin. In addition, Fer-1 pretreatment of cochlear explants substantially protected hair cells from cisplatin-induced damage. Therefore, our results demonstrated that ferroptosis might be involved in cisplatin ototoxicity. Fer-1 administration mitigated cisplatin-induced hair cell damage, further investigations are required to elucidate the molecular mechanisms of its otoprotective effect.
Collapse
Affiliation(s)
- Bing Hu
- Department of Otolaryngology and Institute of Translational Medicine, Shenzhen Second People's Hospital/ the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, China
| | - Yunsheng Liu
- Department of Neurosurgery and Institute of Translational Medicine, Shenzhen Second People's Hospital/ the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, China
| | - Xiaozhu Chen
- Department of Otolaryngology and Institute of Translational Medicine, Shenzhen Second People's Hospital/ the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, China
| | - Jianjun Zhao
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44106, USA
| | - Jinghong Han
- Department of Otolaryngology and Institute of Translational Medicine, Shenzhen Second People's Hospital/ the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, China
| | - Hongsong Dong
- Department of Otolaryngology and Institute of Translational Medicine, Shenzhen Second People's Hospital/ the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, China
| | - Qingyin Zheng
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| | - Guohui Nie
- Department of Otolaryngology and Institute of Translational Medicine, Shenzhen Second People's Hospital/ the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, China.
| |
Collapse
|
38
|
Song X, Long D. Nrf2 and Ferroptosis: A New Research Direction for Neurodegenerative Diseases. Front Neurosci 2020; 14:267. [PMID: 32372896 PMCID: PMC7186402 DOI: 10.3389/fnins.2020.00267] [Citation(s) in RCA: 383] [Impact Index Per Article: 76.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 03/09/2020] [Indexed: 12/19/2022] Open
Abstract
Ferroptosis is a kind of regulated cell death (RCD) caused by the redox state disorder of intracellular microenvironment controlled by glutathione (GSH) peroxidase 4 (GPX4), which is inhibited by iron chelators and lipophilic antioxidants. In addition to classical regulatory mechanisms, new regulatory factors for ferroptosis have been discovered in recent years, such as the P53 pathway, the activating transcription factor (ATF)3/4 pathway, Beclin 1 (BECN1) pathway, and some non-coding RNA. Ferroptosis is closely related to cancer treatment, neurodegenerative diseases, ischemia–reperfusion of organ, neurotoxicity, and others, in particular, in the field of neurodegenerative diseases treatment has aroused people’s interest. The nuclear factor E2 related factor 2 (Nrf2/NFE2L2) has been proved to play a key role in neurodegenerative disease treatment and ferroptosis regulation. Ferroptosis promotes the progression of neurodegenerative diseases, while the expression of Nrf2 and its target genes (Ho-1, Nqo-1, and Trx) has been declined with aging; therefore, there is still insufficient evidence for ferroptosis and Nrf2 regulatory networks in the field of neurodegenerative diseases. In this review, we will provide a brief overview of ferroptosis regulatory mechanisms, as well as an emphasis on the mechanism of Nrf2 regulating ferroptosis. We also highlight the role of ferroptosis and Nrf2 during the process of neurodegenerative diseases and investigate a theoretical basis for further research on the relationship between Nrf2 and ferroptosis in the process of neurodegenerative diseases treatment.
Collapse
Affiliation(s)
- Xiaohua Song
- School of Public Health, University of South China, Hengyang, China
| | - Dingxin Long
- School of Public Health, University of South China, Hengyang, China
| |
Collapse
|
39
|
Han C, Liu Y, Dai R, Ismail N, Su W, Li B. Ferroptosis and Its Potential Role in Human Diseases. Front Pharmacol 2020; 11:239. [PMID: 32256352 PMCID: PMC7090218 DOI: 10.3389/fphar.2020.00239] [Citation(s) in RCA: 197] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/21/2020] [Indexed: 12/26/2022] Open
Abstract
Ferroptosis is a novel regulated cell death pattern discovered when studying the mechanism of erastin-killing RAS mutant tumor cells in 2012. It is an iron-dependent programmed cell death pathway mainly caused by an increased redox imbalance but with distinct biological and morphology characteristics when compared to other known cell death patterns. Ferroptosis is associated with various diseases including acute kidney injury, cancer, and cardiovascular, neurodegenerative, and hepatic diseases. Moreover, activation or inhibition of ferroptosis using a variety of ferroptosis initiators and inhibitors can modulate disease progression in animal models. In this review, we provide a comprehensive analysis of the characteristics of ferroptosis, its initiators and inhibitors, and the potential role of its main metabolic pathways in the treatment and prevention of various diseased states. We end the review with the current knowledge gaps in this area to provide direction for future research on ferroptosis.
Collapse
Affiliation(s)
- Chu Han
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, China
| | - Yuanyuan Liu
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, China
| | - Rongji Dai
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Nafissa Ismail
- Neuroimmunology, Stress and Endocrinology (NISE) Lab, School of Psychology, Faculty of Social Science, University of Ottawa, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Weijun Su
- School of Medicine, Nankai University, Tianjin, China
| | - Bo Li
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, China
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
40
|
Deng G, Li Y, Ma S, Gao Z, Zeng T, Chen L, Ye H, Yang M, Shi H, Yao X, Zeng Z, Chen Y, Song Y, Liu B, Gao L. Caveolin-1 dictates ferroptosis in the execution of acute immune-mediated hepatic damage by attenuating nitrogen stress. Free Radic Biol Med 2020; 148:151-161. [PMID: 31877357 DOI: 10.1016/j.freeradbiomed.2019.12.026] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/16/2019] [Accepted: 12/20/2019] [Indexed: 02/06/2023]
Abstract
Ferroptosis is a new regulated cells death manner defined as results of iron-dependent accumulation of lipid peroxidation. However, the specific mechanisms of regulating ferroptosis remain unclear. In our present study, we demonstrated that Caveolin-1 (Cav-1) played a central role in protecting hepatocytes against ferroptosis in autoimmunity-mediated hepatitis (AIH). The down-regulated Cav-1 in liver tissues, accompanied by ferroptotic events and RNS production, were contributed to the outcome of ConA-induced hepatic damage, which were rescued by ferrostatin-1 (an inhibitor of ferroptosis) in vivo and in vitro. Additionally, Cav-1 deficiency aggravated ConA-induced hepatocellular death and ferroptosis associated with excessive nitrogen stress response. Short hairpin RNA of Cav-1 in hepatocytes promoted ferroptosis and nitrative stress in response to erastin in vitro, which was ameliorated by Cav-1 over-expression. Meanwhile, administration of the iNOS inhibitor (1400W) or ONOO- scavenger (Fe-TMPyP), diminished reactive nitrogen species (RNS), remarkably reduced hepatocytes ferroptosis and attenuated ConA-induced liver damage. Furthermore, immune inhibition by gadolinium chloride (GdCl3), a well-known Kupffer cell depletor, elevated hepatic Cav-1 but inhibited ferroptosis and nitrative stress under ConA exposure. In conclusion, these data revealed a novel molecular mechanism of ferroptosis with the Cav-1 regulation was essential for pathogenesis of ConA-induced hepatitis. Downstream of Cav-1, RNS-mediated ferroptosis was a pivotal step that drives the execution of acute immune-mediated hepatic damage.
Collapse
Affiliation(s)
- Guanghui Deng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yunjia Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Shuoyi Ma
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Zhuowei Gao
- Shunde Hospital, Guangzhou University of Chinese Medicine, Foshan, 528333, Guangdong, China; Shunde Hospital, Southern Medical University, Foshan, 528308, Guangdong, China
| | - Ting Zeng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Limei Chen
- Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Haixin Ye
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Menghan Yang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Hao Shi
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xiaofen Yao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhiyun Zeng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yuyao Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yuhong Song
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China.
| | - Bing Liu
- Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| | - Lei Gao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
41
|
Hu Z, Zhang H, Yang SK, Wu X, He D, Cao K, Zhang W. Emerging Role of Ferroptosis in Acute Kidney Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8010614. [PMID: 31781351 PMCID: PMC6875218 DOI: 10.1155/2019/8010614] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/22/2019] [Accepted: 09/09/2019] [Indexed: 12/12/2022]
Abstract
Acute kidney injury (AKI) is a heterogeneous group of critical disease conditions with high incidence and mortality. Vasoconstriction, oxidative stress, apoptosis, and inflammation are generally thought to be the main pathogenic mechanisms of AKI. Ferroptosis is a type of iron-dependent nonapoptotic cell death characterized by membrane lipid peroxide accumulation and polyunsaturated fatty acid consumption, and it plays essential roles in many diseases, including cancers and neurologic diseases. Recent studies have revealed an emerging role of ferroptosis in the pathophysiological processes of AKI. Here, in the present review, we summarized the most recent discoveries on the role of ferroptosis in the pathogenesis of AKI as well as its therapeutic potential in AKI.
Collapse
Affiliation(s)
- Zhaoxin Hu
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan Province, China
| | - Hao Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan Province, China
| | - Shi-kun Yang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan Province, China
| | - Xueqin Wu
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan Province, China
| | - Dong He
- Department of Respiration, The Second People's Hospital of Hunan Province of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ke Cao
- Department of Oncology, The Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan Province, China
| | - Wei Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan Province, China
| |
Collapse
|
42
|
Larrea tridentata Extract Mitigates Oxidative Stress-Induced Cytotoxicity in Human Neuroblastoma SH-SY5Y Cells. Antioxidants (Basel) 2019; 8:antiox8100427. [PMID: 31557847 PMCID: PMC6827101 DOI: 10.3390/antiox8100427] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 09/19/2019] [Accepted: 09/23/2019] [Indexed: 12/12/2022] Open
Abstract
Creosote bush (Larrea tridentata; LT) leaves extracts were tested for their potential efficacy to mitigate cellular oxidative stress on human SH-SY5Y cells. Here, the differential nuclear staining assay, a bioimager system, and flow cytometric protocols, concurrently with several specific chemicals, were used to measure the percentage of cell viability and several facets implicated in the cytoprotective mechanism of LT extracts. Initially, three LT extracts, prepared with different solvents, ethanol, ethanol:water (e/w), and water, were tested for their capacity to rescue the viability of cells undergoing aggressive H2O2-induced oxidative stress. Results indicate that the LT extract prepared with a mixture of ethanol:water (LT-e/w; 60:40% v/v) displayed the most effective cytoprotection rescue activity. Interestingly, by investigating the LT-e/w mechanism of action, it was found that LT-e/w extract decreases the levels of H2O2-provoked reactive oxidative species (ROS) accumulation, mitochondrial depolarization, phosphatidylserine externalization, caspase-3/7 activation, and poly (ADP-ribose) polymerase (PARP) cleavage significantly, which are hallmarks of apoptosis. Thus, out of the three LT extracts tested, our findings highlight that the LT-e/w extract was the most effective protective reagent on SH-SY5Y cells undergoing oxidative stress in vitro, functioning as a natural anti-apoptotic extract. These findings warrant further LT-e/w extract examination in a holistic context.
Collapse
|
43
|
Stamenkovic A, Pierce GN, Ravandi A. Phospholipid oxidation products in ferroptotic myocardial cell death. Am J Physiol Heart Circ Physiol 2019; 317:H156-H163. [DOI: 10.1152/ajpheart.00076.2019] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cell death is an important component of the pathophysiology of any disease. Myocardial disease is no exception. Understanding how and why cells die, particularly in the heart where cardiomyocyte regeneration is limited at best, becomes a critical area of study. Ferroptosis is a recently described form of nonapoptotic cell death. It is an iron-mediated form of cell death that occurs because of accumulation of lipid peroxidation products. Reactive oxygen species and iron-mediated phospholipid peroxidation is a hallmark of ferroptosis. To date, ferroptosis has been shown to be involved in cell death associated with Alzheimer’s disease, Huntington’s disease, cancer, Parkinson’s disease, and kidney degradation. Myocardial reperfusion injury is characterized by iron deposition as well as reactive oxygen species production. These conditions, therefore, favor the induction of ferroptosis. Currently there is no available treatment for reperfusion injury, which accounts for up to 50% of the final infarct size. This review will summarize the evidence that ferroptosis can induce cardiomyocyte death following reperfusion injury and the potential for this knowledge to open new therapeutic approaches for myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Aleksandra Stamenkovic
- Institute of Cardiovascular Sciences, Saint Boniface Hospital, and Departments of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Grant N. Pierce
- Institute of Cardiovascular Sciences, Saint Boniface Hospital, and Departments of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Amir Ravandi
- Institute of Cardiovascular Sciences, Saint Boniface Hospital, and Departments of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
- Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
44
|
Xia Y, Sun X, Luo Y, Stary CM. Ferroptosis Contributes to Isoflurane Neurotoxicity. Front Mol Neurosci 2019; 11:486. [PMID: 30687003 PMCID: PMC6333734 DOI: 10.3389/fnmol.2018.00486] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 12/14/2018] [Indexed: 01/01/2023] Open
Abstract
The underlying mechanisms of isoflurane neurotoxicity in the developing brain remain unclear. Ferroptosis is a recently characterized form of programmed cell death distinct from apoptosis or autophagy, characterized by iron-dependent reactive oxygen species (ROS) generation secondary to failure of glutathione-dependent antioxidant defenses. The results of the present study are the first to demonstrate in vitro that ferroptosis is a central mechanism contributing to isoflurane neurotoxicity. We observed in embryonic mouse primary cortical neuronal cultures (day-in-vitro 7) that 6 h of 2% isoflurane exposure was associated with decreased transcription and protein expression of the lipid repair enzyme glutathione peroxidase 4. In parallel, isoflurane exposure resulted in increased ROS generation, disruption in mitochondrial membrane potential, and cell death. These effects were significantly attenuated by pre-treatment with the selective ferroptosis inhibitor ferrostatin-1 (Fer-1). Collectively, these observations provide a novel mechanism for isoflurane-induced injury in the developing brain and suggest that pre-treatment with Fer-1 may be a potential clinical intervention for neuroprotection.
Collapse
Affiliation(s)
- Yimeng Xia
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Xiaoyun Sun
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Yan Luo
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Creed M Stary
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
45
|
Weiland A, Wang Y, Wu W, Lan X, Han X, Li Q, Wang J. Ferroptosis and Its Role in Diverse Brain Diseases. Mol Neurobiol 2018; 56:4880-4893. [PMID: 30406908 DOI: 10.1007/s12035-018-1403-3] [Citation(s) in RCA: 353] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/18/2018] [Indexed: 02/07/2023]
Abstract
Ferroptosis is a recently identified, iron-regulated, non-apoptotic form of cell death. It is characterized by cellular accumulation of lipid reactive oxygen species that ultimately leads to oxidative stress and cell death. Although first identified in cancer cells, ferroptosis has been shown to have significant implications in several neurologic diseases, such as ischemic and hemorrhagic stroke, Alzheimer's disease, and Parkinson's disease. This review summarizes current research on ferroptosis, its underlying mechanisms, and its role in the progression of different neurologic diseases. Understanding the role of ferroptosis could provide valuable information regarding treatment and prevention of these devastating diseases.
Collapse
Affiliation(s)
- Abigail Weiland
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yamei Wang
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Neural Regeneration and Repair, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Weihua Wu
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Neural Regeneration and Repair, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xi Lan
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Xiaoning Han
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Qian Li
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Neural Regeneration and Repair, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
- Advanced Innovation Center for Human Brain Protection, Captical Medical University, Beijing, 100069, China.
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
46
|
Limatola A, Eichmann C, Jacob RS, Ben-Nissan G, Sharon M, Binolfi A, Selenko P. Time-Resolved NMR Analysis of Proteolytic α-Synuclein Processing in vitro and in cellulo. Proteomics 2018; 18:e1800056. [PMID: 30260559 DOI: 10.1002/pmic.201800056] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/14/2018] [Indexed: 11/07/2022]
Abstract
Targeted proteolysis of the disordered Parkinson's disease protein alpha-synuclein (αSyn) constitutes an important event under physiological and pathological cell conditions. In this work, site-specific αSyn cleavage by different endopeptidases in vitro and by endogenous proteases in extracts of challenged and unchallenged cells was studied by time-resolved NMR spectroscopy. Specifically, proteolytic processing was monitored under neutral and low pH conditions and in response to Rotenone-induced oxidative stress. Further, time-dependent degradation of electroporation-delivered αSyn in intact SH-SY5Y and A2780 cells was analyzed. Results presented here delineate a general framework for NMR-based proteolysis studies in vitro and in cellulo, and confirm earlier reports pertaining to the exceptional proteolytic stability of αSyn under physiological cell conditions. However, experimental findings also reveal altered protease susceptibilities in selected mammalian cell lines and upon induced cell stress.
Collapse
Affiliation(s)
- Antonio Limatola
- Leibniz Institute of Molecular Pharmacology (FMP-Berlin), In-cell NMR Group,, Robert-Rössle Strasse 10, 13125, Berlin, Germany.,Department of Biology, Stanford University, Stanford, CA, 94305-5430, USA
| | - Cédric Eichmann
- Leibniz Institute of Molecular Pharmacology (FMP-Berlin), In-cell NMR Group,, Robert-Rössle Strasse 10, 13125, Berlin, Germany
| | - Reeba Susan Jacob
- Leibniz Institute of Molecular Pharmacology (FMP-Berlin), In-cell NMR Group,, Robert-Rössle Strasse 10, 13125, Berlin, Germany.,Department of Biological Regulation, Weizmann Institute of Science, 234 Herzl Street, 761000, Rehovot, Israel
| | - Gili Ben-Nissan
- Department of Biomolecular Sciences, Weizmann Institute of Science, 234 Herzl Street, 761000, Rehovot, Israel
| | - Michal Sharon
- Department of Biomolecular Sciences, Weizmann Institute of Science, 234 Herzl Street, 761000, Rehovot, Israel
| | - Andres Binolfi
- Leibniz Institute of Molecular Pharmacology (FMP-Berlin), In-cell NMR Group,, Robert-Rössle Strasse 10, 13125, Berlin, Germany.,Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET) and Plataforma Argentina de Biología Estructural y Metabolómica (PLABEM), Ocampo y Esmeralda, 2000, Rosario, Argentina
| | - Philipp Selenko
- Leibniz Institute of Molecular Pharmacology (FMP-Berlin), In-cell NMR Group,, Robert-Rössle Strasse 10, 13125, Berlin, Germany.,Department of Biological Regulation, Weizmann Institute of Science, 234 Herzl Street, 761000, Rehovot, Israel
| |
Collapse
|
47
|
Assessing the Antioxidant Properties of Larrea tridentata Extract as a Potential Molecular Therapy against Oxidative Stress. Molecules 2018; 23:molecules23071826. [PMID: 30041415 PMCID: PMC6099408 DOI: 10.3390/molecules23071826] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 07/13/2018] [Accepted: 07/18/2018] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress has been linked to neurodegenerative diseases such as Huntington's, Parkinson's, Alzheimer's and amyotrophic lateral sclerosis diseases. Larrea tridentata (LT) also known as Creosote Bush is an evergreen shrub found in the Chihuahuan desert which has been used medicinally by Native American tribes in southwestern North America and the Amerindians of South America. However, studies of the antioxidant capacity of the crude extract of LT towards the discovery of novel molecular therapies bearing antioxidants and drug-like properties are lacking. In this study, we assessed the antioxidant properties of Larrea tridentata, collected specifically from the Chihuahuan desert in the region of El Paso del Norte, TX, USA. LT phytochemicals were obtained from three different extracts (ethanol; ethanol: water (60:40) and water). Then the extracts were evaluated in eight different assays (DPPH, ABTS, superoxide; FRAP activity, nitric oxide, phenolic content, UV visible absorption and cytotoxicity in non-cancerous HS27 cells). The three extracts were not affecting the HS27 cells at concentrations up to 120 µg/mL. Among the three extracts, we found that the mixture of ethanol: water (60:40) LT extract has the most efficient antioxidant properties (IC50 (DPPH at 30 min) = 111.7 ± 3.8 μg/mL; IC50 (ABTS) = 8.49 ± 2.28 μg/mL; IC50 (superoxide) = 0.43 ± 0.17 μg/mL; IC50 (NO) = 230.4 ± 130.4 μg/mL; and the highest phenolic content was estimated to 212.46 ± 7.05 mg GAE/L). In addition, there was a strong correlation between phenolic content and the free-radical scavenging activity assays. HPLC-MS study identified nine compounds from the LT-ethanol: water extract including Justicidin B and Beta peltain have been previously reported as secondary metabolites of Larrea tridentata.
Collapse
|
48
|
Down‐regulation of intracellular anti‐apoptotic proteins, particularly c‐FLIP by therapeutic agents; the novel view to overcome resistance to TRAIL. J Cell Physiol 2018; 233:6470-6485. [PMID: 29741767 DOI: 10.1002/jcp.26585] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/08/2018] [Indexed: 12/24/2022]
|
49
|
Gaschler MM, Hu F, Feng H, Linkermann A, Min W, Stockwell BR. Determination of the Subcellular Localization and Mechanism of Action of Ferrostatins in Suppressing Ferroptosis. ACS Chem Biol 2018. [PMID: 29512999 DOI: 10.1021/acschembio.8b00199] [Citation(s) in RCA: 257] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Ferroptosis is a form of nonapoptotic cell death characterized by the unchecked accumulation of lipid peroxides. Ferrostatin-1 and its analogs (ferrostatins) specifically prevent ferroptosis in multiple contexts, but many aspects of their molecular mechanism of action remain poorly described. Here, we employed stimulated Raman scattering (SRS) microscopy coupled with small vibrational tags to image the distribution of ferrostatins in cells and found that they accumulate in lysosomes, mitochondria, and the endoplasmic reticulum. We then evaluated the functional relevance of lysosomes and mitochondria to ferroptosis suppression by ferrostatins and found that neither is required for effective ferroptosis suppression.
Collapse
Affiliation(s)
- Michael M. Gaschler
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Fanghao Hu
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Huizhong Feng
- Department of Biological Sciences, Columbia University, New York, New York 10027, United States
| | - Andreas Linkermann
- Medical Clinic III, Division of Nephrology, Carl Gustav Carus University Hospital at the Technical University Dresden, Dresden 01309, Germany
| | - Wei Min
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Brent R. Stockwell
- Department of Chemistry, Columbia University, New York, New York 10027, United States
- Department of Biological Sciences, Columbia University, New York, New York 10027, United States
| |
Collapse
|
50
|
Chen MB, Liu YY, Xing ZY, Zhang ZQ, Jiang Q, Lu PH, Cao C. Itraconazole-Induced Inhibition on Human Esophageal Cancer Cell Growth Requires AMPK Activation. Mol Cancer Ther 2018; 17:1229-1239. [PMID: 29592879 DOI: 10.1158/1535-7163.mct-17-1094] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/11/2018] [Accepted: 03/22/2018] [Indexed: 11/16/2022]
Abstract
We here evaluated the antiesophageal cancer cell activity by the antifungal drug itraconazole. Our results show that μg/mL concentrations of itraconazole potently inhibited survival and proliferation of established (TE-1 and Eca-109) and primary human esophageal cancer cells. Itraconazole activated AMPK signaling, which was required for subsequent esophageal cancer cell death. Pharmacologic AMPK inhibition, AMPKα1 shRNA, or dominant negative mutation (T172A) almost completely abolished itraconazole-induced cytotoxicity against esophageal cancer cells. Significantly, itraconazole induced AMPK-dependent autophagic cell death (but not apoptosis) in esophageal cancer cells. Furthermore, AMPK activation by itraconazole induced multiple receptor tyrosine kinases (RTKs: EGFR, PDGFRα, and PDGFRβ), lysosomal translocation, and degradation to inhibit downstream Akt activation. In vivo, itraconazole oral gavage potently inhibited Eca-109 tumor growth in SCID mice. It was yet ineffective against AMPKα1 shRNA-expressing Eca-109 tumors. The in vivo growth of the primary human esophageal cancer cells was also significantly inhibited by itraconazole administration. AMPK activation, RTK degradation, and Akt inhibition were observed in itraconazole-treated tumors. Together, itraconazole inhibits esophageal cancer cell growth via activating AMPK signaling. Mol Cancer Ther; 17(6); 1229-39. ©2018 AACR.
Collapse
Affiliation(s)
- Min-Bin Chen
- Department of Radiotherapy & Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Yuan-Yuan Liu
- Clinical Research and Lab Center, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Zhao-Yu Xing
- The Department of Urology, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Zhi-Qing Zhang
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Qin Jiang
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China.
| | - Pei-Hua Lu
- Department of Medical Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China.
| | - Cong Cao
- Institute of Neuroscience, Soochow University, Suzhou, China. .,The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China.,The Municipal Hospital of Suzhou, North District, Suzhou, China
| |
Collapse
|