1
|
Shen Y, Liu W, Zhou Z, He J, Qi X. FTO-mediated m6A Demethylation of OTUB1 stabilizes SLC7A11 to alleviate Ferroptosis in cerebral ischemia/reperfusion injury. J Stroke Cerebrovasc Dis 2025; 34:108316. [PMID: 40233842 DOI: 10.1016/j.jstrokecerebrovasdis.2025.108316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 04/08/2025] [Accepted: 04/12/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUD Therapeutic strategies for cerebral ischemia/reperfusion (I/R) injury, an important contributor to neurological impairment and disability, exhibit limited efficacy. Reperfusion therapy intensifies neuronal damage by promoting iron deposition, ferroptosis (lipid peroxidation-associated iron-dependent cellular death), and reactive oxygen species (ROS) accumulation. METHODS we investigated the role of the m6A demethylase FTO in modulating ferroptosis during cerebral I/R injury, using middle cerebral artery occlusion/reperfusion (MCAO/R) model rats and neuronal cells subjected to oxygen glucose deprivation/reoxygenation (OGD/R) as in vivo and in vitro experimental platforms, respectively. Neurological scores and cerebral infarction volumes were measured by TTC staining. FTO, OTUB1, and SLC7A11 levels, and FTO demethylase activity, were assessed by qRT-PCR, western blotting, and immunohistochemistry. MeRIP was applied to ascertain the m6A methylation status of OTUB1 mRNA. Apoptotic rates and cell viability were quantitatively aalyzed by flow cytometry and CCK-8 assay, respectively, while brain tissue apoptosis was evaluated using TUNEL staining. RESULTS MCAO/R rat brains and OGD/R cells showed decreased FTO expression and increased OTUB1 m6A methylation. FTO overexpression upregulated OTUB1 by diminishing m6A methylation, consequently stabilizing SLC7A11 and reducing ferroptosis. FTO or OTUB1 silencing increased ferroptosis, while their co-overexpression enhanced neuroprotective effects. FTO overexpression reduced infarct volume and cell apoptosis, and improved neurological outcomes in vivo. CONCLUSIONS FTO enhanced OTUB1 expression via m6A demethylation, stabilizing SLC7A11, and inhibiting ferroptosis to alleviate cerebral I/R injury. The FTO/OTUB1/SLC7A11 pathway is a viable therapeutic target for ischemic stroke, providing novel perspectives on the molecular mechanisms underlying neuroprotection and proposing innovative m6A-based therapies.
Collapse
Affiliation(s)
- Youjin Shen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou City, Guangdong Province, PR China; Department of Neurology, Deqing People's Hospital, Zhaoqing City, Guangdong Province, PR China
| | - Wentao Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou City, Guangdong Province, PR China; Department of Emergency, Huhhot First Hospital, Huhhot City, Inner Mongolia Autonomous Region, PR China
| | - Zonghua Zhou
- Department of Neurology, Deqing People's Hospital, Zhaoqing City, Guangdong Province, PR China
| | - Jianwen He
- Department of Neurology, Deqing People's Hospital, Zhaoqing City, Guangdong Province, PR China
| | - Xiaokun Qi
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou City, Guangdong Province, PR China; Department of Neurology, The Sixth Medical Center of PLA of Chinese General Hospital, Beijing, PR China.
| |
Collapse
|
2
|
Li Z, Xing J. Role of sirtuins in cerebral ischemia-reperfusion injury: Mechanisms and therapeutic potential. Int J Biol Macromol 2025; 310:143591. [PMID: 40300682 DOI: 10.1016/j.ijbiomac.2025.143591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/22/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025]
Abstract
The high incidence and mortality rate of cardiac arrest (CA) establishes it as a critical clinical challenge in emergency medicine globally. Despite continuous advances in advanced life support (ALS) technology, the prognosis for patients experiencing cardiac arrest remains poor, with cerebral ischemia and reperfusion injury (CIRI) being a significant determinant of adverse neurological outcomes and increased mortality. Sirtuins (SIRTs) are a class of highly evolutionarily conserved NAD+-dependent histone deacylenzymes capable of regulating the expression of various cytoprotective genes to play a neuroprotective role in CIRI. SIRTs mainly regulate the levels of downstream proteins such as PGC 1-α, Nrf 2, NLRP 3, FoxOs, and PINK 1 to inhibit inflammatory response, attenuate oxidative stress, improve mitochondrial dysfunction, promote angiogenesis, and inhibit apoptosis while reducing CIRI. Natural active ingredients are widely used in regulating the protein level of SIRTs in the body because of their multi-components, multi-pathway, multi-target, and minimal toxic side effects. However, these naturally active ingredients still face many challenges related to drug targeting, pharmacokinetic properties, and drug delivery. The emergence and vigorous development of new drug delivery systems, such as nanoparticles, micromilk, and exosomes, provide strong support for solving the above problems. In the context of the rapid development of molecular biology technology, non-coding RNA (NcRNA), represented by miRNA and LncRNA, offers great potential for achieving gene-level precision medicine. In the context of multidisciplinary integration, combining SIRTs proteins with biotechnology, omics technologies, artificial intelligence, and material science will strongly promote the deepening of their basic research and expand their clinical application. This review describes the major signaling pathways of targeting SIRTs to mitigate CIRI, as well as the current research status of Chinese and Western medicine and medical means for the intervention level of SIRTs. Meanwhile, the challenges and possible solutions in the clinical application of targeted drugs are summarized. In the context of medical and industrial crossover, the development direction of SIRTs in the future is discussed to provide valuable reference for basic medical researchers and clinicians to improve the clinical diagnosis and treatment effects of CIRI.
Collapse
Affiliation(s)
- Zheng Li
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| | - Jihong Xing
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
3
|
Yin Y, Wang B, Yang Y, Jiang Y, Fu W. Tectorigenin mitigates homocysteine-induced inflammation and ferroptosis in BV-2 microglial cells through promoting the SIRT1/SLC7A11 pathway. Brain Res Bull 2025; 224:111272. [PMID: 40058656 DOI: 10.1016/j.brainresbull.2025.111272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/06/2025] [Accepted: 02/23/2025] [Indexed: 03/20/2025]
Abstract
Ferroptosis and inflammation are central to the pathophysiology of hyperhomocysteinemia (HHcy)-associated neurological disorders. Tectorigenin, a natural flavonoid aglycone extracted from numerous plants, possesses antioxidant, anti-inflammatory, and neuroprotective properties. This study aimed to investigate whether tectorigenin mitigates elevated homocysteine (Hcy)-induced toxicity in BV-2 microglial cells, focusing on its effects on inflammation and ferroptosis. Cell viability, lactate dehydrogenase (LDH) release, and proliferation assays were employed to evaluate cell injury. Inflammatory cytokines levels were determined by ELISA. Ferroptosis markers, including reactive oxygen species (ROS), lipid ROS, malondialdehyde (MDA), 4-hydroxy-nonenal (4-HNE), mitochondrial membrane potential (MMP), ATP, Fe2 + content, antioxidant enzymes (superoxide dismutase [SOD] and catalase [CAT]) activities were evaluated. mRNA and protein expressions were analyzed by qRT-PCR and western blotting, respectively. Our findings revealed that tectorigenin pretreatment significantly alleviated Hcy-induced cell injury and inflammatory response in BV-2 microglial cells. Furthermore, tectorigenin pretreatment reduced lipid peroxidation, enhanced antioxidant capacity, and alleviated ferroptotic cell death in Hcy-treated cells. Importantly, ferroptosis inhibitor Fer-1 also alleviated Hcy-induced cell injury and inflammation. Mechanistically, tectorigenin pretreatment activated the SIRT1/SLC7A11 pathway, and silencing SIRT1 reversed its protective effects. Collectively, these results indicate that tectorigenin attenuates Hcy-induced microglial injury by inhibiting inflammation and ferroptosis through the activation of the SIRT1/SLC7A11 pathway.
Collapse
Affiliation(s)
- Ye Yin
- The First Affiliated Hospital, Department of General Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Bo Wang
- The First Affiliated Hospital, Institute of Anesthesiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Yan Yang
- The First Affiliated Hospital, Institute of Anesthesiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Yichen Jiang
- The First Affiliated Hospital, Institute of Anesthesiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Wan Fu
- The First Affiliated Hospital, Institute of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
4
|
Song J, Chen Y, Chen Y, Qiu M, Xiang W, Ke B, Fang X. Wnt/β-catenin Pathway Aggravates Renal Fibrosis by Activating PUM2 Transcription to Repress YME1L-mediated Mitochondrial Homeostasis. Biochem Genet 2025; 63:1343-1360. [PMID: 38564095 DOI: 10.1007/s10528-024-10756-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 02/23/2024] [Indexed: 04/04/2024]
Abstract
Chronic kidney disease (CKD) affects more than 10% of people worldwide and is a leading cause of death. However, the pathogenesis of CKD remains elusive. The oxidative stress and mitochondrial membrane potential were detected using Enzyme-linked immunosorbent assay and JC-1 assay. Co-immunoprecipitation, dual-luciferase assay, chromatin IP, RNA IP and RNA pull-down were used to validate the interactions among genes. Exploiting a H2O2-induced fibrosis model in vitro, PUM2 expression was upregulated in Human kidney 2 cell (HK-2) cells, along with reduced cell viability, enhanced oxidative stress, impaired mitochondrial potential, and upregulated expressions of fibrosis-associated proteins. While PUM2 knockdown reversed the H2O2-induced injury in HK-2 cells. Mechanically, Wnt/β-catenin pathway activated PUM2 transcription via TCF4. It was further identified that Wnt/β-catenin pathway inhibited YME1L expression through PUM2-mediated destabilizing of its mRNA. PUM2 aggravated H2O2-induced oxidative stress, mitochondrial dysfunction, and renal fibrosis in HK-2 cell via suppressing YME1L expression. Our study revealed that Wnt/β-catenin aggravated renal fibrosis by activating PUM2 transcription to repress YME1L-mediated mitochondrial homeostasis, providing novel insights and potential therapeutic targets for the treatment of kidney fibrosis.
Collapse
Affiliation(s)
- Jianling Song
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yanxia Chen
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yan Chen
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Minzi Qiu
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Wenliu Xiang
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Ben Ke
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| | - Xiangdong Fang
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
5
|
Li Z, Liu J, Ju J, Peng X, Zhao W, Ren J, Jia X, Wang J, Gao F. Cordycepin ameliorates morphine tolerance by inhibiting spinal cord ferroptosis and inflammation via targeting SIRT1. Int J Med Sci 2025; 22:2059-2074. [PMID: 40303502 PMCID: PMC12035829 DOI: 10.7150/ijms.108518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/21/2025] [Indexed: 05/02/2025] Open
Abstract
Morphine tolerance caused by long-term use of morphine is a major medical problem. Neuroinflammation plays an important role in morphine tolerance, and currently no drugs have been found for clinical use to alleviate neuroinflammation during morphine tolerance. Cordycepin is the main active component of fungus cordycepin militaris, has been demonstrated to have anti-oxidative stress and anti-inflammatory properties in various diseases. In this study, we established a rat model of morphine tolerance, examined the effect of cordycepin on the development of morphine tolerance, and evaluated its potential regulatory mechanisms. We found that cordycepin treatment ameliorated the development of morphine tolerance, improved mitochondrial damage associated with ferroptosis, by reducing the levels of reactive oxygen species (ROS), malondialdehyde (MDA) and Fe2+, increasing superoxide dismutase (SOD) and glutathione (GSH) levels, and decreasing the secretion of pro-inflammatory factors (IL-1β, IL-6, and TNF-α). Besides, cordycepin upregulated the expression of SIRT1, SLC7A11 and GPX4. Further research found that the above effects of cordycepin on morphine-tolerant rats were abolished by SIRT1 selective inhibitor EX-527. Thus, these findings indicated that cordycepin could ameliorate the development of morphine tolerance by inhibiting spinal cord ferroptosis and inflammation via targeting SIRT1. Collectively, these results demonstrated the protective effects of cordycepin and highlighted its therapeutic potential as a drug component for morphine tolerance treatment and prevention.
Collapse
Affiliation(s)
- Zheng Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Department of Anesthesiology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Jie Liu
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jie Ju
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Xiaoling Peng
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Wei Zhao
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jihao Ren
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Xiaoqian Jia
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jihong Wang
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Feng Gao
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| |
Collapse
|
6
|
Cao C, Lu J, Lu P, Li L, Zhang F, Li X, Chen G, Bai L, Li H. Disruption of the Pum2 axis Aggravates neuronal damage following cerebral Ischemia-Reperfusion in mice. Brain Res 2025; 1851:149455. [PMID: 39832611 DOI: 10.1016/j.brainres.2025.149455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 12/24/2024] [Accepted: 01/11/2025] [Indexed: 01/22/2025]
Abstract
Stroke remains a leading cause of disability and mortality worldwide, with mitochondrial dysfunction closely linked to ischemic injury. This study explores the Norad-Pum2-Mff axis as a key regulator of mitochondrial function following ischemia-reperfusion (I/R) injury. Using an oxygen-glucose deprivation/reoxygenation (OGD/R) model, Mff protein levels were significantly elevated post-OGD/R, while mRNA levels remained unchanged, suggesting post-transcriptional regulation. Pumilio2 (Pum2), an RNA-binding protein, was shown to inhibit Mff translation, while Norad, a long non-coding RNA, sequestered Pum2, alleviating this inhibition. We observed decreased Pum2 levels and binding capacity to Mff mRNA, alongside increased Norad levels and binding to Pum2 in neurons after OGD/R. Overexpression of Pum2 in neurons reduced Mff levels, mitigated mitochondrial fragmentation, and alleviated neuronal injury. In a mouse model of middle cerebral artery occlusion/reperfusion (MCAO/R), Pum2 overexpression further improved mitochondrial morphology, reduced infarct volume, and enhanced neurobehavioral recovery. These findings suggest that targeting the Norad-Pum2-Mff axis could provide a promising therapeutic strategy for ischemic stroke by restoring mitochondrial function and reducing neuronal damage.
Collapse
Affiliation(s)
- Chang Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, China
| | - Jinxin Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, China
| | - Peng Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, China
| | - Lianxin Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, China
| | | | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, China
| | - Lei Bai
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, China.
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou, 215006, China.
| |
Collapse
|
7
|
Zhang Y, Kong F, Li N, Tao L, Zhai J, Ma J, Zhang S. Potential role of SIRT1 in cell ferroptosis. Front Cell Dev Biol 2025; 13:1525294. [PMID: 40109363 PMCID: PMC11919884 DOI: 10.3389/fcell.2025.1525294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
Ferroptosis is a novel form of cell death that uniquely requires iron and is characterized by iron accumulation, the generation of free radicals leading to oxidative stress, and the formation of lipid peroxides, which distinguish it from other forms of cell death. The regulation of ferroptosis is extremely complex and is closely associated with a spectrum of diseases. Sirtuin 1 (SIRT1), a NAD + -dependent histone deacetylase, has emerged as a pivotal epigenetic regulator with the potential to regulate ferroptosis through a wide array of genes intricately associated with lipid metabolism, iron homeostasis, glutathione biosynthesis, and redox homeostasis. This review provides a comprehensive overview of the specific mechanisms by which SIRT1 regulates ferroptosis and explores its potential therapeutic value in the context of multiple disease pathologies, highlighting the significance of SIRT1-mediated ferroptosis in treatment strategies.
Collapse
Affiliation(s)
- Yueming Zhang
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Jilin, China
| | - Fanxiao Kong
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Li
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Jilin, China
| | - Lina Tao
- Department of Pharmacy, The First Hospital of Jilin University, Jilin, China
| | - Jinghui Zhai
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Jilin, China
| | - Jie Ma
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Jilin, China
| | - Sixi Zhang
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Jilin, China
| |
Collapse
|
8
|
Zhang SY, Yang N, Hao PH, Wen R, Zhang TN. Targeting sirtuins in neurological disorders: A comprehensive review. Int J Biol Macromol 2025; 292:139258. [PMID: 39736297 DOI: 10.1016/j.ijbiomac.2024.139258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 01/01/2025]
Abstract
The sirtuin (SIRT) family is a group of seven conserved nicotinamide adenine dinucleotide-dependent histone deacetylases (SIRT1-SIRT7), which play crucial roles in various fundamental biological processes, including metabolism, aging, stress responses, inflammation, and cell survival. The role of SIRTs in neuro-pathophysiology has recently attracted significant attention. Notably, SIRT1-SIRT3 have been identified as key players in neuroprotection as they reduce neuroinflammation and regulate mitochondrial function. This review summarizes the latest research advancements in the role of the SIRT family in neurological diseases, mainly including neurodegenerative diseases, ischemia-related diseases, bleeding-related diseases, nervous system injury and other nervous system diseases, emphasizing their critical functions and associated signaling pathways, (e.g., AMPK/SIRT1/PGC-1α, AMPK/SIRT1/IL-1β/NF-κB, STAT2-SIRT4-mTOR, SIRT3/FOXO3α, and other signaling pathways in disease progression, particularly their protective roles in neurodegenerative diseases, ischemic injuries, and neural damage. Additionally, this review discusses progress in clinical studies targeting SIRT-specific small-molecule agonists and inhibitors. Further research on SIRTs may provide new insights into potential therapeutic strategies for the prevention and treatment of neurological disorders.
Collapse
Affiliation(s)
- Sen-Yu Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ni Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Peng-Hui Hao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ri Wen
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Tie-Ning Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
9
|
He YL, Zhang Y, Liu Q. Regulation of Postoperative Cognitive Dysfunction by Glutathione Under Various Pathways: A Narrative Review. J Biochem Mol Toxicol 2025; 39:e70154. [PMID: 39925043 DOI: 10.1002/jbt.70154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/05/2025] [Accepted: 01/16/2025] [Indexed: 02/11/2025]
Abstract
Postoperative cognitive dysfunction (POCD) is a common neurological complication after surgery and general anesthesia, and the incidence increases with age. Will have a negative impact on patients, family and society. At present, neuroinflammation and oxidative stress are the main recognized mechanisms. Glutathione (GSH) is a powerful reducing agent and may be related to POCD. DATA COLLECTION Using medical search engines such as PubMed, Web of Science, we analyzed articles on topics such as: POCD, GSH, microglia, astrocyte, oligodendrocyte, ferroptosis, BDNF, Neuroinflammation, oxidative stress. The above topics are searched in databases using Boolean operations. We included original articles, reviews and other article types such as medical books. RESULTS According to the reviewed literature, GSH may be a treatment for POCD. CONCLUSIONS Specific and targeted therapies for POCD still sparse, therefore, the implementation of preventive strategies appears to remain the optimal attitude. Further research is needed for a better understanding of GSH and POCD.
Collapse
Affiliation(s)
- Yan Lin He
- Department of Anesthesiology, He jiang Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, China
| | - Ying Zhang
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- Lu Zhou Key Laboratory of Research for Integrative on Pain and Perioperative Organ Protection, Luzhou, China
| | - Qing Liu
- Department of Anesthesiology, He jiang Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, China
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
10
|
Wang L, Li X, Chen L, Mei S, Shen Q, Liu L, Liu X, Liao S, Zhao B, Chen Y, Hou J. Mitochondrial Uncoupling Protein-2 Ameliorates Ischemic Stroke by Inhibiting Ferroptosis-Induced Brain Injury and Neuroinflammation. Mol Neurobiol 2025; 62:501-517. [PMID: 38874704 DOI: 10.1007/s12035-024-04288-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 06/03/2024] [Indexed: 06/15/2024]
Abstract
Ischemic stroke is a devastating disease in which mitochondrial damage or dysfunction substantially contributes to brain injury. Mitochondrial uncoupling protein-2 (UCP2) is a member of the UCP family, which regulates production of mitochondrial superoxide anion. UCP2 is reported to be neuroprotective for ischemic stroke-induced brain injury. However, the molecular mechanisms of UCP2 in ischemic stroke remain incompletely understood. In this study, we investigated whether and how UCP2 modulates neuroinflammation and regulates neuronal ferroptosis following ischemic stroke in vitro and in vivo. Wild-type (WT) and UCP2 knockout (Ucp2-/-) mice were subjected to middle cerebral artery occlusion (MCAO). BV2 cells (mouse microglial cell line) and HT-22 cells (mouse hippocampal neuronal cell line) were transfected with small interfering (si)-RNA or overexpression plasmids to knockdown or overexpress UCP2 levels. Cells were then exposed to oxygen-glucose deprivation and reoxygenation (OGD/RX) to simulate hypoxic injury in vitro. We found that UCP2 expression was markedly reduced in a time-dependent manner in both in vitro and in vivo ischemic stroke models. In addition, UCP2 was mainly expressed in neurons. UCP2 deficiency significantly enlarged infarct volumes, aggravated neurological deficit scores, and exacerbated cerebral edema in mice after MCAO. In vitro knockdown of Ucp2 and in vivo genetic depletion of Ucp2 (Ucp2-/- mice) increased neuronal ferroptosis-related indicators, including Fe2+, malondialdehyde, glutathione, and lipid peroxidation. Overexpression of UCP2 in neuronal cells resulted in reduced ferroptosis. Moreover, knockdown of UCP2 exacerbated neuroinflammation in BV2 microglia and mouse ischemic stroke models, suggesting that endogenous UCP2 inhibits neuroinflammation following ischemic stroke. Upregulation of UCP2 expression in microglia appeared to decrease the release of pro-inflammatory factors and increase the levels of anti-inflammatory factors. Further investigation showed that UCP2 deletion inhibited expression of AMPKα/NRF1 pathway-related proteins, including p-AMPKα, t-AMPKα, NRF1, and TFAM. Thus, UCP2 protects the brain from ischemia-induced ferroptosis by activating AMPKα/NRF1 signaling. Activation of UCP2 represents an attractive strategy for the prevention and treatment of ischemic stroke.
Collapse
Affiliation(s)
- Lei Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaona Li
- Department of Pain Medicine, Wuhan Fourth Hospital, Wuhan, 430033, China
| | - Lili Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, P.O. Box 430060, Wuhan, 430060, China
| | - Shenglan Mei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, P.O. Box 430060, Wuhan, 430060, China
| | - Qianni Shen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, P.O. Box 430060, Wuhan, 430060, China
| | - Lian Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, P.O. Box 430060, Wuhan, 430060, China
| | - Xuke Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, P.O. Box 430060, Wuhan, 430060, China
| | - Shichong Liao
- Department of Thyroid and Breast Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bo Zhao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, P.O. Box 430060, Wuhan, 430060, China
| | - Yannan Chen
- Department of Endocrinology, Wuhan Fourth Hospital, Wuhan, 430033, China
| | - Jiabao Hou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, P.O. Box 430060, Wuhan, 430060, China.
| |
Collapse
|
11
|
Liu W, Zhang Q, Guo S, Wang H. The role of microRNAs regulation of endoplasmic reticulum stress in ischemia-reperfusion injury: A review. Int J Biol Macromol 2024; 283:137566. [PMID: 39542287 DOI: 10.1016/j.ijbiomac.2024.137566] [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: 07/26/2024] [Revised: 11/06/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
The endoplasmic reticulum (ER) is an important organelle in eukaryotic cells, responsible for a range of biological functions such as the secretion, modification and folding of proteins, maintaining Ca2+ homeostasis and the synthesis of steroids/lipids, secreted proteins and membrane proteins. When cells are affected by internal or external factors, including abnormal energy metabolism, disrupted Ca2+ balance, altered glycosylation, drug toxicity, and so on, the unfolded or misfolded proteins accumulate in the ER, leading to the unfolded protein response (UPR) and ER stress. The abnormal ER stress has been reported to be involved in various pathological processes. MicroRNAs (miRNAs) are non-coding RNAs with the length of approximately 19-25 nucleotides. They control the expression of multiple genes through posttranscriptional gene silencing in eukaryotes or some viruses. Increasing evidence indicates that miRNAs are involved in various cellular functions and biological processes, such as cell proliferation and differentiation, growth and development, and metabolic homeostasis. Hence, miRNAs participate in multiple pathological processes. Recently, many studies have shown that miRNAs play an important role by regulating ER stress in ischemia-reperfusion (I/R) injury, but the relevant mechanisms are not fully understood. In this review, we reviewed the current understanding of ER stress, as well as the biogenesis and function of miRNAs, and focused on the role of miRNAs regulation of ER stress in I/R injury, with the aim of providing new targets for the treatment of I/R injury.
Collapse
Affiliation(s)
- Wanying Liu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Qi Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Shiyun Guo
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Honggang Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
12
|
He Y, Wang J, Ying C, Xu KL, Luo J, Wang B, Gao J, Yin Z, Zhang Y. The interplay between ferroptosis and inflammation: therapeutic implications for cerebral ischemia-reperfusion. Front Immunol 2024; 15:1482386. [PMID: 39582857 PMCID: PMC11583640 DOI: 10.3389/fimmu.2024.1482386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 10/14/2024] [Indexed: 11/26/2024] Open
Abstract
Stroke ranks as the second most significant contributor to mortality worldwide and is a major factor in disability. Ischemic strokes account for 71% of all stroke incidences globally. The foremost approach to treating ischemic stroke prioritizes quick reperfusion, involving methods such as intravenous thrombolysis and endovascular thrombectomy. These techniques can reduce disability but necessitate immediate intervention. After cerebral ischemia, inflammation rapidly arises in the vascular system, producing pro-inflammatory signals that activate immune cells, which in turn worsen neuronal injury. Following reperfusion, an overload of intracellular iron triggers the Fenton reaction, resulting in an excess of free radicals that cause lipid peroxidation and damage to cellular membranes, ultimately leading to ferroptosis. The relationship between inflammation and ferroptosis is increasingly recognized as vital in the process of cerebral ischemia-reperfusion (I/R). Inflammatory processes disturb iron balance and encourage lipid peroxidation (LPO) through neuroglial cells, while also reducing the activity of antioxidant systems, contributing to ferroptosis. Furthermore, the lipid peroxidation products generated during ferroptosis, along with damage-associated molecular patterns (DAMPs) released from ruptured cell membranes, can incite inflammation. Given the complex relationship between ferroptosis and inflammation, investigating their interaction in brain I/R is crucial for understanding disease development and creating innovative therapeutic options. Consequently, this article will provide a comprehensive introduction of the mechanisms linking ferroptosis and neuroinflammation, as well as evaluate potential treatment modalities, with the goal of presenting various insights for alleviating brain I/R injury and exploring new therapeutic avenues.
Collapse
Affiliation(s)
- Yuxuan He
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jingyi Wang
- Faculty of Chinese Medicine of Macau University of Science and
Technology, Macao, Macao SAR, China
| | - Chunmiao Ying
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Kang Li Xu
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jingwen Luo
- Faculty of Chinese Medicine of Macau University of Science and
Technology, Macao, Macao SAR, China
| | - Baiqiao Wang
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jing Gao
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Zaitian Yin
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yunke Zhang
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
13
|
Gao J, Song X, Feng Y, Wu L, Ding Z, Qi S, Yu M, Wu R, Zheng X, Qin Y, Tang Y, Wang M, Feng X, Zhang Q. Electroacupuncture ameliorates depression-like behaviors in rats with post-stroke depression by inhibiting ferroptosis in the prefrontal cortex. Front Neurosci 2024; 18:1422638. [PMID: 39420985 PMCID: PMC11483888 DOI: 10.3389/fnins.2024.1422638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Post-stroke depression (PSD) is the most common complication following a stroke, significantly hindering recovery and rehabilitation in affected patients. Despite its prevalence, the pathogenesis of PSD remains poorly understood. Electroacupuncture (EA) has shown antidepressant effects, yet its neuroprotective properties are not well defined. Ferroptosis, a recently identified form of cell death, is implicated in the pathological processes of stroke and is associated with the development of depression-like behaviors. So we aimed to investigate whether PSD induces ferroptosis, identify potential therapeutic targets within these pathways, and elucidate the underlying mechanisms in this study. Methods Male Sprague-Dawley rats were subjected to middle carotid artery occlusion and chronic unpredictable mild stress to model PSD. To explore the role of ferroptosis in the effects of EA, the ferroptosis inducer erastin was administered into the rats' lateral ventricles, followed by 14 days of EA treatment, with sessions lasting 30 minutes per day. The Zea-Longa score was used to assess neurological deficits, while the sucrose preference test, elevated plus maze test, and open-field test were employed to evaluate depression-like behaviors in the rats. Hematoxylin-eosin, Nissl, and Perl's staining were used to observe the morphological changes and iron deposition in the prefrontal neurons. Transmission electron microscopy provided detailed observations of mitochondrial morphological changes in neurons. We utilized activity assay kits, enzyme-linked immunosorbent assay (ELISA), and Western blotting to explore potential molecular mechanisms underlying the effects of EA. Results EA can reduce neurological deficits and enhance the spontaneous activity and exploration behavior of rats. In addition, EA could inhibit prefrontal cortex neuronal ferroptosis by reducing iron deposition, decreasing lipid peroxidation, and enhancing antioxidation. Discussion EA improved depression-like behaviors, mitigated mitochondrial damage, and inhibited ferroptosis in prefrontal cortex neurons. Notably, the administration of erastin further enhanced these effects. In conclusion, EA appears to improve PSD by inhibiting ferroptosis in the prefrontal cortex.
Collapse
Affiliation(s)
- Jing Gao
- Department of Rehabilitation Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaolei Song
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yixuan Feng
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lihua Wu
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhimin Ding
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Shikui Qi
- Department of Rehabilitation Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Mingyue Yu
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ruonan Wu
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xinyue Zheng
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yanyan Qin
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yuchuang Tang
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Mengyu Wang
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaodong Feng
- Department of Rehabilitation Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- School of Rehabilitation Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Qiongshuai Zhang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
14
|
Sun YW, Zhao BW, Li HF, Zhang GX. Overview of ferroptosis and pyroptosis in acute liver failure. World J Gastroenterol 2024; 30:3856-3861. [PMID: 39350783 PMCID: PMC11438646 DOI: 10.3748/wjg.v30.i34.3856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 09/10/2024] Open
Abstract
In this editorial, we comment on the article by Zhou et al published in a recent issue. We specifically focus on the crucial roles of ferroptosis and pyroptosis in acute liver failure (ALF), a disease with high mortality rates. Ferroptosis is the result of increased intracellular reactive oxygen species due to iron accumulation, glutathione (GSH) depletion, and decreased GSH peroxidase 4 activity, while pyroptosis is a procedural cell death mediated by gasdermin D which initiates a sustained inflammatory process. In this review, we describe the characteristics of ferroptosis and pyroptosis, and discuss the involvement of the two cell death modes in the onset and development of ALF. Furthermore, we summarize several interfering methods from the perspective of ferroptosis and pyroptosis for the alleviation of ALF. These observations might provide new targets and a theoretical basis for the treatment of ALF, which are also crucial for improving the prognosis of patients with ALF.
Collapse
Affiliation(s)
- Ya-Wen Sun
- College of Life Sciences, Shandong Agricultural University, Tai’an 271018, Shandong Province, China
| | - Bo-Wen Zhao
- College of Life Sciences, Shandong Agricultural University, Tai’an 271018, Shandong Province, China
| | - Hai-Fang Li
- College of Life Sciences, Shandong Agricultural University, Tai’an 271018, Shandong Province, China
| | - Guang-Xiao Zhang
- College of Life Sciences, Shandong Agricultural University, Tai’an 271018, Shandong Province, China
| |
Collapse
|
15
|
Meng Y, Chen L, Chai Y, Meng W, Yang G, Ren J, Li H, Qi P, Chen J, Wang N. PUM2 promoted osteoarthritis progression through PTEN-mediated chondrocyte ferroptosis by facilitating NEDD4 mRNA degradation. ENVIRONMENTAL TOXICOLOGY 2024; 39:4318-4332. [PMID: 38733337 DOI: 10.1002/tox.24310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/21/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024]
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint disease with a lack of effective therapeutic. Chondrocyte ferroptosis contributes to the progression of OA. PUM2 is shown to exacerbate ischemia-reperfusion-induced neuroinflammation by promoting ferroptosis, but its role in OA remains unexplored. Here, primary mouse chondrocytes were stimulated with IL-1β to mimic OA chondrocyte injury in vitro. And PUM2 was upregulated in OA cartilage tissues and IL-1β-induced chondrocytes. Silencing PUM2 alleviated IL-1β-induced chondrocyte inflammation and ECM degradation. Mechanistically, PUM2 facilitated the degradation of NEDD4 mRNA by binding to the 3'UTR of NEDD4 mRNA, which in turn inhibited NEDD4 induced PTEN ubiquitination and degradation. Consistently, NEDD4 silencing reversed the ameliorative effect of PUM2 knockdown on chondrocyte injury, and overexpression of PTEN abolished the improved role of NEDD4 in chondrocyte injury. Moreover, PTEN aggravated IL-1β-induced ferroptosis in chondrocytes through the Nrf2/HO-1 pathway by increasing the levels of Fe2+, ROS, MDA, and ACSL4 protein, decreasing the activity of SOD and the levels of GSH and GPX4 protein, and aggravating mitochondrial damage. Additionally, destabilized medial meniscus (DMM) were conducted to establish the OA mouse model, and adenovirus-mediated PUM2 shRNA was administered intra-articularly. Silencing PUM2 attenuated OA-induced cartilage damage in vivo. In conclusion, PUM2 promoted OA progression through PTEN-mediated chondrocyte ferroptosis by facilitating NEDD4 mRNA degradation.
Collapse
Affiliation(s)
- Yu Meng
- Department of Emergency Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Chen
- Department of Emergency Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuxia Chai
- Department of Emergency Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weili Meng
- Department of Emergency Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guohui Yang
- Department of Emergency Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jia Ren
- Department of Emergency Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongshuai Li
- Department of Emergency Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Peiyi Qi
- Department of Emergency Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Juwu Chen
- Department of Emergency Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Nan Wang
- Department of Emergency Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
16
|
Zou D, Liao J, Xiao M, Liu L, Xu M. Melatonin alleviates hyperoxia-induced lung injury through elevating MSC exosomal miR-18a-5p expression to repress PUM2 signaling. FASEB J 2024; 38:e70012. [PMID: 39183539 DOI: 10.1096/fj.202400374r] [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: 02/18/2024] [Revised: 08/07/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024]
Abstract
Mesenchymal stem cells (MSC)-derived exosomes (Exo) are a possible option for hyperoxia-induced lung injury (HLI). We wanted to see if melatonin (MT)-pretreated MSC-derived exosomes (MT-Exo) were more effective against HLI, and we also tried to figure out the underlying mechanism. HLI models were established by hyperoxia exposure. HE staining was adopted to analyze lung pathological changes. MTT and flow cytometry were used to determine cell viability and apoptosis, respectively. The mitochondrial membrane potential (MMP) was analyzed using the JC-1 probe. LDH, ROS, SOD, and GSH-Px levels were examined by the corresponding kits. The interactions between miR-18a-5p, PUM2, and DUB3 were analyzed by molecular interaction experiments. MT-Exo could effectively inhibit hyperoxia-induced oxidative stress, inflammatory injury, and apoptosis in lung epithelial cells, while these effects of MT-Exo were weakened by miR-18a-5p knockdown in MSCs. miR-18a-5p reduced PUM2 expression in MLE-12 cells by directly targeting PUM2. In addition, PUM2 inactivated the Nrf2/HO-1 signaling pathway by promoting DUB3 mRNA decay post-transcriptionally. As expected, PUM2 overexpression or DUB3 knockdown abolished the protective effect of MT-Exo on hyperoxia-induced lung epithelial cell injury. MT-Exo carrying miR-18a-5p reduced hyperoxia-mediated lung injury in mice through activating Nrf2/HO-1 pathway. MT reduced PUM2 expression and subsequently activated the DUB3/Nrf2/HO-1 signal axis by increasing miR-18a-5p expression in MSC-derived exosomes to alleviate HLI.
Collapse
Affiliation(s)
- Dongmei Zou
- The Department of Pediatric, Shenzhen Children's Hospital, China Medical University, Shenzhen, Guangdong, China
| | - Jinwen Liao
- The Department of Pediatric, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong, China
| | - Min Xiao
- The Department of Pediatric, Shenzhen Children's Hospital, China Medical University, Shenzhen, Guangdong, China
| | - Liang Liu
- The Department of Pediatric, Shenzhen Children's Hospital, China Medical University, Shenzhen, Guangdong, China
| | - Mingguo Xu
- The Department of Pediatric, Shenzhen Children's Hospital, China Medical University, Shenzhen, Guangdong, China
- The Department of Pediatric, The Third People's Hospital of Longgang District Shenzhen, Shenzhen, Guangdong, China
| |
Collapse
|
17
|
Liu S, Zhang P, Wu Y, Zhou H, Wu H, Jin Y, Wu D, Wu G. SLC25A19 is a novel prognostic biomarker related to immune invasion and ferroptosis in HCC. Int Immunopharmacol 2024; 136:112367. [PMID: 38823177 DOI: 10.1016/j.intimp.2024.112367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024]
Abstract
SLC25A19 is a mitochondrial thiamine pyrophosphate (TPP) carrier that mediates TPP entry into the mitochondria. SLC25A19 has been recognized to play a crucial role in many metabolic diseases, but its role in cancer has not been clearly reported. Based on clinical data from The Cancer Genome Atlas (TCGA), the following parameters were analyzed among HCC patients: SLC25A19 expression, enrichment analyses, immune infiltration, ferroptosis and prognosis analyses. In vitro, the SLC25A19 high expression was validated by qRT-PCR and Immunohistochemistry. Subsequently, a series of cell function experiments, including CCK8, EdU, clone formation, trans-well and scratch assays, were conducted to illustrate the effect of SLC25A19 on the growth and metastasis of cancer cells. Meanwhile, indicators related to ferroptosis were also detected. SCL25A19 is highly expressed in HCC and predicts a poor prognosis. Elevated SLC25A19 expression in HCC patients was markedly associated with T stage, pathological status (PS), tumor status (TS), histologic grade (HG), and AFP. Our results indicate that SLC25A19 has a generally good prognosis predictive and diagnostic ability. The results of gene enrichment analyses showed that SLC25A19 is significantly correlated with immune infiltration, fatty acid metabolism, and ferroptosis marker genes. In vitro experiments have confirmed that silencing SLC25A19 can significantly inhibit the proliferation and migration ability of cancer cells and induce ferroptosis in HCC. In conclusion, these findings indicate that SLC25A19 is novel prognostic biomarker related to immune invasion and ferroptosis in HCC, and it is an excellent candidate for therapeutic target against HCC.
Collapse
Affiliation(s)
- Shiqi Liu
- Hepatobiliary Surgery Department, First Hospital of China Medical, University, No.155, Nanjingbei Street, 110001 Shenyang, Liaoning, Province, PR China; Key Laboratory of General Surgery of Liaoning Province, the First Affiliated Hospital of China Medical University, No.155, Nanjingbei Street, 110001 Shenyang, Liaoning Province, PR China
| | - Pengjie Zhang
- Hepatobiliary Surgery Department, First Hospital of China Medical, University, No.155, Nanjingbei Street, 110001 Shenyang, Liaoning, Province, PR China; Key Laboratory of General Surgery of Liaoning Province, the First Affiliated Hospital of China Medical University, No.155, Nanjingbei Street, 110001 Shenyang, Liaoning Province, PR China
| | - Yubo Wu
- Hepatobiliary Surgery Department, First Hospital of China Medical, University, No.155, Nanjingbei Street, 110001 Shenyang, Liaoning, Province, PR China; Key Laboratory of General Surgery of Liaoning Province, the First Affiliated Hospital of China Medical University, No.155, Nanjingbei Street, 110001 Shenyang, Liaoning Province, PR China
| | - Haonan Zhou
- Hepatobiliary Surgery Department, First Hospital of China Medical, University, No.155, Nanjingbei Street, 110001 Shenyang, Liaoning, Province, PR China; Key Laboratory of General Surgery of Liaoning Province, the First Affiliated Hospital of China Medical University, No.155, Nanjingbei Street, 110001 Shenyang, Liaoning Province, PR China
| | - Haomin Wu
- Hepatobiliary Surgery Department, First Hospital of China Medical, University, No.155, Nanjingbei Street, 110001 Shenyang, Liaoning, Province, PR China; Key Laboratory of General Surgery of Liaoning Province, the First Affiliated Hospital of China Medical University, No.155, Nanjingbei Street, 110001 Shenyang, Liaoning Province, PR China
| | - Yifan Jin
- Hepatobiliary Surgery Department, First Hospital of China Medical, University, No.155, Nanjingbei Street, 110001 Shenyang, Liaoning, Province, PR China; Key Laboratory of General Surgery of Liaoning Province, the First Affiliated Hospital of China Medical University, No.155, Nanjingbei Street, 110001 Shenyang, Liaoning Province, PR China
| | - Di Wu
- Hepatobiliary Surgery Department, First Hospital of China Medical, University, No.155, Nanjingbei Street, 110001 Shenyang, Liaoning, Province, PR China; Key Laboratory of General Surgery of Liaoning Province, the First Affiliated Hospital of China Medical University, No.155, Nanjingbei Street, 110001 Shenyang, Liaoning Province, PR China
| | - Gang Wu
- Hepatobiliary Surgery Department, First Hospital of China Medical, University, No.155, Nanjingbei Street, 110001 Shenyang, Liaoning, Province, PR China; Key Laboratory of General Surgery of Liaoning Province, the First Affiliated Hospital of China Medical University, No.155, Nanjingbei Street, 110001 Shenyang, Liaoning Province, PR China.
| |
Collapse
|
18
|
Xu Y, Jia B, Li J, Li Q, Luo C. The Interplay between Ferroptosis and Neuroinflammation in Central Neurological Disorders. Antioxidants (Basel) 2024; 13:395. [PMID: 38671843 PMCID: PMC11047682 DOI: 10.3390/antiox13040395] [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: 03/11/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Central neurological disorders are significant contributors to morbidity, mortality, and long-term disability globally in modern society. These encompass neurodegenerative diseases, ischemic brain diseases, traumatic brain injury, epilepsy, depression, and more. The involved pathogenesis is notably intricate and diverse. Ferroptosis and neuroinflammation play pivotal roles in elucidating the causes of cognitive impairment stemming from these diseases. Given the concurrent occurrence of ferroptosis and neuroinflammation due to metabolic shifts such as iron and ROS, as well as their critical roles in central nervous disorders, the investigation into the co-regulatory mechanism of ferroptosis and neuroinflammation has emerged as a prominent area of research. This paper delves into the mechanisms of ferroptosis and neuroinflammation in central nervous disorders, along with their interrelationship. It specifically emphasizes the core molecules within the shared pathways governing ferroptosis and neuroinflammation, including SIRT1, Nrf2, NF-κB, Cox-2, iNOS/NO·, and how different immune cells and structures contribute to cognitive dysfunction through these mechanisms. Researchers' findings suggest that ferroptosis and neuroinflammation mutually promote each other and may represent key factors in the progression of central neurological disorders. A deeper comprehension of the common pathway between cellular ferroptosis and neuroinflammation holds promise for improving symptoms and prognosis related to central neurological disorders.
Collapse
Affiliation(s)
- Yejia Xu
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
- Hebei Key Laboratory of Forensic Medicine, College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Bowen Jia
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
| | - Jing Li
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
| | - Qianqian Li
- NHC Key Laboratory of Drug Addiction Medicine, Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming 650500, China
- School of Forensic Medicine, Wannan Medical College, Wuhu 241002, China
| | - Chengliang Luo
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
- Hebei Key Laboratory of Forensic Medicine, College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China
- NHC Key Laboratory of Drug Addiction Medicine, Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming 650500, China
| |
Collapse
|
19
|
Chen X, Wang Z, Li C, Zhang Z, Lu S, Wang X, Liang Q, Zhu X, Pan C, Wang Q, Ji Z, Wang Y, Piao M, Chi G, Ge P. SIRT1 activated by AROS sensitizes glioma cells to ferroptosis via induction of NAD+ depletion-dependent activation of ATF3. Redox Biol 2024; 69:103030. [PMID: 38181705 PMCID: PMC10791567 DOI: 10.1016/j.redox.2024.103030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/26/2023] [Accepted: 01/02/2024] [Indexed: 01/07/2024] Open
Abstract
Ferroptosis is a type of programmed cell death resulting from iron overload-dependent lipid peroxidation, and could be promoted by activating transcription factor 3 (ATF3). SIRT1 is an enzyme accounting for removing acetylated lysine residues from target proteins by consuming NAD+, but its role remains elusive in ferroptosis and activating ATF3. In this study, we found SIRT1 was activated during the process of RSL3-induced glioma cell ferroptosis. Moreover, the glioma cell death was aggravated by SIRT1 activator SRT2183, but suppressed by SIRT inhibitor EX527 or when SIRT1 was silenced with siRNA. These indicated SIRT1 sensitized glioma cells to ferroptosis. Furthermore, we found SIRT1 promoted RSL3-induced expressional upregulation and nuclear translocation of ATF3. Silence of ATF3 with siRNA attenuated RSL3-induced increases of ferrous iron and lipid peroxidation, downregulation of SLC7A11 and GPX4 and depletion of cysteine and GSH. Thus, SIRT1 promoted glioma cell ferroptosis by inducting ATF3 activation. Mechanistically, ATF3 activation was reinforced when RSL3-induced decline of NAD+ was aggravated by FK866 that could inhibit NAD + synthesis via salvage pathway, but suppressed when intracellular NAD+ was maintained at higher level by supplement of exogenous NAD+. Notably, the NAD + decline caused by RSL3 was enhanced when SIRT1 was further activated by SRT2183, but attenuated when SIRT1 activation was inhibited by EX527. These indicated SIRT1 promoted ATF3 activation via consumption of NAD+. Finally, we found RSL3 activated SIRT1 by inducing reactive oxygen species-dependent upregulation of AROS. Together, our study revealed SIRT1 activated by AROS sensitizes glioma cells to ferroptosis via activation of ATF3-dependent inhibition of SLC7A11 and GPX4.
Collapse
Affiliation(s)
- Xi Chen
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China; Research Center of Neuroscience, First Hospital of Jilin University, Changchun, 130021, China
| | - Zhenchuan Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China; Research Center of Neuroscience, First Hospital of Jilin University, Changchun, 130021, China
| | - Chen Li
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China; Research Center of Neuroscience, First Hospital of Jilin University, Changchun, 130021, China
| | - Zhao Zhang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China; Research Center of Neuroscience, First Hospital of Jilin University, Changchun, 130021, China
| | - Shan Lu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Xuanzhong Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Qi Liang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Xiaoxi Zhu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Chengliang Pan
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Qingxuan Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Zhilin Ji
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Yubo Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Meihua Piao
- Department of Anesthesiology, First Hospital of Jilin University, Changchun, 130021, China
| | - Guangfan Chi
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Pengfei Ge
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, 130021, China; Research Center of Neuroscience, First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
20
|
Hu X, Bao Y, Li M, Zhang W, Chen C. The role of ferroptosis and its mechanism in ischemic stroke. Exp Neurol 2024; 372:114630. [PMID: 38056585 DOI: 10.1016/j.expneurol.2023.114630] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/02/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
Ischemic stroke is an acute cerebrovascular disease with a high morbidity, mortality, and disability rate. Persistent ischemia of brain tissue can cause irreversible damage to neurons, leading to neurological dysfunction and seriously affecting patients' quality of life. However, current clinical therapies are limited and have not achieved satisfactory outcome, due to the incomplete understanding of the mechanism of neuronal damage during ischemic stroke. Recent studies have found that ferroptosis is implicated in the pathophysiology of ischemic stroke. Ferroptosis is an iron-dependent regulated cell death driven by lipid peroxidation. Under normal physiological conditions, GSH/GPX4, FSP1/CoQ10, GCH/BH4 and other anti-ferroptosis pathways can function effectively to suppress the occurrence of ferroptosis. After ischemic stroke, two typical ferroptosis characteristics, lipid peroxidation and iron accumulation, are observed, accompanied by changes in the expression of ferroptosis related genes such as GPX4, ACSL4, and SLC7A11, suggesting that ferroptosis plays a key role in ischemic stroke, which provides a new idea for the clinical treatment of ischemic stroke. This article reviewed the pathological mechanisms of ferroptosis in the occurrence and development of ischemic stroke, as well as the related progress of ferroptosis targeted therapy.
Collapse
Affiliation(s)
- Xiaodan Hu
- School of Clinical Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yutong Bao
- School of Clinical Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Man Li
- Department of Human Anatomy, Histology and embryology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Weiguang Zhang
- Department of Human Anatomy, Histology and embryology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Chunhua Chen
- Department of Human Anatomy, Histology and embryology, School of Basic Medical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
21
|
Feng S, Tang D, Wang Y, Li X, Bao H, Tang C, Dong X, Li X, Yang Q, Yan Y, Yin Z, Shang T, Zheng K, Huang X, Wei Z, Wang K, Qi S. The mechanism of ferroptosis and its related diseases. MOLECULAR BIOMEDICINE 2023; 4:33. [PMID: 37840106 PMCID: PMC10577123 DOI: 10.1186/s43556-023-00142-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/23/2023] [Indexed: 10/17/2023] Open
Abstract
Ferroptosis, a regulated form of cellular death characterized by the iron-mediated accumulation of lipid peroxides, provides a novel avenue for delving into the intersection of cellular metabolism, oxidative stress, and disease pathology. We have witnessed a mounting fascination with ferroptosis, attributed to its pivotal roles across diverse physiological and pathological conditions including developmental processes, metabolic dynamics, oncogenic pathways, neurodegenerative cascades, and traumatic tissue injuries. By unraveling the intricate underpinnings of the molecular machinery, pivotal contributors, intricate signaling conduits, and regulatory networks governing ferroptosis, researchers aim to bridge the gap between the intricacies of this unique mode of cellular death and its multifaceted implications for health and disease. In light of the rapidly advancing landscape of ferroptosis research, we present a comprehensive review aiming at the extensive implications of ferroptosis in the origins and progress of human diseases. This review concludes with a careful analysis of potential treatment approaches carefully designed to either inhibit or promote ferroptosis. Additionally, we have succinctly summarized the potential therapeutic targets and compounds that hold promise in targeting ferroptosis within various diseases. This pivotal facet underscores the burgeoning possibilities for manipulating ferroptosis as a therapeutic strategy. In summary, this review enriched the insights of both investigators and practitioners, while fostering an elevated comprehension of ferroptosis and its latent translational utilities. By revealing the basic processes and investigating treatment possibilities, this review provides a crucial resource for scientists and medical practitioners, aiding in a deep understanding of ferroptosis and its effects in various disease situations.
Collapse
Affiliation(s)
- Shijian Feng
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Dan Tang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yichang Wang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiang Li
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hui Bao
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Chengbing Tang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiuju Dong
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xinna Li
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Qinxue Yang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yun Yan
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zhijie Yin
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Tiantian Shang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Kaixuan Zheng
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiaofang Huang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zuheng Wei
- Chengdu Jinjiang Jiaxiang Foreign Languages High School, Chengdu, People's Republic of China
| | - Kunjie Wang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Shiqian Qi
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
22
|
Zhang Z, Liu C, Zhou X, Zhang X. The Critical Role of Sirt1 in Subarachnoid Hemorrhages: Mechanism and Therapeutic Considerations. Brain Sci 2023; 13:brainsci13040674. [PMID: 37190639 DOI: 10.3390/brainsci13040674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/28/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
The subarachnoid hemorrhage (SAH) is an important cause of death and long-term disability worldwide. As a nicotinamide adenine dinucleotide-dependent deacetylase, silent information regulator 1 (Sirt1) is a multipotent molecule involved in many pathophysiological processes. A growing number of studies have demonstrated that Sirt1 activation may exert positive effects on SAHs by regulating inflammation, oxidative stress, apoptosis, autophagy, and ferroptosis. Thus, Sirt1 agonists may serve as potential therapeutic drugs for SAHs. In this review, we summarized the current state of our knowledge on the relationship between Sirt1 and SAHs and provided an updated overview of the downstream molecules of Sirt1 in SAHs.
Collapse
Affiliation(s)
- Zhonghua Zhang
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Cong Liu
- Department of Ophthalmology, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Xiaoming Zhou
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Xin Zhang
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
23
|
Zeng J, Guo J, Huang S, Cheng Y, Luo F, Xu X, Chen R, Ma G, Wang Y. The roles of sirtuins in ferroptosis. Front Physiol 2023; 14:1131201. [PMID: 37153222 PMCID: PMC10157232 DOI: 10.3389/fphys.2023.1131201] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 04/10/2023] [Indexed: 05/09/2023] Open
Abstract
Ferroptosis represents a novel non-apoptotic form of regulated cell death that is driven by iron-dependent lipid peroxidation and plays vital roles in various diseases including cardiovascular diseases, neurodegenerative disorders and cancers. Plenty of iron metabolism-related proteins, regulators of lipid peroxidation, and oxidative stress-related molecules are engaged in ferroptosis and can regulate this complex biological process. Sirtuins have broad functional significance and are targets of many drugs in the clinic. Recently, a growing number of studies have revealed that sirtuins can participate in the occurrence of ferroptosis by affecting many aspects such as redox balance, iron metabolism, and lipid metabolism. This article reviewed the studies on the roles of sirtuins in ferroptosis and the related molecular mechanisms, highlighting valuable targets for the prevention and treatment of ferroptosis-associated diseases.
Collapse
Affiliation(s)
- Jieqing Zeng
- Maternal and Children’s Health Research Institute, Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
| | - Junhao Guo
- Maternal and Children’s Health Research Institute, Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
| | - Si Huang
- Maternal and Children’s Health Research Institute, Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
| | - Yisen Cheng
- Maternal and Children’s Health Research Institute, Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
| | - Fei Luo
- Maternal and Children’s Health Research Institute, Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
| | - Xusan Xu
- Maternal and Children’s Health Research Institute, Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
| | - Riling Chen
- Maternal and Children’s Health Research Institute, Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
| | - Guoda Ma
- Maternal and Children’s Health Research Institute, Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
- *Correspondence: Guoda Ma, ; Yajun Wang,
| | - Yajun Wang
- Institute of Respiratory, Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
- *Correspondence: Guoda Ma, ; Yajun Wang,
| |
Collapse
|
24
|
Zhou L, Han S, Guo J, Qiu T, Zhou J, Shen L. Ferroptosis-A New Dawn in the Treatment of Organ Ischemia-Reperfusion Injury. Cells 2022; 11:cells11223653. [PMID: 36429080 PMCID: PMC9688314 DOI: 10.3390/cells11223653] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/12/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022] Open
Abstract
Ischemia-reperfusion (I/R) is a common pathological phenomenon that occurs in numerous organs and diseases. It generally results from secondary damage caused by the recovery of blood flow and reoxygenation, followed by ischemia of organ tissues, which is often accompanied by severe cellular damage and death. Currently, effective treatments for I/R injury (IRI) are limited. Ferroptosis, a new type of regulated cell death (RCD), is characterized by iron overload and iron-dependent lipid peroxidation. Mounting evidence has indicated a close relationship between ferroptosis and IRI. Ferroptosis plays a significantly detrimental role in the progression of IRI, and targeting ferroptosis may be a promising approach for treatment of IRI. Considering the substantial progress made in the study of ferroptosis in IRI, in this review, we summarize the pathological mechanisms and therapeutic targets of ferroptosis in IRI.
Collapse
Affiliation(s)
- Linxiang Zhou
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Shangting Han
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Jiayu Guo
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Tao Qiu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Jiangqiao Zhou
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
- Correspondence: (J.Z.); (L.S.)
| | - Lei Shen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
- Correspondence: (J.Z.); (L.S.)
| |
Collapse
|