1
|
Zhu Y, Cao C, Li Z, Xu Z, Qian S, Zhang J, Li M, Hu X, Zhang A, Du N, Pan X, Wang X, Sun Y, Wang J, Huang Y. ASIC1a regulates ferroptosis in hepatic stellate cells via the Hippo/Yap-1 pathway in liver fibrosis. Int Immunopharmacol 2024; 143:113226. [PMID: 39353388 DOI: 10.1016/j.intimp.2024.113226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/28/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Liver fibrosis is a sustained process of liver tissue damage and repair caused by various physiological and pathological factors, with the activation and proliferation of hepatic stellate cells being central. Therefore, understanding and clarifying the relevant mechanisms of hepatic stellate cell activation and death is of great clinical significance for the treatment of liver fibrosis diseases. METHODS In vivo, recombinant adeno-associated virus was used to infect the liver of experimental mice, overexpressing ASIC1a, and based on this, a liver fibrosis model treated with sorafenib was constructed. In vitro, using RNA plasmid technology to transfect HSC-T6 cells, ASIC1a was overexpressed or silenced in the cells, and on this basis, PDGF-BB and Sorafenib were used to stimulate HSC-T6 cells, causing activated HSC-T6 to undergo ferroptosis. RESULTS The ferroptosis inducers Sorafenib and erastin can induce ferroptosis in HSCs, effectively inhibiting or reversing the progression of liver fibrosis. We found that the expression level of ASIC1a was significantly reduced in the livers of mice with liver fibrosis treated with Sorafenib. After treatment with an adeno-associated virus overexpressing ASIC1a, the therapeutic effect of Sorafenib was inhibited, and the level of ferroptosis induced by Sorafenib was also inhibited. The induction of ferroptosis in hepatic stellate cells in vitro depends on the presence of ASIC1a. By further exploring the potential mechanism, we observed that the overexpression of ASIC1a can promote an increase in YAP nuclear translocation, thereby regulating the activity of Hippo/YAP pathway signaling. After treatment with Sorafenib, the influx of Ca2+ significantly increased when ASIC1a was overexpressed, and BAPTA-AM intervention eliminated the intracellular Ca2+ accumulation induced by ASIC1a overexpression. CONCLUSIONS This indicated that the activation of YAP depends on the calcium ion influx induced by ASIC1a, which regulates ferroptosis in hepatic stellate cells by regulating the calcium ion-dependent Hippo/YAP pathway.
Collapse
Affiliation(s)
- Yueqin Zhu
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Office of Drug Clinical Trial Institutions, Anhui Provincial Cancer Hospital, Hefei 230031, China
| | - Chun Cao
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Zihao Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Zhou Xu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Shishun Qian
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Jingrong Zhang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Mengxue Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Xiaojie Hu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Anqi Zhang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Na Du
- Shanghai Songjiang District Central Hospital, Shanghai 201600, China
| | - Xuesheng Pan
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xinchen Wang
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Office of Drug Clinical Trial Institutions, Anhui Provincial Cancer Hospital, Hefei 230031, China
| | - Yancai Sun
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Office of Drug Clinical Trial Institutions, Anhui Provincial Cancer Hospital, Hefei 230031, China
| | - Jiajia Wang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| | - Yan Huang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
2
|
He C, Zeng Z, Yang Y, Ye S, Wu Q, Liu X, Liu C, Zeng W, Liu S. Silencing of CircTRIM25/miR-138-5p/CREB1 axis promotes chondrogenesis in osteoarthritis. Autoimmunity 2024; 57:2361749. [PMID: 39007896 DOI: 10.1080/08916934.2024.2361749] [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: 12/14/2023] [Accepted: 05/26/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Dysregulated circular RNAs (circRNAs) are involved in osteoarthritis (OA) progression. OBJECTIVE We aimed to explore the effect of hsa_circ_0044719 (circTRIM25) on the ferroptosis of chondrocytes. METHODS Chondrocytes were treated with interleukin (IL)-1β to generate cell model. Cellular behaviours were measured using cell counting kit-8, enzyme-linked immunosorbent assay, relevant kits, propidium iodide staining, and immunofluorescence assay. Quantitative real-time polymerase chain reaction was performed to examine the expression of circTRIM25, miR-138-5p, and cAMP responsive element binding protein 1 (CREB1), and their interactions were assessed using luciferase reporter analysis and RNA pull-down assay. RESULTS CircTRIM25 was upregulated in OA tissues and IL-1β-stimulated chondrocytes. Knockdown of circTRIM25 facilitated the viability and suppressed ferroptosis and inflammation of IL-1β-induced cells. CircTRIM25 served as a sponge of miR-138-5p, which directly targets CREB1. Downregulation of miR-138-5p abrogated the effect induced by knockdown of circTRIM25. Furthermore, enforced CREB1 reversed the miR-138-5p induced effect. Moreover, knockdown of circTRIM25 attenuated cartilage injury in vivo. CONCLUSION Silencing of circTRIM25 inhibited ferroptosis of chondrocytes via the miR-138-5p/CREB axis and thus attenuated OA progression.
Collapse
Affiliation(s)
- Chunlei He
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | | | - Yadong Yang
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Shanshan Ye
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Qiang Wu
- Gannan Medical University, Ganzhou, China
| | - Xunzhi Liu
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Chenghong Liu
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Wanhui Zeng
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Sheng Liu
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
3
|
Zhang X, Liu Z, Li Z, Qi L, Huang T, Li F, Li M, Wang Y, Ma Z, Gao Y. Ferroptosis pathways: Unveiling the neuroprotective power of cistache deserticola phenylethanoid glycosides. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118465. [PMID: 38944360 DOI: 10.1016/j.jep.2024.118465] [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: 05/03/2024] [Revised: 05/22/2024] [Accepted: 06/14/2024] [Indexed: 07/01/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cistanche deserticola is a kind of parasitic plant living in the roots of desert trees. It is a rare Chinese medicine, which has the effect of tonifying kidney Yang, benefiting essence and blood and moistening the intestinal tract. Cistache deserticola phenylethanoid glycoside (PGS), an active component found in Cistanche deserticola Ma, have potential kidney tonifying, intellectual enhancing, and neuroprotective effects. Cistanche total glycoside capsule has been marketed to treat vascular dementia disease. AIM OF THE STUDY To identify the potential renal, intellectual enhancing and neuroprotective effects of PGS and explore the exact targets and mechanisms of PGS. MATERIALS AND METHODS This study systematically investigated the four types of pathways leading to ferroptosis through transcriptome, metabolome, ultrastructure and molecular biology techniques and explored the molecular mechanism by which multiple PGS targets and pathways synergistically exert neuroprotective effects on hypoxia. RESULTS PGS alleviated learning and memory dysfunction and pathological injury in mice exposed to hypobaric hypoxia by attenuating hypobaric hypoxia-induced hippocampal histopathological damage, impairing blood‒brain barrier integrity, increasing oxidative stress levels, and increasing the expression of cognitive proteins. PGS reduced the formation of lipid peroxides and improved ferroptosis by upregulating the GPX-4/SCL7A311 axis and downregulating the ACSL4/LPCAT3/LOX axis. PGS also reduced ferroptosis by facilitating cellular Fe2+ efflux and regulating mitochondrial Fe2+ transport and effectively antagonized cell ferroptosis induced by erastin (a ferroptosis inducer). CONCLUSIONS This study demonstrated the mechanism by which PGS prevents hypobaric hypoxic nerve injury through four types of ferroptosis pathways, achieved neuroprotective effects and alleviated learning and memory dysfunction in hypobaric hypoxia mice. This study provides a theoretical basis for the development and application of PGS.
Collapse
Affiliation(s)
- Xianxie Zhang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Zuoxu Liu
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Zhihui Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Ling Qi
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Tianke Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Fang Li
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Maoxing Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Yuguang Wang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Zengchun Ma
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China
| | - Yue Gao
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006, Guangzhou, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 100850, Beijing, China.
| |
Collapse
|
4
|
Chen J, Yan L, Zhang Y, Liu X, Wei Y, Zhao Y, Li K, Shi Y, Liu H, Lai W, Tian L, Lin B. Maternal exposure to nanopolystyrene induces neurotoxicity in offspring through P53-mediated ferritinophagy and ferroptosis in the rat hippocampus. J Nanobiotechnology 2024; 22:651. [PMID: 39438901 DOI: 10.1186/s12951-024-02911-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024] Open
Abstract
There are increasing concerns regarding the rapid expansion of polystyrene nanoplastics (PS-NPs), which could impact human health. Previous studies have shown that nanoplastics can be transferred from mothers to offspring through the placenta and breast milk, resulting in cognitive deficits in offspring. However, the neurotoxic effects of maternal exposure on offspring and its mechanisms remain unclear. In this study, PS-NPs (50 nm) were gavaged to female rats throughout gestation and lactation to establish an offspring exposure model to study the neurotoxicity and behavioral changes caused by PS-NPs on offspring. Neonatal rat hippocampal neuronal cells were used to investigate the pathways through which NPs induce neurodevelopmental toxicity in offspring rats, using iron inhibitors, autophagy inhibitors, reactive oxygen species (ROS) scroungers, P53 inhibitors, and NCOA4 inhibitors. We found that low PS-NPs dosages can cause ferroptosis in the hippocampus of the offspring, resulting in a decline in the cognitive, learning, and memory abilities of the offspring. PS-NPs induced NOCA4-mediated ferritinophagy and promoted ferroptosis by inciting ROS production to activate P53-mediated ferritinophagy. Furthermore, the levels of the antioxidant factors glutathione peroxidase 4 (GPX4) and glutathione (GSH), responsible for ferroptosis, were reduced. In summary, this study revealed that consumption of PS-NPs during gestation and lactation can cause ferroptosis and damage the hippocampus of offspring. Our results can serve as a basis for further research into the neurodevelopmental effects of nanoplastics in offspring.
Collapse
Affiliation(s)
- Jiang Chen
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
- School of Public Health, North China University of Science and Technology, Tangshan, 063200, China
| | - Licheng Yan
- School of Public Health, North China University of Science and Technology, Tangshan, 063200, China
| | - Yaping Zhang
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Xuan Liu
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Yizhe Wei
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Yiming Zhao
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
- School of Public Health, North China University of Science and Technology, Tangshan, 063200, China
| | - Kang Li
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Yue Shi
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Huanliang Liu
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Wenqing Lai
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Lei Tian
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China.
| | - Bencheng Lin
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China.
| |
Collapse
|
5
|
Lai Z, Wei Y, He M, Lin C, Ouyang W, Liu X. Toxicity and related molecular mechanisms of Sb(III) in the embryos and larvae of zebrafish (Danio rerio). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 359:124551. [PMID: 39004205 DOI: 10.1016/j.envpol.2024.124551] [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: 05/12/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/16/2024]
Abstract
Antimony (Sb) pollution poses a severe threat to humans and ecosystems due to the extensive use of Sb in various fields. However, little is known about the toxic effects of Sb and its aquatic ecotoxicological mechanism. This study aimed to reveal the toxicity and related molecular mechanisms of trivalent Sb (Sb(III)) in zebrafish embryos/larvae. Sb(III) accumulated in larvae, which correlated with the exposure concentration. Although no significant lethal or teratogenic effects were observed, normal growth and development were affected. Exposure to 10 or 20 mg/L Sb(III) increased the levels of reactive oxygen species in the larvae while enhancing catalase activity and increasing cell apoptosis. Transcriptomic analysis revealed that Sb(III) promoted glutathione metabolism and the ferroptosis pathway. In addition, symptoms associated with ferroptosis, including mitochondrial damage, biochemical levels of related molecules and increased tissue iron content, were detected. Quantitative polymerase chain reaction (qPCR) analyses further confirmed that Sb(III) significantly altered the transcription levels of genes related to the ferroptosis pathway by disrupting iron homeostasis. Furthermore, ferrostatin-1 (Fer-1) mitigated the toxic effects induced by Sb(III) in zebrafish. Our research fills the gap in the literature on the toxicity and mechanism of Sb(III) in aquatic organisms, which is highly important for understanding the ecological risks associated with Sb.
Collapse
Affiliation(s)
- Ziyang Lai
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing, 100875, China
| | - Yihan Wei
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing, 100875, China
| | - Mengchang He
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing, 100875, China.
| | - Chunye Lin
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing, 100875, China
| | - Wei Ouyang
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing, 100875, China; Advanced Interdisciplinary Institute of Environment and Ecology, Beijing Normal University, Zhuhai, 519087, China
| | - Xitao Liu
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing, 100875, China
| |
Collapse
|
6
|
Stojkovic L, Djordjevic A, Stefanovic M, Stankovic A, Dincic E, Djuric T, Zivkovic M. Circulatory Indicators of Lipid Peroxidation, the Driver of Ferroptosis, Reflect Differences between Relapsing-Remitting and Progressive Multiple Sclerosis. Int J Mol Sci 2024; 25:11024. [PMID: 39456806 PMCID: PMC11507982 DOI: 10.3390/ijms252011024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Ferroptosis, a lipid peroxidation- and iron-mediated type of regulated cell death, relates to both neuroinflammation, which is common in relapsing-remitting multiple sclerosis (RRMS), and neurodegeneration, which is prevalent in progressive (P)MS. Currently, findings related to the molecular markers proposed in this paper in patients are scarce. We analyzed circulatory molecular indicators of the main ferroptosis-related processes, comprising lipid peroxidation (malondialdehyde (MDA), 4-hydroxynonenal (4-HNE), and hexanoyl-lysine adduct (HEL)), glutathione-related antioxidant defense (total glutathione (reduced (GSH) and oxidized (GSSG)) and glutathione peroxidase 4 (GPX4)), and iron metabolism (iron, transferrin and ferritin) to estimate their contributions to the clinical manifestation of MS and differences between RRMS and PMS disease course. In 153 patients with RRMS and 69 with PMS, plasma/serum lipid peroxidation indicators and glutathione were quantified using ELISA and colorimetric reactions, respectively. Iron serum concentrations were determined using spectrophotometry, and transferrin and ferritin were determined using immunoturbidimetry. Compared to those with RRMS, patients with PMS had decreased 4-HNE (median, 1368.42 vs. 1580.17 pg/mL; p = 0.03). Interactive effects of MS course (RRMS/PMS) and disease-modifying therapy status on MDA (p = 0.009) and HEL (p = 0.02) levels were detected. In addition, the interaction of disease course and self-reported fatigue revealed significant impacts on 4-HNE levels (p = 0.01) and the GSH/GSSG ratio (p = 0.04). The results also show an association of MS course (p = 0.03) and EDSS (p = 0.04) with GSH levels. No significant changes were observed in the serum concentrations of iron metabolism indicators between the two patient groups (p > 0.05). We suggest circulatory 4-HNE as an important parameter related to differences between RRMS and PMS. Significant interactions of MS course and other clinically relevant parameters with changes in redox processes associated with ferroptosis support the further investigation of MS with a larger sample while taking into account both circulatory and central nervous system estimation.
Collapse
Affiliation(s)
- Ljiljana Stojkovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia; (A.D.); (A.S.); (T.D.)
| | - Ana Djordjevic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia; (A.D.); (A.S.); (T.D.)
| | - Milan Stefanovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia; (A.D.); (A.S.); (T.D.)
| | - Aleksandra Stankovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia; (A.D.); (A.S.); (T.D.)
| | - Evica Dincic
- Clinic for Neurology, Military Medical Academy, 11000 Belgrade, Serbia;
- Medical Faculty of the Military Medical Academy, University of Defence, 11000 Belgrade, Serbia
| | - Tamara Djuric
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia; (A.D.); (A.S.); (T.D.)
| | - Maja Zivkovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia; (A.D.); (A.S.); (T.D.)
| |
Collapse
|
7
|
Cheng X, Tan Y, Li H, Zhang Z, Hui S, Zhang Z, Peng W. Mechanistic Insights and Potential Therapeutic Implications of NRF2 in Diabetic Encephalopathy. Mol Neurobiol 2024; 61:8253-8278. [PMID: 38483656 DOI: 10.1007/s12035-024-04097-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/04/2024] [Indexed: 09/21/2024]
Abstract
Diabetic encephalopathy (DE) is a complication of diabetes, especially type 2 diabetes (T2D), characterized by damage in the central nervous system and cognitive impairment, which has gained global attention. Despite the extensive research aimed at enhancing our understanding of DE, the underlying mechanism of occurrence and development of DE has not been established. Mounting evidence has demonstrated a close correlation between DE and various factors, such as Alzheimer's disease-like pathological changes, insulin resistance, inflammation, and oxidative stress. Of interest, nuclear factor erythroid 2-related factor 2 (NRF2) is a transcription factor with antioxidant properties that is crucial in maintaining redox homeostasis and regulating inflammatory responses. The activation and regulatory mechanisms of NRF2 are a relatively complex process. NRF2 is involved in the regulation of multiple metabolic pathways and confers neuroprotective functions. Multiple studies have provided evidence demonstrating the significant involvement of NRF2 as a critical transcription factor in the progression of DE. Additionally, various molecules capable of activating NRF2 expression have shown potential in ameliorating DE. Therefore, it is intriguing to consider NRF2 as a potential target for the treatment of DE. In this review, we aim to shed light on the role and the possible underlying mechanism of NRF2 in DE. Furthermore, we provide an overview of the current research landscape and address the challenges associated with using NRF2 activators as potential treatment options for DE.
Collapse
Affiliation(s)
- Xin Cheng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Mental Disorder, Changsha, 410011, China
| | - Yejun Tan
- School of Mathematics, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Hongli Li
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, Hunan, 410011, People's Republic of China
- National Clinical Research Center for Mental Disorder, Changsha, 410011, China
| | - Zhen Zhang
- YangSheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Shan Hui
- Department of Geratology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, China
| | - Zheyu Zhang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, Hunan, 410011, People's Republic of China.
- National Clinical Research Center for Mental Disorder, Changsha, 410011, China.
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, No.139 Middle Renmin Road, Changsha, Hunan, 410011, People's Republic of China.
- National Clinical Research Center for Mental Disorder, Changsha, 410011, China.
| |
Collapse
|
8
|
Hu H, Gao H, Wang K, Jin Z, Zheng W, Wang Q, Yang Y, Yu C, Xu K, Gao C. Effective treatment of traumatic brain injury by injection of a selenium-containing ointment. Acta Biomater 2024; 187:161-171. [PMID: 39236795 DOI: 10.1016/j.actbio.2024.08.051] [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: 03/29/2024] [Revised: 08/22/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
Traumatic brain injury (TBI) is an incurable and overwhelming disease accompanied with serve disability and huge financial burden, where the overproduced reactive oxygen species (ROS) can exacerbate the secondary injury, leading to massive apoptosis of neurons. In this study, β-cyclodextrin (CD)-capped hyperbranched polymers containing selenium element (HSE-CD) were crosslinked with CD-modified hyaluronic acid (HA-CD) and amantadine-modified hyaluronic acid (HA-AD) to obtain a ROS-responsive ointment (R-O). The structures of synthesized polymers were characterized with 1H nuclear magnetic resonance, and the properties of ointment were investigated with rheology and antioxidation. Compared to non-ROS-responsive ointment (N-O), the R-O ointment had stronger efficiency in decreasing the ROS level in BV2 cells in vitro. In a controlled rat cortical impact (CCI) model, the R-O ointment could relieve the DNA damage and decrease apoptosis in injured area via reducing the ROS level. Besides, after the R-O treatment, the rats showed significantly less activated astrocytes and microglia, a lower level of pro-inflammatory cytokines and a higher ratio of M2/M1 macrophage and microglia. Moreover, compared to the TBI group the R-O ointment promoted the doublecortin (DCX) expression and tissue structure integrity around the cavity, and promoted the recovery of nerve function post TBI. STATEMENT OF SIGNIFICANCE: Traumatic brain injury (TBI) is an incurable and overwhelming disease, leading to severe disability and huge social burden, where reactive oxygen species (ROS) are considered as one of the most significant factors in the secondary injury of TBI. A ROS responsive supramolecular ointment containing di-selenide bonds was injected in rats with controlled cortical impact. It relieved the DNA damage and decreased apoptosis in the injured area via reducing the ROS levels, downregulated neuroinflammation, and improved neurological recovery of TBI in vivo. This designed self-adaptive biomaterial effectively regulated the pathological microenvironment in injured tissue, and achieved better therapeutic effect.
Collapse
Affiliation(s)
- Haijun Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Huan Gao
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310027, China
| | - Kai Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Zeyuan Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Weiwei Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Qiaoxuan Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yufang Yang
- Key Laboratory of Biomedical Engineering of Education Ministry, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Chaonan Yu
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310027, China
| | - Kedi Xu
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310027, China; Key Laboratory of Biomedical Engineering of Education Ministry, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China.
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China; Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing 312099, China.
| |
Collapse
|
9
|
Zhang W, Uyemura R, Zhong K, Guo R, Zhong L. Current Advances and Future Perspectives on Mesenchymal Stem Cell-Derived Extracellular Vesicles in Alzheimer's Disease. Aging Dis 2024; 15:2015-2027. [PMID: 38270122 PMCID: PMC11346404 DOI: 10.14336/ad.2023.1206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/06/2023] [Indexed: 01/26/2024] Open
Abstract
The incidence of Alzheimer's disease (AD) has been increasing in recent years as the world's population ages, which poses a significant challenge to public health. Due to the complexity of pathogenesis of AD, currently there is no effective treatment for it. In recent years, cell and gene therapy has attracted widespread attention in the treatment of neurodegenerative diseases. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) represent a novel cell-free therapy with numerous advantages over cell-based therapies owing to their low immunogenicity and high safety profile. We summarize recent progress in the application of EVs for treating AD and the specific mechanisms and outline the underlying mechanisms. We also explore various methods for optimizing the function of MSC-EVs, including gene editing, modifying stem cell culture conditions and peptide modification. In addition, we discuss the therapeutic potentials of MSC-EVs, as well as the obstacles that currently impede their clinical utilization.
Collapse
Affiliation(s)
- Wenjing Zhang
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Russell Uyemura
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766, USA
| | - Kun Zhong
- American Center of Stem Cell Research and Regenerative Medicine, Farmington Hills, Michigan 48336, USA
| | - Rui Guo
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Li Zhong
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766, USA
| |
Collapse
|
10
|
Liu F, Chen Y, Huang K. Electro-acupuncture Suppresses Ferroptosis to Alleviate Cerebral Ischemia-Reperfusion Injury Through KAT3B-Mediated Succinylation of ACSL4. Appl Biochem Biotechnol 2024:10.1007/s12010-024-05063-6. [PMID: 39340629 DOI: 10.1007/s12010-024-05063-6] [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] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
Electro-acupuncture (EA) is identified as an effective therapeutic method for cerebral ischemia/reperfusion injury (CIRI), which is a combination of Chinese traditional acupuncture and modern electro-therapy. However, the downstream molecular mechanisms of EA in CIRI process remains largely unknown. The purpose of the present study is to unveil the therapeutic effect of EA on CIRI rat and its regulatory mechanisms. At first, we constructed middle cerebral artery occlusion (MCAO) rat models and then treated them with EA to observe the pathological changes. The results indicated that EA decreased the infarct volume (43.81 ± 3.34 vs 15.96 ± 2.22) and the neurological scores (3.33 ± 0.52 vs 1.67 ± 0.52) and suppressed the apoptosis in MCAO model rats. For ferroptosis analysis, EA decreased the Fe2 + (0.08 ± 0.01 vs 0.06 ± 0.01), MDA (36.61 ± 4.29 vs 21.72 ± 2.79), and LPS (5.25 ± 0.69 vs 2.89 ± 0.42) contents and increased the GSH (4.94 ± 1.04 vs 11.69 ± 1.88) content in MCAO model rats. We next detected whether succinylation mediated EA-treated I/R injury. According to immunoprecipitation and western blot analysis, EA treatment could lower both levels of succinylation and KAT3B in MCAO rats. Moreover, mechanism experiments unveiled that KAT3B promoted the succinylation of the ferroptosis-related protein ACSL4 at K661 site and thus stabilizing ACSL4. Finally, EA-treated MCAO rats were further injected with KAT3B expression vector. The results showed that KAT3B overexpression increased the infarct volume (31.44 ± 3.92 vs 7.94 ± 2.84) and the neurological scores (2.67 ± 0.51 vs 1.33 ± 0.51) and promoted the apoptosis in EA treated MCAO model rats. For ferroptosis analysis, KAT3B overexpression increased the Fe2 + (0.08 ± 0.01 vs 0.05 ± 0.01), MDA (29.24 ± 4.30 vs 22.06 ± 1.89), and LPO (5.07 ± 0.45 vs 2.88 ± 0.49) contents and decreased the GSH (7.86 ± 1.09 vs 11.06 ± 1.76) content in EA treated MCAO model rats. Collectively, our study demonstrates that EA plays a therapeutic role in CIRI through suppressing KAT3B-induced stabilization of ACSL4 to inhibit ferroptosis. These findings contribute to our understanding of the molecular mechanisms underlying the neuroprotective effects of EA and open new avenues for the development of innovative therapeutic strategies for CIRI.
Collapse
Affiliation(s)
- Fang Liu
- Department of Chinese Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Ying Chen
- Department of Chinese Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Kangbai Huang
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou, 510405, China.
| |
Collapse
|
11
|
Jiang X, He X, Wong J, Scheeff S, Hau SCK, Wong TH, Qin Y, Fan CH, Ma B, Chung NL, Huang J, Zhao J, Yan Y, Xiao M, Song X, Hui TKC, Zuo Z, Wu WKK, Ko H, Chow KHM, Ng BWL. Lactone-to-Lactam Editing Alters the Pharmacology of Bilobalide. JACS AU 2024; 4:3537-3546. [PMID: 39328759 PMCID: PMC11423332 DOI: 10.1021/jacsau.4c00416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 09/28/2024]
Abstract
Precise transformations of natural products (NPs) can fine-tune their physicochemical properties while preserving inherently complex and evolutionarily optimized parent scaffolds. Here, we report an unprecedented lactone-to-lactam transformation on bilobalide, thus improving its stability and paving the way for biological exploration of previously inaccessible chemical space that is highly representative of the parent structure. This late-stage molecular editing of bilobalide enables facile access to a unique library of lactam analogues with altered pharmacology. Through phenotypic screening, we identify BB10 as a hit compound with unexpected inhibition of ferroptotic cell death. We further reveal that BB10 suppresses ferroptosis by restoring the expression of glutathione peroxidase 4 (GPX4) in brain cells. This study highlights that even subtle changes on NP scaffolds can confer new pharmacological properties, inspiring the exploration of simple yet critical transformations on complex NPs.
Collapse
Affiliation(s)
- Xiaoding Jiang
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Xu He
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Jonathan Wong
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Stephan Scheeff
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Sam Chun-Kit Hau
- Department
of Chemistry, Faculty of Science, The Chinese
University of Hong Kong, Hong Kong SAR, China
| | - Tak Hin Wong
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Yao Qin
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Chi Hang Fan
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Bowen Ma
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Ngai Lam Chung
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department
of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Junzhe Huang
- Division
of Neurology, Department of Medicine and Therapeutics, Margaret K.L.
Cheung Research Centre for Management of Parkinsonism, Faculty of
Medicine, The Chinese University of Hong
Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Jiajia Zhao
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Yu Yan
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Min Xiao
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Xueqin Song
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Tony K. C. Hui
- Primemax
Biotech Ltd., Wayson
Commercial House, 68-70 Lockhard Road, Wan Chai, Hong Kong SAR, China
| | - Zhong Zuo
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - William Ka-Kei Wu
- Department
of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Li
Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Ho Ko
- Division
of Neurology, Department of Medicine and Therapeutics, Margaret K.L.
Cheung Research Centre for Management of Parkinsonism, Faculty of
Medicine, The Chinese University of Hong
Kong, Shatin, New Territories, Hong Kong SAR, China
- Li
Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Peter
Hung Pain Research Institute, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Gerald Choa
Neuroscience Institute, The Chinese University
of Hong Kong, Hong Kong SAR, China
| | - Kim Hei-Man Chow
- School
of Life Sciences, Faculty of Science, The
Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Gerald Choa
Neuroscience Institute, The Chinese University
of Hong Kong, Hong Kong SAR, China
| | - Billy Wai-Lung Ng
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Li
Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Peter
Hung Pain Research Institute, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| |
Collapse
|
12
|
Tao J, Li J, Fan X, Jiang C, Wang Y, Qin M, Nikfard Z, Nikfard F, Wang Y, Zhao T, Xing N, Zille M, Wang J, Zhang J, Chen X, Wang J. Unraveling the protein post-translational modification landscape: Neuroinflammation and neuronal death after stroke. Ageing Res Rev 2024; 101:102489. [PMID: 39277050 DOI: 10.1016/j.arr.2024.102489] [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: 08/07/2024] [Revised: 08/31/2024] [Accepted: 09/01/2024] [Indexed: 09/17/2024]
Abstract
The impact of stroke on global health is profound, with both high mortality and morbidity rates. This condition can result from cerebral ischemia, intracerebral hemorrhage (ICH), and subarachnoid hemorrhage (SAH). The pathophysiology of stroke involves secondary damage and irreversible loss of neuronal function. Post-translational modifications (PTMs) have been recognized as crucial regulatory mechanisms in ischemic and hemorrhagic stroke-induced brain injury. These PTMs include phosphorylation, glycosylation, ubiquitination, SUMOylation, acetylation, and succinylation. This comprehensive review delves into recent research on the PTMs landscape associated with neuroinflammation and neuronal death specific to cerebral ischemia, ICH, and SAH. This review aims to explain the role of PTMs in regulating pathologic mechanisms and present critical techniques and proteomic strategies for identifying PTMs. This knowledge helps us comprehend the underlying mechanisms of stroke injury and repair processes, leading to the development of innovative treatment strategies. Importantly, this review underscores the significance of exploring PTMs to understand the pathophysiology of stroke.
Collapse
Affiliation(s)
- Jin Tao
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Jiaxin Li
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China
| | - Chao Jiang
- Department of Neurology, People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P. R. China
| | - Yebin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Mengzhe Qin
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Zahra Nikfard
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Fatemeh Nikfard
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China
| | - Yunchao Wang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450000, P. R. China
| | - Ting Zhao
- Department of Neurology, People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P. R. China
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P. R. China
| | - Marietta Zille
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna 1090, Austria
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China.
| | - Jiewen Zhang
- Department of Neurology, People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P. R. China.
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China.
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P. R. China.
| |
Collapse
|
13
|
Wang Y, Song X, Wang R, Xu X, Du Y, Chen G, Mei J. Genome-Wide Mendelian Randomization Identifies Ferroptosis-Related Drug Targets for Alzheimer's Disease. J Alzheimers Dis Rep 2024; 8:1185-1197. [PMID: 39247875 PMCID: PMC11380310 DOI: 10.3233/adr-240062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/15/2024] [Indexed: 09/10/2024] Open
Abstract
Background Alzheimer's disease (AD) currently lacks effective disease-modifying treatments. Recent research suggests that ferroptosis could be a potential therapeutic target. Mendelian randomization (MR) is a widely used method for identifying novel therapeutic targets. Objective Employ genetic information to evaluate the causal impact of ferroptosis-related genes on the risk of AD. Methods 564 ferroptosis-related genes were obtained from FerrDb. We derived genetic instrumental variables for these genes using four brain quantitative trait loci (QTL) and two blood QTL datasets. Summary-data-based Mendelian randomization (SMR) and two-sample MR methods were applied to estimate the causal effects of ferroptosis-related genes on AD. Using extern transcriptomic datasets and triple-transgenic mouse model of AD (3xTg-AD) to further validate the gene targets identified by the MR analysis. Results We identified 17 potential AD risk gene targets from GTEx, 13 from PsychENCODE, and 22 from BrainMeta (SMR p < 0.05 and HEIDI test p > 0.05). Six overlapping ferroptosis-related genes associated with AD were identified, which could serve as potential therapeutic targets (PEX10, CDC25A, EGFR, DLD, LIG3, and TRIB3). Additionally, we further pinpointed risk genes or proteins at the blood tissue and pQTL levels. Notably, EGFR demonstrated significant dysregulation in the extern transcriptomic datasets and 3xTg-AD models. Conclusions This study provides genetic evidence supporting the potential therapeutic benefits of targeting the six druggable genes for AD treatment, especially for EGFR (validated by transcriptome and 3xTg-AD), which could be useful for prioritizing AD drug development in the field of ferroptosis.
Collapse
Affiliation(s)
- Ying Wang
- Department of Neurology, Traditional Chinese and Western Medicine Hospital of Wuhan/Wuhan First Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinhua Song
- Department of Neurology, Wuhan First Hospital, Hubei University of Chinese Medicine, Wuhan, China
| | - Rui Wang
- Department of Neurology, Wuhan First Hospital, Hubei University of Chinese Medicine, Wuhan, China
| | - Xinzi Xu
- Department of Neurology, Wuhan First Hospital, Hubei University of Chinese Medicine, Wuhan, China
| | - Yaming Du
- Department of Neurology, Wuhan First Hospital, Hubei University of Chinese Medicine, Wuhan, China
| | - Guohua Chen
- Department of Neurology, Traditional Chinese and Western Medicine Hospital of Wuhan, Hubei University of Chinese Medicine, Wuhan, China
| | - Junhua Mei
- Department of Neurology, Traditional Chinese and Western Medicine Hospital of Wuhan, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
14
|
Wang S, Qin M, Fan X, Jiang C, Hou Q, Ye Z, Zhang X, Yang Y, Xiao J, Wallace K, Rastegar-Kashkooli Y, Peng Q, Jin D, Wang J, Wang M, Ding R, Tao J, Kim YT, Bhawal UK, Wang J, Chen X, Wang J. The role of metal ions in stroke: Current evidence and future perspectives. Ageing Res Rev 2024; 101:102498. [PMID: 39243890 DOI: 10.1016/j.arr.2024.102498] [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: 03/18/2024] [Revised: 07/24/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Metal ions play a pivotal role in maintaining optimal brain function within the human body. Nevertheless, the accumulation of these ions can result in irregularities that lead to brain damage and dysfunction. Disruptions of metal ion homeostasis can result in various pathologies, including inflammation, redox dysregulation, and blood-brain barrier disruption. While research on metal ions has chiefly focused on neurodegenerative diseases, little attention has been given to their involvement in the onset and progression of stroke. Recent studies have identified cuproptosis and confirmed ferroptosis as significant factors in stroke pathology, underscoring the importance of metal ions in stroke pathology, including abnormal ion transport, neurotoxicity, blood-brain barrier damage, and cell death. Additionally, it provides an overview of contemporary metal ion chelators and detection techniques, which may offer novel approaches to stroke treatment.
Collapse
Affiliation(s)
- Shaoshuai Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Non-commissioned Officer School of Army Medical University, Shijiazhuang, Hebei 050000, China
| | - Mengzhe Qin
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Chao Jiang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Qingchuan Hou
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ziyi Ye
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xinru Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yunfan Yang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jingyu Xiao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Kevin Wallace
- College of Mathematical and Natural Sciences, University of Maryland, College Park, MD 20742, USA
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Qinfeng Peng
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Dongqi Jin
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Junyang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Menglu Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ruoqi Ding
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jin Tao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yun Tai Kim
- Division of Functional Food Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea; Department of Food Biotechnology, Korea University of Science & Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Ujjal K Bhawal
- Center for Global Health Research, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India; Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba 271-8587, Japan
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
15
|
Yu X, Wang S, Wang X, Li Y, Dai Z. Melatonin improves stroke by inhibiting autophagy-dependent ferroptosis mediated by NCOA4 binding to FTH1. Exp Neurol 2024; 379:114868. [PMID: 38901754 DOI: 10.1016/j.expneurol.2024.114868] [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: 10/16/2023] [Revised: 05/30/2024] [Accepted: 06/16/2024] [Indexed: 06/22/2024]
Abstract
Ischemic stroke is a disease associated with high morbidity and disability rates; however, its pathogenesis remains elusive, and treatment options are limited. Ferroptosis, an iron-dependent form of cell death, represents a novel avenue for investigation. The objective of this study was to explore the role of melatonin in MCAO-induced ferroptosis and elucidate its underlying molecular mechanism. To simulate brain damage and neuronal injury caused by ischemic stroke, we established a mouse model of MCAO and an HT-22 cell model of OGD/R. The therapeutic efficacy of melatonin was assessed through measurements of infarct size, brain edema, and neurological scores. Additionally, qRT-PCR, WB analysis, and Co-IP assays were employed to investigate the impact of melatonin on ferroptosis markers such as NCOA4 and FTH1 expression levels. Confocal microscopy was utilized to confirm the colocalization between ferritin and lysosomes. Furthermore, we constructed a SIRT6 siRNA model to validate the regulatory effect exerted by SIRT6 on NCOA4 as well as their binding interaction. The present study provides initial evidence that melatonin possesses the ability to mitigate neuronal damage induced by MCAO and OGD/R. Assessment of markers for oxidative damage and ferroptosis revealed that melatonin effectively inhibits intracellular Fe2+ levels, thereby suppressing ferroptosis. Additionally, our findings demonstrate that melatonin modulates the interaction between FTH1 and NCOA4 via SIRT6, influencing ferritin autophagy without affecting cellular macroautophagy. These findings provide reliable data support for the promotion and application of melatonin in clinical practice.
Collapse
Affiliation(s)
- Xiang Yu
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China
| | - Shan Wang
- Department of Echocardiography & Noninvasive Cardiology Laboratory, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China
| | - Xu Wang
- Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 2 Ward of Hepatobiliary Surgery, Chengdu 610072, Sichuan, China
| | - Yi Li
- Emergency Department, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China.
| | - Zheng Dai
- Emergency Department, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China.
| |
Collapse
|
16
|
Ma Y, Liu C, Ren L, Li J, Xu Y, Liang J, Wang P. β-1,4-Galactosyltransferase 1 protects against cerebral ischemia injury in mice by suppressing ferroptosis via the TAZ/Nrf2/HO-1 signaling pathway. CNS Neurosci Ther 2024; 30:e70030. [PMID: 39233353 PMCID: PMC11374693 DOI: 10.1111/cns.70030] [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: 07/16/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Ischemic stroke leads a primary cause of mortality in human diseases, with a high disability rate worldwide. This study aims to investigate the function of β-1,4-galactosyltransferase 1 (B4galt1) in mouse brain ischemia/reperfusion (I/R) injury. METHODS Recombinant human B4galt1 (rh-B4galt1) was intranasally administered to the mice model of middle cerebral artery occlusion (MCAO)/reperfusion. In this study, the impact of rh-B4galt1 on cerebral injury assessed using multiple methods, including the neurological disability status scale, 2,3,5-triphenyltetrazolium chloride (TTC), Nissl and TUNEL staining. This study utilized laser speckle Doppler flowmeter to monitor the cerebral blood flow. Western blotting was performed to assess the protein expression levels, and fluorescence-labeled dihydroethidium method was performed to determine the superoxide anion generation. Assay kits were used for the measurement of iron, malondialdehyde (MDA) and glutathione (GSH) levels. RESULTS We demonstrated that rh-B4galt1 markedly improved neurological function, reduced cerebral infarct volume and preserved the completeness of blood-brain barrier (BBB) for preventing damage. These findings further illustrated that rh-B4galt1 alleviated oxidative stress, lipid peroxidation, as well as iron deposition induced by I/R. The vital role of ferroptosis was proved in brain injury. Furthermore, the rh-B4galt1 could increase the levels of TAZ, Nrf2 and HO-1 after I/R. And TAZ-siRNA and ML385 reversed the neuroprotective effects of rh-B4galt1. CONCLUSIONS The results indicated that rh-B4galt1 implements neuroprotective effects by modulating ferroptosis, primarily via upregulating TAZ/Nrf2/HO-1 pathway. Thus, B4galt1 could be seen as a promising novel objective for ischemic stroke therapy.
Collapse
Affiliation(s)
- Yao Ma
- Department of Neurobiology, School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Chang Liu
- Department of Neurobiology, School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Lili Ren
- Department of Neurobiology, School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Jiachen Li
- Department of Neurobiology, School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yunhao Xu
- Department of Neurobiology, School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Jia Liang
- Institute of Life Science, Jinzhou Medical University, Jinzhou, Liaoning, China
- Liaoning Provincial Key Laboratory of Neurodegenerative Diseases, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Peng Wang
- Department of Neurobiology, School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
- Liaoning Provincial Key Laboratory of Neurodegenerative Diseases, Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
17
|
Maheshwari S, Singh A, Verma A. Ferroptosis: A Frontier in Osteoporosis. Horm Metab Res 2024; 56:625-632. [PMID: 38307092 DOI: 10.1055/a-2230-2664] [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: 02/04/2024]
Abstract
Reduced bone mass and degeneration of the microarchitecture of bone tissue are the hallmarks of osteoporosis, a bone metabolic disease that increases skeletal fragility and fracture susceptibility. Osteoporosis is primarily caused by unbalanced bone remodeling, in which bone synthesis is outpaced by bone resorption caused by osteoclasts. Along with the bone-building vitamins calcium and vitamin D, typical medications for treating osteoporosis include bisphosphonates and calcitonin. The present therapies effectively stop osteoclast activation that is too high, however they come with varying degrees of negative effects. Numerous factors can contribute to osteoporosis, which is characterized by a loss of bone mass and density due to the deterioration of the bone's microstructure, which makes the bone more fragile. As a result, it is a systemic bone condition that makes patients more likely to fracture. Interest in the function of ferroptosis in the pathophysiology of osteoporosis is developing. In this review, we go through the shape of the cell, the fundamental mechanisms of ferroptosis, the relationship between osteoclasts and osteoblasts, the association between ferroptosis and diabetic osteoporosis, steroid-induced osteoporosis, and the relationship between ferroptosis and postmenopausal osteoporosis. The functions of ferroptosis and osteoporosis in cellular function, signaling cascades, pharmacological inhibition, and gene silencing have been better understood thanks to recent advances in biomedical research.
Collapse
Affiliation(s)
- Shubhrat Maheshwari
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, India
- Faculty of Pharmaceutical Sciences, Rama University, Kanpur, India
| | - Aditya Singh
- Faculty of Pharmacy, Integral University, Lucknow, India
| | - Amita Verma
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, India
| |
Collapse
|
18
|
Joshi DC, Joshi N, Kumar A, Maheshwari S. Recent Advances in Molecular Pathways and Therapeutic Implications for Peptic Ulcer Management: A Comprehensive Review. Horm Metab Res 2024; 56:615-624. [PMID: 38467155 DOI: 10.1055/a-2256-6592] [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: 03/13/2024]
Abstract
Peptic ulcers, recognized for their erosive impact on the gastrointestinal mucosa, present a considerable challenge in gastroenterology. Epidemiological insights underscore the global prevalence of peptic ulcers, affecting 5-10+% of individuals, with a yearly incidence of 0.3 to 1.9 cases per thousand. Recent decades have witnessed a decline in complications, attributed to improved diagnostics and therapeutic advancements. The review deepens into H. pylori-associated and NSAID-induced ulcers, emphasizing their distinct prevalence in developing and industrialized nations, respectively. Despite advancements, managing peptic ulcers remains challenging, notably in H. pylori-infected individuals facing recurrence and the rise of antibiotic resistance. The pathophysiology unravels the delicate balance between protective and destructive factors, including the intricate molecular mechanisms involving inflammatory mediators such as TNF-α, ILs, and prostaglandins. Genetic and ethnic factors, rare contributors, and recent molecular insights further enhance our understanding of peptic ulcer development. Diagnostic approaches are pivotal, with upper gastrointestinal endoscopy standing as the gold standard. Current treatment strategies focus on H. pylori eradication, NSAID discontinuation, and proton pump inhibitors. Surgical options become imperative for refractory cases, emphasizing a comprehensive approach. Advances include tailored H. pylori regimens, the emergence of vonoprazan, and ongoing vaccine development. Challenges persist, primarily in antibiotic resistance, side effects of acid suppressants, and translating natural compounds into standardized therapies. Promising avenues include the potential H. pylori vaccine and the exploration of natural compounds, with monoterpenes showing therapeutic promise. This review serves as a compass, guiding healthcare professionals, researchers, and policymakers through the intricate landscape of peptic ulcer management.
Collapse
Affiliation(s)
- Deepak Chandra Joshi
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Ajmer, India
| | - Nirmal Joshi
- Department of Pharmacology, Amrapali Institute of Pharmacy and Sciences, Haldwani, India
| | - Ajeet Kumar
- Faculty of Pharmaceutical Sciences, Rama University, Kanpur, India
| | | |
Collapse
|
19
|
Zhu YW, Liu ZT, Tang AQ, Liang XY, Wang Y, Liu YF, Jin YQ, Gao W, Yuan H, Wang DY, Ji XY, Wu DD. The Emerging Roles of Hydrogen Sulfide in Ferroptosis. Antioxid Redox Signal 2024. [PMID: 39041626 DOI: 10.1089/ars.2023.0535] [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] [Indexed: 07/24/2024]
Abstract
Significance: Ferroptosis, a form of regulated cell death characterized by a large amount of lipid peroxidation-mediated membrane damage, joins the evolution of multisystem diseases, for instance, neurodegenerative diseases, chronic obstructive pulmonary disease, acute respiratory distress syndrome, osteoporosis, osteoarthritis, and so forth. Since being identified as the third gasotransmitter in living organisms, the intricate role of hydrogen sulfide (H2S) in ferroptosis has emerged at the forefront of research. Recent Advances: Novel targets in the relevant metabolic pathways have been found, including transferrin receptor 1, cystine/glutamate antiporter, and others, coupled with the exploration of new signaling pathways, particularly the p53 signaling pathway, the nitric oxide/nuclear factor erythroid 2-related factor 2 signaling pathway, and so on. Many diseases such as emphysema and airway inflammation, myocardial diseases, endothelial dysfunction in aging arteries, and traumatic brain injury have recently been found to be alleviated directly by H2S inhibition of ferroptosis. Safe, effective, and tolerable novel H2S donors have been developed and have shown promising results in phase I clinical trials. Critical Issues: Complicated cross talk between the ferroptosis signaling pathway and oncogenic factors results in the risk of cancer when inhibiting ferroptosis. Notably, targeted delivery of H2S is still a challenging task. Future Directions: Discovering more reliable and stable novel H2S donors and achieving their targeted delivery will enable further clinical trials for diseases associated with ferroptosis inhibition by H2S, determining their safety, efficacy, and tolerance.
Collapse
Affiliation(s)
- Yi-Wen Zhu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
| | - Zi-Tao Liu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
| | - Ao-Qi Tang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
| | - Xiao-Yi Liang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
| | - Yan Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
| | - Ya-Fang Liu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
| | - Yu-Qing Jin
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
| | - Wei Gao
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
| | - Hang Yuan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
| | - Da-Yong Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
- The First Affiliated Hospital of Henan University, Kaifeng, China
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, China
- Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, China
| |
Collapse
|
20
|
Kim JW, Nam SA, Koh ES, Kim HW, Kim S, Woo JJ, Kim YK. The Impairment of Endothelial Autophagy Accelerates Renal Senescence by Ferroptosis and NLRP3 Inflammasome Signaling Pathways with the Disruption of Endothelial Barrier. Antioxidants (Basel) 2024; 13:886. [PMID: 39199133 PMCID: PMC11351978 DOI: 10.3390/antiox13080886] [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: 06/18/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 09/01/2024] Open
Abstract
Autophagy is a cellular process that degrades damaged cytoplasmic components and regulates cell death. The homeostasis of endothelial cells (ECs) is crucial for the preservation of glomerular structure and function in aging. Here, we investigated the precise mechanisms of endothelial autophagy in renal aging. The genetic deletion of Atg7 in the ECs of Atg7flox/flox;Tie2-Cre mice accelerated aging-related glomerulopathy and tubulointerstitial fibrosis. The EC-specific Atg7 deletion in aging mice induced the detachment of EC with the disruption of glomerular basement membrane (GBM) assembly and increased podocyte loss resulting in microalbuminuria. A Transwell co-culture system of ECs and kidney organoids showed that the iron and oxidative stress induce the disruption of the endothelial barrier and increase vascular permeability, which was accelerated by the inhibition of autophagy. This resulted in the leakage of iron through the endothelial barrier into kidney organoids and increased oxidative stress, which led to ferroptotic cell death. The ferritin accumulation was increased in the kidneys of the EC-specific Atg7-deficient aging mice and upregulated the NLRP3 inflammasome signaling pathway. The pharmacologic inhibition of ferroptosis with liproxstatin-1 recovered the disrupted endothelial barrier and reversed the decreased expression of GPX4, as well as NLRP3 and IL-1β, in endothelial autophagy-deficient aged mice, which attenuated aging-related renal injury including the apoptosis of renal cells, abnormal structures of GBM, and tubulointerstitial fibrosis. Our data showed that endothelial autophagy is essential for the maintenance of the endothelial barrier during renal aging and the impairment of endothelial autophagy accelerates renal senescence by ferroptosis and NLRP3 inflammasome signaling pathways. These processes may be attractive therapeutic targets to reduce cellular injury from renal aging.
Collapse
Affiliation(s)
- Jin Won Kim
- Department of Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sun Ah Nam
- Department of Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Eun-Sil Koh
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hyung Wook Kim
- Department of Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, St. Vincent’s Hospital, Suwon 16247, Republic of Korea
| | - Sua Kim
- Department of Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jin Ju Woo
- Department of Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Yong Kyun Kim
- Department of Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, St. Vincent’s Hospital, Suwon 16247, Republic of Korea
| |
Collapse
|
21
|
Hagar FF, Abbas SH, Atef E, Abdelhamid D, Abdel-Aziz M. Benzimidazole scaffold as a potent anticancer agent with different mechanisms of action (2016-2023). Mol Divers 2024:10.1007/s11030-024-10907-8. [PMID: 39031290 DOI: 10.1007/s11030-024-10907-8] [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: 03/24/2024] [Accepted: 05/30/2024] [Indexed: 07/22/2024]
Abstract
Benzimidazole scaffolds have potent anticancer activity due to their structure similarity to nucleoside. In addition, benzimidazoles could function as hydrogen donors or acceptors and bind to different drug targets that participate in cancer progression. The literature had many anticancer agents containing benzimidazole cores that gained much interest. Provoked by our endless interest in benzimidazoles as anticancer agents, we summarized the successful trials of the benzimidazole scaffolds in this concern. Moreover, we discuss the substantial opportunities in cancer treatment using benzimidazole-based drugs that may direct medicinal chemists for a compelling future design of more active chemotherapeutic agents with potential clinical applications. The uniqueness of this work lies in the highlighted benzimidazole scaffold hybridization with different molecules and benzimidazole-metal complexes, detailed mechanisms of action, and the IC50 of the developed compounds determined by different laboratories after 2015.
Collapse
Affiliation(s)
- Fatma Fouad Hagar
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Samar H Abbas
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt.
| | - Eman Atef
- College of Pharmacy, West Coast University, Los Angeles, CA, USA
| | - Dalia Abdelhamid
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt.
- Raabe College of Pharmacy, Ohio Northern University, Ohio, USA.
| | - Mohamed Abdel-Aziz
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| |
Collapse
|
22
|
Yang Y, Li Z, Fan X, Jiang C, Wang J, Rastegar-Kashkooli Y, Wang TJ, Wang J, Wang M, Cheng N, Yuan X, Chen X, Jiang B, Wang J. Nanozymes: Potential Therapies for Reactive Oxygen Species Overproduction and Inflammation in Ischemic Stroke and Traumatic Brain Injury. ACS NANO 2024; 18:16450-16467. [PMID: 38897929 DOI: 10.1021/acsnano.4c03425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Nanozymes, which can selectively scavenge reactive oxygen species (ROS), have recently emerged as promising candidates for treating ischemic stroke and traumatic brain injury (TBI) in preclinical models. ROS overproduction during the early phase of these diseases leads to oxidative brain damage, which has been a major cause of mortality worldwide. However, the clinical application of ROS-scavenging enzymes is limited by their short in vivo half-life and inability to cross the blood-brain barrier. Nanozymes, which mimic the catalytic function of natural enzymes, have several advantages, including cost-effectiveness, high stability, and easy storage. These advantages render them superior to natural enzymes for disease diagnosis and therapeutic interventions. This review highlights recent advancements in nanozyme applications for ischemic stroke and TBI, emphasizing their potential to mitigate the detrimental effect of ROS overproduction, oxidative brain damage, inflammation, and blood-brain barrier compromise. Therefore, nanozymes represent a promising treatment modality for ROS overproduction conditions in future medical practices.
Collapse
Affiliation(s)
- Yunfan Yang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan, P. R. China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Zixiang Li
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan, P. R. China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan, P. R. China
| | - Chao Jiang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou 450000, Henan, P. R. China
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
- School of International Education, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Tom J Wang
- Program in Behavioral Biology, The Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Junyang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Menglu Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Nannan Cheng
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Xiqian Yuan
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Bing Jiang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan, P. R. China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, P. R. China
| |
Collapse
|
23
|
Chai Z, Zheng J, Shen J. Mechanism of ferroptosis regulating ischemic stroke and pharmacologically inhibiting ferroptosis in treatment of ischemic stroke. CNS Neurosci Ther 2024; 30:e14865. [PMID: 39042604 PMCID: PMC11265528 DOI: 10.1111/cns.14865] [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: 04/11/2024] [Revised: 06/27/2024] [Accepted: 07/05/2024] [Indexed: 07/25/2024] Open
Abstract
Ferroptosis is a newly discovered form of programmed cell death that is non-caspase-dependent and is characterized by the production of lethal levels of iron-dependent lipid reactive oxygen species (ROS). In recent years, ferroptosis has attracted great interest in the field of cerebral infarction because it differs morphologically, physiologically, and genetically from other forms of cell death such as necrosis, apoptosis, autophagy, and pyroptosis. In addition, ROS is considered to be an important prognostic factor for ischemic stroke, making it a promising target for stroke treatment. This paper summarizes the induction and defense mechanisms associated with ferroptosis, and explores potential treatment strategies for ischemic stroke in order to lay the groundwork for the development of new neuroprotective drugs.
Collapse
Affiliation(s)
- Zhaohui Chai
- Department of NeurosurgeryFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou CityChina
| | - Jiesheng Zheng
- Department of NeurosurgeryFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou CityChina
| | - Jian Shen
- Department of NeurosurgeryFirst Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou CityChina
| |
Collapse
|
24
|
Li C, Jiang M, Chen Z, Hu Q, Liu Z, Wang J, Yin X, Wang J, Wu M. The neuroprotective effects of normobaric oxygen therapy after stroke. CNS Neurosci Ther 2024; 30:e14858. [PMID: 39009510 PMCID: PMC11250159 DOI: 10.1111/cns.14858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/29/2024] [Accepted: 07/03/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Stroke, including ischemic and hemorrhagic stroke, is a severe and prevalent acute cerebrovascular disease. The development of hypoxia following stroke can trigger a cascade of pathological events, including mitochondrial dysfunction, energy deficiency, oxidative stress, neuroinflammation, and excitotoxicity, all of which are often associated with unfavorable prognosis. Nonetheless, a noninvasive intervention, referred to as normobaric hyperoxia (NBO), is known to have neuroprotective effects against stroke. RESULTS NBO can exert neuroprotective effects through various mechanisms, such as the rescue of hypoxic tissues, preservation of the blood-brain barrier, reduction of brain edema, alleviation of neuroinflammation, improvement of mitochondrial function, mitigation of oxidative stress, reduction of excitotoxicity, and inhibition of apoptosis. These mechanisms may help improve the prognosis of stroke patients. CONCLUSIONS This review summarizes the mechanism by which hypoxia causes brain injury and how NBO can act as a neuroprotective therapy to treat stroke. We conclude that NBO has significant potential for treating stroke and may represent a novel therapeutic strategy.
Collapse
Affiliation(s)
- Chuan Li
- Department of Medical LaboratoryAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Min Jiang
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Zhiying Chen
- Department of NeurologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Qiongqiong Hu
- Department of NeurologyZhengzhou Central Hospital, Zhengzhou UniversityZhengzhouHenanChina
| | - Ziying Liu
- Department of Medical LaboratoryAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Junmin Wang
- Department of Human AnatomySchool of Basic Medical Sciences, Zhengzhou UniversityZhengzhouHenanChina
| | - Xiaoping Yin
- Department of NeurologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Jian Wang
- Department of Human AnatomySchool of Basic Medical Sciences, Zhengzhou UniversityZhengzhouHenanChina
| | - Moxin Wu
- Department of Medical LaboratoryAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| |
Collapse
|
25
|
Dong H, Ma YP, Cui MM, Qiu ZH, He MT, Zhang BG. Recent advances in potential therapeutic targets of ferroptosis‑associated pathways for the treatment of stroke (Review). Mol Med Rep 2024; 30:128. [PMID: 38785160 PMCID: PMC11134507 DOI: 10.3892/mmr.2024.13252] [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: 02/02/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
Stroke is a severe neurological disease that is associated with high rates of morbidity and mortality, and the underlying pathological processes are complex. Ferroptosis fulfills a significant role in the progression and treatment of stroke. It is well established that ferroptosis is a type of programmed cell death that is distinct from other forms or types of cell death. The process of ferroptosis involves multiple signaling pathways and regulatory mechanisms that interact with mechanisms inherent to stroke development. Inducers and inhibitors of ferroptosis have been shown to exert a role in the onset of this cell death process. Furthermore, it has been shown that interfering with ferroptosis affects the occurrence of stroke, indicating that targeting ferroptosis may offer a promising therapeutic approach for treating patients of stroke. Hence, the present review aimed to summarize the latest progress that has been made in terms of using therapeutic interventions for ferroptosis as treatment targets in cases of stroke. It provides an overview of the relevant pathways and molecular mechanisms that have been investigated in recent years, highlighting the roles of inducers and inhibitors of ferroptosis in stroke. Additionally, the intervention potential of various types of Traditional Chinese Medicine is also summarized. In conclusion, the present review provides a comprehensive overview of the potential therapeutic targets afforded by ferroptosis‑associated pathways in stroke, offering new insights into how ferroptosis may be exploited in the treatment of stroke.
Collapse
Affiliation(s)
- Hao Dong
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261042, P.R. China
| | - Ya-Ping Ma
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261042, P.R. China
| | - Mei-Mei Cui
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261042, P.R. China
| | - Zheng-Hao Qiu
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261042, P.R. China
| | - Mao-Tao He
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261042, P.R. China
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
| | - Bao-Gang Zhang
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261042, P.R. China
| |
Collapse
|
26
|
Wu M, Chen L, Wang Y, Li Y, An Y, Wu R, Zhang Y, Gao J, Su K, Feng X. The Effect of Acupuncture on Brain Iron Deposition and Body Iron Metabolism in Vascular Cognitive Impairment: Protocol for a Randomized Controlled Trial. JMIR Res Protoc 2024; 13:e56484. [PMID: 38885500 PMCID: PMC11217710 DOI: 10.2196/56484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/12/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Vascular cognitive impairment (VCI) persistently impairs cognition and the ability to perform activities of daily living, seriously compromising patients' quality of life. Previous studies have reported that disorders of serum iron metabolism and iron deposition in the brain can lead to inflammation, abnormal protein aggregation and degeneration, and massive neuronal apoptosis in the central nervous system, which in turn leads to a progressive decline in cognitive processes. Our previous clinical studies have found acupuncture to be a safe and effective intervention for treating VCI, but the specific mechanisms require further exploration. OBJECTIVE The objective of the trial is to evaluate the clinical efficacy of Tongdu Xingshen acupuncture and to investigate whether it can improve VCI by regulating brain iron deposition and body iron metabolism. METHODS In total, 42 patients with VCI and 21 healthy individuals will participate in this clinical trial. The 42 patients with VCI will be randomized into acupuncture and control groups, while the 21 healthy individuals will be in the healthy control group. Both the control and acupuncture groups will receive conventional medical treatment and cognitive rehabilitation training. In addition, the acupuncture group will receive electroacupuncture treatment with Tongdu Xingshen for 30 minutes each time, 6 times a week for 4 weeks. Meanwhile, the healthy control group will not receive any intervention. All 3 groups will undergo baseline assessments of brain iron deposition, serum iron metabolism, and neuropsychological tests after enrollment. The acupuncture and control groups will be evaluated again at the end of 4 weeks of treatment, as described earlier. By comparing neuropsychological test scores between groups, we will examine the efficacy of Tongdu Xingshen acupuncture in treating VCI. Additionally, we will test the correlations between neuropsychological test scores, brain iron deposition, and body iron metabolism indexes to explore the possible mechanisms of Tongdu Xingshen acupuncture in treating VCI. RESULTS Participants are currently being recruited. The first participant was enrolled in June 2023, which marked the official start of the experiment. As of the submission of the paper, there were 23 participants. The recruitment process is expected to continue until June 2025, at which point the processing and analysis of data will begin. As of May 15, 2024, up to 30 people have been enrolled in this clinical trial. CONCLUSIONS This study will provide data on the effects of Tongdu Xingshen acupuncture on cerebral iron deposition as well as somatic iron metabolism in patients with VCI. These results will help to prove whether Tongdu Xingshen acupuncture can improve VCI by regulating brain iron deposition and body iron metabolism, which will provide the clinical and theoretical basis for the wide application of acupuncture therapy in VCI rehabilitation. TRIAL REGISTRATION China Clinical Registration Agency ChiCTR2300072188; https://tinyurl.com/5fcydtkv. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) PRR1-10.2196/56484.
Collapse
Affiliation(s)
- Mingli Wu
- Rehabilitation Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, China
- Medical College of Rehabilitation, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lulu Chen
- Medical College of Rehabilitation, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yamin Wang
- Medical College of Rehabilitation, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yunpeng Li
- Medical College of Rehabilitation, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yuqi An
- Medical College of Rehabilitation, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ruonan Wu
- Medical College of Rehabilitation, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yuhan Zhang
- Medical College of Rehabilitation, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jing Gao
- Rehabilitation Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Kaiqi Su
- Rehabilitation Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaodong Feng
- Rehabilitation Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, China
- Medical College of Rehabilitation, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
27
|
Balusu S, De Strooper B. The necroptosis cell death pathway drives neurodegeneration in Alzheimer's disease. Acta Neuropathol 2024; 147:96. [PMID: 38852117 PMCID: PMC11162975 DOI: 10.1007/s00401-024-02747-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
Although apoptosis, pyroptosis, and ferroptosis have been implicated in AD, none fully explains the extensive neuronal loss observed in AD brains. Recent evidence shows that necroptosis is abundant in AD, that necroptosis is closely linked to the appearance of Tau pathology, and that necroptosis markers accumulate in granulovacuolar neurodegeneration vesicles (GVD). We review here the neuron-specific activation of the granulovacuolar mediated neuronal-necroptosis pathway, the potential AD-relevant triggers upstream of this pathway, and the interaction of the necrosome with the endo-lysosomal pathway, possibly providing links to Tau pathology. In addition, we underscore the therapeutic potential of inhibiting necroptosis in neurodegenerative diseases such as AD, as this presents a novel avenue for drug development targeting neuronal loss to preserve cognitive abilities. Such an approach seems particularly relevant when combined with amyloid-lowering drugs.
Collapse
Affiliation(s)
- Sriram Balusu
- Laboratory for the Research of Neurodegenerative Diseases, VIB Center for Brain and Disease Research, 3000, Leuven, Belgium.
- Leuven Brain Institute, KU Leuven, 3000, Leuven, Belgium.
| | - Bart De Strooper
- Laboratory for the Research of Neurodegenerative Diseases, VIB Center for Brain and Disease Research, 3000, Leuven, Belgium.
- Leuven Brain Institute, KU Leuven, 3000, Leuven, Belgium.
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK.
| |
Collapse
|
28
|
Lu L, Jifu C, Xia J, Wang J. E3 ligases and DUBs target ferroptosis: A potential therapeutic strategy for neurodegenerative diseases. Biomed Pharmacother 2024; 175:116753. [PMID: 38761423 DOI: 10.1016/j.biopha.2024.116753] [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: 03/14/2024] [Revised: 04/30/2024] [Accepted: 05/10/2024] [Indexed: 05/20/2024] Open
Abstract
Ferroptosis is a form of cell death mediated by iron and lipid peroxidation (LPO). Recent studies have provided compelling evidence to support the involvement of ferroptosis in the pathogenesis of various neurodegenerative diseases (NDDs), such as Alzheimer's disease (AD), Parkinson's disease (PD). Therefore, understanding the mechanisms that regulate ferroptosis in NDDs may improve disease management. Ferroptosis is regulated by multiple mechanisms, and different degradation pathways, including autophagy and the ubiquitinproteasome system (UPS), orchestrate the complex ferroptosis response by directly or indirectly regulating iron accumulation or lipid peroxidation. Ubiquitination plays a crucial role as a protein posttranslational modification in driving ferroptosis. Notably, E3 ubiquitin ligases (E3s) and deubiquitinating enzymes (DUBs) are key enzymes in the ubiquitin system, and their dysregulation is closely linked to the progression of NDDs. A growing body of evidence highlights the role of ubiquitin system enzymes in regulating ferroptosis sensitivity. However, reports on the interaction between ferroptosis and ubiquitin signaling in NDDs are scarce. In this review, we first provide a brief overview of the biological processes and roles of the UPS, summarize the core molecular mechanisms and potential biological functions of ferroptosis, and explore the pathophysiological relevance and therapeutic implications of ferroptosis in NDDs. In addition, reviewing the roles of E3s and DUBs in regulating ferroptosis in NDDs aims to provide new insights and strategies for the treatment of NDDs. These include E3- and DUB-targeted drugs and ferroptosis inhibitors, which can be used to prevent and ameliorate the progression of NDDs.
Collapse
Affiliation(s)
- Linxia Lu
- College of Basic Medicine, Jiamusi University, Jiamusi 154007, People's Republic of China
| | - Cili Jifu
- College of Basic Medicine, Jiamusi University, Jiamusi 154007, People's Republic of China
| | - Jun Xia
- College of Basic Medicine, Jiamusi University, Jiamusi 154007, People's Republic of China
| | - Jingtao Wang
- College of Basic Medicine, Jiamusi University, Jiamusi 154007, People's Republic of China.
| |
Collapse
|
29
|
Yang Q, Ye W, Luo D, Xing J, Xiao Q, Wu H, Yao Y, Wang G, Yang L, Guo D, Wang K, He Y, Ye X, Zhang J, Jin Z, Fan Z, Wen X, Mao J, Chen X, Zhao Q. Neuroprotective effects of anti-TRAIL-ICG nanoagent and its multimodal imaging evaluation in cerebral ischemia-reperfusion injury. Mater Today Bio 2024; 26:101094. [PMID: 38854952 PMCID: PMC11157279 DOI: 10.1016/j.mtbio.2024.101094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 05/08/2024] [Accepted: 05/19/2024] [Indexed: 06/11/2024] Open
Abstract
Cerebral ischemia-reperfusion injury (CIRI) is a major challenge to neuronal survival in acute ischemic stroke (AIS). However, effective neuroprotective agents remain to be developed for the treatment of CIRI. In this work, we have developed an Anti-TRAIL protein-modified and indocyanine green (ICG)-responsive nanoagent (Anti-TRAIL-ICG) to target ischemic areas and then reduce CIRI and rescue the ischemic penumbra. In vitro and in vivo experiments have demonstrated that the carrier-free nanoagent can enhance drug transport across the blood-brain barrier (BBB) in stroke mice, exhibiting high targeting ability and good biocompatibility. Anti-TRAIL-ICG nanoagent played a better neuroprotective role by reducing apoptosis and ferroptosis, and significantly improved ischemia-reperfusion injury. Moreover, the multimodal imaging platform enables the dynamic in vivo examination of multiple morphofunctional information, so that the dynamic molecular events of nanoagent can be detected continuously and in real time for early treatment in transient middle cerebral artery occlusion (tMCAO) models. Furthermore, it has been found that Anti-TRAIL-ICG has great potential in the functional reconstruction of neurovascular networks through optical coherence tomography angiography (OCTA). Taken together, our work effectively alleviates CIRI after stoke by blocking multiple cell death pathways, which offers an innovative strategy for harnessing the apoptosis and ferroptosis against CIRI.
Collapse
Affiliation(s)
- Qiong Yang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Center for Molecular Imaging and Translational Medicine, Department of Vascular & Tumor Interventional Radiology, The First Affiliated Hospital of Xiamen University, School of Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Wenxuan Ye
- The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Doudou Luo
- The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Jiwei Xing
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Center for Molecular Imaging and Translational Medicine, Department of Vascular & Tumor Interventional Radiology, The First Affiliated Hospital of Xiamen University, School of Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Qingqing Xiao
- Department of Vascular Intervention, Guilin Medical College Affiliated Hospital, Guilin Medical College, Guilin, 541000, China
| | - Huiling Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Center for Molecular Imaging and Translational Medicine, Department of Vascular & Tumor Interventional Radiology, The First Affiliated Hospital of Xiamen University, School of Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Youliang Yao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Center for Molecular Imaging and Translational Medicine, Department of Vascular & Tumor Interventional Radiology, The First Affiliated Hospital of Xiamen University, School of Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Guangxing Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Center for Molecular Imaging and Translational Medicine, Department of Vascular & Tumor Interventional Radiology, The First Affiliated Hospital of Xiamen University, School of Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Luyao Yang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Center for Molecular Imaging and Translational Medicine, Department of Vascular & Tumor Interventional Radiology, The First Affiliated Hospital of Xiamen University, School of Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Dongbei Guo
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Center for Molecular Imaging and Translational Medicine, Department of Vascular & Tumor Interventional Radiology, The First Affiliated Hospital of Xiamen University, School of Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Kun Wang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Normal University, Fuzhou, 350117, China
| | - Yaqin He
- Department of Oncology Surgery, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| | - Xiaofeng Ye
- Department of Oncology Surgery, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| | - Jinde Zhang
- Institute of Advanced Science Facilities, Shenzhen, Guangdong, 518107, China
| | - Zhaokui Jin
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhongxiong Fan
- School of Pharmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Urumqi, 830017, China
| | - Xiaofei Wen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Center for Molecular Imaging and Translational Medicine, Department of Vascular & Tumor Interventional Radiology, The First Affiliated Hospital of Xiamen University, School of Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Jingsong Mao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Center for Molecular Imaging and Translational Medicine, Department of Vascular & Tumor Interventional Radiology, The First Affiliated Hospital of Xiamen University, School of Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- Department of Vascular Intervention, Guilin Medical College Affiliated Hospital, Guilin Medical College, Guilin, 541000, China
| | - Xiaoyuan Chen
- Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 117597, Singapore
| | - Qingliang Zhao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Center for Molecular Imaging and Translational Medicine, Department of Vascular & Tumor Interventional Radiology, The First Affiliated Hospital of Xiamen University, School of Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
30
|
Wang S, Guo Q, Zhou L, Xia X. Ferroptosis: A double-edged sword. Cell Death Discov 2024; 10:265. [PMID: 38816377 PMCID: PMC11139933 DOI: 10.1038/s41420-024-02037-9] [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: 11/08/2023] [Revised: 05/19/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024] Open
Abstract
Ferroptosis represents a form of programmed cell death that is propelled by iron-dependent lipid peroxidation, thereby being distinguished by the prominent features of iron accumulation and lipid peroxidation. Ferroptosis has been implicated in numerous physiological and pathological phenomena, with mounting indications that it holds significant implications for cancer and other medical conditions. On one side, it demonstrates anti-cancer properties by triggering ferroptosis within malignant cells, and on the other hand, it damages normal cells causing other diseases. Therefore, in this paper, we propose to review the paradoxical regulation of ferroptosis in tumors and other diseases. First, we introduce the development history, concept and mechanism of ferroptosis. The second part focuses on the methods of inducing ferroptosis in tumors. The third section emphasizes the utilization of ferroptosis in different medical conditions and strategies to inhibit ferroptosis. The fourth part elucidates the key contradictions in the control of ferroptosis. Finally, potential research avenues in associated domains are suggested.
Collapse
Affiliation(s)
- Shengmei Wang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Qiuyan Guo
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Lili Zhou
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Xinhua Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| |
Collapse
|
31
|
Torres A, Michea MA, Végvári Á, Arce M, Pérez V, Alcota M, Morales A, Vernal R, Budini M, Zubarev RA, González FE. A multi-platform analysis of human gingival crevicular fluid reveals ferroptosis as a relevant regulated cell death mechanism during the clinical progression of periodontitis. Int J Oral Sci 2024; 16:43. [PMID: 38802345 PMCID: PMC11130186 DOI: 10.1038/s41368-024-00306-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: 11/05/2023] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 05/29/2024] Open
Abstract
Ferroptosis is implicated in the pathogenesis of numerous chronic-inflammatory diseases, yet its association with progressive periodontitis remains unexplored. To investigate the involvement and significance of ferroptosis in periodontitis progression, we assessed sixteen periodontitis-diagnosed patients. Disease progression was clinically monitored over twelve weeks via weekly clinical evaluations and gingival crevicular fluid (GCF) collection was performed for further analyses. Clinical metrics, proteomic data, in silico methods, and bioinformatics tools were combined to identify protein profiles linked to periodontitis progression and to explore their potential connection with ferroptosis. Subsequent western blot analyses validated key findings. Finally, a single-cell RNA sequencing (scRNA-seq) dataset (GSE164241) for gingival tissues was analyzed to elucidate cellular dynamics during periodontitis progression. Periodontitis progression was identified as occurring at a faster rate than traditionally thought. GCF samples from progressing and non-progressing periodontal sites showed quantitative and qualitatively distinct proteomic profiles. In addition, specific biological processes and molecular functions during progressive periodontitis were revealed and a set of hub proteins, including SNCA, CA1, HBB, SLC4A1, and ANK1 was strongly associated with the clinical progression status of periodontitis. Moreover, we found specific proteins - drivers or suppressors - associated with ferroptosis (SNCA, FTH1, HSPB1, CD44, and GCLC), revealing the co-occurrence of this specific type of regulated cell death during the clinical progression of periodontitis. Additionally, the integration of quantitative proteomic data with scRNA-seq analysis suggested the susceptibility of fibroblasts to ferroptosis. Our analyses reveal proteins and processes linked to ferroptosis for the first time in periodontal patients, which offer new insights into the molecular mechanisms of progressive periodontal disease. These findings may lead to novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Alfredo Torres
- Laboratory of Experimental Immunology & Cancer, Faculty of Dentistry, University of Chile, Santiago, Chile
- Department of Conservative Dentistry, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - M Angélica Michea
- Department of Conservative Dentistry, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Ákos Végvári
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Marion Arce
- Department of Conservative Dentistry, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Valentina Pérez
- Laboratory of Experimental Immunology & Cancer, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Marcela Alcota
- Department of Conservative Dentistry, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Alicia Morales
- Department of Conservative Dentistry, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Rolando Vernal
- Department of Conservative Dentistry, Faculty of Dentistry, University of Chile, Santiago, Chile
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Mauricio Budini
- Laboratory of Cellular and Molecular Pathology, Institute for Research in Dental Sciences, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Roman A Zubarev
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Fermín E González
- Laboratory of Experimental Immunology & Cancer, Faculty of Dentistry, University of Chile, Santiago, Chile.
- Department of Conservative Dentistry, Faculty of Dentistry, University of Chile, Santiago, Chile.
| |
Collapse
|
32
|
Xia N, Xu L, Xu D, Huang M, Li Y, Mei Z, Yu Z. Neuroprotective effect of emodin on Aβ 25-35-induced cytotoxicity in PC12 cells involves Nrf2/GPX4 and TLR4/p-NF-κB/NLRP3 pathways. Brain Res 2024; 1841:149019. [PMID: 38795791 DOI: 10.1016/j.brainres.2024.149019] [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: 03/25/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024]
Abstract
The present study aims to investigate the neuroprotective effects of emodin in Alzheimer's disease (AD). PC12 cells were used to explore the underlying mechanism and were incubated with Aβ25-35 for 24 h as the model group, incubated with emodin at different concentrations (2.5, 5, 10 μM) as the drug administration groups. The content of MDA and the enzymic activities of CAT, GSH-Px were detected by the corresponding commercial kits. The ROS level in Aβ25-35 induced cells was decreased by emodin dose-dependently, but the MMP in these cells were elevated. The expressions of AChE, TLR4, p-NF-κB, NLRP3, IL-1β, and TNF-α in PC12 cells were increased by Aβ25-35 treatment, the expressions of Nrf2, HO-1, GPX4, xCT were decreased, all the levels of expressions were reversed by emodin. Besides, ultraviolet spectrophotometry and infrared spectrophotometry were ultilized to ascertain the production of emodin-Fe (Ⅱ) complex. The FerroOrange results showed that emodin reduced free Fe2+ in cells. The immunofluorescent intensities of Nrf2, GPX4, and p-NF-κB offered direct visible evidence for emodin's multi-targets in AD treatment. Collectively, emodin could inhibit the activity of AChE and exert neuroprotective effects against AD through antioxidant, anti-ferroptotic, anti-inflammatory properties via Nrf2/GPX4 and TLR4/p-NF-κB/NLRP3 pathways.
Collapse
Affiliation(s)
- Nengyin Xia
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Lingyun Xu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Dengrui Xu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Mengyuan Huang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yang Li
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Zhinan Mei
- College of Plant Science & Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zejun Yu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China.
| |
Collapse
|
33
|
Lou T, Wu H, Feng M, Liu L, Yang X, Pan M, Wei Z, Zhang Y, Shi L, Qu B, Yang H, Cong S, Chen K, Liu J, Li Y, Jia Z, Xiao H. Integration of metabolomics and transcriptomics reveals that Da Chuanxiong Formula improves vascular cognitive impairment via ACSL4/GPX4 mediated ferroptosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117868. [PMID: 38325668 DOI: 10.1016/j.jep.2024.117868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/02/2024] [Accepted: 02/04/2024] [Indexed: 02/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Da Chuanxiong Formula (DCX) is a traditional herbal compound composed of Gastrodia elata Bl. and Ligusticum chuanxiong Hort, which could significantly enhance blood circulation and neuroprotection, showing promise in treating Vascular Cognitive Impairment (VCI). AIM OF STUDY This study aims to elucidate the potential of DCX in treating VCI and its underlying mechanism. MATERIALS AND METHODS Firstly, the cognitive behavior level, blood flow changes, and brain pathology changes were evaluated through techniques such as the Morris water maze, step-down, laser speckle, coagulation analysis, and pathological staining to appraise the DCX efficacy. Then, the DCX targeting pathways were decoded by merging metabolomics with transcriptomics. Finally, the levels of reactive oxygen species (ROS), Fe2+, and lipid peroxidation related to the targeting signaling pathways of DCX were detected by kit, and the expression levels of mRNAs or proteins related to ferroptosis were determined by qPCR or Western blot assays respectively. RESULTS DCX improved cognitive abilities and cerebral perfusion significantly, and mitigated pathological damage in the hippocampal region of VCI model rats. Metabolomics revealed that DCX was able to call back 33 metabolites in plasma and 32 metabolites in brain samples, and the majority of the differential metabolites are phospholipid metabolites. Transcriptomic analysis revealed that DCX regulated a total of 3081 genes, with the ferroptosis pathway exhibiting the greatest impact. DCX inhibited ferroptosis of VCI rates by decreasing the levels of ferrous iron, ROS, and malondialdehyde (MDA) while increasing the level of superoxide dismutase (SOD) and glutathione (GSH) in VCI rats. Moreover, the mRNA and protein levels of ACSL4, LPCAT3, ALOX15, and GPX4, which are related to lipid metabolism in ferroptosis, were also regulated by DCX. CONCLUSION Our research findings indicated that DCX could inhibit ferroptosis through the ACSL4/GPX4 signaling pathway, thereby exerting its therapeutic benefits on VCI.
Collapse
Affiliation(s)
- Tianyu Lou
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Hao Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Menghan Feng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Lirong Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoqin Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Mingxia Pan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zuying Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yinhuan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China; Department of Pharmacy, China-Japan Friendship Hospital, Beijing, China
| | - Lixia Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Biqiong Qu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Haolan Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shiyu Cong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Kui Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Liu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yueting Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhixin Jia
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hongbin Xiao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China; Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
34
|
Shang J, Jiao J, Wang J, Yan M, Li Q, Shabuerjiang L, Huang G, Song Q, Wen Y, Zhang X, Wu K, Cui Y, Liu X. Chrysin inhibits ferroptosis of cerebral ischemia/reperfusion injury via regulating HIF-1α/CP loop. Biomed Pharmacother 2024; 174:116500. [PMID: 38555815 DOI: 10.1016/j.biopha.2024.116500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/18/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024] Open
Abstract
Chrysin is a natural flavonoid with powerful neuroprotective capacity. Cerebral ischemia/reperfusion injury (CIRI) is associated with oxidative stress and ferroptosis. Hypoxia-inducible factor 1α (HIF-1α) and ceruloplasmin (CP) are the critical targets for oxidation reactions and iron transport. But the regulatory mechanism between them is still unclear. Transient middle cerebral artery occlusion (tMCAO) model in rats and oxygen and glucose deprivation/re-oxygenation (OGD/R) model in PC12 cells were applied. Pathological tissue staining and biochemical kit were used to evaluate the effect of chrysin. The relationship between HIF-1α and CP was verified by transcriptomics, qRT-PCR and Western blot. In CIRI, HIF-1α/CP loop was discovered to be the regulatory pathway of ferroptosis. CIRI led to activation and nuclear translocation of HIF-1α, which promoted CP transcription and translation, and downstream ferroptosis. Inhibition of HIF-1α had opposite effect on CP and ferroptosis regulation. Overexpression of CP increased the expression of HIF-1α, nevertheless, inhibited the nuclear translocation of HIF-1α and alleviated CIRI. Silencing CP promoted HIF-1α elevation in nucleus and aggravated CIRI. Mechanistically, chrysin restrained HIF-1α nuclear translocation, thereby inhibiting CP transcription and translation, which in turn reduced downstream HIF-1α expression and mitigated ferroptosis in CIRI. Our results highlight chrysin restrains ferroptosis in CIRI through HIF-1α/CP loop.
Collapse
Affiliation(s)
- Jinfeng Shang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiakang Jiao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jingyi Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Mingxue Yan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Qiannan Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Lizha Shabuerjiang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Guijinfeng Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Qi Song
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yinlian Wen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaolu Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Kai Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yiran Cui
- Department of pharmacy, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing 100010, China
| | - Xin Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
35
|
Tan Q, Wu D, Lin Y, Ai H, Xu J, Zhou H, Gu Q. Identifying eleven new ferroptosis inhibitors as neuroprotective agents from FDA-approved drugs. Bioorg Chem 2024; 146:107261. [PMID: 38460336 DOI: 10.1016/j.bioorg.2024.107261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/24/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
With increasing evidence that ferroptosis is associated with diverse neurological disorders, targeting ferroptosis offers a promising avenue for developing effective pharmaceutical agents for neuroprotection. In this study, we identified ferroptosis inhibitors as neuroprotective agents from US Food and Drug Administration (FDA)-approved drugs. 1176 drugs have been screened against erastin-induced ferroptosis in HT22 cells, resulting in 89 ferroptosis inhibitors. Among them, 26 drugs showed significant activity with EC50 below10 μM. The most active ferroptosis inhibitor is lumateperone tosylate at nanomolar level. 11 drugs as ferroptosis inhibitors were not reported previously. Further mechanistic studies revealed that their mechanisms of actions involve free radical scavenging, Fe2+ chelation, and 15-lipoxygenase inhibition. Notably, the active properties of some drugs were firstly revealed here. These ferroptosis inhibitors increase the chemical diversity of ferroptosis inhibitors, and offer new therapeutic possibilities for the treatments of related neurological diseases.
Collapse
Affiliation(s)
- Qingyun Tan
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Deyin Wu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yating Lin
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Haopeng Ai
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Jun Xu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Huihao Zhou
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Qiong Gu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China.
| |
Collapse
|
36
|
Xu K, Liu Z, Pan S, Zhang N, Wu S, Yang G, Song X, Teng Y, Tong X. BMSCs attenuate radiation-induced brain injury induced hippocampal neuronal apoptosis through a PI3K/Akt/Bax/Bcl-2 signaling pathway. Brain Res 2024; 1829:148795. [PMID: 38331376 DOI: 10.1016/j.brainres.2024.148795] [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: 10/07/2023] [Revised: 01/26/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND Bone marrow mesenchymal stem cell (BMSCs) -based therapies represent a promising treatment for neurological disorders. However, therapeutic effects and mechanisms of BMSCs transplantation for radiation-induced brain injury (RIBI) have not been fully disclosed. In this article, we explored the functions of BMSCs transplantation on RIBI and investigated the protective effects of BMSCS on hippocampal neurons in RIBI as well as the related molecular mechanisms. MATERIALS AND METHODS 6-8 weeks-old rats were used to build a RIBI model. Rats in BMSC group were treated with a 3 × 106 BMSCs injection through the tail vein on the 1st day and 8th day after irradiation; rats in both control and RIBI groups were injected with an equivalent volume of physiological saline for comparisons. The Morris water maze was applied to detect the variations in cognitive function after RIBI. MRS was performed to test changes in NAA/Cr, indicating neuronal apoptosis after RIBI. TUNEL was conducted to detect apoptosis of rat hippocampal neurons, and HE staining was carried out to show pathological variations in the hippocampal region of rats. Protein levels of PI3K, P-PI3K, AKT, P-AKT, Bcl-2, and Bax proteins of rats in the hippocampal area were all determined by Western blot. RESULTS Cognitive function was reduced and hippocampal neurons underwent apoptosis in the rats of the RIBI group, and cognitive abilities, histopathological alterations, and apoptosis of hippocampal neurons were significantly improved after BMSCs treatment; the expression of PI3K, P-PI3K, AKT, P-AKT, and Bcl-2 proteins, in the hippocampal region of the rat, was up-regulated, and Bax proteins were down-regulated. CONCLUSIONS BMCSs can inhibit hippocampal neuronal apoptosis in RIBI, and the mechanism may be associated with the up-regulation of Bcl-2 and down-regulation of Bax by the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Kaina Xu
- College of Medical Technology, Qiqihar Medical University, No.333 Bukui Street, Jianhua District, Qiqihar, Heilongjiang 161006, China
| | - Zhanhong Liu
- College of Medical Technology, Qiqihar Medical University, No.333 Bukui Street, Jianhua District, Qiqihar, Heilongjiang 161006, China
| | - Shichao Pan
- College of Medical Technology, Qiqihar Medical University, No.333 Bukui Street, Jianhua District, Qiqihar, Heilongjiang 161006, China
| | - Na Zhang
- College of Medical Technology, Qiqihar Medical University, No.333 Bukui Street, Jianhua District, Qiqihar, Heilongjiang 161006, China
| | - Shuang Wu
- Department of Radiotherapy, The Third Affiliated Hospital of Qiqihar Medical University, No.27, Taishun Street, Tiefeng District, Qiqihar, Heilongjiang 161006, China
| | - Guangrun Yang
- Department of Radiotherapy, The Third Affiliated Hospital of Qiqihar Medical University, No.27, Taishun Street, Tiefeng District, Qiqihar, Heilongjiang 161006, China
| | - Xue Song
- Department of Radiotherapy, The Third Affiliated Hospital of Qiqihar Medical University, No.27, Taishun Street, Tiefeng District, Qiqihar, Heilongjiang 161006, China
| | - Ye Teng
- Department of Radiotherapy, The Third Affiliated Hospital of Qiqihar Medical University, No.27, Taishun Street, Tiefeng District, Qiqihar, Heilongjiang 161006, China
| | - Xu Tong
- Department of Radiotherapy, The Third Affiliated Hospital of Qiqihar Medical University, No.27, Taishun Street, Tiefeng District, Qiqihar, Heilongjiang 161006, China.
| |
Collapse
|
37
|
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
|
38
|
ZHANG X, HUANG Z, HUANG P, YANG M, ZHANG Z, NI G. Mechanism of acupuncture in attenuating cerebral ischaemia-reperfusion injury based on nuclear receptor coactivator 4 mediated ferritinophagy. J TRADIT CHIN MED 2024; 44:345-352. [PMID: 38504540 PMCID: PMC10927404 DOI: 10.19852/j.cnki.jtcm.20240203.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 05/09/2023] [Indexed: 03/21/2024]
Abstract
OBJECTIVE To explore the effect of acupuncture treatment on cerebral ischaemia-reperfusion injury (CIRI) and reveal the underlying mechanism of the effect based on nuclear receptor coactivator 4 (NCOA4) mediated ferritinophagy. METHODS Sprague-Dawley male rats were divided into four groups: the sham group, model group, acupuncture group, and sham acupuncture group. After 2 h of middle cerebral artery occlusion (MCAO), reperfusion was performed for 24 h to induce CIRI. The rats were treated with acupuncture at the Neiguan (PC6) and Shuigou (GV26) acupoints. Their neurological function was evaluated by taking their Bederson scores at 2 h after ischaemia and 24 h after reperfusion. Triphenyltetrazolium chloride staining was applied to assess the cerebral infarct volume at 24 h after reperfusion. The malondialdehyde (MDA) and ferrous iron (Fe2+) levels were observed after 24 h of reperfusion using an assay kit. Western blotting was performed to detect the expression of NCOA4 and ferritin heavy chain 1 (FTH1) at 24 h after reperfusion. Moreover, the colocalization of ferritin with neurons, NCOA4 with microtubule-associated protein 1 light chain 3 (LC3), and NCOA4 with ferritin was visualized using immunofluorescence staining. RESULTS Acupuncture significantly improved neurological function and decreased cerebral infarct volume in the acupuncture group. Following CIRI, the expression of NCOA4, LC3 and FTH1 was increased, which enhanced ferritinophagy and induced an inappropriate accumulation of Fe2+ and MDA in the ischaemic brain. However, acupuncture dramatically downregulated the expression of NCOA4, LC3 and FTH1, inhibited the overactivation of ferritinophagy, and decreased the levels of MDA and Fe2+. CONCLUSIONS Acupuncture can inhibit NCOA4-mediated ferritinophagy and protect neurons against CIRI in a rat model.
Collapse
Affiliation(s)
- Xinchang ZHANG
- 1 College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing 210023, China
- 2 Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zheng HUANG
- 1 College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing 210023, China
- 2 Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Peiyan HUANG
- 1 College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing 210023, China
- 2 Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mengning YANG
- 1 College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing 210023, China
- 2 Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhihui ZHANG
- 1 College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing 210023, China
- 2 Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Guangxia NI
- 1 College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing 210023, China
- 2 Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
39
|
Kang L, Piao M, Liu N, Gu W, Feng C. Sevoflurane Exposure Induces Neuronal Cell Ferroptosis Initiated by Increase of Intracellular Hydrogen Peroxide in the Developing Brain via ER Stress ATF3 Activation. Mol Neurobiol 2024; 61:2313-2335. [PMID: 37874483 DOI: 10.1007/s12035-023-03695-z] [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: 05/11/2023] [Accepted: 10/04/2023] [Indexed: 10/25/2023]
Abstract
Neuronal cell death is acknowledged as the primary pathological basis underlying developmental neurotoxicity in response to sevoflurane exposure, but the exact mechanism remains unclear. Ferroptosis is a form of programmed cell death characterized by iron-dependent lipid peroxidation that is driven by hydrogen peroxide (H2O2) and ferrous iron through the Fenton reaction and participates in the pathogenesis of multiple neurological diseases. As stress response factor, activating transcription factor 3 (ATF3) can be activated by the PERK/ATF4 pathway during endoplasmic reticulum (ER) stress, followed by increased intracellular H2O2, which is involved in regulation of apoptosis, autophagy, and ferroptosis. Here, we investigated whether ferroptosis and ATF3 activation were implicated in sevoflurane-induced neuronal cell death in the developing brain. The results showed that sevoflurane exposure induced neuronal death as a result of iron-dependent lipid peroxidation damage secondary to H2O2 accumulation and ferrous iron increase, which was consistent with the criteria for ferroptosis. Furthermore, we observed that increases in iron and H2O2 induced by sevoflurane exposure were associated with the upregulation and nuclear translocation of ATF3 in response to ER stress. Knockdown of ATF3 expression alleviated iron-dependent lipid peroxidation, which prevented sevoflurane-induced neuronal ferroptosis. Mechanistically, ATF3 promoted sevoflurane-induced H2O2 accumulation by activating NOX4 and suppressing catalase, GPX4, and SLC7A11 expression. Additionally, an increase in H2O2 was accompanied by the upregulation of TFR and TF and downregulation of FPN, which linked iron overload to ferroptosis induced by sevoflurane. Taken together, our results demonstrated that ER stress-mediated ATF3 activation contributed to sevoflurane-induced neuronal ferroptosis via H2O2 accumulation and the resultant iron overload.
Collapse
Affiliation(s)
- Liheng Kang
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun, 130021, China
| | - Meihua Piao
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun, 130021, China
| | - Nan Liu
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun, 130021, China
| | - Wanping Gu
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun, 130021, China
| | - Chunsheng Feng
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun, 130021, China.
| |
Collapse
|
40
|
Koshiishi I. [What is the Initiating Reaction for the Lipid Radical Chain Reaction System That Can Induce Ferroptotic Cell Death at the Lower Oxygen Content?]. YAKUGAKU ZASSHI 2024; 144:431-439. [PMID: 38246655 DOI: 10.1248/yakushi.23-00207] [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] [Indexed: 01/23/2024]
Abstract
The neural cell death in cerebral infarction is suggested to be ferroptosis-like cell death, involving the participation of 15-lipoxygenase (15-LOx). Ferroptosis is induced by lipid radical species generated through the one-electron reduction of lipid hydroperoxides, and it has been shown to propagate intracellularly and intercellularly. At lower oxygen concentration, it appeared that both regiospecificity and stereospecificity of conjugated diene moiety in lipoxygenase-catalysed lipid hydroperoxidation are drastically lost. As a result, in the reaction with linoleic acid, the linoleate 9-peroxyl radical-ferrous lipoxygenase complex dissolves into the linoleate 9-peroxyl radical and ferrous 15-lipoxygenase. Subsequently, the ferrous 15-lipoxygenase then undergoes one-electron reduction of 13-hydroperoxy octadecadienoic acid, generating an alkoxyl radical (pseudoperoxidase reaction). A part of the produced lipid alkoxyl radicals undergoes cleavage of C-C bonds, liberating small molecular hydrocarbon radicals. Particularly, in ω-3 polyunsaturated fatty acids, which are abundant in the vascular and nervous systems, the liberation of small molecular hydrocarbon radicals was more pronounced compared to ω-6 polyunsaturated fatty acids. The involvement of these small molecular hydrocarbon radicals in the propagation of membrane lipid damage is suggested.
Collapse
Affiliation(s)
- Ichiro Koshiishi
- Department of Laboratory Sciences, Graduate School of Health Sciences, Gunma University
| |
Collapse
|
41
|
Lv B, Fu P, Wang M, Cui L, Bao L, Wang X, Yu L, Zhou C, Zhu M, Wang F, Pang Y, Qi S, Zhang Z, Cui G. DMT1 ubiquitination by Nedd4 protects against ferroptosis after intracerebral hemorrhage. CNS Neurosci Ther 2024; 30:e14685. [PMID: 38634270 PMCID: PMC11024684 DOI: 10.1111/cns.14685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/20/2024] [Accepted: 02/06/2024] [Indexed: 04/19/2024] Open
Abstract
OBJECTIVE Neuronal precursor cells expressed developmentally down-regulated 4 (Nedd4) are believed to play a critical role in promoting the degradation of substrate proteins and are involved in numerous biological processes. However, the role of Nedd4 in intracerebral hemorrhage (ICH) remains unknown. This study aims to investigate the regulatory role of Nedd4 in the ICH model. METHODS Male C57BL/6J mice were induced with ICH. Subsequently, the levels of glutathione peroxidase 4 (GPX4), malondialdehyde (MDA) concentration, iron content, mitochondrial morphology, as well as the expression of divalent metal transporter 1 (DMT1) and Nedd4 were assessed after ICH. Furthermore, the impact of Nedd4 overexpression was evaluated through analyses of hematoma area, ferroptosis, and neurobehavioral function. The mechanism underlying Nedd4-mediated degradation of DMT1 was elecidated using immunoprecipitation (IP) after ICH. RESULTS Upon ICH, the level of DMT1 in the brain increased, but decreased when Nedd4 was overexpressed using Lentivirus, suggesting a negative correlation between Nedd4 and DMT1. Additionally, the degradation of DMT1 was inhibited after ICH. Furthermore, it was found that Nedd4 can interact with and ubiquitinate DMT1 at lysine residues 6, 69, and 277, facilitating the degradation of DMT1. Functional analysis indicated that overexpression of Nedd4 can alleviate ferroptosis and promote recovery following ICH. CONCLUSION The results demonstrated that ferroptosis occurs via the Nedd4/DMT1 pathway during ICH, suggesting it potential as a valuable target to inhibit ferroptosis for the treatment of ICH.
Collapse
Affiliation(s)
- Bingchen Lv
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Ping Fu
- School of Life Sciences, Nanjing UniversityNanjingChina
| | - Miao Wang
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Department of GeriatricsThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Likun Cui
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Lei Bao
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Xingzhi Wang
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Lu Yu
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Chao Zhou
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Mengxin Zhu
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Fei Wang
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Ye Pang
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Suhua Qi
- School of Medical Technology, Xuzhou Medical UniversityXuzhouChina
| | - Zuohui Zhang
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Guiyun Cui
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| |
Collapse
|
42
|
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
|
43
|
Zhang X, Hu Y, Wang B, Yang S. Ferroptosis: Iron-mediated cell death linked to disease pathogenesis. J Biomed Res 2024; 38:1-23. [PMID: 38808552 PMCID: PMC11461536 DOI: 10.7555/jbr.37.20230224] [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: 09/19/2023] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 05/30/2024] Open
Abstract
Ferroptosis is an iron-mediated regulatory cell death pattern characterized by oxidative damage. The molecular regulating mechanisms are related to iron metabolism, lipid peroxidation, and glutathione metabolism. Additionally, some immunological signaling pathways, such as the cyclic GMP-AMP synthase-stimulator ofinterferon genes axis, Janus kinase-signal transducer and activator of transcription 1 axis, and transforming growth factor beta 1-Smad3 axis may also participate in the regulation of ferroptosis. Studies have shown that ferroptosis is closely related to many diseases such as cancer, neurodegenerative diseases, inflammatory diseases, and autoimmune diseases. Considering the pivotal role of ferroptosis-regulating signaling in the pathogenesis of diverse diseases, the development of ferroptosis inducers or inhibitors may have significant clinical potential for the treatment of the aforementioned conditions.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yingchao Hu
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Bingwei Wang
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Shuo Yang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
44
|
Ye Y, Dai L, Mugaanyi J, Fu W, Hu F. Novel insights into the pathogenesis of thyroid eye disease through ferroptosis-related gene signature and immune infiltration analysis. Aging (Albany NY) 2024; 16:6008-6034. [PMID: 38536014 PMCID: PMC11042930 DOI: 10.18632/aging.205685] [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: 10/18/2023] [Accepted: 02/13/2024] [Indexed: 04/23/2024]
Abstract
Thyroid eye disease (TED) has brought great physical and mental trauma to patients worldwide. Although a few potential signaling pathways have been reported, knowledge of TED remains limited. Our objective is to explore the fundamental mechanism of TED and identify potential therapeutic targets using diverse approaches. To perform a range of bioinformatic analyses, such as identifying differentially expressed genes (DEGs), conducting enrichment analysis, establishing nomograms, analyzing weighted gene correlation network analysis (WGCNA), and studying immune infiltration, the datasets GSE58331, GSE105149, and GSE9340 were integrated. Further validation was conducted using qPCR, western blot, and immunohistochemistry techniques. Eleven ferroptosis-related DEGs derived from the lacrimal gland were originally screened. Their high diagnostic value was proven, and diagnostic prediction nomogram models with high accuracy and robustness were established by using machine learning. A total of 15 hub gene-related DEGs were identified by WGCNA. Through CIBERSORTx, we uncovered five immune cells highly correlated with TED and found several special associations between these immune cells and the above DEGs. Furthermore, EGR2 from the thyroid sample was revealed to be closely negatively correlated with most DEGs from the lacrimal gland. High expression of APOD, COPB2, MYH11, and MYCN, as well as CD4/CD8 T cells and B cells, was verified in the periorbital adipose tissues of TED patients. To summarize, we discovered a new gene signature associated with ferroptosis that has a critical impact on the development of TED and provides valuable insights into immune infiltration. These findings might highlight the new direction and therapeutic strategies of TED.
Collapse
Affiliation(s)
- Yunyan Ye
- Department of Ophthalmology, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China
| | - Lei Dai
- Department of Hepato-Pancreato-Biliary Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China
| | - Joseph Mugaanyi
- Department of Hepato-Pancreato-Biliary Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China
| | - Weina Fu
- Department of Ophthalmology, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China
| | - Feng Hu
- Department of Ophthalmology, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China
| |
Collapse
|
45
|
Wei J, Wang G, Lai M, Zhang Y, Li F, Wang Y, Tan Y. Faecal Microbiota Transplantation Alleviates Ferroptosis after Ischaemic Stroke. Neuroscience 2024; 541:91-100. [PMID: 38296019 DOI: 10.1016/j.neuroscience.2024.01.021] [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: 11/06/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/10/2024]
Abstract
Ischaemic stroke can induce changes in the abundance of gut microbiota constituents, and the outcome of stroke may also be influenced by the gut microbiota. This study aimed to determine whether gut microbiota transplantation could rescue changes in the gut microbiota and reduce ferroptosis after stroke in rats. Male Sprague-Dawley rats (6 weeks of age) were subjected to ischaemic stroke by middle cerebral artery occlusion (MCAO). Fecal samples were collected for 16S ribosomal RNA (rRNA) sequencing to analyze the effects of FMT on the gut microbiota. Neurological deficits were evaluated using the Longa score. triphenyl tetrazolium chloride (TTC) staining was performed in the brain, and kits were used to measure malondialdehyde (MDA), iron, and glutathione (GSH) levels in the ipsilateral brain of rats. Western blotting was used to detect the protein expression levels of glutathione peroxidase 4 (GPX4), solute carrier family 7 member 11 (SLC7A11), and the transferrin receptor 2 (TFR2) in the ipsilateral brain of rats. Stroke induced significant changes in the gut microbiota, and FMT ameliorated these changes. TTC staining results showed that FMT reduced cerebral infarct volume. In addition, FMT diminished MDA and iron levels and elevated GSH levels in the ipsilateral brain. Western blot analysis showed that FMT increased GPX4 and SLC7A11 protein expression and decreased TFR2 protein expression in the ipsilateral brain after stroke. FMT can reverse gut microbiota dysbiosis, reduce cerebellar infarct volume, and decrease ferroptosis after stroke.
Collapse
Affiliation(s)
- Jinzhen Wei
- Department of Anesthesiology, Affiliated Hospital of Guilin Medical University, Guilin 541000, China
| | - Gang Wang
- Department of Anesthesiology, Affiliated Hospital of Guilin Medical University, Guilin 541000, China
| | - Min Lai
- Department of Anesthesiology, Affiliated Hospital of Guilin Medical University, Guilin 541000, China
| | - Yipin Zhang
- Department of Anesthesiology, Affiliated Hospital of Guilin Medical University, Guilin 541000, China
| | - Fengru Li
- Department of Anesthesiology, Affiliated Hospital of Guilin Medical University, Guilin 541000, China
| | - Yongwang Wang
- Department of Anesthesiology, Affiliated Hospital of Guilin Medical University, Guilin 541000, China.
| | - Yongxing Tan
- Guilin Municipal Hospital of Traditional Chinese Medicine, Guilin 541000, China.
| |
Collapse
|
46
|
Han X, Zhang M, Yan L, Fu Y, Kou H, Shang C, Wang J, Liu H, Jiang C, Wang J, Cheng T. Role of dendritic cells in spinal cord injury. CNS Neurosci Ther 2024; 30:e14593. [PMID: 38528832 PMCID: PMC10964036 DOI: 10.1111/cns.14593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/15/2023] [Accepted: 12/10/2023] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Inflammation can worsen spinal cord injury (SCI), with dendritic cells (DCs) playing a crucial role in the inflammatory response. They mediate T lymphocyte differentiation, activate microglia, and release cytokines like NT-3. Moreover, DCs can promote neural stem cell survival and guide them toward neuron differentiation, positively impacting SCI outcomes. OBJECTIVE This review aims to summarize the role of DCs in SCI-related inflammation and identify potential therapeutic targets for treating SCI. METHODS Literature in PubMed and Web of Science was reviewed using critical terms related to DCs and SCI. RESULTS The study indicates that DCs can activate microglia and astrocytes, promote T-cell differentiation, increase neurotrophin release at the injury site, and subsequently reduce secondary brain injury and enhance functional recovery in the spinal cord. CONCLUSIONS This review highlights the repair mechanisms of DCs and their potential therapeutic potential for SCI.
Collapse
Affiliation(s)
- Xiaonan Han
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Mingkang Zhang
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Liyan Yan
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yikun Fu
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Hongwei Kou
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Chunfeng Shang
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Junmin Wang
- Department of Anatomy, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Hongjian Liu
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Chao Jiang
- Department of NeurologyThe Fifth Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Jian Wang
- Department of Anatomy, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Tian Cheng
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
47
|
Fan B, Zhang Y, Luo Q, Hao C, Liao W. Physical and social environmental enrichment alleviate ferroptosis and inflammation with inhibition of TLR4/MyD88/p38MAPK pathway in chronic cerebral hypoperfusion rats. Brain Res Bull 2024; 208:110897. [PMID: 38340777 DOI: 10.1016/j.brainresbull.2024.110897] [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: 11/07/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
A typical enriched environment (EE), which combines physical activity and social interaction, has been proven to mitigate cognitive impairment caused by chronic cerebral hypoperfusion (CCH). However, it remains unclear how the different components of EE promote cognitive recovery after CCH. This study stripped out the different components of EE into physical environmental enrichment (PE) and social environmental enrichment (SE), and compared the neuroprotective effects of PE, SE and typical EE (PSE) in CCH. The results of novel object recognition and Morris water maze tests showed that PE, SE, and PSE improved cognitive function in CCH rats. Additionally, Nissl and TUNEL staining revealed that three EEs reduced neuronal loss in the hippocampus. PSE exhibited superior neuroprotective and functional improvement effects compared to PE and SE, while there was no significant difference between PE and SE. Furthermore, three EEs reduced lipid peroxidation in the hippocampus with decreasing the levels of MDA and increasing the activities of SOD and GSH. The expression of SLC7A11 and GPX4 was increased, while the level of p53 was reduced in three EEs. This suggested that three EEs inhibited ferroptosis by maintaining the redox homeostasis in the hippocampus. Three EEs reduced the levels of IL-β, TNF-α, and IL-6, thereby inhibiting neuroinflammation. Additionally, Western blotting and immunofluorescence results indicated that three EEs also inhibited the TLR4/MyD88/p38MAPK signaling pathway. These findings collectively demonstrated that the three EEs alleviated hippocampal ferroptosis and neuroinflammation in CCH rats, thereby reducing neuronal loss, which might be associated with the inhibition of the TLR4/MyD88/p38MAPK signaling pathway. Moreover, the study results supported that it is only through the combination of physical exercise and social interaction that the optimal neuroprotective effects can be achieved. These findings provided valuable insights for the prevention and treatment of vascular cognitive impairment.
Collapse
Affiliation(s)
- Bin Fan
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ying Zhang
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qihang Luo
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chizi Hao
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Weijing Liao
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
48
|
Wu Q, Jiang N, Wang Y, Song G, Li P, Fang Y, Xu L, Wang W, Xie M. Soluble epoxide hydrolase inhibitor (TPPU) alleviates ferroptosis by regulating CCL5 after intracerebral hemorrhage in mice. Biomed Pharmacother 2024; 172:116301. [PMID: 38377737 DOI: 10.1016/j.biopha.2024.116301] [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: 12/06/2023] [Revised: 02/16/2024] [Accepted: 02/17/2024] [Indexed: 02/22/2024] Open
Abstract
Soluble epoxide hydrolase (sEH) inhibition has been shown multiple beneficial effects against brain injuries of Intracerebral hemorrhage (ICH). However, the underlying mechanism of its neuroprotective effects after ICH has not been explained fully. Ferroptosis, a new form of iron-dependent programmed cell death, has been shown to be implicated in the secondary injuries after ICH. In this study, We examined whether sEH inhibition can alleviate brain injuries of ICH through inhibiting ferroptosis. Expression of several markers for ferroptosis was observed in the peri-hematomal brain tissues in mice after ICH. lip-1, a ferroptosis inhibitor, alleviated iron accumulation, lipid peroxidation and the secondary damages post-ICH in mice model. Intraperitoneal injection of 1-Trifluoromethoxyphenyl-3- (1-propionylpiperidin-4-yl)urea (TPPU), a highly selective sEH inhibitor, could inhibit ferroptosis and alleviate brain damages in ICH mice. Furthermore, RNA-sequencing was applied to explore the potential regulatory mechanism underlying the effects of TPPU in ferroptosis after ICH. C-C chemokine ligand 5 (CCL5) may be the key factor by which TPPU regulated ferroptosis after ICH since CCL5 antagonist could mimic the effects of TPPU and CCL5 reversed the inhibitive effect of TPPU on ferroptosis and the neuroprotective effects of TPPU on secondary damage after ICH. Taken together, these data indicate that ferroptosis is a key pathological feature of ICH and Soluble epoxide hydrolase inhibitor can exert neuroprotective effect by preventing ferroptosis after ICH.
Collapse
Affiliation(s)
- Qiao Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Na Jiang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Yao Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Guini Song
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Ping Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Yongkang Fang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Li Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, PR China.
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, PR China.
| |
Collapse
|
49
|
Mohan S, Alhazmi HA, Hassani R, Khuwaja G, Maheshkumar VP, Aldahish A, Chidambaram K. Role of ferroptosis pathways in neuroinflammation and neurological disorders: From pathogenesis to treatment. Heliyon 2024; 10:e24786. [PMID: 38314277 PMCID: PMC10837572 DOI: 10.1016/j.heliyon.2024.e24786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 02/06/2024] Open
Abstract
Ferroptosis is a newly discovered non-apoptotic and iron-dependent type of cell death. Ferroptosis mainly takes place owing to the imbalance of anti-oxidation and oxidation in the body. It is regulated via a number of factors and pathways both inside and outside the cell. Ferroptosis is closely linked with brain and various neurological disorders (NDs). In the human body, the brain contains the highest levels of polyunsaturated fatty acids, which are known as lipid peroxide precursors. In addition, there is also a connection of glutathione depletion and lipid peroxidation with NDs. There is growing evidence regarding the possible link between neuroinflammation and multiple NDs, such as Alzheimer's disease, amyotrophic lateral sclerosis, Parkinson's disease, Huntington's disease, and stroke. Recent studies have demonstrated that disruptions of lipid reactive oxygen species (ROS), glutamate excitatory toxicity, iron homeostasis, and various other manifestations linked with ferroptosis can be identified in various neuroinflammation-mediated NDs. It has also been reported that damage-associated molecular pattern molecules including ROS are generated during the events of ferroptosis and can cause glial activation via activating neuroimmune pathways, which subsequently leads to the generation of various inflammatory factors that play a role in various NDs. This review summarizes the regulation pathways of ferroptosis, the link between ferroptosis as well as inflammation in NDs, and the potential of a range of therapeutic agents that can be used to target ferroptosis and inflammation in the treatment of neurological disorders.
Collapse
Affiliation(s)
- Syam Mohan
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Hassan A Alhazmi
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Rym Hassani
- Department of Mathematics, University College AlDarb, Jazan University, Jazan, Saudi Arabia
| | - Gulrana Khuwaja
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - V P Maheshkumar
- Department of Pharmacy, Annamalai University, Annamalai Nagar 608002, Tamil Nadu, India
| | - Afaf Aldahish
- Department of Pharmacology and Toxicology, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Kumarappan Chidambaram
- Department of Pharmacology and Toxicology, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| |
Collapse
|
50
|
Chen A, Li Q, Huang Y, Li Y, Chuang YN, Hu X, Guo S, Wu Y, Guo Y, Bian J. Feasibility of Identifying Factors Related to Alzheimer's Disease and Related Dementia in Real-World Data. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.10.24302621. [PMID: 38405723 PMCID: PMC10889002 DOI: 10.1101/2024.02.10.24302621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
A comprehensive view of factors associated with AD/ADRD will significantly aid in studies to develop new treatments for AD/ADRD and identify high-risk populations and patients for prevention efforts. In our study, we summarized the risk factors for AD/ADRD by reviewing existing meta-analyses and review articles on risk and preventive factors for AD/ADRD. In total, we extracted 477 risk factors in 10 categories from 537 studies. We constructed an interactive knowledge map to disseminate our study results. Most of the risk factors are accessible from structured Electronic Health Records (EHRs), and clinical narratives show promise as information sources. However, evaluating genomic risk factors using RWD remains a challenge, as genetic testing for AD/ADRD is still not a common practice and is poorly documented in both structured and unstructured EHRs. Considering the constantly evolving research on AD/ADRD risk factors, literature mining via NLP methods offers a solution to automatically update our knowledge map.
Collapse
Affiliation(s)
- Aokun Chen
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| | - Qian Li
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| | - Yu Huang
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| | - Yongqiu Li
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| | - Yu-neng Chuang
- Department of Computer Science, George R. Brown School of Engineering, Rice University, 6100 Main St., Houston, TX 77005
| | - Xia Hu
- Department of Computer Science, George R. Brown School of Engineering, Rice University, 6100 Main St., Houston, TX 77005
| | - Serena Guo
- Department of Pharmaceutical Outcomes & Policy, College of Pharmacy, University of Florida, 1225 Center Drive, Gainesville, FL 32610
| | - Yonghui Wu
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| | - Yi Guo
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| | - Jiang Bian
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 1889 Museum Rd, Suite 7000, Gainesville, FL 32610
| |
Collapse
|