1
|
Bai X, Liu Y, Liu J, Guo K, Guan H. ADSCs-derived exosomes suppress macrophage ferroptosis via the SIRT1/NRF2 signaling axis to alleviate acute lung injury in sepsis. Int Immunopharmacol 2025; 146:113914. [PMID: 39732105 DOI: 10.1016/j.intimp.2024.113914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/08/2024] [Accepted: 12/18/2024] [Indexed: 12/30/2024]
Abstract
Acute lung injury being one of the earliest and most severe complications during sepsis and macrophages play a key role in this process. To investigate the regulatory effects and potential mechanisms of adipose mesenchymal stem cell derived-exosomes (ADSC-exo) on macrophages and septic mice, ADSCs-exo was administrated to both LPS-induced macrophage and cecal ligation and puncture (CLP) induced sepsis mice. ADSCs-exo was confirmed to inhibit M1 polarization of macrophages and to reduce excessive inflammation. The use of ADSCs-exo in CLP mice and in LPS-induced macrophages relieved oxidative stress, cellular damage, and acute lung injury. During this process, ADSCs-exo increased the nuclear translocation of Nrf2, significantly upregulating the activation of the antioxidant pathway Nrf2/HO-1. Concurrently, they enhanced the expression of SIRT1 in macrophages. Further SIRT1 interference experiments demonstrated that ADSCs-exo mitigated macrophage inflammatory responses and LPS-induced ferroptosis by upregulating SIRT1. In the LPS-induced macrophage model, after SIRT1 was interfered with, ADSCs-exo failed to upregulate the Nrf2/HO-1 signaling pathway, leading to enhanced ferroptosis. Finally, in a CLP sepsis mouse model with myeloid-specific SIRT1 knockout, ADSCs-exo was observed to reduce lung tissue injury, oxidative stress damage, and ferroptosis. Still, these beneficial effects were reversed due to the myeloid-specific knockout of SIRT1, while co-administration of a ferroptosis inhibitor rescued this situation, alleviating lung injury and significantly reducing tissue levels of oxidative stress. In conclusion, this study elucidated a novel potential therapeutic mechanism wherein ADSCs-exo upregulates the levels of SIRT1 in macrophages through a non-delivery approach.
Collapse
Affiliation(s)
- Xiaozhi Bai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi 710032, China
| | - Yang Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi 710032, China
| | - Jiaqi Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi 710032, China
| | - Kai Guo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi 710032, China
| | - Hao Guan
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
2
|
Fu Y, Wang Q, Wang D, Li Y. Dexmedetomidine Inhibits Ferroptosis to Alleviate Hypoxia/Reoxygenation-Induced Cardiomyocyte Injury by Regulating the HDAC2/FPN Pathway. Cardiovasc Drugs Ther 2025:10.1007/s10557-024-07664-z. [PMID: 39747742 DOI: 10.1007/s10557-024-07664-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 01/04/2025]
Abstract
PURPOSE Myocardial ischemia/reperfusion injury (MIRI) is closely associated with ferroptosis. Dexmedetomidine (Dex) has good therapeutic effects on MIRI. This study investigates whether dexmedetomidine (Dex) regulates ferroptosis during MIRI by affecting ferroportin1 (FPN) levels and elucidates the underlying mechanisms. METHODS A murine MIRI model was established using male C57BL/6 J mice subjected to 30 min of left anterior descending coronary artery ligation followed by 48 h of reperfusion. In vitro, cardiomyocyte hypoxia/reoxygenation (H/R) models were created with 16 h of hypoxia and 8 h of reoxygenation. Triphenyltetrazolium chloride (TTC) staining was employed to determine infarct size. The pathological changes in myocardial tissues were assessed using hematoxylin-eosin (HE) staining. Lipid reactive oxygen species (ROS) level was detected using BODIPY™ 581/591 C11, and ferrous iron (Fe2+) and malondialdehyde (MDA) levels were measured using the kits. Cardiomyocyte viability was examined using cell counting kit-8 (CCK8) assay. The histone H3 lysine 27 acetylation (H3K27Ac) level in the FPN promoter region was determined using DNA pulldown assay. Chromatin immunoprecipitation (ChIP) assay was used to investigate the relationship between histone deacetylase 2 (HDAC2) and FPN promoter. RESULTS Dex alleviated ferroptosis in cardiomyocytes by upregulating FPN levels, which mitigated H/R-induced oxidative damage. FPN knockdown abolished the protective effects of Dex, confirming its dependence on FPN expression. Additionally, HDAC2 knockdown alleviated I/R-induced myocardial injury and ferroptosis in mice. Moreover, H/R-induced HDAC2 upregulation transcriptionally inhibited FPN expression by reducing the H3K27Ac level in the FPN promoter region, but Dex therapy restored this impact via inhibition of HDAC2. As expected, HDAC2 overexpression partially reversed the inhibitory effect of Dex on H/R-mediated cardiomyocyte ferroptosis. CONCLUSION Dex alleviated H/R-mediated cardiomyocyte ferroptosis through regulating the HDAC2/FPN axis. Our findings lend theoretical support to the use of Dex in MIRI therapy.
Collapse
Affiliation(s)
- Yueqi Fu
- Jiamusi University, Jiamusi, 154002, Heilongjiang Province, China
| | - QingDong Wang
- Department of Anesthesiology, The First Affiliated Hospital of Jiamusi University, No.348 Dexiang Street, Xiangyang District, Jiamusi, 154002, Heilongjiang Province, China
| | - DongWei Wang
- Department of Anesthesiology, The First Affiliated Hospital of Jiamusi University, No.348 Dexiang Street, Xiangyang District, Jiamusi, 154002, Heilongjiang Province, China.
| | - Yicong Li
- Department of Anesthesiology, Hainan Hosiptal of Chinese PLA General Hospital, No.80 Jianglin Street, Haitang District, Sanya City, Hainan Province, China.
| |
Collapse
|
3
|
Liu Z, Liu Q, Zhang X, Li G. Dexmedetomidine Inhibits Ferroptosis by Regulating the SRY-Box Transcription Factor 9/Divalent Metal Transporter-1 Axis to Alleviate Cerebral Ischemia/Reperfusion Injury. Chem Biol Drug Des 2025; 105:e70022. [PMID: 39757387 DOI: 10.1111/cbdd.70022] [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: 03/20/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 01/07/2025]
Abstract
Cerebral ischemia/reperfusion injury (IRI) is pathologically associated with ferroptosis. Dexmedetomidine (Dex) exerts neuroprotective activity after cerebral IRI. Our work focused on probing the pharmacologic effect of Dex on ferroptosis during cerebral IRI and the mechanisms involved. Cerebral IRI models were established by oxygen-glucose deprivation/reoxygenation (OGD/R) and middle cerebral artery occlusion (MCAO). 2,3,5-Triphenyltetrazolium chloride (TTC) staining was utilized to detect cerebral infarct size and mNSS was performed to evaluate neurologic deficits. Brain pathologic changes were analyzed by HE staining. Lipid peroxidation level was detected by C11-BODIPY staining, and Fe2+ and MDA levels were measured using the kits. Cell vitality was examined by CCK-8 assay. Dual-luciferase reporter and ChIP assays were adopted to determine the interaction between SOX9 and DMT1 promoter. Dex ameliorated ferroptosis and neuronal death induced by MCAO and OGD/R. SOX9 upregulation abolished the inhibitory effect of Dex on OGD/R-induced ferroptosis and neuronal death in SH-SY5Y cells. Our further trials showed that SOX9 transcriptionally activated DMT1 expression. As expected, DMT1 overexpression prevented Dex-induced decrease in ferroptosis and neuronal death in OGD/R-treated SH-SY5Y cells. Dex inhibited ferroptosis to exert neuroprotection effects on cerebral IRI by inactivating the SOX9/DMT1 axis.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Qionghua Liu
- Department of Anesthesiology, Nanchang Hongdu Hospital of Traditional Chinese Medicine, Nanchang, Jiangxi Province, China
| | - Xuekang Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Gan Li
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
4
|
Luan X, Chen P, Miao L, Yuan X, Yu C, Di G. Ferroptosis in organ ischemia-reperfusion injuries: recent advancements and strategies. Mol Cell Biochem 2025; 480:19-41. [PMID: 38556592 DOI: 10.1007/s11010-024-04978-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/24/2024] [Indexed: 04/02/2024]
Abstract
Ferroptosis is a newly discovered type of regulated cell death participated in multiple diseases. Different from other classical cell death programs such as necrosis and apoptosis, ferroptosis involving iron-catalyzed lipid peroxidation is characterized by Fe2+ accumulation and mitochondria alterations. The phenomenon of oxidative stress following organ ischemia-reperfusion (I/R) has recently garnered attention for its connection to the onset of ferroptosis and subsequent reperfusion injuries. This article provides a comprehensive overview underlying the mechanisms of ferroptosis, with a further focus on the latest research progress regarding interference with ferroptotic pathways in organ I/R injuries, such as intestine, lung, heart, kidney, liver, and brain. Understanding the links between ferroptosis and I/R injury may inform potential therapeutic strategies and targeted agents.
Collapse
Affiliation(s)
- Xiaoyu Luan
- School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Peng Chen
- School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
- Institute of Stem Cell and Regenerative Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Longyu Miao
- School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Xinying Yuan
- School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Chaoqun Yu
- School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Guohu Di
- School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China.
- Institute of Stem Cell and Regenerative Medicine, School of Basic Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
5
|
Zheng ZL, Ma JW, Luo Y, Liang GJ, Lei SJ, Yan KJ, Meng HB, Liu XJ. Mechanism of dexmedetomidine protection against cisplatin induced acute kidney injury in rats. Ren Fail 2024; 46:2337287. [PMID: 38627212 PMCID: PMC11022910 DOI: 10.1080/0886022x.2024.2337287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 03/27/2024] [Indexed: 04/19/2024] Open
Abstract
OBJECTIVE This study explored the molecular mechanisms by which dexmedetomidine (Dex) alleviates cisplatin (CP)-induced acute kidney injury (AKI) in rats. METHODS CP-induced AKI models were established, and Dex was intraperitoneally injected at different concentrations into rats in the model groups. Subsequently, rats were assigned to the control, CP, CP + Dex 10 μg/kg, and CP + Dex 25 μg/kg groups. After weighing the kidneys of the rats, the kidney arterial resistive index was calculated, and CP-induced AKI was evaluated. In addition, four serum biochemical indices were measured: histopathological damage in rat kidneys was detected; levels of inflammatory factors, interleukin (IL)-1β, IL-18, IL-6, and tumor necrosis factor alpha, in kidney tissue homogenate of rats were assessed through enzyme-linked immunosorbent assay (ELISA); and levels of NLRP-3, caspase-1, cleaved caspase-1, gasdermin D (GSDMD), and GSDMD-N in kidney tissues of rats were determined via western blotting. RESULTS Dex treatment reduced nephromegaly and serum clinical marker upregulation caused by CP-induced AKI. In addition, hematoxylin and eosin staining revealed that Dex treatment relieved CP-induced kidney tissue injury in AKI rats. ELISA analyses demonstrated that Dex treatment reduced the upregulated levels of proinflammatory cytokines in the kidney tissue of AKI rats induced by CP, thereby alleviating kidney tissue injury. Western blotting indicated that Dex alleviated CP-induced AKI by inhibiting pyroptosis mediated by NLRP-3 and caspase-1. CONCLUSION Dex protected rats from CP-induced AKI, and the mechanism may be related to NLRP-3/Caspase-1-mediated pyroptosis.
Collapse
Affiliation(s)
- Zeng-lu Zheng
- Department of Anesthesiology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Jun-wei Ma
- Department of Nephrology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Yi Luo
- Department of Respiratory, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Gui-jin Liang
- Department of Anesthesiology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Shi-jie Lei
- Department of Proctology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Ke-jin Yan
- Department of Proctology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Hai-bing Meng
- Department of Anesthesiology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Xiu-juan Liu
- Department of Nephrology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| |
Collapse
|
6
|
Bai C, Xiao P, Chen Y, Chu F, Jiao Y, Fan J, Zhang Y, Liu J, Jiang J, Yu S. GPX4 Promoter Hypermethylation Induced by Ischemia/Reperfusion Injury Regulates Hepatocytic Ferroptosis. J Clin Transl Hepatol 2024; 12:917-929. [PMID: 39544244 PMCID: PMC11557362 DOI: 10.14218/jcth.2024.00135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 09/11/2024] [Accepted: 09/29/2024] [Indexed: 11/17/2024] Open
Abstract
Background and Aims Glutathione peroxidase 4 (GPX4) is a key factor in ferroptosis, which is involved in ischemia-reperfusion injury. However, little is known about its role in hepatic ischemia-reperfusion injury (HIRI). This study aimed to investigate the role of GPX4 methylation in ferroptosis during HIRI. Methods For the in vitro experiments, an oxygen and glucose deprivation cell model was established. For the in vivo experiments, an ischemia-reperfusion model was created by subjecting mice to simulated HIRI. Ferroptosis occurrence, GPX4 promoter methylation, and global methylation levels were then assessed. Results Ferroptosis was observed in oxygen and glucose deprivation, characterized by a significant decrease in cellular viability (P < 0.05), an increase in lipid peroxidation (P < 0.01), iron overload (P < 0.05), and down-regulation of GPX4 (P < 0.05). This ferroptosis was exacerbated by GPX4 knockdown (P < 0.01) and mitigated by exogenous glutathione (P < 0.01). Similarly, ferroptosis was evident in mice subjected to HIRI, with a down-regulation of GPX4 mRNA and protein expression (all P < 0.01), and an upregulation of acyl-CoA synthetase long-chain family member 4 mRNA and protein (all P < 0.01), as well as prostaglandin-endoperoxide synthase 2 mRNA and protein expression (all P < 0.05). Methylation levels increased, evidenced by upregulation of DNA methylation transferase expression (P < 0.05) and down-regulation of Ten-eleven translocation family demethylases (P < 0.01), along with an upregulation of GPX4 promoter methylation. Conclusions Ferroptosis may be the primary mode of cell death in hepatocytes following ischemia-reperfusion injury. The methylation of the GPX4 promoter and elevated levels of global hepatic methylation are involved in the regulation of ferroptosis.
Collapse
Affiliation(s)
| | | | - Yuting Chen
- Department of Anatomy, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| | - Fangfang Chu
- Department of Anatomy, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| | - Yue Jiao
- Department of Anatomy, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| | - Jiaqi Fan
- Department of Anatomy, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| | - Yuexia Zhang
- Department of Anatomy, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| | - Jiao Liu
- Department of Anatomy, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| | - Jiying Jiang
- Department of Anatomy, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| | - Shuna Yu
- Department of Anatomy, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| |
Collapse
|
7
|
Zheng Y, Sun J, Luo Z, Li Y, Huang Y. Emerging mechanisms of lipid peroxidation in regulated cell death and its physiological implications. Cell Death Dis 2024; 15:859. [PMID: 39587094 PMCID: PMC11589755 DOI: 10.1038/s41419-024-07244-x] [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: 08/09/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024]
Abstract
Regulated cell death (RCD) refers to the form of cell death that can be regulated by various biomacromolecules. Each cell death modalities have their distinct morphological changes and molecular mechanisms. However, intense evidences suggest that lipid peroxidation can be the common feature that initiates and propagates the cell death. Excessive lipid peroxidation alters the property of membrane and further damage the proteins and nucleic acids, which is implicated in various human pathologies. Here, we firstly review the classical chain process of lipid peroxidation, and further clarify the current understanding of the myriad roles and molecular mechanisms of lipid peroxidation in various RCD types. We also discuss how lipid peroxidation involves in diseases and how such intimate association between lipid peroxidation-driven cell death and diseases can be leveraged to develop rational therapeutic strategies.
Collapse
Affiliation(s)
- Yongxin Zheng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou, China
| | - Junlu Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Zhiting Luo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Yimin Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China.
- State Key Laboratory of Respiratory Diseases, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou, China.
| | - Yongbo Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China.
- State Key Laboratory of Respiratory Diseases, Guangzhou, China.
| |
Collapse
|
8
|
Wang J, Li Q, Huo Y, Liu X, Shi Y, Xie B. ASPP2 deficiency promotes the progression of metabolic dysfunction-associated steatohepatitis via ACSL4 upregulation. Sci Rep 2024; 14:29177. [PMID: 39587161 PMCID: PMC11589572 DOI: 10.1038/s41598-024-80415-0] [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: 07/24/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024] Open
Abstract
As a member of the p53-binding protein family, apoptosis-stimulating protein p53 2 (ASPP2) is closely related to autophagy and apoptosis. However, the mechanistic role of ASPP2 in the development of metabolic dysfunction-associated steatohepatitis (MASH) remains elusive. Therefore, we investigated the role and underlying mechanisms of ASPP2 in MASH progression in a mouse model of MASH and a cellular model of metabolic dysfunction-associated fatty liver disease. ASPP2 deficiency significantly promoted the inflammatory response, steatosis, and MASH progression in mice. Through transcriptomic analysis, increased ACSL4 expression was identified as a potential key factor. Further elucidation of the underlying mechanisms demonstrated that ASPP2 deficiency increased lipid accumulation and inhibited mitochondrial respiration capacity in HepG2 cells induced by oleic acid. However, silencing of ACSL4 reversed these effects. Thus, our study indicates that ASPP2 is an important regulator of MASH progression through ACSL4 upregulation, highlighting its potential as an alternative approach to MASH treatment.
Collapse
Affiliation(s)
- Jinming Wang
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Quanwei Li
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Yunfei Huo
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Xiaoni Liu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Ying Shi
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| | - Bangxiang Xie
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
9
|
Chen Y, Guan S, Liu M, Lang L, Peng H, Lu J. 1,3-Dichloro-2-propanol Induced Renal Cell Ferroptosis via the Circadian Clock Protein BMAL1 Targeting GPX4. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 39561408 DOI: 10.1021/acs.jafc.4c05676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
1,3-Dichloro-2-propanol (1,3-DCP), a representative chloropropyl alcohol contaminant in food, has shown toxic effects on the kidney. Ferroptosis is a newly identified cell death driven by iron-dependent lipid peroxidation that is associated with renal injury. However, the role of 1,3-DCP in ferroptosis in renal cells remains unclear. In this study, we found that ferroptosis was involved in a 1,3-DCP-induced renal injury. Mechanistically, we revealed that 1,3-DCP triggered ferroptosis by inhibiting GPX4 activity and disturbing iron homeostasis in NRK-52E cells. The circadian clock is crucial in modulating physiological cellular functions through the regulation of various downstream proteins. Furthermore, our findings also showed that 1,3-DCP triggered ferroptosis through interference with the circadian clock. The data showed that the expression of GPX4 was regulated by clock core protein BMAL1. 1,3-DCP interfered with GPX4 rhythmic expression through disordering BMAL1 and led to lipid peroxidation, ultimately inducing ferroptosis. In conclusion, our study uncovered that BMAL1 was responsible for controlling GPX4 to mediate 1,3-DCP-induced ferroptosis. The BMAL1/GPX4 axis may be a potentially novel pathway for ferroptosis. Our work may offer a fresh perspective for toxicological research examining the interactions between food pollutants, circadian clock, and ferroptosis.
Collapse
Affiliation(s)
- Yuelin Chen
- College of Food science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Shuang Guan
- College of Food science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Meitong Liu
- College of Food science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Lingxi Lang
- College of Food science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Huanhuan Peng
- College of Food science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Jing Lu
- College of Food science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| |
Collapse
|
10
|
Wu CYC, Zhang Y, Howard P, Huang F, Lee RHC. ACSL3 is a promising therapeutic target for alleviating anxiety and depression in Alzheimer's disease. GeroScience 2024:10.1007/s11357-024-01424-5. [PMID: 39532829 DOI: 10.1007/s11357-024-01424-5] [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: 08/15/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD), the leading cause of dementia, affects over 55 million people worldwide and is often accompanied by depression and anxiety. Both significantly impact patients' quality of life and impose substantial societal and economic burdens on healthcare systems. Identifying the complex regulatory mechanisms that contribute to the psychological and emotional deficits in AD will provide promising therapeutic targets. Biosynthesis of omega-3 (ω3) and omega-6 fatty acids (ω6-FA) through long-chain acyl-CoA synthetases (ACSL) is crucial for cell function and survival. This is due to ω3/6-FA's imperative role in modulating the plasma membrane, energy production, and inflammation. While ACSL dysfunction is known to cause heart, liver, and kidney diseases, the role of ACSL in pathological conditions in the central nervous system (e.g., depression and anxiety) remains largely unexplored. The impact of ACSLs on AD-related depression and anxiety was investigated in a mouse model of Alzheimer's disease (3xTg-AD). ACSL3 levels were significantly reduced in the hippocampus of aged 3xTg-AD mice (via capillary-based immunoassay). This reduction in ACAL3 was closely associated with increased depression and anxiety-like behavior (via forced swim, tail suspension, elevated plus maze, and sucrose preference test). Upregulation of ACSL3 via adenovirus in aged 3xTg-AD mice led to increased protein levels of brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor C (VEGF-C) (via brain histology, capillary-based immunoassay), resulting in alleviation of depression and anxiety symptoms. The present study highlights a novel neuroprotective role of ACSL3 in the brain. Targeting ACSL3 will offer an innovative approach for treating AD-related depression and anxiety.
Collapse
Affiliation(s)
- Celeste Yin-Chieh Wu
- Department of Neurology, Louisiana State University Health, LSU Health Sciences Center Shreveport, 1501 Kings Hwy, Shreveport, LA, 71103-3932, USA.
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA.
| | - Yulan Zhang
- Department of Neurology, Louisiana State University Health, LSU Health Sciences Center Shreveport, 1501 Kings Hwy, Shreveport, LA, 71103-3932, USA
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA
| | - Peyton Howard
- Department of Neurology, Louisiana State University Health, LSU Health Sciences Center Shreveport, 1501 Kings Hwy, Shreveport, LA, 71103-3932, USA
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA
| | - Fang Huang
- Department of Neurology, Louisiana State University Health, LSU Health Sciences Center Shreveport, 1501 Kings Hwy, Shreveport, LA, 71103-3932, USA
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA
| | - Reggie Hui-Chao Lee
- Department of Neurology, Louisiana State University Health, LSU Health Sciences Center Shreveport, 1501 Kings Hwy, Shreveport, LA, 71103-3932, USA
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Cellular Biology and Anatomy, Louisiana State University Health, Shreveport, LA, USA
| |
Collapse
|
11
|
Wu Q, Chen Q, Liang S, Nie J, Wang Y, Fan C, Liu Z, Zhang X. Dexmedetomidine alleviates intestinal ischemia/reperfusion injury by modulating intestinal neuron autophagy and mitochondrial homeostasis via Nupr1 regulation. Mol Med 2024; 30:203. [PMID: 39508252 PMCID: PMC11542338 DOI: 10.1186/s10020-024-00952-2] [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: 06/19/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Intestinal ischemia/reperfusion injury (I/R) is a common yet challenging-to-treat condition, presenting a significant clinical challenge. This study aims to investigate the protective mechanisms of Dexmedetomidine (Dex) against I/R injury, with a particular focus on its role in regulating autophagy activity in intestinal neurons and maintaining mitochondrial homeostasis. Experimental findings demonstrate that Dex can mitigate intestinal damage induced by I/R through the modulation of autophagy activity and mitochondrial function in intestinal neurons by suppressing the expression of Nupr1. This discovery sheds light on a new molecular mechanism underlying the potential efficacy of Dex in treating intestinal I/R injury, offering valuable insights for clinical therapy.
Collapse
Affiliation(s)
- Qiong Wu
- Department of Surgery and Anesthesia, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Qiuhong Chen
- Department of Surgery and Anesthesia, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Sisi Liang
- Department of Surgery and Anesthesia, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Jinping Nie
- Department of Surgery and Anesthesia, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Yingjie Wang
- Department of Surgery and Anesthesia, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Chenlu Fan
- Department of Surgery and Anesthesia, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Zhen Liu
- Department of Surgery and Anesthesia, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Xuekang Zhang
- Department of Surgery and Anesthesia, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwai Zhengjie, Donghu District, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
12
|
Zhang S, Feng X, Yang G, Tan H, Cheng X, Tang Q, Yang H, Zhao Y, Ding X, Li S, Dou X, Li J, Kang H, Li X, Ji Y, Hou Q, An Q, Fang H, Fan H. Dexmedetomidine ameliorates acute kidney injury by regulating mitochondrial dynamics via the α2-AR/SIRT1/PGC-1α pathway activation in rats. Mol Med 2024; 30:184. [PMID: 39455916 PMCID: PMC11505563 DOI: 10.1186/s10020-024-00964-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: 08/22/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Sepsis-associated acute kidney injury (AKI) is a serious complication of systemic infection with high morbidity and mortality in patients. However, no effective drugs are available for AKI treatment. Dexmedetomidine (DEX) is an alpha 2 adrenal receptor agonist with antioxidant and anti-apoptotic effects. This study aimed to investigate the therapeutic effects of DEX on sepsis-associated AKI and to elucidate the role of mitochondrial dynamics during this process. METHODS A lipopolysaccharide (LPS)-induced AKI rat model and an NRK-52E cell model were used in the study. This study investigated the effects of DEX on sepsis-associated AKI and the molecular mechanisms using histologic assessment, biochemical analyses, ultrastructural observation, western blotting, immunofluorescence, immunohistochemistry, qRT-PCR, flow cytometry, and si-mRNA transfection. RESULTS In rats, the results showed that administration of DEX protected kidney structure and function from LPS-induced septic AKI. In addition, we found that DEX upregulated the α2-AR/SIRT1/PGC-1α pathway, protected mitochondrial structure and function, and decreased oxidative stress and apoptosis compared to the LPS group. In NRK-52E cells, DEX regulated the mitochondrial dynamic balance by preventing intracellular Ca2+ overloading and activating CaMKII. CONCLUSIONS DEX ameliorated septic AKI by reducing oxidative stress and apoptosis in addition to modulating mitochondrial dynamics via upregulation of the α2-AR/SIRT1/PGC-1α pathway. This is a confirmatory study about DEX pre-treatment to ameliorate septic AKI. Our research reveals a novel mechanistic molecular pathway by which DEX provides nephroprotection.
Collapse
Affiliation(s)
- Shuai Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xiujing Feng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin, China
| | - Guiyan Yang
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Haoyang Tan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xin Cheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Qichao Tang
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin, China
| | - Haotian Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yuan Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xuanpan Ding
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Siyao Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xinyi Dou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Junfeng Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Huijie Kang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xingxing Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yaxin Ji
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Qingdian Hou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Qiuyue An
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hao Fang
- College of Optoelectronic Engineering, Chongqing University, Chongqing, China
| | - Honggang Fan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| |
Collapse
|
13
|
Luo RR, Yang J, Sun YL, Zhou BY, Zhou SX, Zhang GX, Yang AX. Dexmedetomidine attenuates ferroptosis by Keap1-Nrf2/HO-1 pathway in LPS-induced acute kidney injury. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7785-7796. [PMID: 38722344 DOI: 10.1007/s00210-024-03125-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 04/26/2024] [Indexed: 10/04/2024]
Abstract
Previous research has demonstrated that Dexmedetomidine (DEX), an α2 adrenergic agonist commonly used for its sedative and analgesic properties, can attenuate lipopolysaccharide (LPS)-induced acute kidney injury (AKI). This study explores the possibility that DEX's protective effects in LPS-induced AKI are mediated through the inhibition of ferroptosis, a form of regulated cell death characterized by iron-dependent lipid peroxidation, and the activation of the antioxidant response through the Keap1/Nrf2/HO-1 signaling pathway. We induced AKI in 42 mice using LPS and divided them into six groups: saline control, LPS, LPS + DEX, LPS + Ferrostatin-1 (LPS + Fer-1; a ferroptosis inhibitor), LPS + DEX with α2-receptor antagonist Altipamizole (LPS + DEX + ATI), and LPS + DEX with Nrf2 inhibitor ML385 (LPS + DEX + ML385). After 24 h, we analyzed blood and kidney tissues. LPS exposure resulted in AKI, with increased serum creatinine, BUN, and cystatin C, and tubular damage, which DEX and Fer-1 ameliorated. However, Altipamizole and ML385 negated these improvements. The LPS group exhibited elevated oxidative stress markers and mitochondrial damage, reduced by DEX and Fer-1, but not when α2-adrenergic or Nrf2 pathways were blocked. Nrf2 and HO-1 expression declined in the LPS group, rebounded with LPS + DEX and LPS + Fer-1, and fell again with inhibitors; inversely, Keap1 expression varied. Our results demonstrate that DEX may protect against LPS-induced AKI, at least partially by regulating ferroptosis and the α2-adrenergic receptor/Keap1/Nrf2/HO-1 pathway, suggesting a potential therapeutic role for DEX in AKI management by modulating cell death and antioxidant defenses.
Collapse
Affiliation(s)
- Rui-Rui Luo
- Department of Intensive Care Unit, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guang-Ji Road, Gusu District, Suzhou, 215008, China
- Suzhou Clinical Medical Center of Critical Care Medicine, Suzhou, 215001, China
| | - Jing Yang
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yan-Lin Sun
- Department of Intensive Care Unit, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guang-Ji Road, Gusu District, Suzhou, 215008, China
- Suzhou Clinical Medical Center of Critical Care Medicine, Suzhou, 215001, China
| | - Bi-Ying Zhou
- Department of Intensive Care Unit, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guang-Ji Road, Gusu District, Suzhou, 215008, China
- Suzhou Clinical Medical Center of Critical Care Medicine, Suzhou, 215001, China
| | - Si-Xuan Zhou
- Department of Intensive Care Unit, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guang-Ji Road, Gusu District, Suzhou, 215008, China
- Suzhou Clinical Medical Center of Critical Care Medicine, Suzhou, 215001, China
| | - Guo-Xing Zhang
- Department of Physiology and Neurosciences, Medical College of Soochow University, 199 Ren-Ai Road, Suzhou Industrial Park, Suzhou, 215123, China.
| | - Ai-Xiang Yang
- Department of Intensive Care Unit, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guang-Ji Road, Gusu District, Suzhou, 215008, China.
- Suzhou Clinical Medical Center of Critical Care Medicine, Suzhou, 215001, China.
| |
Collapse
|
14
|
Tan Y, Wang Q, Guo Y, Zhang N, Xu Y, Bai X, Liu J, Bi X. Dexmedetomidine mitigates lidocaine-induced spinal cord injury by repressing ferritinophagy-mediated ferroptosis by increasing CISD2 expression in rat models. J Bioenerg Biomembr 2024; 56:517-530. [PMID: 39168950 PMCID: PMC11455791 DOI: 10.1007/s10863-024-10034-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
Dexmedetomidine (DEX) has been confirmed to exert neuroprotective effects in various nerve injury models by regulating ferroptosis, including spinal cord injury (SCI). Although it has been established that CDGSH iron sulfur domain 2 (CISD2) can regulate ferroptosis, whether DEX can regulate ferroptosis by CISD2 in SCI remains unclear. Lidocaine was used to induce PC12 cells and stimulate rats to establish SCI models in vitro and in vivo. MTT assays were performed to analyze cell viability. Ferroptosis was assessed by determining the levels of cellular reactive axygen species (ROS), malondialdehyde (MDA), glutathione (GSH), and Fe2+. Ferritinophagy was analyzed by LysoTracker staining, FerroOrange staining, and immunofluorescence. Western blotting was carried out to quantify the levels of several proteins. Fluorescence microscopy was also used to observe cell autophagy. The morphology of mitochondria within the tissue was observed under transmission electron microscopy (TEM). DEX treatment weakened lidocaine-induced elevation of ROS, Fe2+, and MDA and reduced GSH in PC12 cells, indicating that DEX treatment weakened lidocaine-induced ferroptosis in PC12 cells. Similarly, lidocaine promoted autophagy, Fe2+, and microtubule-associated protein 1 light chain 3 (LC3) in PC12 cells and suppressed ferritin and p62 protein levels, indicating that DEX could weaken lidocaine-induced ferritinophagy in PC12 cells. DEX treatment improved the BBB score, reduced tissue damage, increased the number of neurons, and alleviated mitochondrial damage by inhibiting ferroptosis and ferritinophagy in lidocaine-induced SCI rat models. The decreased CISD2, ferritin heavy chain 1 (FTH1), solute carrier family 7-member 11-glutathione (SLC7A11), and glutathione peroxidase 4 (GPX4) protein levels and the elevated nuclear receptor coactivator 4 (NCOA4) protein levels in rat models in the lidocaine group were weakened by DEX treatment. Moreover, CISD2 inhibition reversed the inhibitory effects of DEX treatment on lidocaine-induced ferroptosis and ferritinophagy in PC12 cells significantly. Taken together, DEX treatment could impair lidocaine-induced SCI by inhibiting ferroptosis and ferritinophagy by upregulating CISD2 in rat models.
Collapse
Affiliation(s)
- Yonghong Tan
- Department of Anesthesiology, Liuzhou Hospital, Guangzhou Women and Children's Medical Center, No. 50 Boyuan Avenue, Yufeng District, Liuzhou, Guangxi, 545000, China
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Qiong Wang
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yubing Guo
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Na Zhang
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yingyi Xu
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xue Bai
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Jianhua Liu
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xiaobao Bi
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
| |
Collapse
|
15
|
Fan CH, Zeng XQ, Feng RM, Yi HW, Xia R. Comprehensive review of perioperative factors influencing ferroptosis. Biomed Pharmacother 2024; 179:117375. [PMID: 39278186 DOI: 10.1016/j.biopha.2024.117375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/18/2024] Open
Abstract
The perioperative period encompasses all phases of patient care from the decision to perform surgery until full recovery. Ferroptosis, a newly identified type of regulated cell death, influences a wide array of diseases, including those affecting the prognosis and regression of surgical patients, such as ischemia-reperfusion injury and perioperative cognitive dysfunction. This review systematically examines perioperative factors impacting ferroptosis such as surgical trauma-induced stress, tissue hypoxia, anesthetics, hypothermia, and blood transfusion. By analyzing their intrinsic relationships, we aim to improve intraoperative management, enhance perioperative safety, prevent complications, and support high-quality postoperative recovery, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Cheng-Hui Fan
- Department of Anaesthesiology, the First Affiliated Hospital of Yangtze University, Jingzhou 434000, PR China
| | - Xiao-Qin Zeng
- Department of Anaesthesiology, The Second People's Hospital of Jingzhou, Jingzhou 434020, PR China
| | - Rui-Min Feng
- Laboratory Department, the First Affiliated Hospital of Yangtze University, Jingzhou 434000, PR China
| | - Hua-Wei Yi
- Laboratory Department, the First Affiliated Hospital of Yangtze University, Jingzhou 434000, PR China.
| | - Rui Xia
- Department of Anaesthesiology, the First Affiliated Hospital of Yangtze University, Jingzhou 434000, PR China.
| |
Collapse
|
16
|
Tian L, Liu Q, Wang X, Chen S, Li Y. Fighting ferroptosis: Protective effects of dexmedetomidine on vital organ injuries. Life Sci 2024; 354:122949. [PMID: 39127318 DOI: 10.1016/j.lfs.2024.122949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Vital organ injury is one of the leading causes of global mortality and socio-economic burdens. Current treatments have limited efficacy, and new strategies are needed. Dexmedetomidine (DEX) is a highly selective α2-adrenergic receptor that protects multiple organs by reducing inflammation and preventing cell death. However, its exact mechanism is not yet fully understood. Understanding the underlying molecular mechanisms of its protective effects is crucial as it could provide a basis for designing highly targeted and more effective drugs. Ferroptosis is the primary mode of cell death during organ injury, and recent studies have shown that DEX can protect vital organs from this process. This review provides a detailed analysis of preclinical in vitro and in vivo studies and gains a better understanding of how DEX protects against vital organ injuries by inhibiting ferroptosis. Our findings suggest that DEX can potentially protect vital organs mainly by regulating iron metabolism and the antioxidant defense system. This is the first review that summarizes all evidence of ferroptosis's role in DEX's protective effects against vital organ injuries. Our work aims to provide new insights into organ therapy with DEX and accelerate its translation from the laboratory to clinical settings.
Collapse
Affiliation(s)
- Lei Tian
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Qian Liu
- Department of Anesthesiology, Zigong First People's Hospital, Zigong, China
| | - Xing Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Suheng Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Yulan Li
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
17
|
Cao S, Wei Y, Yue Y, Chen Y, Qian J, Wang D, Xiong A, Liu P, Zeng H. Rosiglitazone retards the progression of iron overload-induced osteoarthritis by impeding chondrocyte ferroptosis. iScience 2024; 27:110526. [PMID: 39224514 PMCID: PMC11366908 DOI: 10.1016/j.isci.2024.110526] [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: 04/13/2024] [Revised: 06/03/2024] [Accepted: 07/12/2024] [Indexed: 09/04/2024] Open
Abstract
Ferroptosis is implicated in several diseases, including iron overload-induced osteoarthritis (IOOA), which is marked by oxidative stress, iron imbalance, and lipid peroxidation. Given rosiglitazone's (RSG) ability to inhibit lipid peroxidation and ferroptosis, this study aims to assess its therapeutic potential for treating IOOA. Our in vitro results show that RSG targets acyl-CoA synthetase long-chain family member 4 to mitigate impairments induced by interleukin-1 beta and ferric ammonium citrate, including cell apoptosis, senescence, inflammatory responses, extracellular matrix degradation, and ferroptosis. RSG reduced intracellular iron content, alleviated oxidative stress and lipid peroxidation, mitigated damage to membrane-bound organelles, and enhanced glucose transport. Additionally, pre-treatment with RSG imparted anti-ferroptotic properties to chondrocytes. In vivo, RSG alleviated cartilage degradation, inflammatory responses, and ferroptosis in mice with IOOA. In conclusion, RSG exhibits chondroprotective and anti-ferroptotic effects by suppressing lipid peroxidation and restoring iron homeostasis, highlighting its potential for treating IOOA.
Collapse
Affiliation(s)
- Siyang Cao
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
| | - Yihao Wei
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
| | - Yaohang Yue
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
| | - Yingqi Chen
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
| | - Junyu Qian
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
| | - Deli Wang
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
| | - Ao Xiong
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
| | - Peng Liu
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
| | - Hui Zeng
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People's Republic of China
| |
Collapse
|
18
|
Mishra S, Shelke V, Gaikwad AB. Acyl-CoA Synthetase Long-Chain Isoenzymes in Kidney Diseases: Mechanistic Insights and Therapeutic Implications. Cell Biochem Funct 2024; 42:e4114. [PMID: 39210707 DOI: 10.1002/cbf.4114] [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: 07/14/2024] [Revised: 08/07/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Long-chain acyl-CoA synthetases (ACSLs) are pivotal enzymes in fatty acid metabolism, essential for maintaining cellular homeostasis and energy production. Recent research has uncovered their significant involvement in the pathophysiology of various kidney diseases, including acute kidney injury (AKI), chronic kidney disease (CKD), diabetic kidney disease (DKD), and renal cell carcinoma (RCC). While ACSL1, ACSL3, ACSL4, and ACSL5 have been extensively studied for their roles in processes such as ferroptosis, lipid peroxidation, renal fibrosis, epithelial-mesenchymal transition, and tumor progression, the role of ACSL6 in kidney diseases remain largely unexplored. Notably, these isoenzymes exhibit distinct functions in different kidney diseases. Therefore, to provide a comprehensive understanding of their involvement, this review highlights the molecular pathways influenced by ACSLs and their roles in modulating cell death, inflammation, and fibrosis during kidney disease progression. By examining these mechanisms in detail, this review underscores the potential of ACSLs as biomarkers and therapeutic targets, advocating for further research to elucidate the precise roles of individual ACSL isoenzymes in kidney disease progression. Understanding these mechanisms opens new avenues for developing targeted interventions and improving therapeutic outcomes for patients with kidney diseases.
Collapse
Affiliation(s)
- Swati Mishra
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, India
| | - Vishwadeep Shelke
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, India
| | - Anil Bhanudas Gaikwad
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, India
| |
Collapse
|
19
|
Yao X, Liu Y, Sui Y, Zheng M, Zhu L, Li Q, Irwin MG, Yang L, Zhan Q, Xiao J. Dexmedetomidine facilitates autophagic flux to promote liver regeneration by suppressing GSK3β activity in mouse partial hepatectomy. Biomed Pharmacother 2024; 177:117038. [PMID: 39002441 DOI: 10.1016/j.biopha.2024.117038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/16/2024] [Accepted: 06/25/2024] [Indexed: 07/15/2024] Open
Abstract
INTRODUCTION Dexmedetomidine (DEX), a highly selective α2-adrenergic receptor agonist, is widely used for sedation and anesthesia in patients undergoing hepatectomy. However, the effect of DEX on autophagic flux and liver regeneration remains unclear. OBJECTIVES This study aimed to determine the role of DEX in hepatocyte autophagic flux and liver regeneration after PHx. METHODS In mice, DEX was intraperitoneally injected 5 min before and 6 h after PHx. In vitro, DEX was co-incubated with culture medium for 24 h. Autophagic flux was detected by LC3-II and SQSTM1 expression levels in primary mouse hepatocytes and the proportion of red puncta in AML-12 cells transfected with FUGW-PK-hLC3 plasmid. Liver regeneration was assessed by cyclinD1 expression, Edu incorporation, H&E staining, ki67 immunostaining and liver/body ratios. Bafilomycin A1, si-GSK3β and Flag-tagged GSK3β, α2-ADR antagonist, GSK3β inhibitor, AKT inhibitor were used to identify the role of GSK3β in DEX-mediated autophagic flux and hepatocyte proliferation. RESULTS Pre- and post-operative DEX treatment promoted liver regeneration after PHx, showing 12 h earlier than in DEX-untreated mice, accompanied by facilitated autophagic flux, which was completely abolished by bafilomycin A1 or α2-ADR antagonist. The suppression of GSK3β activity by SB216763 and si-GSK3β enhanced the effect of DEX on autophagic flux and liver regeneration, which was abolished by AKT inhibitor. CONCLUSION Pre- and post-operative administration of DEX facilitates autophagic flux, leading to enhanced liver regeneration after partial hepatectomy through suppression of GSK3β activity in an α2-ADR-dependent manner.
Collapse
Affiliation(s)
- Xueya Yao
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China; Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China.
| | - Yingxiang Liu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China; Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China.
| | - Yongheng Sui
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China; Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China.
| | - Miao Zheng
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China; Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China.
| | - Ling Zhu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China; Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China.
| | - Quanfu Li
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| | | | - Liqun Yang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China; Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China.
| | - Qionghui Zhan
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China; Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China.
| | - Jie Xiao
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China; Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation, Shanghai, China.
| |
Collapse
|
20
|
Chu J, Wang K, Lu L, Zhao H, Hu J, Xiao W, Wu Q. Advances of Iron and Ferroptosis in Diabetic Kidney Disease. Kidney Int Rep 2024; 9:1972-1985. [PMID: 39081773 PMCID: PMC11284386 DOI: 10.1016/j.ekir.2024.04.012] [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: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 08/02/2024] Open
Abstract
Diabetes mellitus presents a significant threat to human health because it disrupts energy metabolism and gives rise to various complications, including diabetic kidney disease (DKD). Metabolic adaptations occurring in the kidney in response to diabetes contribute to the pathogenesis of DKD. Iron metabolism and ferroptosis, a recently defined form of cell death resulting from iron-dependent excessive accumulation of lipid peroxides, have emerged as crucial players in the progression of DKD. In this comprehensive review, we highlight the profound impact of adaptive and maladaptive responses regulating iron metabolism on the progression of kidney damage in diabetes. We summarize the current understanding of iron homeostasis and ferroptosis in DKD. Finally, we propose that precise manipulation of iron metabolism and ferroptosis may serve as potential strategies for kidney management in diabetes.
Collapse
Affiliation(s)
- Jiayi Chu
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| | - Kewu Wang
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| | - Lulu Lu
- Department of Nutrition and Toxicology, Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines of Zhejiang Province, School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Hui Zhao
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| | - Jibo Hu
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| | - Wenbo Xiao
- Department of Radiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China
| | - Qian Wu
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| |
Collapse
|
21
|
Yu Y, Zhang L, Zhang D, Dai Q, Hou M, Chen M, Gao F, Liu XL. The role of ferroptosis in acute kidney injury: mechanisms and potential therapeutic targets. Mol Cell Biochem 2024:10.1007/s11010-024-05056-3. [PMID: 38943027 DOI: 10.1007/s11010-024-05056-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024]
Abstract
Acute kidney injury (AKI) is one of the most common and severe clinical renal syndromes with high morbidity and mortality. Ferroptosis is a form of programmed cell death (PCD), is characterized by iron overload, reactive oxygen species accumulation, and lipid peroxidation. As ferroptosis has been increasingly studied in recent years, it is closely associated with the pathophysiological process of AKI and provides a target for the treatment of AKI. This review offers a comprehensive overview of the regulatory mechanisms of ferroptosis, summarizes its role in various AKI models, and explores its interaction with other forms of cell death, it also presents research on ferroptosis in AKI progression to other diseases. Additionally, the review highlights methods for detecting and assessing AKI through the lens of ferroptosis and describes potential inhibitors of ferroptosis for AKI treatment. Finally, the review presents a perspective on the future of clinical AKI treatment, aiming to stimulate further research on ferroptosis in AKI.
Collapse
Affiliation(s)
- Yanxin Yu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Lei Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Die Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Qiangfang Dai
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Mingzheng Hou
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Meini Chen
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Feng Gao
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Xiao-Long Liu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China.
| |
Collapse
|
22
|
张 方, 刘 刚. [Dexmedetomidine inhibits ferroptosis of human renal tubular epithelial cells by activating the Nrf2/HO-1/GPX4 pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1135-1140. [PMID: 38977343 PMCID: PMC11237299 DOI: 10.12122/j.issn.1673-4254.2024.06.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Indexed: 07/10/2024]
Abstract
OBJECTIVE To investigate the protective effect of dexmedetomidine (DEX) against erastin-induced ferroptosis in human renal tubular epithelial cells (HK-2 cells) and explore the underlying mechanism. METHODS HK-2 cells were treated with erastin alone or in combination with different concentrations (2.5, 5.0 and 10 μmol/L) of DEX, and the changes in cell viability were observed using CCK-8 assay. To explore the mechanism by which DEX inhibits erastin-induced ferroptosis, HK-2 cells were treated with erastin, erastin+10 μmol/L DEX, or erastin+10 μmol/L DEX+ML385 (a Nrf2 inhibitor), after which the cell viability was assessed. The level of intracellular Fe2+ was detected by cell ferrous iron colorimetric assay kit, and flow cytometry was performed to detect reactive oxygen species (ROS); MDA and reduced glutathione assay kits were used to detect the contents of MDA and GSH in the cells; The expressions of Nrf2, HO-1 and GPX4 proteins were detected by Western blotting. RESULTS Erastin treatment significantly inhibited the viability of the cells, decreased GSH content, and increased intracellular levels of Fe2+, ROS and MDA. The combined treatment with 10 μmol/L DEX markedly increased the viability of the cells, increased GSH content, reduced the levels of Fe2+, ROS and MDA, and upregulated the protein expressions of Nrf2, HO-1 and GPX4 in the cells. The application of ML385 obviously blocked the protective effect of DEX and caused significant inhibition of the Nrf2/HO-1/GPX4 pathway, decreased the cell viability and GSH content, and increased the levels of Fe2+, ROS and MDA in HK-2 cells. CONCLUSION The protective effect of DEX against erastin-induced ferroptosis of HK-2 cells is probably mediated by activation of the Nrf2/HO-1/GPX4 pathway to inhibit oxidative stress.
Collapse
|
23
|
Zhu M, Yuan Z, Wen C, Wei X. DEX Inhibits H/R-induced Cardiomyocyte Ferroptosis by the miR-141-3p/lncRNA TUG1 Axis. Thorac Cardiovasc Surg 2024. [PMID: 38889747 DOI: 10.1055/s-0044-1787691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
BACKGROUND Ferroptosis is emerging as a critical pathway in ischemia/reperfusion (I/R) injury, contributing to compromised cardiac function and predisposing individuals to sepsis and myocardial failure. The study investigates the underlying mechanism of dexmedetomidine (DEX) in hypoxia/reoxygenation (H/R)-induced ferroptosis in cardiomyocytes, aiming to identify novel targets for myocardial I/R injury treatment. METHODS H9C2 cells were subjected to H/R and treated with varying concentrations of DEX. Additionally, H9C2 cells were transfected with miR-141-3p inhibitor followed by H/R treatment. Levels of miR-141-3p, long noncoding RNA (lncRNA) taurine upregulated 1 (TUG1), Fe2+, glutathione (GSH), and malondialdehyde were assessed. Reactive oxygen species (ROS) generation was measured via fluorescent labeling. Expression of ferroptosis-related proteins glutathione peroxidase 4 (GPX4) and acyl-CoA synthetase long-chain family member 4 (ACSL4) was determined using Western blot. The interaction between miR-141-3p and lncRNA TUG1 was evaluated through RNA pull-down assay and dual-luciferase reporter gene assays. The stability of lncRNA TUG1 was assessed using actinomycin D. RESULTS DEX ameliorated H/R-induced cardiomyocyte injury and elevated miR-141-3p expression in cardiomyocytes. DEX treatment increased cell viability, Fe2+, and ROS levels while decreasing ACSL4 protein expression. Furthermore, DEX upregulated GSH and GPX4 protein levels. miR-141-3p targeted lncRNA TUG1, reducing its stability and overall expression. Inhibition of miR-141-3p or overexpression of lncRNA TUG1 partially reversed the inhibitory effect of DEX on H/R-induced ferroptosis in cardiomyocytes. CONCLUSION DEX mitigated H/R-induced ferroptosis in cardiomyocytes by upregulating miR-141-3p expression and downregulating lncRNA TUG1 expression, unveiling a potential therapeutic strategy for myocardial I/R injury.
Collapse
Affiliation(s)
- Mei Zhu
- Department of Anesthesiology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, People's Republic of China
| | - Zhiguo Yuan
- Department of Anesthesiology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, People's Republic of China
| | - Chuanyun Wen
- Department of Anesthesiology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, People's Republic of China
| | - Xiaojia Wei
- Department of Anesthesiology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou, People's Republic of China
| |
Collapse
|
24
|
Guo F, Lu Y, Du L, Guo X, Xie J, Cai X. ACLS4 could be a potential therapeutic target for severe acute pancreatitis. Sci Rep 2024; 14:13457. [PMID: 38862656 PMCID: PMC11166934 DOI: 10.1038/s41598-024-63898-9] [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: 02/26/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024] Open
Abstract
Acute pancreatitis (AP) is currently among the most prevalent digestive diseases. The pathogenesis of AP remains elusive, and there is no specific treatment. Therefore, identifying novel therapeutic targets is imperative for effective management and prevention of AP. In this study, we conducted a comprehensive transcriptomic analysis of peripheral blood from patients with AP and the pancreatic tissue from a mouse model of AP. Our analyses revealed that mouse model of AP exhibited a higher enrichment of mitogen-activated protein kinase signaling, endocytosis, apoptosis and tight junction pathways than the control. Subsequent weighted gene co-expression network analysis identified 15 gene modules, containing between 50 and 1000 genes each, which demonstrated significant correlations within samples from patients with AP. Further screening identified four genes (ACSL4, GALNT3, WSB1, and IL1R1) that were significantly upregulated in severe acute pancreatitis (SAP) in both human and mouse samples. In mouse models of SAP, ACSL4 was significantly upregulated in the pancreas, whereas GALNT3, WSB1, and IL1R1 were not. Lastly, we found that a commercially available ACSL4 inhibitor, PRGL493, markedly reduced IL-6 and TNFα expression, alleviated pancreatic edema and necrosis, and diminished the infiltration of inflammatory cells. In conclusion, this study comprehensively depicts the key genes and signaling pathways implicated in AP and suggests the potential of ACSL4 as a novel therapeutic target for SAP. These findings provide valuable insights for further exploration of therapeutic strategies for SAP.
Collapse
Affiliation(s)
- Feng Guo
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, People's Republic of China
| | - Yunkun Lu
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Lijun Du
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Xiuliu Guo
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, People's Republic of China
| | - Jinyan Xie
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, People's Republic of China.
| | - Xiujun Cai
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Hangzhou, People's Republic of China.
| |
Collapse
|
25
|
Zhuang K, Yang HT, Long YQ, Liu H, Ji FH, Peng K. Dexmedetomidine and acute kidney injury after non-cardiac surgery: A meta-analysis with trial sequential analysis. Anaesth Crit Care Pain Med 2024; 43:101359. [PMID: 38395357 DOI: 10.1016/j.accpm.2024.101359] [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: 11/05/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Acute kidney injury (AKI) is a common complication after surgery and is associated with detrimental outcomes. This systematic review and meta-analysis evaluated perioperative dexmedetomidine on AKI and renal function after non-cardiac surgery. METHODS PubMed, Embase, and Cochrane Library databases were searched until August 2023 for randomised trials comparing dexmedetomidine with normal saline on AKI and renal function in adults undergoing non-cardiac surgery. The primary outcome was the incidence of AKI (according to Kidney Disease Improving Global Outcomes or Acute Kidney Injury Network criteria). Meta-analysis was performed using a random-effect model. We conducted sensitivity analysis, trial sequential analysis (TSA), and Grading of Recommendations Assessment, Development and Evaluation level of evidence. RESULTS Twenty-three trials involving 2440 patients were included. Dexmedetomidine administration, as compared to normal saline, significantly reduced the incidence of AKI (7.4% vs. 13.2%; risk ratio = 0.57, 95% CI = 0.40-0.83, P = 0.003, I2 = 0%; a high level of evidence); TSA and sensitivity analyses suggested the robustness of this outcome. For the renal function and inflammation parameters, dexmedetomidine decreased serum creatinine, blood urea nitrogen, cystatin C, tumour necrosis factor-α, and interleukin-6, and increased urine output and estimated glomerular filtration rate. Additionally, dexmedetomidine reduced postoperative nausea and vomiting and length of hospital stay. Dexmedetomidine was associated with an increased rate of bradycardia, but not hypotension. CONCLUSION Dexmedetomidine administration reduced the incidence of AKI and improved renal function after non-cardiac surgery. Based on a high level of evidence, dexmedetomidine is recommended as a component of perioperative renoprotection. REGISTRATION International Prospective Register of Systematic Reviews; Registration number: CRD42022299252.
Collapse
Affiliation(s)
- Kai Zhuang
- Department of Anaesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Institute of Anaesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Hao-Tian Yang
- Department of Anaesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Institute of Anaesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Yu-Qin Long
- Department of Anaesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Institute of Anaesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Hong Liu
- Department of Anaesthesiology and Pain Medicine, University of California Davis Health, Sacramento, CA, USA
| | - Fu-Hai Ji
- Department of Anaesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Institute of Anaesthesiology, Soochow University, Suzhou, Jiangsu, China
| | - Ke Peng
- Department of Anaesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Institute of Anaesthesiology, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
26
|
Tan Q, Dong W, Wang Q, Gao L. Dexmedetomidine alleviates Hypoxia/reoxygenation-induced mitochondrial dysfunction in cardiomyocytes via activation of Sirt3/Prdx3 pathway. Daru 2024; 32:189-196. [PMID: 38407745 PMCID: PMC11087443 DOI: 10.1007/s40199-024-00504-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/18/2024] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Myocardial ischemia/reperfusion injury (MIRI) seriously threatens the health of people. The mitochondrial dysfunction in cardiomyocytes can promote the progression of MIRI. Dexmedetomidine (Dex) could alleviate the myocardial injury, which was known to reverse mitochondrial dysfunction in lung injury. However, the function of Dex in mitochondrial dysfunction during MIRI remains unclear. OBJECTIVE To assess the function of Dex in mitochondrial dysfunction during MIRI. METHODS To investigate the function of Dex in MIRI, H9C2 cells were placed in condition of hypoxia/reoxygenation (H/R). CCK8 assay was performed to test the cell viability, and the mitochondrial membrane potential was evaluated by JC-1 staining. In addition, the binding relationship between Sirt3 and Prdx3 was explored by Co-IP assay. Furthermore, the protein expressions were examined using western blot. RESULTS Dex could abolish H/R-induced mitochondrial dysfunction in H9C2 cells. In addition, H/R treatment significantly inhibited the expression of Sirt3, while Dex partially restored this phenomenon. Knockdown of Sirt3 or Prdx3 obviously reduced the protective effect of Dex on H/R-induced mitochondrial injury. Meanwhile, Sirt3 could enhance the function of Prdx3 via deacetylation of Prdx3. CONCLUSION Dex was found to attenuate H/R-induced mitochondrial dysfunction in cardiomyocytes via activation of Sirt3/Prdx3 pathway. Thus, this study might shed new lights on exploring new strategies for the treatment of MIRI.
Collapse
Affiliation(s)
- Qingyun Tan
- Department of Anesthesiology, The First Affiliated Hospital of Jiamusi University, No.348, Dexiang Street, Xiangyang District, Jiamusi, 154002, Heilongjiang Province, People's Republic of China
| | - Wenming Dong
- Department of Anesthesiology, Shenzhen Baoan Hospital of TCM, Shenzhen, 518133, Guangdong Province, People's Republic of China
| | - Qingdong Wang
- Department of Anesthesiology, The First Affiliated Hospital of Jiamusi University, No.348, Dexiang Street, Xiangyang District, Jiamusi, 154002, Heilongjiang Province, People's Republic of China.
| | - Li Gao
- Department of Anesthesiology, The First Affiliated Hospital of Jiamusi University, No.348, Dexiang Street, Xiangyang District, Jiamusi, 154002, Heilongjiang Province, People's Republic of China.
| |
Collapse
|
27
|
Wang J, Sun J, Yu H, Hu C, Wu J, Hu C. Donor Inhalation of Nebulized Dexmedetomidine Alleviates Ischemia-Reperfusion Injury in Rat Lung Transplantation. Pharmacology 2024; 109:293-304. [PMID: 38806015 DOI: 10.1159/000539528] [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: 10/22/2023] [Accepted: 05/23/2024] [Indexed: 05/30/2024]
Abstract
INTRODUCTION The occurrence of lung ischemia-reperfusion injury (LIRI) after lung transplantation results in primary graft dysfunction (PGD) in more than 50% of cases, which seriously affects the prognosis of recipients. Currently, donor lung protection is the focus of research on improving graft survival in lung transplant recipients. Dexmedetomidine (Dex) is a widely used general anesthesia adjuvant in clinical practice to alleviate ischemia-reperfusion injury in the lungs, liver, heart, kidneys, and brain. However, intravenous infusion of Dex can cause negative effects on the cardiovascular system. Inhaling nebulized Dex can directly act on the alveolar tissue and alleviate its cardiovascular inhibitory effect by reducing drug intake. This study aimed to investigate the effect of donor nebulized Dex inhalation on LIRI after lung transplantation in rats. METHODS We randomly divided the male Sprague-Dawley rats into donor rats and recipient rats, and allowed the donor rats to inhale nebulized Dex or physiological saline 15 min before surgery. The donor lung was refrigerated for 8 h before each single-lung transplant. After 2 h of reperfusion of the transplanted lung, serum and transplanted lung tissue were collected. The wet-to-dry weight ratio of the lung tissue was measured, arterial blood gas was detected, and histopathology changes, oxidative stress, inflammatory reactions, and apoptosis were evaluated. RESULTS Pretransplant inhalation of Dex through the donor's lung reduced the injury of the transplanted lung, increased the levels of malondialdehyde and myeloperoxidase, and decreased the levels of superoxide dismutase and glutathione in the lung tissue. Moreover, nebulized Dex inhalation of the donor lung inhibited LIRI-induced tumor necrosis factor-α, interleukin-6, and inducible nitric oxide synthase expression and also suppressed nuclear factor kappa B phosphorylation. Nebulized Dex inhalation reduced the rate of cell apoptosis in the transplanted lung tissue by inhibiting the upregulation of Bax, downregulation of Bcl-2, and increase in caspase-3 lysis caused by LIRI. CONCLUSION Inhalation of atomized Dex is a potential donor lung protection strategy, which can be used to reduce LIRI after lung transplantation and may be helpful to improve the occurrence of PGD and prognosis of lung transplant recipients.
Collapse
Affiliation(s)
- Jing Wang
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China,
- Department of Anesthesiology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China,
| | - Jiaojiao Sun
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Huizhi Yu
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Chunlan Hu
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Jinbo Wu
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Chunxiao Hu
- Department of Transplantation Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| |
Collapse
|
28
|
Li C, Yu Y, Zhu S, Hu Y, Ling X, Xu L, Zhang H, Guo K. The emerging role of regulated cell death in ischemia and reperfusion-induced acute kidney injury: current evidence and future perspectives. Cell Death Discov 2024; 10:216. [PMID: 38704372 PMCID: PMC11069531 DOI: 10.1038/s41420-024-01979-4] [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: 12/25/2023] [Revised: 04/14/2024] [Accepted: 04/18/2024] [Indexed: 05/06/2024] Open
Abstract
Renal ischemia‒reperfusion injury (IRI) is one of the main causes of acute kidney injury (AKI), which is a potentially life-threatening condition with a high mortality rate. IRI is a complex process involving multiple underlying mechanisms and pathways of cell injury and dysfunction. Additionally, various types of cell death have been linked to IRI, including necroptosis, apoptosis, pyroptosis, and ferroptosis. These processes operate differently and to varying degrees in different patients, but each plays a role in the various pathological conditions of AKI. Advances in understanding the underlying pathophysiology will lead to the development of new therapeutic approaches that hold promise for improving outcomes for patients with AKI. This review provides an overview of the recent research on the molecular mechanisms and pathways underlying IRI-AKI, with a focus on regulated cell death (RCD) forms such as necroptosis, pyroptosis, and ferroptosis. Overall, targeting RCD shows promise as a potential approach to treating IRI-AKI.
Collapse
Affiliation(s)
- Chenning Li
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Ying Yu
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Shuainan Zhu
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yan Hu
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Xiaomin Ling
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Liying Xu
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| | - Kefang Guo
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| |
Collapse
|
29
|
Wang LL, Kang ML, Liu CW, Liu L, Tang B. Panax notoginseng Saponins Activate Nuclear Factor Erythroid 2-Related Factor 2 to Inhibit Ferroptosis and Attenuate Inflammatory Injury in Cerebral Ischemia-Reperfusion. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:821-839. [PMID: 38699996 DOI: 10.1142/s0192415x24500332] [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: 05/05/2024]
Abstract
Panax notoginseng saponins (PNS), the primary medicinal ingredient of Panax notoginseng, mitigates cerebral ischemia-reperfusion injury (CIRI) by inhibiting inflammation, regulating oxidative stress, promoting angiogenesis, and improving microcirculation. Moreover, PNS activates nuclear factor erythroid 2-related factor 2 (Nrf2), which is known to inhibit ferroptosis and reduce inflammation in the rat brain. However, the molecular regulatory roles of PNS in CIRI-induced ferroptosis remain unclear. In this study, we aimed to investigate the effects of PNS on ferroptosis and inflammation in CIRI. We induced ferroptosis in SH-SY5Y cells via erastin stimulation and oxygen glucose deprivation/re-oxygenation (OGD/R) in vitro. Furthermore, we determined the effect of PNS treatment in a rat model of middle cerebral artery occlusion/reperfusion and assessed the underlying mechanism. We also analyzed the changes in the expression of ferroptosis-related proteins and inflammatory factors in the established rat model. OGD/R led to an increase in the levels of ferroptosis markers in SH-SY5Y cells, which were reduced by PNS treatment. In the rat model, combined treatment with an Nrf2 agonist, Nrf2 inhibitor, and PNS-Nrf2 inhibitor confirmed that PNS promotes Nrf2 nuclear localization and reduces ferroptosis and inflammatory responses, thereby mitigating brain injury. Mechanistically, PNS treatment facilitated Nrf2 activation, thereby regulating the expression of iron overload and lipid peroxidation-related proteins and the activities of anti-oxidant enzymes. This cascade inhibited ferroptosis and mitigated CIRI. Altogether, these results suggest that the ferroptosis-mediated activation of Nrf2 by PNS reduces inflammation and is a promising therapeutic approach for CIRI.
Collapse
Affiliation(s)
- Lin-Lin Wang
- Key Laboratory of Vascular Biology and Translational Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P. R. China
| | - Man-Lin Kang
- Key Laboratory of Vascular Biology and Translational Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P. R. China
| | - Can-Wen Liu
- Key Laboratory of Vascular Biology and Translational Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P. R. China
| | - Liang Liu
- People's Hospital of Ningxiang City, Hunan University of Chinese Medicine, Changsha, Hunan 410600, P. R. China
| | - Biao Tang
- Key Laboratory of Vascular Biology and Translational Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P. R. China
- National Key Laboratory Cultivation Base of Chinese Medicinal Powder & Innovative Medicinal Jointly Established by Province and Ministry, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P. R. China
- People's Hospital of Ningxiang City, Hunan University of Chinese Medicine, Changsha, Hunan 410600, P. R. China
| |
Collapse
|
30
|
Yang YY, Deng RR, Xiang DX. Naodesheng Pills Ameliorate Cerebral Ischemia Reperfusion-Induced Ferroptosis via Inhibition of the ERK1/2 Signaling Pathway. Drug Des Devel Ther 2024; 18:1499-1514. [PMID: 38716368 PMCID: PMC11074533 DOI: 10.2147/dddt.s443479] [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: 11/10/2023] [Accepted: 04/23/2024] [Indexed: 06/05/2024] Open
Abstract
Background Ferroptosis plays a crucial role in the occurrence and development of cerebral ischemia-reperfusion (I/R) injury and is regulated by mitogen-activated protein kinase 1/2 (ERK1/2). In China, Naodesheng Pills (NDSP) are prescribed to prevent and treat cerebrosclerosis and stroke. However, the protective effects and mechanism of action of NDSP against cerebral I/R-induced ferroptosis remain unclear. We investigated whether NDSP exerts its protective effects against I/R injury by regulating ferroptosis and aimed to elucidate the underlying mechanisms. Methods The efficacy of NDSP was evaluated using a Sprague-Dawley rat model of middle cerebral artery occlusion and an in vitro oxygen-glucose deprivation/reoxygenation (OGD/R) model. Brain injury was assessed using 2,3,5-triphenyltetrazolium chloride (TTC), hematoxylin and eosin staining, Nissl staining, and neurological scoring. Western blotting was performed to determine the expression levels of glutathione peroxidase 4 (GPX4), divalent metal-ion transporter-1 (DMT1), solute carrier family 7 member 11 (SLC7A11), and transferrin receptor 1 (TFR1). Iron levels, oxidative stress, and mitochondrial morphology were also evaluated. Network pharmacology was used to assess the associated mechanisms. Results NDSP (1.08 g/kg) significantly improved cerebral infarct area, cerebral water content, neurological scores, and cerebral tissue damage. Furthermore, NDSP inhibited I/R- and OGD/R-induced ferroptosis, as evidenced by the increased protein expression of GPX4 and SLC7A11, suppression of TFR1 and DMT1, and an overall reduction in oxidative stress and Fe2+ levels. The protective effects of NDSP in vitro were abolished by the GPX4 inhibitor RSL3. Network pharmacology analysis revealed that ERK1/2 was the core target gene and that NDSP reduced the amount of phosphorylated ERK1/2. Conclusion NDSP exerts its protective effects against I/R by inhibiting cerebral I/R-induced ferroptosis, and this mechanism is associated with the regulation of ferroptosis via the ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Yong-Yu Yang
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Provincial Engineering Research Central of Translational Medical and Innovative Drug, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
| | - Rong-Rong Deng
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, People’s Republic of China
| | - Da-Xiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Provincial Engineering Research Central of Translational Medical and Innovative Drug, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
31
|
Wang H, Li Y, Liu X, Wu Y. Identification and validation of ferroptosis-related gene SLC2A1 as a novel prognostic biomarker in AKI. Aging (Albany NY) 2024; 16:5634-5650. [PMID: 38517368 PMCID: PMC11006501 DOI: 10.18632/aging.205669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Emerging evidence reveals the key role of ferroptosis in the pathophysiological process of acute kidney injury (AKI). Our study aimed to investigate the potential ferroptosis-related gene in AKI through bioinformatics and experimental validation. METHODS The AKI single-cell sequencing dataset was retrieved from the GEO database and ferroptosis-related genes were extracted from the GENECARD website. The potential differentially expressed ferroptosis-related genes of AKI were selected. Functional enrichment analysis was performed. Machine learning algorithms were used to identify key ferroptosis-related genes associated with AKI. A multi-factor Cox regression analysis was used to construct a risk score model. The accuracy of the risk score model was validated using receiver operating characteristic (ROC) curve analysis. We extensively explored the immune landscape of AKI using CIBERSORT tool. Finally, expressions of ferroptosis DEGs were validated in vivo and in vitro by Western blot, ICH and transfection experiments. RESULTS Three hub genes (BAP1, MDM4, SLC2A1) were identified and validated by constructing drug regulatory network and subsequent screening using experimentally determined interactions. The risk mode showed the low-risk group had significantly better prognosis compared to high-risk group. The risk score was independently associated with overall survival. The ROC curve analysis showed that the prognosis model had good predictive ability. Additionally, CIBERSORT immune infiltration analysis suggest that the hub gene may influence cell recruitment and infiltration in AKI. Validation experiments revealed that SLC2A1 functions by regulating ferroptosis. CONCLUSIONS In summary, our study not only identifies SLC2A1 as diagnostic biomarker for AKI, but also sheds light on the role of it in AKI progression, providing novel insights for the clinical diagnosis and treatment of AKI.
Collapse
Affiliation(s)
- Huaying Wang
- Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, PR China
| | - Yuanyuan Li
- Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, PR China
| | - Xinran Liu
- Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, PR China
| | - Yonggui Wu
- Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, PR China
- Center for Scientific Research of Anhui Medical University, Hefei, Anhui 230022, PR China
| |
Collapse
|
32
|
Ni C, Xu W, Mu B, Li H, Geng J, Qu Y, Tian Y, Yu J, Tian N, Wang X, Chen C, Jin X, Zheng H. The feasibility of dexmedetomidine-led anesthesia maintenance strategy during major abdominal surgery. Heliyon 2024; 10:e26983. [PMID: 38444477 PMCID: PMC10912630 DOI: 10.1016/j.heliyon.2024.e26983] [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: 02/22/2024] [Accepted: 02/22/2024] [Indexed: 03/07/2024] Open
Abstract
Background Dexmedetomidine is known for its selective action on α2-adrenoceptor sites and is recognized for its neuroprotective capabilities. It can improve postoperative cognitive function. Commonly used anesthetics, such as sevoflurane and propofol, have been reported to affect postoperative cognitive function. Therefore, it could be valuable to explore dexmedetomidine-led anesthesia strategy. This study was designed to assess the performance, safety, and effective infusion rate in anesthesia maintenance, to explore a feasible dexmedetomidine-led anesthesia maintenance protocol, and to provide a foundation for potential combined anesthesia. Methods Thirty patients aged 18-60 years, classified as ASA I or II, undergoing abdominal surgery were involved. The anesthesia maintenance was achieved with dexmedetomidine, remifentanil and rocuronium. Dixon up-and-down sequential methodology was utilized to ascertain the ED50 of dexmedetomidine for maintaining Patient State Index (PSI) 25-40 (depth of stage III anesthesia). Intraoperative HR, BP and depth of anesthesia were monitored and controlled. The wake-up time from anesthesia, the incidence of intraoperative awareness and postoperative delirium, and the patients' satisfaction were assessed. Results The results indicated that dexmedetomidine-led anesthesia could maintain the depth of stage III anesthesia during abdominal surgery. The ED50 and ED95 of dexmedetomidine infusion rates during anesthesia maintenance were 2.298 μg/kg·h (95%CI: 2.190-2.404 μg/kg·h) and 3.765 μg/kg·h (95%CI: 3.550-4.050 μg/kg·h). Continuous infusion of dexmedetomidine and 0.1-0.3 μg/kg·min remifentanil could maintain PSI 25-40, and provide appropriate anesthesia depth for abdominal surgery. Perioperative bradycardia and hypertension could be rapidly corrected with atropine and nitroglycerin. The median wake-up time after anesthesia was 4.8 min, the perioperative maximum HR had significant correlation with wake-up time and intraoperative dexmedetomidine dose. No intraoperative awareness and postoperative delirium occurred; the patients were satisfied with dexmedetomidine-led anesthesia. Conclusions dexmedetomidine-led strategy could maintain stable depth of anesthesia throughout surgery, and the ED50 of dexmedetomidine infusion rates was 2.298 μg/kg·h. Intraoperative HR, BP and depth of anesthesia require monitoring, the bradycardia and hypertension could be rapidly corrected.
Collapse
Affiliation(s)
- Cheng Ni
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Wenjie Xu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Bing Mu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Hongyi Li
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Jiao Geng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Yinyin Qu
- Department of Anesthesiology, Peking University Third Hospital, Beijing, PR China
| | - Yi Tian
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Jie Yu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Naiyuan Tian
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Xiaoxiao Wang
- Clinical Epidemiology Research Center, Peking University Third Hospital, Beijing, PR China
| | - Chan Chen
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, PR China
| | - Xu Jin
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| |
Collapse
|
33
|
Zhang CH, Yan YJ, Luo Q. The molecular mechanisms and potential drug targets of ferroptosis in myocardial ischemia-reperfusion injury. Life Sci 2024; 340:122439. [PMID: 38278348 DOI: 10.1016/j.lfs.2024.122439] [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: 11/23/2023] [Revised: 01/07/2024] [Accepted: 01/14/2024] [Indexed: 01/28/2024]
Abstract
Myocardial ischemia-reperfusion injury (MIRI), caused by the initial interruption and subsequent restoration of coronary artery blood, results in further damage to cardiac function, affecting the prognosis of patients with acute myocardial infarction. Ferroptosis is an iron-dependent, superoxide-driven, non-apoptotic form of regulated cell death that is involved in the pathogenesis of MIRI. Ferroptosis is characterized by the accumulation of lipid peroxides (LOOH) and redox disequilibrium. Free iron ions can induce lipid oxidative stress as a substrate of the Fenton reaction and lipoxygenase (LOX) and participate in the inactivation of a variety of lipid antioxidants including CoQ10 and GPX4, destroying the redox balance and causing cell death. The metabolism of amino acid, iron, and lipids, including associated pathways, is considered as a specific hallmark of ferroptosis. This review systematically summarizes the latest research progress on the mechanisms of ferroptosis and discusses and analyzes the therapeutic approaches targeting ferroptosis to alleviate MIRI.
Collapse
Affiliation(s)
- Chen-Hua Zhang
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Yu-Jie Yan
- School of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Qi Luo
- School of Basic Medical Science, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
34
|
Gao X, Wu Y. Perioperative acute kidney injury: The renoprotective effect and mechanism of dexmedetomidine. Biochem Biophys Res Commun 2024; 695:149402. [PMID: 38159412 DOI: 10.1016/j.bbrc.2023.149402] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/10/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
Dexmedetomidine (DEX) is a highly selective and potent α2-adrenoceptor (α2-AR) agonist that is widely used as a clinical anesthetic to induce anxiolytic, sedative, and analgesic effects. In recent years, a growing body of evidence has demonstrated that DEX protects against acute kidney injury (AKI) caused by sepsis, drugs, surgery, and ischemia-reperfusion (I/R) in organs or tissues, indicating its potential role in the prevention and treatment of AKI. In this review, we summarized the evidence of the renoprotective effects of DEX on different models of AKI and explored the mechanism. We found that the renoprotective effects of DEX mainly involved antisympathetic effects, reducing inflammatory reactions and oxidative stress, reducing apoptosis, increasing autophagy, reducing ferroptosis, protecting renal tubular epithelial cells (RTECs), and inhibiting renal fibrosis. Thus, the use of DEX is a promising strategy for the management and treatment of perioperative AKI. The aim of this review is to further clarify the renoprotective mechanism of DEX to provide a theoretical basis for its use in basic research in various AKI models, clinical management, and the treatment of perioperative AKI.
Collapse
Affiliation(s)
- Xiong Gao
- Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Yaohua Wu
- Department of Anesthesiology, Huanggang Central Hospital, Huanggang, Hube, China.
| |
Collapse
|
35
|
Hu Y, Li Q, Wang Y. Serum ACSL4 levels in patients with ST-segment elevation myocardial infarction (STEMI) and its association with one-year major adverse cardiovascular events (MACE): A prospective cohort study. Medicine (Baltimore) 2024; 103:e36870. [PMID: 38215103 PMCID: PMC10783377 DOI: 10.1097/md.0000000000036870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/15/2023] [Indexed: 01/14/2024] Open
Abstract
In the present prospective cohort research, we aimed to explore the serum levels of Acyl-CoA synthetase long-chain family member 4 (ACSL4) in patients with ST-segment elevation myocardial infarction (STEMI) and its association with 1-year major adverse cardiovascular events (MACE). This prospective cohort study recruited 507 patients who underwent percutaneous coronary intervention for the treatment of STEMI at our hospital during August 2019 to July 2022. The serum ACSL4, tumor necrosis factor-α, interleukin (IL)-6, IL-1β, and C-reactive protein levels were measured by enzyme-linked immunosorbent assay. Demographic and clinical statistics were also collected. In addition, all patients were followed up for 1 year, and patients with MACE were defined as poor prognosis group. All data used SPSS 26.0 to statistical analyses. The poor prognosis group had significantly higher age and low-density leptin cholesterol (LDLC) levels compared to the favorable prognosis group (P < .05). STEMI patients exhibited significantly elevated serum levels of ACSL4, tumor necrosis factor-α, IL-6, IL-1β, and C-reactive protein (P < .05). Serum ACSL4 and IL-1β levels in the poor prognosis group were remarkably enhanced compared to the favorable prognosis group. Curvilinear regression analysis demonstrated that ACSL4 was associated with LDLC and IL-1β. Moreover, ACSL4 (B = 0.138, 95% CI 1.108-1.189, P < .001), LDLC (B = 2.317, 95% CI 5.253-19.603, P < .001), and IL-1β (B = 0.061, 95%CI 1.008-1.122, P = .025) levels were the risk factors for STEMI patients with 1-year MACE. This study showed that the serum ACSL4 levels was remarkably elevated in STEMI patients. This study might provide new targets and a comprehensive approach to cardiovascular protection in STEMI patients.
Collapse
Affiliation(s)
- Yun Hu
- Emergency Department, Wuhan Dongxihu District People’s Hospital, Wuhan, China
| | - Qingye Li
- Emergency Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Yinglin Wang
- Emergency Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
36
|
Wang B, Yang LN, Yang LT, Liang Y, Guo F, Fu P, Ma L. Fisetin ameliorates fibrotic kidney disease in mice via inhibiting ACSL4-mediated tubular ferroptosis. Acta Pharmacol Sin 2024; 45:150-165. [PMID: 37696989 PMCID: PMC10770410 DOI: 10.1038/s41401-023-01156-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 08/23/2023] [Indexed: 09/13/2023] Open
Abstract
Kidney fibrosis is the hallmark of chronic kidney disease (CKD) progression, whereas no effective anti-fibrotic therapies exist. Recent evidence has shown that tubular ferroptosis contributes to the pathogenesis of CKD with persistent proinflammatory and profibrotic responses. We previously reported that natural flavonol fisetin alleviated septic acute kidney injury and protected against hyperuricemic nephropathy in mice. In this study, we investigated the therapeutic effects of fisetin against fibrotic kidney disease and the underlying mechanisms. We established adenine diet-induced and unilateral ureteral obstruction (UUO)-induced CKD models in adult male mice. The two types of mice were administered fisetin (50 or 100 mg·kg-1·d-1, i.g.) for 3 weeks or 7 days, respectively. At the end of the experiments, the mice were euthanized, and blood and kidneys were gathered for analyzes. We showed that fisetin administration significantly ameliorated tubular injury, inflammation, and tubulointerstitial fibrosis in the two types of CKD mice. In mouse renal tubular epithelial (TCMK-1) cells, treatment with fisetin (20 μM) significantly suppressed adenine- or TGF-β1-induced inflammatory responses and fibrogenesis, and improved cell viability. By quantitative real-time PCR analysis of ferroptosis-related genes, we demonstrated that fisetin treatment inhibited ferroptosis in the kidneys of CKD mice as well as in injured TCMK-1 cells, as evidenced by decreased ACSL4, COX2, and HMGB1, and increased GPX4. Fisetin treatment effectively restored ultrastructural abnormalities of mitochondrial morphology and restored the elevated iron, the reduced GSH and GSH/GSSG as well as the increased lipid peroxide MDA in the kidneys of CKD mice. Notably, abnormally high expression of the ferroptosis key marker ACSL4 was verified in the renal tubules of CKD patients (IgAN, MN, FSGS, LN, and DN) as well as adenine- or UUO-induced CKD mice, and in injured TCMK-1 cells. In adenine- and TGF-β1-treated TCMK-1 cells, ACSL4 knockdown inhibited tubular ferroptosis, while ACSL4 overexpression blocked the anti-ferroptotic effect of fisetin and reversed the cytoprotective, anti-inflammatory, and anti-fibrotic effects of fisetin. In summary, we reveal a novel aspect of the nephroprotective effect of fisetin, i.e. inhibiting ACSL4-mediated tubular ferroptosis against fibrotic kidney diseases.
Collapse
Affiliation(s)
- Bo Wang
- Kidney Research Institute, Division of Nephrology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Li-Na Yang
- Kidney Research Institute, Division of Nephrology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Le-Tian Yang
- Kidney Research Institute, Division of Nephrology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yan Liang
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fan Guo
- Kidney Research Institute, Division of Nephrology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Ping Fu
- Kidney Research Institute, Division of Nephrology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Liang Ma
- Kidney Research Institute, Division of Nephrology, West China Hospital of Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
37
|
Guo S, Jia D, Liu X, Gao L, Wang H, Chen C, Wu Y. The positive efficacy of dexmedetomidine on the clinical outcomes of patients undergoing renal transplantation: evidence from meta-analysis. Aging (Albany NY) 2023; 15:14192-14209. [PMID: 38085644 PMCID: PMC10756127 DOI: 10.18632/aging.205296] [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: 06/15/2023] [Accepted: 11/02/2023] [Indexed: 12/21/2023]
Abstract
INTRODUCTION Whether dexmedetomidine (DEX), an anesthetic adjuvant, can improve renal transplant outcomes is not clear. METHODS We systematically identified clinical trials in which DEX was administered in renal transplantation (RT). On November 1, 2022, we searched The Cochrane Library, MEDLINE, EMBASE and https://www. CLINICALTRIALS gov/. The main outcomes were delayed graft function and acute rejection. RESULTS A total of seven studies were included in the meta-analysis. The results showed that compared with the control, DEX significantly reduced the occurrence of delayed graft function (RR 0.76; 95% CI 0.60-0.98), short-term serum creatinine [postoperative day (POD) 2: (MD -22.82; 95% CI -42.01 - -3.64)] and blood urea nitrogen [POD 2: (MD -2.90; 95% CI -5.10 - -0.70); POD 3: (MD 2.07; 95% CI -4.12 - -0.02)] levels, postoperative morphine consumption (MD -4.27; 95% CI -5.92 - -2.61) and the length of hospital stay (MD -0.85; 95% CI-1.47 - -0.23). However, DEX did not reduce the risk of postoperative acute rejection (RR 0.75; 95% CI 0.45-1.23). The results of the subgroup analysis showed that country type, donor type, and average age had a certain impact on the role of DEX. CONCLUSIONS DEX may improve the short-term clinical outcome of RT and shorten the length of hospital stay of patients.
Collapse
Affiliation(s)
- Shanshan Guo
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, PR China
| | - Degong Jia
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Xueqi Liu
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, PR China
| | - Li Gao
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, PR China
| | - Huaying Wang
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, PR China
| | - Chaoyi Chen
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, PR China
| | - Yonggui Wu
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, PR China
- Center for Scientific Research of Anhui Medical University, Hefei, Anhui 230022, PR China
| |
Collapse
|
38
|
Wang Y, Hu J, Wu S, Fleishman JS, Li Y, Xu Y, Zou W, Wang J, Feng Y, Chen J, Wang H. Targeting epigenetic and posttranslational modifications regulating ferroptosis for the treatment of diseases. Signal Transduct Target Ther 2023; 8:449. [PMID: 38072908 PMCID: PMC10711040 DOI: 10.1038/s41392-023-01720-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/16/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
Ferroptosis, a unique modality of cell death with mechanistic and morphological differences from other cell death modes, plays a pivotal role in regulating tumorigenesis and offers a new opportunity for modulating anticancer drug resistance. Aberrant epigenetic modifications and posttranslational modifications (PTMs) promote anticancer drug resistance, cancer progression, and metastasis. Accumulating studies indicate that epigenetic modifications can transcriptionally and translationally determine cancer cell vulnerability to ferroptosis and that ferroptosis functions as a driver in nervous system diseases (NSDs), cardiovascular diseases (CVDs), liver diseases, lung diseases, and kidney diseases. In this review, we first summarize the core molecular mechanisms of ferroptosis. Then, the roles of epigenetic processes, including histone PTMs, DNA methylation, and noncoding RNA regulation and PTMs, such as phosphorylation, ubiquitination, SUMOylation, acetylation, methylation, and ADP-ribosylation, are concisely discussed. The roles of epigenetic modifications and PTMs in ferroptosis regulation in the genesis of diseases, including cancers, NSD, CVDs, liver diseases, lung diseases, and kidney diseases, as well as the application of epigenetic and PTM modulators in the therapy of these diseases, are then discussed in detail. Elucidating the mechanisms of ferroptosis regulation mediated by epigenetic modifications and PTMs in cancer and other diseases will facilitate the development of promising combination therapeutic regimens containing epigenetic or PTM-targeting agents and ferroptosis inducers that can be used to overcome chemotherapeutic resistance in cancer and could be used to prevent other diseases. In addition, these mechanisms highlight potential therapeutic approaches to overcome chemoresistance in cancer or halt the genesis of other diseases.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Jing Hu
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300060, PR China
| | - Shuang Wu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, 430000, PR China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Yulin Li
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Yinshi Xu
- Department of Outpatient, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Wailong Zou
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Jinhua Wang
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| | - Yukuan Feng
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, PR China.
| | - Jichao Chen
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China.
| | - Hongquan Wang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, PR China.
| |
Collapse
|
39
|
Qiao O, Wang X, Wang Y, Li N, Gong Y. Ferroptosis in acute kidney injury following crush syndrome: A novel target for treatment. J Adv Res 2023; 54:211-222. [PMID: 36702249 PMCID: PMC10703611 DOI: 10.1016/j.jare.2023.01.016] [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: 11/20/2022] [Revised: 12/29/2022] [Accepted: 01/16/2023] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Crush syndrome (CS) is a kind of traumatic and ischemic injury that seriously threatens life after prolonged compression. It is characterized by systemic inflammatory reaction, myoglobinuria, hyperkalemia and acute kidney injury (AKI). Especially AKI, it is the leading cause of death from CS. There are various cell death forms in AKI, among which ferroptosis is a typical form of cell death. However, the role of ferroptosis has not been fully revealed in CS-AKI. AIM OF REVIEW This review aimed to summarize the evidence of ferroptosis in CS-AKI and its related molecular mechanism, discuss the therapeutic significance of ferroptosis in CS-AKI, and open up new ideas for the treatment of CS-AKI. KEY SCIENTIFIC CONCEPTS OF REVIEW One of the main pathological manifestations of CS-AKI is renal tubular epithelial cell dysfunction and cell death, which has been attributed to massive deposition of myoglobin. Large amounts of myoglobin released from damaged muscle deposited in the renal tubules, impeding the normal renal tubules function and directly damaging the tubules with oxidative stress and elevated iron levels. Lipid peroxidation damage and iron overload are the distinguishing features of ferroptosis. Moreover, high levels of pro-inflammatory cytokines and damage-associated molecule pattern molecules (HMGB1, double-strand DNA, and macrophage extracellular trap) in renal tissue have been shown to promote ferroptosis. However, how ferroptosis occurs in CS-AKI and whether it can be a therapeutic target remains unclear. In our current work, we systematically reviewed the occurrence and underlying mechanism of ferroptosis in CS-AKI.
Collapse
Affiliation(s)
- Ou Qiao
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Xinyue Wang
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Yuru Wang
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Ning Li
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China.
| | - Yanhua Gong
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China.
| |
Collapse
|
40
|
Ouyang ZQ, Shao LS, Wang WP, Ke TF, Chen D, Zheng GR, Duan XR, Chu JX, Zhu Y, Yang L, Shan HY, Huang L, Liao CD. Low intensity pulsed ultrasound ameliorates Adriamycin-induced chronic renal injury by inhibiting ferroptosis. Redox Rep 2023; 28:2251237. [PMID: 37652897 PMCID: PMC10472869 DOI: 10.1080/13510002.2023.2251237] [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: 09/02/2023] Open
Abstract
OBJECTIVE It is very important to develop a new therapeutic strategy to cope with the increasing morbidity and mortality of chronic kidney disease (CKD). As a kind of physical therapy, low intensity pulsed ultrasound (LIPUS) has remarkable anti-inflammatory and repair-promoting effects and is expected to become a new therapeutic method for CKD. This study aims to clarify the treatment effect of LIPUS on CKD-related renal inflammation and fibrosis, and to further explore the potential signal network of LIPUS treatment for ameliorating chronic renal injury. METHODS A rat model simulating the progress of CKD was established by twice tail-vein injection of Adriamycin (ADR). Under anesthesia, bilateral kidneys of CKD rats were continuously stimulated by LIPUS for four weeks. The parameters of LIPUS were 1.0 MHz, 60 mW/cm2, 50% duty cycle and 20 min/d. RESULTS LIPUS treatment effectively inhibited ADR-induced renal inflammation and fibrosis, and improved CKD-related to oxidative stress and ferroptosis. In addition, the therapeutic effect of LIPUS is closely related to the regulation of TGF-β1/Smad and Nrf2/keap1/HO-1 signalling pathways. DISCUSSION This study provides a new direction for further mechanism research and lays an important foundation for clinical trials.
Collapse
Affiliation(s)
- Zhi-Qiang Ouyang
- Department of Radiology, Yan` an Hospital of Kunming City (Yanan Hospital Affiliated to Kunming Medical University), Kunming, People’s Republic of China
| | - Li-shi Shao
- Department of Radiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Wei-peng Wang
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Teng-fei Ke
- Department of Radiology, Yunnan Cancer Hospital (The Third Affiliated Hospital of Kunming Medical University), Kunming, People’s Republic of China
| | - Dong Chen
- Department of Ultrasound, Yunnan Cancer Hospital (The Third Affiliated Hospital of Kunming Medical University), Kunming, People’s Republic of China
| | - Guang-rong Zheng
- Department of Radiology, Yan` an Hospital of Kunming City (Yanan Hospital Affiliated to Kunming Medical University), Kunming, People’s Republic of China
| | - Xi-rui Duan
- Department of Radiology, Yunnan Cancer Hospital (The Third Affiliated Hospital of Kunming Medical University), Kunming, People’s Republic of China
| | - Ji-xiang Chu
- Department of Radiology, Yunnan Cancer Hospital (The Third Affiliated Hospital of Kunming Medical University), Kunming, People’s Republic of China
| | - Yu Zhu
- Department of Radiology, Yunnan Cancer Hospital (The Third Affiliated Hospital of Kunming Medical University), Kunming, People’s Republic of China
| | - Lu Yang
- Department of Radiology, Yunnan Cancer Hospital (The Third Affiliated Hospital of Kunming Medical University), Kunming, People’s Republic of China
| | - Hai-yan Shan
- Department of Radiology, Yan` an Hospital of Kunming City (Yanan Hospital Affiliated to Kunming Medical University), Kunming, People’s Republic of China
| | - Lin Huang
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Cheng-de Liao
- Department of Radiology, Yan` an Hospital of Kunming City (Yanan Hospital Affiliated to Kunming Medical University), Kunming, People’s Republic of China
| |
Collapse
|
41
|
Liu J, Han X, Zhou J, Leng Y. Molecular Mechanisms of Ferroptosis and Their Involvement in Acute Kidney Injury. J Inflamm Res 2023; 16:4941-4951. [PMID: 37936596 PMCID: PMC10627075 DOI: 10.2147/jir.s427505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/10/2023] [Indexed: 11/09/2023] Open
Abstract
Ferroptosis is a novel way of regulating cell death, which occurs in a process that is closely linked to intracellular iron metabolism, lipid metabolism, amino acid metabolism, and multiple signaling pathways. The latest research shows that ferroptosis plays a key role in the pathogenesis of acute kidney injury (AKI). Ferroptosis may be an important target for treating AKI caused by various reasons, such as ischemia-reperfusion injury, rhabdomyolysis syndrome, sepsis, and nephrotoxic drugs. This paper provides a review on the regulatory mechanisms of ferroptosis and its role in AKI, which may help to provide new research ideas for the treatment of AKI and future research.
Collapse
Affiliation(s)
- Jie Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, People’s Republic of China
| | - Xiaoxia Han
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, People’s Republic of China
| | - Jia Zhou
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, People’s Republic of China
| | - Yufang Leng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, People’s Republic of China
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, People’s Republic of China
| |
Collapse
|
42
|
Maciel L. Pharmacological Conditioning is Salvageable by Dexmedetomidine: How a Sedative Can Mitigate I/R Injuries? Arq Bras Cardiol 2023; 120:e20230696. [PMID: 38055537 DOI: 10.36660/abc.20230696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 10/18/2023] [Indexed: 12/08/2023] Open
Affiliation(s)
- Leonardo Maciel
- Instituto de Biofísica Carlos Chagas Filho Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ - Brasil
- Campus Professor Geraldo Cidade Universidade Federal do Rio de Janeiro, Duque de Caxias, RJ - Brasil
| |
Collapse
|
43
|
Ouyang Z, Zhang G, Wang W, Shao L, Du X, Li G, Tan N, Zhou X, Yang J, Huang L, Liao C. Transcriptome profile analysis revealed the potential mechanism of LIPUS treatment for Adriamycin-induced chronic kidney disease rat. Heliyon 2023; 9:e21531. [PMID: 38027717 PMCID: PMC10663852 DOI: 10.1016/j.heliyon.2023.e21531] [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: 07/14/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Background Developing effective therapeutic strategies to delay the progression of chronic kidney disease (CKD) remains a significant challenge. Low-intensity pulsed ultrasound (LIPUS) has demonstrated potential for treating CKD, but the underlying molecular mechanisms are still elusive. This study aimed to evaluate the therapeutic efficacy of LIPUS and to elucidate the involved genes and signaling pathways. Methods The CKD model was established in rats using Adriamycin (ADR). The bilateral kidneys of CKD rats were continuously stimulated with LIPUS for a period of four weeks. The therapeutic efficacy was defined by renal function and histopathological evaluation. RNA sequencing was employed to profile the transcriptome of rat kidneys in each group. Cluster analysis was utilized to identify differentially expressed genes (DEGs), followed by enrichment analysis of their associated pathways using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. Results LIPUS treatment improved ADR-induced renal dysfunction in the CKD group. Renal fibrosis and pathological damages were also alleviated in the ADR + LIPUS group compared to the ADR group. Cluster analysis identified 844 DEGs. GO enrichment analysis revealed enrichment in inflammatory response terms, while KEGG enrichment analysis highlighted the nuclear factor kappa B (NF-κB) signaling and ferroptosis-related pathways. Conclusion Continuous LIPUS treatment improved ADR-induced renal fibrosis and dysfunction. The therapeutic effect of LIPUS was primarily due to its ability to suppress the CKD-related inflammation, which was associated with the modulation of the NF-κB and ferroptosis signaling pathways. These findings provide a new insight into the potential molecular mechanisms of LIPUS in treating CKD. Further research is necessary to confirm these findings and to identify potential therapeutic targets within these pathways.
Collapse
Affiliation(s)
- Zhiqiang Ouyang
- Department of Radiology, Yan′an Hospital of Kunming City (Yanan Hospital Affiliated to Kunming Medical University), Kunming 650051, China
| | - Guodong Zhang
- Department of Resource Management, Yunnan Cancer Hospital (The Third Affiliated Hospital of Kunming Medical University), Kunming 650100, China
| | - Weipeng Wang
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Lishi Shao
- Department of Radiology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650033, China
| | - Xiaolan Du
- Department of Radiology, Yan′an Hospital of Kunming City (Yanan Hospital Affiliated to Kunming Medical University), Kunming 650051, China
| | - Guocheng Li
- Department of Radiology, Yan′an Hospital of Kunming City (Yanan Hospital Affiliated to Kunming Medical University), Kunming 650051, China
| | - Na Tan
- Department of Radiology, Yan′an Hospital of Kunming City (Yanan Hospital Affiliated to Kunming Medical University), Kunming 650051, China
| | - Xinyan Zhou
- Department of Radiology, Yan′an Hospital of Kunming City (Yanan Hospital Affiliated to Kunming Medical University), Kunming 650051, China
| | - Jun Yang
- Department of Radiology, Yunnan Cancer Hospital (The Third Affiliated Hospital of Kunming Medical University), Kunming 650100, China
| | - Lin Huang
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Chengde Liao
- Department of Radiology, Yan′an Hospital of Kunming City (Yanan Hospital Affiliated to Kunming Medical University), Kunming 650051, China
| |
Collapse
|
44
|
Wang Q, Fan X, Sheng Q, Yang M, Zhou P, Lu S, Gao Y, Kong Z, Shen N, Lv Z, Wang R. N6-methyladenosine methylation in kidney injury. Clin Epigenetics 2023; 15:170. [PMID: 37865763 PMCID: PMC10590532 DOI: 10.1186/s13148-023-01586-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/11/2023] [Indexed: 10/23/2023] Open
Abstract
Multiple mechanisms are involved in kidney damage, among which the role of epigenetic modifications in the occurrence and development of kidney diseases is constantly being revealed. However, N6-methyladenosine (M6A), a well-known post-transcriptional modification, has been regarded as the most prevalent epigenetic modifications in higher eukaryotic, which is involved in various biological processes of cells such as maintaining the stability of mRNA. The role of M6A modification in the mechanism of kidney damage has attracted widespread attention. In this review, we mainly summarize the role of M6A modification in the progression of kidney diseases from the following aspects: the regulatory pattern of N6-methyladenosine, the critical roles of N6-methyladenosine in chronic kidney disease, acute kidney injury and renal cell carcinoma, and then reveal its potential significance in the diagnosis and treatment of various kidney diseases. A better understanding of this field will be helpful for future research and clinical treatment of kidney diseases.
Collapse
Affiliation(s)
- Qimeng Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Xiaoting Fan
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Qinghao Sheng
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Meilin Yang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Ping Zhou
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Shangwei Lu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Ying Gao
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Zhijuan Kong
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Ning Shen
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Zhimei Lv
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
| | - Rong Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
| |
Collapse
|
45
|
Chen LD, Huang YP, Chen JZ, Huang JF, Xu QZ, Chen GP, Lin QC. Nrf2 plays protective role during intermittent hypoxia-induced ferroptosis in rat liver (BRL-3A) cells. Sleep Breath 2023; 27:2069-2076. [PMID: 36856923 DOI: 10.1007/s11325-023-02801-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 02/24/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023]
Abstract
PURPOSE Ferroptosis is reported to be involved in the chronic intermittent hypoxia (CIH)-related liver damage in vivo. Nuclear factor E2-related factor 2 (Nrf2) has an essential role in the regulation of ferroptosis. This study tested the hypothesis that intermittent hypoxia (IH) could lead to hepatocyte ferroptosis in vitro and the function of Nrf2 in IH-induced hepatocyte ferroptosis. METHODS BRL-3A cells (rat liver cells) were exposed to normoxia or IH. The protocol of IH consisted of 32 cycles of 60-min hypoxic exposure with 30-min reoxygenation phase (nadir of 1% oxygen to peak of 20% oxygen). Ferroptosis was evaluated by cell viability, iron concentration, lipid reactive oxygen species (ROS), protein content of ferritin heavy chain (FTH1), and glutathione peroxidase 4 (GPX4). Both ferrostatin-1 (a ferroptosis inhibitor) and Nrf2 interfering RNA were applied to treat BRL-3A cells, respectively. RESULTS IH exposure induced ferroptosis in BRL-3A cells with decreased cell viability and increased total iron content and lipid ROS levels. The protein contents of GPX4 and FTH1 in IH group were markedly lower than that in normoxic control. Ferroptosis inhibitor ferrostatin-1 alleviated IH-induced ferroptosis in BRL-3A cells. IH treatment enhanced expression of Nrf2, and Nrf2 knockdown augmented IH-induced ferroptosis in BRL-3A cells. CONCLUSIONS The results revealed that Nrf2 played a protective role during IH-induced ferroptosis in BRL-3A cells. The finding provides a therapeutic target for obstructive sleep apnea-related liver injury.
Collapse
Affiliation(s)
- Li-Da Chen
- Department of Respiratory and Critical Care Medicine, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian Province, China
| | - Ya-Ping Huang
- Department of Respiratory and Critical Care Medicine, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian Province, China
| | - Ji-Zhi Chen
- Department of Emergency Medicine, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian Province, China
| | - Jie-Feng Huang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Fujian Medical University, No 20, Chazhong road, Taijiang district, Fuzhou, 350005, Fujian Province, China
- Fujian Provincial Sleep-Disordered Breathing Clinic Center, Fuzhou, Fujian Province, China
- Laboratory of Respiratory Disease of the Fujian Medical University, Fuzhou, Fujian Province, China
| | - Qiao-Zhen Xu
- Department of Respiratory and Critical Care Medicine, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian Province, China
| | - Gong-Ping Chen
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Fujian Medical University, No 20, Chazhong road, Taijiang district, Fuzhou, 350005, Fujian Province, China
- Fujian Provincial Sleep-Disordered Breathing Clinic Center, Fuzhou, Fujian Province, China
- Laboratory of Respiratory Disease of the Fujian Medical University, Fuzhou, Fujian Province, China
| | - Qi-Chang Lin
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Fujian Medical University, No 20, Chazhong road, Taijiang district, Fuzhou, 350005, Fujian Province, China.
- Fujian Provincial Sleep-Disordered Breathing Clinic Center, Fuzhou, Fujian Province, China.
- Laboratory of Respiratory Disease of the Fujian Medical University, Fuzhou, Fujian Province, China.
| |
Collapse
|
46
|
Abudurousuli G, Xu S, Che J, Ding X, Gui B, Zhu L. Role of ferroptosis in effects of anesthetics on multiple organ diseases: A literature review. Heliyon 2023; 9:e20405. [PMID: 37780755 PMCID: PMC10539942 DOI: 10.1016/j.heliyon.2023.e20405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/03/2023] Open
Abstract
Anesthesiologists are often faced with patients combined with a series of organ injuries, such as acute lung injury, myocardial ischemia-reperfusion injury, and neurodegenerative diseases. With the in-depth study of these diseases, we are more aware of the choice and rational use of anesthetics for the prognosis of these patients. Ferroptosis is a new type of programmed cell death. This unique pattern of cell death, driven by an imbalance between oxides and antioxidants, is regulated by multiple cellular metabolic events, including redox homeostasis, iron handling, mitochondrial activity, and lipids peroxidation. Numerous studies confirmed that anesthetics modulate ferroptosis by interfering its machineries such as cystine-import-glutathione-glutathione peroxidase 4 axis, Heme oxygenase 1, nuclear factor erythroid 2-related factor 2, and iron homeostasis system. In this literature review, we systemically illustrated possible involvement of ferroptosis in effects of anesthetics and adjuvant drugs on multiple organ diseases, hoping our work may serve as a basis for further studies on regulating ferroptosis through anesthetics related pharmacological modulation and promoting the rational use of anesthetics.
Collapse
Affiliation(s)
- Gulibositan Abudurousuli
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Siyang Xu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
- Department of Anesthesiology, Jiangsu Province Official Hospital, Nanjing, China
| | - Jinxing Che
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
- Department of Anesthesiology, The Huai'an Maternity and Child Healthcare Hospital, Huai'an, China
| | - Xiahao Ding
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Bo Gui
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Linjia Zhu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| |
Collapse
|
47
|
Guan C, Li C, Shen X, Yang C, Liu Z, Zhang N, Xu L, Zhao L, Zhou B, Man X, Luo C, Luan H, Che L, Wang Y, Xu Y. Hexarelin alleviates apoptosis on ischemic acute kidney injury via MDM2/p53 pathway. Eur J Med Res 2023; 28:344. [PMID: 37710348 PMCID: PMC10500723 DOI: 10.1186/s40001-023-01318-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/27/2023] [Indexed: 09/16/2023] Open
Abstract
INTRODUCTION Hexarelin exhibits significant protection against organ injury in models of ischemia/reperfusion (I/R)-induced injury (IRI). Nevertheless, the impact of Hexarelin on acute kidney injury (AKI) and its underlying mechanism remains unclear. In this study, we investigated the therapeutic potential of Hexarelin in I/R-induced AKI and elucidated its molecular mechanisms. METHODS We assessed the protective effects of Hexarelin through both in vivo and in vitro experiments. In the I/R-induced AKI model, rats were pretreated with Hexarelin at 100 μg/kg/d for 7 days before being sacrificed 24 h post-IRI. Subsequently, kidney function, histology, and apoptosis were assessed. In vitro, hypoxia/reoxygenation (H/R)-induced HK-2 cell model was used to investigate the impact of Hexarelin on apoptosis in HK-2 cells. Then, we employed molecular docking using a pharmmapper server and autodock software to identify potential target proteins of Hexarelin. RESULTS In this study, rats subjected to I/R developed severe kidney injury characterized by tubular necrosis, tubular dilatation, increased serum creatinine levels, and cell apoptosis. However, pretreatment with Hexarelin exhibited a protective effect by mitigating post-ischemic kidney pathological changes, improving renal function, and inhibiting apoptosis. This was achieved through the downregulation of conventional apoptosis-related genes, such as Caspase-3, Bax and Bad, and the upregulation of the anti-apoptotic protein Bcl-2. Consistent with the in vivo results, Hexarelin also reduced cell apoptosis in post-H/R HK-2 cells. Furthermore, our analysis using GSEA confirmed the essential role of the apoptosis pathway in I/R-induced AKI. Molecular docking revealed a strong binding affinity between Hexarelin and MDM2, suggesting the potential mechanism of Hexarelin's anti-apoptosis effect at least partially through its interaction with MDM2, a well-known negative regulator of apoptosis-related protein that of p53. To validate these findings, we evaluated the relative expression of MDM2 and p53 in I/R-induced AKI with or without Hexarelin pre-administration and observed a significant suppression of MDM2 and p53 by Hexarelin in both in vivo and in vitro experiments. CONCLUSION Collectively, Hexarelin was identified as a promising medication in protecting apoptosis against I/R-induced AKI.
Collapse
Affiliation(s)
- Chen Guan
- Department of Nephrology, the Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Chenyu Li
- Medizinische Klinik Und Poliklinik IV, Klinikum Der Universität, LMU München, Munich, Germany
| | - Xuefei Shen
- Department of Nephrology, the Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Chengyu Yang
- Department of Nephrology, the Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Zengying Liu
- Department of Nephrology, the Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Ningxin Zhang
- Department of Nephrology, the Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Lingyu Xu
- Department of Nephrology, the Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Long Zhao
- Department of Nephrology, the Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Bin Zhou
- Department of Nephrology, the Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Xiaofei Man
- Department of Nephrology, the Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Congjuan Luo
- Department of Nephrology, the Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Hong Luan
- Department of Nephrology, the Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Lin Che
- Department of Nephrology, the Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Yanfei Wang
- Department of Nephrology, the Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
| | - Yan Xu
- Department of Nephrology, the Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China.
| |
Collapse
|
48
|
Chen P, Wang D, Xiao T, Gu W, Yang H, Yang M, Wang H. ACSL4 promotes ferroptosis and M1 macrophage polarization to regulate the tumorigenesis of nasopharyngeal carcinoma. Int Immunopharmacol 2023; 122:110629. [PMID: 37451020 DOI: 10.1016/j.intimp.2023.110629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/29/2023] [Accepted: 07/08/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is a head and neck malignant tumor with a high incidence and recurrence rate. The crosstalk between ferroptosis and tumor-associated macrophages (TAMs) is thought to have major implications in interfering with cancers. We intended to explore the effect of acyl-CoA synthetase long-chain family member 4 (ACSL4) on the pathogenesis of NPC via ferroptosis and TAMs. METHODS Differential genes in NPC patients were analyzed using publicly available databases, and the ferroptosis-related gene ACSL4 was identified. Expression of ACSL4 in NPC cell lines and xenografted mice was examined. Colony formation, cell proliferation, migration, and invasion were assessed. The abundance of epithelial-mesenchymal transition (EMT) markers (E-cadherin, N-cadherin, and Vimentin) was confirmed. Lipid peroxidation levels and related markers were measured. Clophosome was administered to determine the role of TAMs in NPC mice. RESULTS Low levels of ACSL4 were observed in NPC patients and CNE-2 and 5-8F cells. Erastin (a ferroptosis inducer) and ACSL4 increased lipid peroxidation, decreased cell viability, colony formation, cell proliferation, migration and invasion, and inhibited EMT. Moreover, Erastin and ACSL4 promoted M2 to M1 macrophage polarization. The effects of erastin and ACSL4 were additive. Ferrostatin-1, an inhibitor of ferroptosis, exerted the opposite effect and reversed the beneficial effects of ACSL4 overexpression. In xenograft mice, ACSL4 and clophosome hindered the growth of NPC, and extra clophosome slightly enhanced the antitumor effect of ACSL4. CONCLUSION Our findings indicated that ACSL4 inhibited the pathogenesis of NPC, at least through crosstalk between ferroptosis and macrophages, providing potential direction for NPC therapy.
Collapse
Affiliation(s)
- Pan Chen
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410031, Hunan, China
| | - Dan Wang
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China; Hunan Clinical Research Center of Pediatric Cancer, Changsha 410013, Hunan, China
| | - Tengfei Xiao
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410031, Hunan, China
| | - Wangning Gu
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410031, Hunan, China
| | - Hongmin Yang
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410031, Hunan, China
| | - Minghua Yang
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China; Hunan Clinical Research Center of Pediatric Cancer, Changsha 410013, Hunan, China.
| | - Hui Wang
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410031, Hunan, China.
| |
Collapse
|
49
|
Liu X, Pan B, Wang X, Xu J, Wang X, Song Z, Zhang E, Wang F, Wang W. Ischemia/reperfusion-activated ferroptosis in the early stage triggers excessive inflammation to aggregate lung injury in rats. Front Med (Lausanne) 2023; 10:1181286. [PMID: 37425328 PMCID: PMC10327590 DOI: 10.3389/fmed.2023.1181286] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/05/2023] [Indexed: 07/11/2023] Open
Abstract
Objective Lung ischemia/reperfusion injury (LIRI) is a clinical syndrome of acute lung injury that occurs after lung transplantation or remote organ ischemia. Ferroptosis and inflammation are involved in the pathogenesis of LIRI according to the results of several studies on animal models. However, the interactive mechanisms between ferroptosis and inflammation contributing to LIRI remain unclear. Methods HE staining and indicators of oxidative stress were used to evaluated the lung injury. The reactive oxygen species (ROS) level was examined by DHE staining. The quantitative Real-time PCR (qRT-PCR) and western blot analysis were employed to detect the level of inflammation and ferroptosis, and deferoxamine (DFO) was used to assess the importance of ferroptosis in LIRI and its effect on inflammation. Results In the present study, the link of ferroptosis with inflammation was evaluated at reperfusion 30-, 60- and 180-minute time points, respectively. As the results at reperfusion 30-minute point shown, the pro-ferroptotic indicators, especially cyclooxygenase (COX)-2 and acyl-CoA synthetase long-chain family member 4 (ACSL4), were upregulated while the anti-ferroptotic factors glutathione peroxidase 4 (GPX4), cystine-glumate antiporter (XCT) and ferritin heavy chain (FTH1) were downregulated. Meanwhile, the increased level of interleukin (IL)-6, tumor necrosis factor alpha (TNF-α) and IL-1β were observed beginning at reperfusion 60-minute point but mostly activated at reperfusion 180-minute point. Furthermore, deferoxamine (DFO) was employed to block ferroptosis, which can alleviate lung injury. Expectedly, the survival rate of rats was increased and the lung injury was mitigated containing the improvement of type II alveolar cells ultrastructure and ROS production. In addition, at the reperfusion 180-minute point, the inflammation was observed to be dramatically inhibited after DFO administration as verified by IL-6, TNF-α and IL-1β detection. Conclusion These findings suggest that ischemia/reperfusion-activated ferroptosis plays an important role as the trigger for inflammation to further deteriorate lung damages. Inhibiting ferroptosis may have therapeutic potential for LIRI in clinical practice.
Collapse
Affiliation(s)
- Xiujie Liu
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
- Institute of Ischemia/Reperfusion Injury, Wenzhou Medical University, Wenzhou, China
| | - Binhui Pan
- Nephrology Department, Wenzhou Central Hospital, Wenzhou, China
| | - Xiaoting Wang
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
- Institute of Ischemia/Reperfusion Injury, Wenzhou Medical University, Wenzhou, China
| | - Junpeng Xu
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
- Institute of Ischemia/Reperfusion Injury, Wenzhou Medical University, Wenzhou, China
| | - Xinyu Wang
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
- Institute of Ischemia/Reperfusion Injury, Wenzhou Medical University, Wenzhou, China
| | - Zhengyang Song
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
- Institute of Ischemia/Reperfusion Injury, Wenzhou Medical University, Wenzhou, China
| | - Eryao Zhang
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fangyan Wang
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
- Institute of Ischemia/Reperfusion Injury, Wenzhou Medical University, Wenzhou, China
| | - Wantie Wang
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
- Institute of Ischemia/Reperfusion Injury, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
50
|
Chen F, Kang R, Liu J, Tang D. The ACSL4 Network Regulates Cell Death and Autophagy in Diseases. BIOLOGY 2023; 12:864. [PMID: 37372148 DOI: 10.3390/biology12060864] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/05/2023] [Accepted: 06/11/2023] [Indexed: 06/29/2023]
Abstract
Lipid metabolism, cell death, and autophagy are interconnected processes in cells. Dysregulation of lipid metabolism can lead to cell death, such as via ferroptosis and apoptosis, while lipids also play a crucial role in the regulation of autophagosome formation. An increased autophagic response not only promotes cell survival but also causes cell death depending on the context, especially when selectively degrading antioxidant proteins or organelles that promote ferroptosis. ACSL4 is an enzyme that catalyzes the formation of long-chain acyl-CoA molecules, which are important intermediates in the biosynthesis of various types of lipids. ACSL4 is found in many tissues and is particularly abundant in the brain, liver, and adipose tissue. Dysregulation of ACSL4 is linked to a variety of diseases, including cancer, neurodegenerative disorders, cardiovascular disease, acute kidney injury, and metabolic disorders (such as obesity and non-alcoholic fatty liver disease). In this review, we introduce the structure, function, and regulation of ACSL4; discuss its role in apoptosis, ferroptosis, and autophagy; summarize its pathological function; and explore the potential implications of targeting ACSL4 in the treatment of various diseases.
Collapse
Affiliation(s)
- Fangquan Chen
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 511436, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jiao Liu
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 511436, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|