1
|
Liu H, Xue H, Guo Q, Xue X, Yang L, Zhao K, Liu Y. Ferroptosis meets inflammation: A new frontier in cancer therapy. Cancer Lett 2025; 620:217696. [PMID: 40189012 DOI: 10.1016/j.canlet.2025.217696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, has emerged as a critical player in cancer pathogenesis. Concurrently, inflammation, a key biological response to tissue injury or infection, significantly influences cancer development and progression. The interplay between ferroptosis and inflammation represents a promising yet underexplored area of research. This review synthesizes recent advances in understanding the molecular mechanisms governing their interaction, emphasizing how ferroptosis triggers inflammatory responses and how inflammatory mediators, such as TNF-α, regulate ferroptosis through iron metabolism and lipid peroxidation pathways. Key molecular targets within the ferroptosis-inflammation axis, including GPX4, ACSL4, and the NF-κB signaling pathway, offer therapeutic potential for cancer treatment. By modulating these targets, it may be possible to enhance ferroptosis and fine-tune inflammatory responses, thereby improving therapeutic outcomes. Additionally, this review explores the broader implications of targeting the ferroptosis-inflammation interplay in disease treatment, highlighting opportunities for developing innovative strategies to combat cancer. By bridging the gap in current knowledge, this review provides a comprehensive resource for researchers and clinicians, offering insights into the therapeutic potential of this intricate biological relationship.
Collapse
Affiliation(s)
- Hu Liu
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Hui Xue
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Qian Guo
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xutong Xue
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Lixue Yang
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China.
| | - Kaijun Zhao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Yu'e Liu
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA; Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
2
|
Zhang H, Tian L, Wang P, Li L, Wang K, Li Y, Zhang Y, Feng L, Yao S, Guan H, Ren W. Ferrostatin-1 mitigates acute lung injury by reducing ferroptosis levels in gas explosions. Tissue Cell 2025; 94:102773. [PMID: 39954560 DOI: 10.1016/j.tice.2025.102773] [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: 11/26/2024] [Revised: 01/15/2025] [Accepted: 02/01/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Gas explosion injuries are a severe form of trauma with high incidence and mortality rates, both in daily life and industrial settings. Acute lung injury (ALI) is one of the most serious complications of gas explosion injuries and is a leading cause of mortality in such cases. However, the mechanisms underlying gas explosion-induced ALI have not been fully elucidated, and the treatment process consumes a significant amount of medical resources. Therefore, it is crucial to conduct research on the injury mechanisms of gas explosion injuries, especially the mechanisms of gas explosion-induced ALI, which can effectively improve the treatment rate of this condition. In this study, we analyzed the relationship between a novel form of cell death, ferroptosis, and gas explosion-induced ALI, and explored its specific mechanisms. METHODS We established ALI rat model by Shock tube biological injury system, and detected lung injury-related indexes as well as ferroptosis related indexes, such as glutathione peroxidase 4(GPX4), 4-hydroxynonenal(4HNE), Malondialdehyde(MDA), Fe2 + . We also investigated the therapeutic effects of the ferroptosis inhibitor ferrostatin-1 (Fer-1) in ALI induced by gas explosion, as well as its specific mechanisms of action. RESULTS A rat ALI model by gas explosion was successfully established. After the gas explosion treatment, we observed that the systemic inflammatory reaction of rats was increased, and lung function, liver function, kidney function and cardiac function were damaged to different degrees. The inflammatory infiltration in the lung tissue was more severe, and the degree of lung injury and pulmonary edema increased. The ferroptosis markers GPX4 was decreased, while the levels of 4HNE, MDA and Fe2 + were increased. Treatment with Fer-1 significantly ameliorated gas explosion ALI damage and down-regulated the expression level of ferroptosis. CONCLUSIONS Gas explosion-induced ALI in rats is characterized by enhanced inflammatory responses and reduced antioxidant capacity in lung tissues. The specific mechanism of injury involves ferroptosis. Fer-1 has been shown to mitigate the severity of ALI caused by gas explosion by suppressing ferroptosis expression levels in lung tissues via the Nrf2/GPX4 axis.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; Department of Orthopedics of the General Hospital of Western Theater Command, Chengdu 610083, China
| | - Linqiang Tian
- Institutes of Health Central Plain, Xinxiang Medical University, Jinhui Road #601, Xinxiang, Henan 453003, China; Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China; Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, China
| | - Peng Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Long Li
- Institutes of Health Central Plain, Xinxiang Medical University, Jinhui Road #601, Xinxiang, Henan 453003, China; Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China; Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, China
| | - Kunxi Wang
- Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Yanyan Li
- Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Yue Zhang
- Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Lili Feng
- Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Sanqiao Yao
- Institutes of Health Central Plain, Xinxiang Medical University, Jinhui Road #601, Xinxiang, Henan 453003, China; School of Public Health, Xinxiang Medical University, Xinxiang 453003, China.
| | - Hao Guan
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Wenjie Ren
- Institutes of Health Central Plain, Xinxiang Medical University, Jinhui Road #601, Xinxiang, Henan 453003, China; Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China; Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, China.
| |
Collapse
|
3
|
He Z, Long H, Ma H, Ran W, Song Y, Shang W, Wang J, Qin Y, Deng Z. 4, 9-dihydroxy-α-lapachone as a potent antiproliferation agent for triple-negative breast cancer via ferroptosis. Fitoterapia 2025; 183:106567. [PMID: 40280251 DOI: 10.1016/j.fitote.2025.106567] [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: 02/18/2025] [Revised: 04/07/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and malignant breast cancer. Ferroptosis is an oxidative, iron-dependent form of regulated cell death. Ferroptosis-targeted therapies is a promising approach to improving treatment outcomes of TNBC. Combining death pathway inhibitors with relevant indices for ferroptosis and LipROS, this study uncovered that a natural product of 4, 9-dihydroxy-α-lapachone (DLN) from Catalpa bungei "jinsi" exhibited in vitro and in vivo inhibitory activity against TNBC via ferroptosis. The molecular mechanism is an activation of the FTH1 led to iron overload, and then inhibition of cysteine-glutamate antiporter (system Xc-) and GPX4, which further sensitized TNBC cells to ferroptosis. This study clarified the pathway of DLN-induced cell death in TNBC treatment and exhibited its potential as therapeutic agent for TNBC.
Collapse
Affiliation(s)
- Zhong He
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China; State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hui Long
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Hongyue Ma
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Wanrong Ran
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China
| | - Yinhong Song
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China; Institute of Infection and Inflammation, China Three Gorges University, Yichang 443002, China.
| | - Wenjuan Shang
- Medical College, Hubei Three Gorges Polytechnic, Yichang 443000, China
| | - Junhui Wang
- State Key Laboratory of Tree Genetics and Breeding, Key Laboratory of Tree Breeding and Cultivation of State Forestry Administration, Research Institute of Forestry, Chinese Academy of Forestry, Beijing 100091, China.
| | - Ye Qin
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China; State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Zhangshuang Deng
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China.
| |
Collapse
|
4
|
Fan X, Ai Y, Mu G, Wang F, Feng J, Jia J, Chen Y, Zhou J. Celastrol attenuates ferroptosis-mediated intestinal ischemia/reperfusion-induced acute lung injury via Hippo-YAP signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156865. [PMID: 40414049 DOI: 10.1016/j.phymed.2025.156865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/17/2025] [Accepted: 05/14/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Acute lung injury commonly arises as a secondary complication following intestinal ischemia/reperfusion (II/R) injury. Celastrol (CEL), recognized for its therapeutic effects on inflammation-related conditions such as acute lung injury. Its protective efficacy against II/R-induced acute lung injury remains insufficiently investigated. The Hippo-YAP signaling pathway regulates ferroptosis and plays a pivotal role in II/R injury. PURPOSE To evaluate whether CEL can activate the Hippo-YAP signaling pathway, suppress ferroptosis, and mitigate II/R-induced acute lung injury. METHODS Firstly, an II/R model in mice was established, Immunofluorescence staining and Western blot were used to evaluate the effects of CEL on the Hippo signaling pathway and ferroptosis regulation. Network pharmacology predicted the relevance of the Hippo-YAP signaling pathway in CEL's improvement of acute lung injury. Molecular docking experiment indicated that CEL binds effectively to yes-associated protein (YAP), and overexpression of YAP significantly alleviated both lung injury and ferroptosis. Furthermore, the oxygen-glucose deprivation/recovery (OGD/R) model of MLE-12 cells was developed to further confirm CEL's inhibition of ferroptosis via the Hippo-YAP signaling pathway. RESULTS CEL ameliorated II/R-induced acute lung injury and inhibited inflammation. In vivo and in vitro studies further revealed that CEL significantly reduced ferroptosis and reactive oxygen species (ROS) accumulation in the lung epithelial cells. CONCLUSION CEL effectively mitigated ferroptosis and II/R-induced acute lung injury through elevating YAP protein level, reducing lipid peroxidation, and decreasing intracellular iron accumulation. This study highlights CEL's therapeutic potential for inhibiting ferroptosis, provides mechanistic insights to support CEL's broader therapeutic utility.
Collapse
Affiliation(s)
- Xin Fan
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Yinxin Ai
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Guo Mu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Feixiang Wang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jianguo Feng
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jing Jia
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Ye Chen
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China; Department of Traditional Chinese Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
5
|
Lei Y, Jiang S, Kong C, Pang P, Shan H. Ferroptosis: Therapeutic Potential and Strategies in Non-Small Cell Lung Cancer. BIOLOGY 2025; 14:545. [PMID: 40427734 PMCID: PMC12108931 DOI: 10.3390/biology14050545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 05/09/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025]
Abstract
Non-small cell lung cancer (NSCLC) is the most common subtype of lung cancer and a leading cause of cancer-related morbidity and mortality worldwide. Despite advancements in therapeutic strategies, the prognosis for NSCLC patients remains unfavorable. The effective treatment of NSCLC remains challenging due to its aggressive metastatic and invasive properties. Therefore, there is an urgent need to explore novel treatment strategies. In recent years, different from apoptosis and necrosis, ferroptosis has garnered increasing attention since its initial identification in 2012. It is increasingly recognized as a key factor in the development and progression of various cancers. In this review, we summarize the distinctive morphological and biochemical characteristics of ferroptosis and its regulatory mechanisms. Furthermore, we discuss the genetic regulation of ferroptosis in NSCLC, highlighting key biomarkers that may serve as potential therapeutic targets. We also evaluate emerging therapeutic strategies targeting ferroptosis, including gene therapy, natural compounds, chemical agents, combination therapies, and nanoparticle-based approaches. Based on current evidence, the limitations and future prospects of ferroptosis-based therapies for NSCLC are discussed. This review aims to provide novel insights into the potential of ferroptosis-based therapies for NSCLC and its implications for the development of novel treatments.
Collapse
Affiliation(s)
| | - Shuxia Jiang
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China; (Y.L.); (C.K.); (P.P.)
| | | | | | - Hongli Shan
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China; (Y.L.); (C.K.); (P.P.)
| |
Collapse
|
6
|
Wang T, Huang X, Zhang X, Li N, Lu K, Zeng Y. Unveiling taurine's protective role in ischemic stroke: insights from bidirectional Mendelian randomization and LC-MS/MS analysis. GENES & NUTRITION 2025; 20:10. [PMID: 40361025 PMCID: PMC12076942 DOI: 10.1186/s12263-025-00769-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/25/2025] [Indexed: 05/15/2025]
Abstract
Ischemic stroke remains a leading cause of mortality and disability globally, emphasizing the urgent need for innovative preventative and therapeutic strategies. Taurine, a critical amino sulfonic acid, has garnered attention for its neuroprotective effects, yet its precise role in ischemic stroke remains elusive. This study utilized a bidirectional Mendelian Randomization (MR) approach to explore the causal relationship between plasma taurine levels and ischemic stroke risk, employing genome-wide association study (GWAS) datasets. In parallel, a novel high-sensitivity liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed to quantify plasma taurine levels in ischemic stroke patients and healthy controls. Our findings reveal a significant inverse association between taurine levels and stroke risk, with IVW analysis showing beta = -0.001 and P = 0.0085. Furthermore, LC-MS/MS analysis demonstrated that plasma taurine levels in patients with ischemic stroke were notably lower at 36.07 ± 5.37 μmol/L compared to controls at 108.66 ± 25.11 μmol/L, confirming taurine's potential as a protective factor. These results suggest taurine as a promising biomarker and therapeutic target for stroke prevention and recovery. This study not only highlights the importance of taurine in cerebrovascular health but also provides a foundation for personalized intervention strategies.
Collapse
Affiliation(s)
- Tianyi Wang
- Beijing Institute of Heart, Lung, and Blood Vessel Disease, Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xuyang Huang
- Department of Neurology Central Hospital Affiliated to Shenyang Medical College, Liaoning, China
| | - Xinyue Zhang
- Department of Pediatrics, Liaoning Provincial People's Hospital, Liaoning, China
| | - Na Li
- Mass Spectrometry Research Institute, Beijing Gobroad Hospital, Beijing, China
| | - Kaizhi Lu
- Mass Spectrometry Research Institute, Beijing Gobroad Hospital, Beijing, China
| | - Yong Zeng
- Beijing Institute of Heart, Lung, and Blood Vessel Disease, Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
7
|
Guo Z, Chai T, Yu L, Liu K, Qu Z, Zhang Z, Liu M, Qiu J, Wang G, Li N. Duck House Inhalable Particulate Matter Induces Lung Injury by Activating Ferroptosis. Poult Sci 2025; 104:105169. [PMID: 40403547 DOI: 10.1016/j.psj.2025.105169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 04/12/2025] [Accepted: 04/15/2025] [Indexed: 05/24/2025] Open
Abstract
The particulate matter (PM) generated during poultry farming is characterized by its complex composition and substantial emission levels. However, researches on the respiratory damage caused by poultry house PM and the underlying mechanisms remain limited. In this study, inhalable PM collected from duck houses was administered to experimental mice through inhalation exposure. After 10 days of short-term exposure and 30 days of long-term exposure, mice samples were collected for lung histopathological analysis and inflammatory cytokines detection. The results showed that inhalation of duck house PM induced pulmonary and systemic inflammatory responses in both groups of mice, with significant upregulation of IL-6 and CXCL2. Compared to short-term exposure, long-term exposure resulted in more severe microscopic lesions in the lungs. In addition, the concentrations of malondialdehyde (MDA) and glutathione (GSH) increased in mice, indicating that duck house PM could trigger oxidative stress in lungs, we also found duck house PM induced ferroptosis in mice. Furthermore, it was confirmed that duck house PM caused cell damage and increased intracellular iron levels in MLE-12 cells, and PM reduced GSH in a dose-dependent manner. Notably, ferroptosis inhibitor treatment effectively alleviated PM-induced cell damage. These findings indicated that duck house PM can induce ferroptosis in both mice and cells, and ferroptosis plays a critical role in duck house PM-induced lung damage. These results laid a solid foundation for further exploring the mechanism of PM-induced lung injury, and providing a new insight for targeting ferroptosis to treat such damage.
Collapse
Affiliation(s)
- Zhiyun Guo
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271000, Shandong Province, PR China; Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271018, Shandong Province, PR China; Sino-German Cooperative Research Centre for Zoonosis of Animal Origin Shandong Province, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, PR China.
| | - Tongjie Chai
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271000, Shandong Province, PR China; Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271018, Shandong Province, PR China; Sino-German Cooperative Research Centre for Zoonosis of Animal Origin Shandong Province, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, PR China.
| | - Luna Yu
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271000, Shandong Province, PR China; Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271018, Shandong Province, PR China.
| | - Kuihao Liu
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271000, Shandong Province, PR China; Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271018, Shandong Province, PR China.
| | - Zhengxiu Qu
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271000, Shandong Province, PR China; Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271018, Shandong Province, PR China.
| | - Zhaopeng Zhang
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271000, Shandong Province, PR China; Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271018, Shandong Province, PR China.
| | - Meiling Liu
- College of Veterinary Medicine, Shan Dong Vocational Animal Science and Veterinary College, 88 Shengli East Street, Weifang City 261000, Shandong Province, PR China.
| | - Jianhua Qiu
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271000, Shandong Province, PR China; Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271018, Shandong Province, PR China.
| | - Gang Wang
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271000, Shandong Province, PR China; Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271018, Shandong Province, PR China.
| | - Ning Li
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271000, Shandong Province, PR China; Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, 271018, Shandong Province, PR China; Sino-German Cooperative Research Centre for Zoonosis of Animal Origin Shandong Province, Shandong Agricultural University, 61 Daizong Street, Tai'an City, 271018, Shandong Province, PR China.
| |
Collapse
|
8
|
Wei YY, Chen TT, Zhang DW, Zhang Y, Li F, Ding YC, Wang MY, Zhang L, Chen KG, Fei GH. Microplastics exacerbate ferroptosis via mitochondrial reactive oxygen species-mediated autophagy in chronic obstructive pulmonary disease. Autophagy 2025:1-27. [PMID: 40114310 DOI: 10.1080/15548627.2025.2481126] [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: 12/02/2024] [Revised: 03/09/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025] Open
Abstract
Microplastics (MPs) induce mitochondrial dysfunction and iron accumulation, contributing to mitochondrial macroautophagy/autophagy and ferroptosis, which has increased susceptibility to the exacerbation of chronic obstructive pulmonary disease (COPD); however, the underlying mechanism remains unclear. We demonstrated that MPs intensified inflammation in COPD by enhancing autophagy-dependent ferroptosis (ADF) in vitro and in vivo. In the lung tissues of patients with COPD, the concentrations of MPs, especially polystyrene microplastics (PS-MPs), were significantly higher than that of the control group, as detected by pyrolysis gas chromatography mass spectrometry (Py-GCMS), with increased iron accumulation. The exposure to PS-MPs, 2 μm in size, resulted in their being deposited in the lungs of COPD model mice detected by optical in vivo imaging, and observed in bronchial epithelial cells traced by GFP-labeled PS-MPs. There were mitochondrial impairments accompanied by mitochondrial reactive oxygen species (mito-ROS) overproduction and significantly increased levels of lysosome biogenesis and acidification in pDHBE cells with PS-MP stimulation, triggering occurrence of ferritinophagy and enhancing ADF in COPD, which triggered acute exacerbation of COPD (AECOPD). Reestablishing autophagy-dependent ferroptosis via mitochondria-specific ROS scavenging or ferroptosis inhibition alleviated excessive inflammation and ameliorated AECOPD induced by PS-MPs. Collectively, our data initially revealed that MPs exacerbate ferroptosis via mito-ROS-mediated autophagy in COPD, which sheds light on further hazard assessments of MPs on human respiratory health and potential therapeutic agents for patients with COPD.Abbreviations: ADF: autophagy-dependent ferroptosis; AECOPD: acute exacerbation of chronic obstructive pulmonary disease; Cchord: static compliance; COPD: chronic obstructive pulmonary disease; CQ: chloroquine; CS: cigarette smoke; DEGs: differentially expressed genes; Fer-1: ferrostatin-1; FEV 0.1: forced expiratory volume in first 100 ms; FVC: forced vital capacity; GSH: glutathione; HE: hematoxylin and eosin; IL1B/IL-1β: interleukin 1 beta; IL6: interleukin 6; MDA: malondialdehyde; Mito-ROS: mitochondrial reactive oxygen species; MMA: methyl methacrylate; MMF: maximal mid-expiratory flow curve; MMP: mitochondrial membrane potential; MOI: multiplicity of infection; MPs: microplastics; MV: minute volume; PA: polyamide; PBS: phosphate-buffered saline; PC: polycarbonate; pDHBE: primary human bronchial epithelial cell from COPD patients; PET: polyethylene terephthalate; PIF: peak inspiratory flow; PLA: polylactic acid; pNHBE: primary normal human bronchial epithelial cell; PS-MPs: polystyrene microplastics; PVA: polyvinyl acetate; PVC: polyvinyl chloride; Py-GCMS: pyrolysis gas chromatography mass spectrometry; SEM: scanning electron microscopy; Te: expiratory times; Ti: inspiratory times; TNF/TNF-α: tumor necrosis factor.
Collapse
Affiliation(s)
- Yuan Yuan Wei
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
| | - Ting Ting Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
| | - Da Wei Zhang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
| | - Ying Zhang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
| | - Fang Li
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
| | - Yi Chuan Ding
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
| | - Ming Yu Wang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
| | - Ling Zhang
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Ke Gong Chen
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
- Department of Thoracic Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Guang He Fei
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
| |
Collapse
|
9
|
Zhang B, Meng C, Quan L, Duan L, Kang J, Zhou H. Exosomes derived from bone marrow mesenchymal stem cells alleviate lung ischemia-reperfusion injury in rats through miRNA-335/ SIRT3 pathway. Drug Deliv Transl Res 2025:10.1007/s13346-025-01844-6. [PMID: 40180761 DOI: 10.1007/s13346-025-01844-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2025] [Indexed: 04/05/2025]
Abstract
Lung ischemia-reperfusion injury (IRI) is a clinically challenging problem. Exosomes (EXOs) derived from bone marrow mesenchymal stem cells (BMSC-EXOs) can alleviate multiple organs IRI, but few reports on lung IRI. MiRNA-335 is a kind of miRNA in EXOs, which was also shown protective effects on lung IRI. This study hypothesizes that BMSC-EXOs might alleviate lung IRI through miRNA-335, and further to explore its mechanism. The Sprague-Dawley male rats were divided into sham, IRI, phosphate buffer saline (PBS), and EXO groups (n = 6). In the sham group, rats were underwent anesthesia without IRI model establishment. In the IRI, PBS, and EXO groups, rats were established lung IRI model and with no treatment, 30 µl PBS, or 20 µg EXOs (in 30 µl PBS), respectively. The miRNA-335 inhibitor and miRNA-335 mimic processed EXOs were also given to observe the effects of miRNA-335. The oxidative index, lung static compliance, inflammation response, oxidative stress injury, apoptosis, and mitochondrial were observed. The expression of miRNA-335 and silent matching type information regulation 2 homolog 3 (SIRT3) were also detected. The oxidative index, lung static compliance, inflammation response, oxidative stress injury, apoptosis, and mitochondrial injury were significantly deteriorated in the IRI group compared with those in the sham group, while those indicators have significantly improved in the EXO group, and the miRNA-335 and SIRT3 expressions increased (P < 0.05). And the miRNA-335 inhibitor processed EXOs suppressed the SIRT3 expression significantly, but the miRNA-335 mimic processed EXOs enhanced the SIRT3 expression significantly (P < 0.05). In conclusion, BMSC-EXOs maintained mitochondrial structural stability, and alleviated rat lung IRI by inhibiting lung inflammation, oxidative stress injury, and apoptosis, improved lung oxygenation capacity and static compliance, which might be achieved through the miRNA335/SIRT3 pathway.
Collapse
Affiliation(s)
- Bing Zhang
- Department of Pain, Fourth Affiliated Hospital of Harbin Medical University, No. 37, Yiyuan Street, Nangang District, Harbin City, Hei Longjiang Province, 150001, China
| | - Chao Meng
- Department of Pain Management, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Lini Quan
- Department of Pain, Fourth Affiliated Hospital of Harbin Medical University, No. 37, Yiyuan Street, Nangang District, Harbin City, Hei Longjiang Province, 150001, China
| | - Le Duan
- Department of Pain, Fourth Affiliated Hospital of Harbin Medical University, No. 37, Yiyuan Street, Nangang District, Harbin City, Hei Longjiang Province, 150001, China
| | - Jiyu Kang
- Department of Pain, Fourth Affiliated Hospital of Harbin Medical University, No. 37, Yiyuan Street, Nangang District, Harbin City, Hei Longjiang Province, 150001, China
| | - Huacheng Zhou
- Department of Pain, Fourth Affiliated Hospital of Harbin Medical University, No. 37, Yiyuan Street, Nangang District, Harbin City, Hei Longjiang Province, 150001, China.
| |
Collapse
|
10
|
Zhang M, Zhang W, Chen Z, He L, Chen Q, Lan P, Li L, Wu X, Wu X, Xu J. LncRNA ENSSSCG00000035331 Alleviates Hippocampal Neuronal Ferroptosis and Brain Injury Following Porcine Cardiopulmonary Resuscitation by Regulating the miR-let7a/GPX4 Axis. CNS Neurosci Ther 2025; 31:e70377. [PMID: 40237277 PMCID: PMC12001066 DOI: 10.1111/cns.70377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 03/13/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Following successful cardiopulmonary resuscitation, those survivors of cardiac arrest (CA) often suffer from severe brain injury, and the latter can result in significant mortality and morbidity. Emerging evidence implicates that ferroptosis is involved in the pathogenesis of post-resuscitation brain injury, and its regulatory mechanisms remain to be investigated. Recently, some studies manifested that long noncoding RNAs could be critical regulators of cell ferroptosis in diverse ischemia-reperfusion injuries of vital organs. This study was designed to explore the role and mechanism of a newly screened long noncoding RNA ENSSSCG00000035331 in alleviating post-resuscitation hippocampal neuronal ferroptosis and further investigate its potential regulation by a novel antioxidant sulforaphane. METHODS AND RESULTS Healthy male pigs and mice were used to establish the models of CA and resuscitation in vivo. A hypoxia/reoxygenation (H/R) model using primary porcine hippocampal neurons was constructed to replicate post-resuscitation brain injury in vitro. We found that the expression of ENSSSCG00000035331 was significantly decreased in the post-resuscitation impaired hippocampus using RNA sequencing analysis and verification. Subsequently, ENSSSCG00000035331 overexpression significantly reduced ferroptosis-related ferrous iron and reactive oxygen species production while markedly increased glutathione and further alleviated post-resuscitation brain injury. Mechanistically, ENSSSCG00000035331 interacted with miR-let7a, then inhibited its binding with glutathione peroxidase 4 (GPX4) mRNA and finally promoted the recovery of the latter's translation after H/R stimulation. In addition, sulforaphane treatment significantly increased ENSSSCG00000035331 and GPX4 expression while markedly decreased miR-let7a expression and hippocampal neuronal ferroptosis and finally alleviated post-resuscitation brain injury. CONCLUSIONS Our findings highlighted that ENSSSCG00000035331 was a critical regulator of hippocampal neuronal ferroptosis after CA and resuscitation by targeting the miR-let7a/GPX4 axis, and additionally, sulforaphane might be a promising therapeutic agent for alleviating post-resuscitation brain injury by regulating the signaling axis mentioned above.
Collapse
Affiliation(s)
- Mao Zhang
- Department of Emergency Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Key Laboratory of Trauma, Burn, and Medical RescueHangzhouChina
- Zhejiang Province Clinical Research Center for Emergency and Critical Care MedicineHangzhouChina
| | - Wenbin Zhang
- Department of Emergency Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Key Laboratory of Trauma, Burn, and Medical RescueHangzhouChina
- Zhejiang Province Clinical Research Center for Emergency and Critical Care MedicineHangzhouChina
| | - Ziwei Chen
- Department of Emergency Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Key Laboratory of Trauma, Burn, and Medical RescueHangzhouChina
- Zhejiang Province Clinical Research Center for Emergency and Critical Care MedicineHangzhouChina
| | - Lu He
- Department of Emergency Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Key Laboratory of Trauma, Burn, and Medical RescueHangzhouChina
- Zhejiang Province Clinical Research Center for Emergency and Critical Care MedicineHangzhouChina
| | - Qijiang Chen
- Department of Intensive Care MedicineThe First Hospital of NinghaiNingboChina
| | - Pin Lan
- Department of Emergency MedicineFifth Affiliated Hospital of Wenzhou Medical University, Lishui Central HospitalLishuiChina
| | - Lulu Li
- Department of Emergency Medicine, First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Xianlong Wu
- Department of Emergency MedicineTaizhou First People's HospitalTaizhouChina
| | - Xingui Wu
- Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect DiseaseGuangzhouChina
- Guangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
| | - Jiefeng Xu
- Department of Emergency Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Key Laboratory of Trauma, Burn, and Medical RescueHangzhouChina
- Zhejiang Province Clinical Research Center for Emergency and Critical Care MedicineHangzhouChina
| |
Collapse
|
11
|
Yang Y, Li C, Lu Z, Cao X, Wu Q. METTL3-mediated m6A Modification Promotes miR-221-3p Expression to Exacerbate Ischemia/Reperfusion-Induced Acute Lung Injury. J Biochem Mol Toxicol 2025; 39:e70235. [PMID: 40127211 DOI: 10.1002/jbt.70235] [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: 06/21/2024] [Revised: 01/19/2025] [Accepted: 03/13/2025] [Indexed: 03/26/2025]
Abstract
Ischemia/reperfusion (I/R)-induced acute lung injury (ALI) represents a prevalent pulmonary pathology. The N6-methyladenosine (m6A) RNA modification is integral in regulating numerous biological processes across various human diseases through the modulation of gene expression. Nevertheless, the precise role and underlying molecular mechanisms of m6A modifications in ALI remain inadequately understood. This study aimed to elucidate the impact of RNA methyltransferase 3 (METTL3)-mediated m6A modification of miR-221-3p on the progression of I/R-induced ALI. Our initial findings demonstrated an upregulation of m6A levels and METTL3 expression in I/R-induced ALI in murine models and hypoxia/reoxygenation (H/R)-induced murine lung epithelial (MLE)-12 cells. Inhibition of METTL3 was observed to reverse H/R-induced apoptotic cell death, oxidative stress, and inflammatory cytokine secretion. Furthermore, METTL3 was found to enhance the expression of miR-221-3p in an m6A-dependent manner, thereby contributing to ALI pathogenesis. In addition, miR-221-3p was shown to negatively regulate PTEN expression, while METTL3 facilitated phosphorylated AKT expression via the miR-221-3p/PTEN axis. Functional experiments further revealed that the downregulation of PTEN negated the inhibitory effects of METTL3 knockdown in H/R-treated MLE-12 cells. In conclusion, our study demonstrates that the METTL3-mediated m6A modification of miR-221-3p exacerbates ALI through modulation of the PTEN/AKT pathway. Therapeutic strategies aimed at targeting the METTL3/m6A/miR-221-3p/PTEN/AKT axis may offer a promising approach to mitigate I/R-induced ALI.
Collapse
Affiliation(s)
- Yang Yang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chenlu Li
- Department of Nursing, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ziwang Lu
- Department of Cardiovascular Medicine, Bao Ji People's Hospital, Bao ji, Shaanxi, China
| | - Xiantong Cao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qifei Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
12
|
Luo L, Zhang S, Gong J, Zhang J, Xie P, Yin J, Zhang M, Zhang C, Chen H, Liu Y, Ni B, Li C, Tian Z. 3-D Sustained-Release Culture Carrier Alleviates Rat Intervertebral Disc Degeneration by Targeting STING in Transplanted Skeletal Stem Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410151. [PMID: 39985222 PMCID: PMC12005824 DOI: 10.1002/advs.202410151] [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: 08/23/2024] [Revised: 01/14/2025] [Indexed: 02/24/2025]
Abstract
The hypoxic and high-pressure microenvironment of the intervertebral discs poses a major challenge to the survival and therapeutic efficiency of exogenous stem cells. Therefore, improving the utilization efficiency and therapeutic effect of exogenous stem cells to delay intervertebral disc degeneration (IVDD) is of great importance. Here, hypoxic induction studies are conducted in vivo and in vitro using rat costal cartilage-derived skeletal stem cells (SSCs) and find that hypoxia activates the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)/stimulator of interferon genes (STING) signaling pathway and increased reactive oxygen species (ROS) accumulation, triggering ferroptosis in SSCs through hypoxia-inducible factor-1 alpha-dependent mitophagy. Progressive hypoxia preconditioning reduce STING expression and ROS accumulation, inducing SSCs differentiation into nucleus pulposus-like cells via the Wnt signaling pathway. Considering this, a 3-D sustained-release culture carrier is generated by mixing SSCs with methacrylated hyaluronic acid and polydopamine nanoparticles coated with the STING inhibitor C-176 and evaluated its inhibitory effect on IVDD. This carrier is demonstrated to inhibit the cGAS/STING pathway and prevent ROS accumulation by continuously releasing C-176-coated polydopamine nanoparticles, thereby reducing ferroptosis, promoting differentiation, and ultimately attenuating IVDD, suggesting its potential as a novel treatment strategy.
Collapse
Affiliation(s)
- Liwen Luo
- Department of OrthopaedicsXinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingP. R. China
- State Key Laboratory of Trauma and Chemical PoisoningArmy Medical University (Third Military Medical University)ChongqingP. R. China
| | - Shiyu Zhang
- Department of OrthopaedicsXinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingP. R. China
| | - Junfeng Gong
- Department of General SurgeryThe Armed Police Corps Hospital of AnhuiHefeiP. R. China
| | - Ji Zhang
- Institute of ImmunologyPLAArmy Medical University (Third Military Medical University)ChongqingP. R. China
| | - Peng Xie
- Department of Military BiosafetyCollege of Basic MedicineArmy Medical UniversityChongqingP. R. China
| | - Jun Yin
- Department of PathophysiologyCollege of High Altitude Military MedicineArmy Military Medical UniversityChongqingP. R. China
| | - MengJie Zhang
- Department of PathophysiologyCollege of High Altitude Military MedicineArmy Military Medical UniversityChongqingP. R. China
| | - Cong Zhang
- Department of Laboratory Animal ScienceCollege of Basic MedicineArmy Medical UniversityChongqingP. R. China
| | - Hong Chen
- Department of Orthopedics903 Hospital of Joint Logistic Support Force of The People's Liberation ArmyHangzhouP. R. China
| | - Yao Liu
- Department of PharmacyDaping HospitalArmy Medical University (Third Military Medical University)ChongqingP. R. China
| | - Bing Ni
- Department of PathophysiologyCollege of High Altitude Military MedicineArmy Military Medical UniversityChongqingP. R. China
| | - Changqing Li
- Department of OrthopaedicsXinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingP. R. China
- State Key Laboratory of Trauma and Chemical PoisoningArmy Medical University (Third Military Medical University)ChongqingP. R. China
| | - Zhiqiang Tian
- Institute of ImmunologyPLAArmy Medical University (Third Military Medical University)ChongqingP. R. China
| |
Collapse
|
13
|
Liu L, Li J, Xue Y, Xie S, Dong N, Chen C. Dihydroartemisinin attenuates PM-induced lung injury by inhibiting inflammation and regulating autophagy. Front Public Health 2025; 13:1548224. [PMID: 40124417 PMCID: PMC11925939 DOI: 10.3389/fpubh.2025.1548224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
Objective The study investigates the effects and mechanisms of dihydroartemisinin (DHA) in mitigating lung injury induced by particulate matter (PM). Methods The lung injury model was induced by PM particles in vivo and in vitro. Hematoxylin and Eosin (H&E) staining was utilized for the detection of the thickening of airway wall and the infiltration of inflammatory cells in mouse lung tissue. The expressions of inflammatory factors were detected in alveolar lavage fluid and cell supernatant. TUNEL (Terminal deoxynucleotidyl transferase dUTP nick end labeling) staining, Caspase-1, Bcl-2-associated X protein (Bax), B-cell lymphoma 2 (Bcl-2), microtubule-associated protein 1 light chain 3-II (LC3-II) and Belcin-1 were used to observe the apoptosis and autophagy related expressions in mouse lung tissue, and p-p65 was detected by immunofluorescence. Results H&E staining revealed DHA alleviates PM-induced lung injury in vivo. Moreover, DHA reduced IL-6, IL-8, and IL-1β levels by ~50% (p < 0.05), highlighting its anti-inflammatory effects. Furthermore, immunohistochemistry showed that DHA treatment inhibited the pro-apoptotic expression of Bax/BCL2 and cleaved-Caspase-3, respectively. In addition, immunofluorescence staining revealed that the LC3-II and Beclin-1 levels dramatically increased in the PM group compared to Control group, but greatly reduced by DHA. Further, we found that DHA inhibited the activation of the NF-KB signaling pathway. Conclusion DHA protects against PM-induced lung injury through anti-inflammatory, anti-apoptotic, and autophagy-regulating mechanisms, offering a potential drug option for improving PM-induced lung injury.
Collapse
Affiliation(s)
- Lingjing Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jingli Li
- Department of Pulmonary and Critical Care Medicine, Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Yincong Xue
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shuying Xie
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Nian Dong
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chengshui Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
14
|
Zheng Z, Qiao X, Yin J, Kong J, Han W, Qin J, Meng F, Tian G, Feng X. Advancements in omics technologies: Molecular mechanisms of acute lung injury and acute respiratory distress syndrome (Review). Int J Mol Med 2025; 55:38. [PMID: 39749711 PMCID: PMC11722059 DOI: 10.3892/ijmm.2024.5479] [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: 09/06/2024] [Accepted: 12/09/2024] [Indexed: 01/04/2025] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is an inflammatory response arising from lung and systemic injury with diverse causes and associated with high rates of morbidity and mortality. To date, no fully effective pharmacological therapies have been established and the relevant underlying mechanisms warrant elucidation, which may be facilitated by multi‑omics technology. The present review summarizes the application of multi‑omics technology in identifying novel diagnostic markers and therapeutic strategies of ALI/ARDS as well as its pathogenesis.
Collapse
Affiliation(s)
- Zhihuan Zheng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Xinyu Qiao
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Junhao Yin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Junjie Kong
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Wanqing Han
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Jing Qin
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Fanda Meng
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Ge Tian
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271000, P.R. China
| | - Xiujing Feng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
- Department of Immunology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
15
|
Bu X, Wang L. Iron metabolism and the tumor microenvironment: A new perspective on cancer intervention and therapy (Review). Int J Mol Med 2025; 55:39. [PMID: 39749705 PMCID: PMC11722052 DOI: 10.3892/ijmm.2024.5480] [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: 09/09/2024] [Accepted: 10/29/2024] [Indexed: 01/04/2025] Open
Abstract
Iron metabolism plays a crucial role in the tumor microenvironment, influencing various aspects of cancer cell biology and tumor progression. This review discusses the regulatory mechanisms of iron metabolism within the tumor microenvironment and highlights how tumor cells and associated stromal cells manage iron uptake, accumulation and regulation. The sources of iron within tumors and the biological importance of ferroptosis in cancer were explored, focusing on its mechanisms, biological effects and, in particular, its tumor‑suppressive properties. Furthermore, the protective strategies employed by cancer cells to evade ferroptosis were examined. This review also delves into the intricate relationship between iron metabolism and immune modulation within the tumor microenvironment, detailing the impact on tumor‑associated immune cells and immune evasion. The interplay between ferroptosis and immunotherapy is discussed and potential strategies to enhance cancer immunotherapy by modulating iron metabolism are presented. Finally, the current ferroptosis‑based cancer therapeutic approaches were summarized and future directions for therapies that target iron metabolism were proposed.
Collapse
Affiliation(s)
- Xiaorui Bu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Lufang Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
16
|
Liu H, Zheng S, Hou G, Dai J, Zhao Y, Yang F, Xiang Z, Zhang W, Wang X, Gong Y, Li L, Zhang N, Hu Y. AKAP1/PKA-mediated GRP75 phosphorylation at mitochondria-associated endoplasmic reticulum membranes protects cancer cells against ferroptosis. Cell Death Differ 2025; 32:488-505. [PMID: 39537840 PMCID: PMC11893801 DOI: 10.1038/s41418-024-01414-2] [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: 05/06/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
Emerging evidence suggests that signaling pathways can be spatially regulated to ensure rapid and efficient responses to dynamically changing local cues. Ferroptosis is a recently defined form of lipid peroxidation-driven cell death. Although the molecular mechanisms underlying ferroptosis are emerging, spatial aspects of its signaling remain largely unexplored. By analyzing a public database, we found that a mitochondrial chaperone protein, glucose-regulated protein 75 (GRP75), may have a previously undefined role in regulating ferroptosis. This was subsequently validated. Interestingly, under ferroptotic conditions, GRP75 translocated from mitochondria to mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs) and the cytosol. Further mechanistic studies revealed a highly spatial regulation of GRP75-mediated antiferroptotic signaling. Under ferroptotic conditions, lipid peroxidation predominantly accumulated at the ER, which activated protein kinase A (PKA) in a cAMP-dependent manner. In particular, a signaling microdomain, the outer mitochondrial membrane protein A-kinase anchor protein 1 (AKAP1)-anchored PKA, phosphorylated GRP75 at S148 in MAMs. This caused GRP75 to be sequestered outside the mitochondria, where it competed with Nrf2 for Keap1 binding through a conserved high-affinity RGD-binding motif, ETGE. Nrf2 was then stabilized and activated, leading to the transcriptional activation of a panel of antiferroptotic genes. Blockade of the PKA/GRP75 axis dramatically increased the responses of cancer cells to ferroptosis both in vivo and in vitro. Our identification a localized signaling cascade involved in protecting cancer cells from ferroptosis broadens our understanding of cellular defense mechanisms against ferroptosis and also provides a new target axis (AKAP1/PKA/GRP75) to improve the responses of cancer cells to ferroptosis.
Collapse
Affiliation(s)
- Hao Liu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, Henan Province, 450000, China
| | - Shanliang Zheng
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
| | - Guixue Hou
- BGI-SHENZHEN, Shenzhen, Guangdong Province, 518083, China
| | - Junren Dai
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
| | - Yanan Zhao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
| | - Fan Yang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
| | - Zhiyuan Xiang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, Henan Province, 450000, China
| | - Wenxin Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, Henan Province, 450000, China
| | - Xingwen Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
| | - Yafan Gong
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, Henan Province, 450000, China
| | - Li Li
- The third affiliated hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150040, China
| | - Ning Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
| | - Ying Hu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China.
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, Henan Province, 450000, China.
| |
Collapse
|
17
|
Xie H, Wei C, Xiong C, Huang Z, Chen C, Xiao X, Zhang L, Lin Z, Yao W, Zhao T, Hei Z. pH-responsive cationic polymer-functionalized poly-ε-caprolactone microspheres scavenge cell-free-DNA to alleviate intestinal ischemia/reperfusion injury by inhibiting M1 macrophage polarization. J Nanobiotechnology 2025; 23:153. [PMID: 40016777 PMCID: PMC11869592 DOI: 10.1186/s12951-025-03231-2] [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: 12/14/2024] [Accepted: 02/11/2025] [Indexed: 03/01/2025] Open
Abstract
Intestinal ischemia/reperfusion (I/R) injury is a common life-threatening condition. Inflammatory dysregulation plays a crucial role in the pathological progression of intestinal I/R injury, indicating that controlling excessive inflammatory responses can be an effective strategy for mitigating I/R injury. Herein, after establishing a correlation between cell-free DNA (cfDNA) levels and postoperative inflammatory factors in samples from patients with intestinal I/R, we tested a cfDNA-scavenging approach for the treatment of intestinal I/R injury. Poly-ε-caprolactone (PCL) microspheres (Micro DEA2k) functionalized with a pH-responsive cationic polymer (DEA2k) to efficiently scavenge cfDNA were synthesized and evaluated.These microspheres exhibited enhanced cfDNA adsorption under inflammation-induced acidic conditions, along with low toxicity, reduced non-specific protein binding, and extended peritoneal retention. In a mouse model of intestinal I/R injury, the intraperitoneal injection Micro DEA2k effectively bound cfDNA, regulated the mononuclear phagocytic system, decreased the number of M1 macrophages, suppressed inflammation, and significantly improved the survival rate of the mice. These findings suggest that cfDNA scavenging using cationic microspheres has considerable potential for alleviating intestinal I/R injury.
Collapse
Affiliation(s)
- Hanbin Xie
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510635, China
| | - Cong Wei
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, Guangdong, 510275, China
| | - Chang Xiong
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510635, China
| | - Ziyan Huang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
| | - Chaojin Chen
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510635, China
| | - Xue Xiao
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510635, China
| | - Linan Zhang
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510635, China
| | - Zhenjia Lin
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510635, China
| | - Weifeng Yao
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510635, China.
| | - Tianyu Zhao
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, Guangdong, 510275, China.
| | - Ziqing Hei
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510635, China.
- Zhaoqing Campus of the Third Affiliated Hospital of Sun Yat Sen University, Zhaoqing, Guangdong, 526000, China.
| |
Collapse
|
18
|
Wang H, Xie Y. Advances in Ferroptosis Research: A Comprehensive Review of Mechanism Exploration, Drug Development, and Disease Treatment. Pharmaceuticals (Basel) 2025; 18:334. [PMID: 40143112 PMCID: PMC11944624 DOI: 10.3390/ph18030334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 03/28/2025] Open
Abstract
In recent years, ferroptosis, as an emerging modality of programmed cell death, has captured significant attention within the scientific community. This comprehensive review meticulously canvasses the pertinent literature of the past few years, spanning multiple facets. It delves into the intricate mechanisms underpinning ferroptosis, tracks the evolution of its inducers and inhibitors, and dissects its roles in a diverse array of diseases, as well as the resultant therapeutic implications. A profound exploration is conducted of the functional mechanisms of ferroptosis-related molecules, intracellular pathways, metabolic cascades, and signaling transduction routes. Novel ferroptosis inducers and inhibitors are introduced in detail, covering their design blueprints, synthetic methodologies, and bioactivity profiles. Moreover, an exhaustive account is provided regarding the involvement of ferroptosis in malignancies, neurodegenerative disorders, cardiovascular ailments, and other pathologies. By highlighting the pivotal status and potential therapeutic regimens of ferroptosis in various diseases, this review aspires to furnish a thorough and profound reference framework for future investigations and clinical translations in the ferroptosis domain.
Collapse
Affiliation(s)
- Haojie Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yuanyuan Xie
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceutical, Zhejiang University of Technology, Hangzhou 310014, China
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, Hangzhou 310014, China
| |
Collapse
|
19
|
Fernández-Acosta R, Vintea I, Koeken I, Hassannia B, Vanden Berghe T. Harnessing ferroptosis for precision oncology: challenges and prospects. BMC Biol 2025; 23:57. [PMID: 39988655 PMCID: PMC11849278 DOI: 10.1186/s12915-025-02154-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/12/2025] [Indexed: 02/25/2025] Open
Abstract
The discovery of diverse molecular mechanisms of regulated cell death has opened new avenues for cancer therapy. Ferroptosis, a unique form of cell death driven by iron-catalyzed peroxidation of membrane phospholipids, holds particular promise for targeting resistant cancer types. This review critically examines current literature on ferroptosis, focusing on its defining features and therapeutic potential. We discuss how molecular profiling of tumors and liquid biopsies can generate extensive multi-omics datasets, which can be leveraged through machine learning-based analytical approaches for patient stratification. Addressing these challenges is essential for advancing the clinical integration of ferroptosis-driven treatments in cancer care.
Collapse
Affiliation(s)
- Roberto Fernández-Acosta
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Iuliana Vintea
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Biobix, Lab of Bioinformatics and Computational Genomics, Department of Mathematical Modelling, Statistics and Bioinformatics, Ghent University, Ghent, Belgium
| | - Ine Koeken
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Behrouz Hassannia
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Tom Vanden Berghe
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
20
|
Shen G, Wen H, Li H, Zhang X, Lan B, Dong X, Ge P, Luo Y, Chen H. Emodin protects against severe acute pancreatitis-associated acute lung injury by activating Nrf2/HO-1/GPX4 signal and inhibiting ferroptosis in vivo and in vitro. BMC Gastroenterol 2025; 25:57. [PMID: 39910464 PMCID: PMC11800461 DOI: 10.1186/s12876-025-03660-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/30/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Severe acute pancreatitis (SAP) has high morbidity, a complicated and dangerous course, and many complications, including severe pulmonary complications. SAP-associated acute lung injury (SAP-ALI) is still a significant challenge for surgeons because of its high mortality. Therefore, more effective treatment methods are urgently needed. Emodin (EMO) has shown tremendous potential in treating many refractory diseases. However, its protection mechanism in SAP-ALI needs to be further clarified. This study was undertaken to investigate the protective effects of EMO against lung injury in SAP rats and alveolar epithelial cells, with a particular focus on the classical ferroptosis pathway. METHODS In an in vivo study, forty SD rats were evenly split into five groups: sham operation (SO) group, the biliopancreatic duct was retrogradely injected with 5% sodium taurocholate (STC) to create the SAP group, SAP + EMO group was administered EMO via gavage to the rats following the modeling, SAP + ML385 group (a given inhibitor of nuclear factor erythroid 2-related factor 2 (Nrf2)), SAP + ML385 + EMO group. In an in vitro study, alveolar epithelial A549 cell lines were exposed to lipopolysaccharide (LPS) and treated with EMO. ML385 was also used to inhibit the expression of Nrf2. Pancreatic and lung tissue damage was evaluated using histological examination and molecular experiments. Enzyme-linked immunosorbent assays (ELISA) were used to assess the levels of pro-inflammatory cytokines, Fe2+, and associated oxidative stress indicators in the serum and cell supernatant. Real-time polymerase chain reaction (PCR), Western blot (WB), and immunofluorescence were used to find the expressions of related mRNAs and proteins in the lung tissue or A549 cells. RESULTS The findings demonstrated that suppressing Nrf2 expression exacerbated the inflammatory response brought on by SAP and the pathological alterations of SAP-ALI. Emodin treatment reversed this pathological change by activating the Nrf2/Heme Oxygenase-1 (HO-1)/glutathione peroxidase 4 (GPX4) signal path. Moreover, these results also showed that EMO, contrary to the effects of ML385, suppressed the ferroptosis response, which manifested as up-regulated glutathione (GSH) and GPX4 levels in vivo and in vitro and down-regulated malondialdehyde (MDA), superoxide dismutase (SOD), Fe2+, and reactive oxygen species (ROS) levels. CONCLUSIONS Our results demonstrated that EMO effectively inhibited ferroptosis both in vivo and in vitro, while also modulating the Nrf2/HO-1/GPX4 signaling pathway to provide protection against SAP-ALI.
Collapse
Affiliation(s)
- Gang Shen
- Department of General Surgery, Pancreatic-Biliary Center, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhongshan Road, Dalian, Liaoning Province, 116011, People's Republic of China
- Dalian Women and Children's Medical Center (Group), Dalian Medical University, Dalian Liaoning, 116011, People's Republic of China
| | - Haiyun Wen
- Department of General Surgery, Pancreatic-Biliary Center, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhongshan Road, Dalian, Liaoning Province, 116011, People's Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian Liaoning, 116011, People's Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian Liaoning, 116011, People's Republic of China
| | - Huijuan Li
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian Liaoning, 116011, People's Republic of China
- The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People's Republic of China
| | - Xuetao Zhang
- Department of General Surgery, Pancreatic-Biliary Center, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhongshan Road, Dalian, Liaoning Province, 116011, People's Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian Liaoning, 116011, People's Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian Liaoning, 116011, People's Republic of China
| | - Bowen Lan
- Department of General Surgery, Pancreatic-Biliary Center, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhongshan Road, Dalian, Liaoning Province, 116011, People's Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian Liaoning, 116011, People's Republic of China
| | - Xuanchi Dong
- Department of General Surgery, Pancreatic-Biliary Center, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhongshan Road, Dalian, Liaoning Province, 116011, People's Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian Liaoning, 116011, People's Republic of China
| | - Peng Ge
- Department of General Surgery, Pancreatic-Biliary Center, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhongshan Road, Dalian, Liaoning Province, 116011, People's Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian Liaoning, 116011, People's Republic of China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian Liaoning, 116011, People's Republic of China
| | - Yalan Luo
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian Liaoning, 116011, People's Republic of China
| | - Hailong Chen
- Department of General Surgery, Pancreatic-Biliary Center, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhongshan Road, Dalian, Liaoning Province, 116011, People's Republic of China.
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian Liaoning, 116011, People's Republic of China.
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian Liaoning, 116011, People's Republic of China.
| |
Collapse
|
21
|
Yang L, Tang H, Wang J, Xu D, Xuan R, Xie S, Xu P, Li X. O-GlcNAcylation attenuates ischemia-reperfusion-induced pulmonary epithelial cell ferroptosis via the Nrf2/G6PDH pathway. BMC Biol 2025; 23:32. [PMID: 39901237 PMCID: PMC11792224 DOI: 10.1186/s12915-025-02126-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: 07/24/2024] [Accepted: 01/09/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Lung ischemia-reperfusion (I/R) injury is a common clinical pathology associated with high mortality. The pathophysiology of lung I/R injury involves ferroptosis and elevated protein O-GlcNAcylation levels, while the effect of O-GlcNAcylation on lung I/R injury remains unclear. This research aimed to explore the effect of O-GlcNAcylation on reducing ferroptosis in pulmonary epithelial cells caused by I/R. RESULTS First, we identified O-GlcNAc transferase 1 (Ogt1) as a differentially expressed gene in lung epithelial cells of acute lung injury/acute respiratory distress syndrome (ALI/ARDS) patients, using single-cell sequencing, and Gene Ontology analysis (GO analysis) revealed the enrichment of the ferroptosis process. We found a time-dependent dynamic alteration in lung O-GlcNAcylation during I/R injury. Proteomics analysis identified the differentially expressed proteins enriched in ferroptosis and multiple redox-related pathways based on KEGG annotation. Thus, we generated Ogt1-conditional knockout mice and found that Ogt1 deficiency aggravated ferroptosis, as evidenced by lipid reactive oxygen species (lipid ROS), malondialdehyde (MDA), Fe2+, as well as alterations in critical protein expression glutathione peroxidase 4 (GPX4) and solute carrier family 7 member 11 (SLC7A11). Consistently, we found that elevated O-GlcNAcylation inhibited ferroptosis sensitivity in hypoxia/reoxygenation (H/R) injury-induced TC-1 cells via O-GlcNAcylated NF-E2-related factor-2 (Nrf2). Furthermore, both the chromatin immunoprecipitation (ChIP) assay and the dual-luciferase reporter assay indicated that Nrf2 could bind with translation start site (TSS) of glucose-6-phosphate dehydrogenase (G6PDH) and promote its transcriptional activity. As an important rate-limiting enzyme in the pentose phosphate pathway (PPP), elevated G6PDH provided a mass of nicotinamide adenine dinucleotide phosphate (NADPH) to improve the redox state of glutathione (GSH) and eventually led to ferroptosis resistance. Rescue experiments proved that Nrf2 knockdown or Nrf2-T334A (O-GlcNAcylation site) mutation abolished the protective effect of ferroptosis resistance. CONCLUSIONS In summary, we revealed that O-GlcNAcylation could protect against I/R lung injury by reducing ferroptosis sensitivity via the Nrf2/G6PDH pathway. Our work will provide a new basis for clinical therapeutic strategies for pulmonary ischemia-reperfusion-induced acute lung injury.
Collapse
Affiliation(s)
- Liuqing Yang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, China, 430071
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Hexiao Tang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Jin Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, China, 430071
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Dawei Xu
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Rui Xuan
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Songping Xie
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430071, China.
| | - Pengfei Xu
- Department of Hepatobiliary and Pancreatic Surgery, School of Pharmaceutical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China.
| | - Xinyi Li
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China.
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, China, 430071.
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China.
| |
Collapse
|
22
|
Zheng H, Ou J, Han H, Lu Q, Shen Y. SS-31@Fer-1 Alleviates ferroptosis in hypoxia/reoxygenation cardiomyocytes via mitochondrial targeting. Biomed Pharmacother 2025; 183:117832. [PMID: 39848110 DOI: 10.1016/j.biopha.2025.117832] [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: 09/04/2024] [Revised: 01/02/2025] [Accepted: 01/09/2025] [Indexed: 01/25/2025] Open
Abstract
PURPOSE Targeting mitochondrial ferroptosis presents a promising strategy for mitigating myocardial ischemia-reperfusion (I/R) injury. This study aims to evaluate the efficacy of the mitochondrial-targeted ferroptosis inhibitor SS-31@Fer-1 (elamipretide@ferrostatin1) in reducing myocardial I/R injury. METHODS SS-31@Fer-1 was synthesized and applied to H9C2 cells subjected to hypoxia/reoxygenation (H/R) to assess its protective effects. Cytotoxicity was evaluated using a cell counting kit-8 (CCK-8) assay, with lactate dehydrogenase (LDH) and creatine kinase isoenzyme (CK-MB) levels measured. Mitochondrial reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) were assessed using Mito-SOX and JC-1 fluorescent dyes, respectively. Lipid peroxidation products, malondialdehyde (MDA) and glutathione (GSH), were quantified. Mitochondrial structure, mt-cytochrome b (mt-Cytb), and mt-ATP synthase membrane subunit 6 (mt-ATP6) were analyzed. Additionally, iron homeostasis and ferroptosis markers were evaluated. RESULTS SS-31@Fer-1 significantly improved H/R-induced cardiomyocyte viability and reduced LDH and CK-MB levels. Compared to the Fer-1 group, SS-31@Fer-1 reduced GSH and increased MDA levels, enhancing mitochondrial integrity and function. Notably, it increased mitochondrial ROS and decreased MMP, indicating a mitigation of H/R-induced cardiomyocyte cytotoxicity. Furthermore, SS-31@Fer-1 maintained cellular iron homeostasis, as evidenced by increased expression of FTH, FTMT, FPN, and ABCB8. Elevated levels of GPX4 and Nrf2 were observed, while ACSL4 and PTGS2 levels were reduced in the SS-31@Fer-1 group. CONCLUSIONS SS-31@Fer-1 effectively suppressed ferroptosis in H/R-induced cardiomyocytes by maintaining cellular iron homeostasis, improving mitochondrial function, and inhibiting oxidative stress. These findings provide novel insights and opportunities for alleviating myocardial I/R injury.
Collapse
Affiliation(s)
- Hao Zheng
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No.87, Dingjiaqiao, Gulou District, Nanjing 210009, China; Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Jinbo Ou
- Departments of Cardiology, Fudan University Zhongshan Hospital, Qingpu Branch, 1158 Park East Road, Shanghai 60518120, China
| | - Hui Han
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qizheng Lu
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Guangdong Second Provincial General Hospital, No. 466 Xingang Middle Road, Haizhu district, Guangzhou 510317, China
| | - Yunli Shen
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| |
Collapse
|
23
|
Xia Q, Liu G, Zhang L, Xie B, Deng L. Anemonin suppresses sepsis-induced acute lung injury by inactivation of nuclear factor-kappa B and activation of nuclear factor erythroid 2-related factor-2/heme oxygenase-1 pathway. FASEB J 2025; 39:e70328. [PMID: 39825692 DOI: 10.1096/fj.202401987rr] [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: 08/29/2024] [Revised: 12/27/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025]
Abstract
Sepsis-induced acute lung injury (ALI) is a common acute and severe reason of death in the intensive care unit. Although the pathogenesis is complicated and multifactorial, elevated inflammation and oxidative stress are considered as fundamental mechanisms for the progression of ALI. Anemonin is a natural compound with diverse biological properties including anti-inflammatory and anti-oxidative effects. To identify whether anemonin has protective effects on sepsis-induced ALI, a mouse sepsis-induced ALI model and cellular models using the mouse alveolar macrophage MH-S cells and mouse lung epithelial MLE-12 cells were established. Our results showed that anemonin reduced lipopolysaccharide (LPS)-induced mortality, and improved sepsis-induced ALI in the mouse model, as shown by improved histopathological changes, decreased lung wet/dry weight ratio, and myeloperoxidase activity. Anemonin alleviated LPS-induced secretion of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) in bronchoalveolar lavage fluid samples, as well as reversed the LPS-caused increase in malondialdehyde (MDA) content and decrease in activities of the antioxidant enzymes superoxide dismutase (SOD) and catalase (CAT) in lung tissues. In the cellular model, anemonin inhibited the LPS-induced inflammatory responses and oxidative stress in MH-S and MLE-12 cells. In addition, anemonin inhibited LPS-induced nuclear factor-kappa B (NF-κB) pathway, while enhancing the activation of nuclear factor erythroid 2-related factor-2 (Nrf2) in lung tissues, MH-S, and MLE-12 cells. NF-κB inhibition enhanced the anti-inflammatory and anti-oxidative effects of anemonin, while Nrf2 knockdown attenuated these effects of anemonin, implying the critical roles of NF-κB and Nrf2. These results indicated that anemonin suppressed sepsis-induced acute lung injury by inhibition of NF-κB and activation of Nrf2/heme oxygenase-1 pathway, suggesting that anemonin might be developed as a new therapeutic agent for the treatment of sepsis-induced ALI.
Collapse
Affiliation(s)
- Qingping Xia
- Department of Science and Education, Gaozhou People's Hospital, Maoming, China
| | - Guohao Liu
- Department of Medical Imaging, Affiliated Hospital of Jilin Medical University, Jilin, China
| | - Liangqing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Baodong Xie
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Li Deng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Cardiovascular Surgery, Gaozhou People's Hospital, Maoming, China
| |
Collapse
|
24
|
Wang Z, Zhang Z, Yan T, Wang Y, Li L, Li J, Zhou W. Network pharmacology-based strategy to reveal Acacetin against lipopolysaccharide-induced lung injury. Int Immunopharmacol 2025; 146:113843. [PMID: 39721450 DOI: 10.1016/j.intimp.2024.113843] [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: 10/10/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Acacetin, a flavonoid isolated from Agastache rugosa, exhibits diverse biological activities, such as anti-tumor, anti-inflammatory and antioxidant activities. Its role in treating Lipopolysaccharide (LPS)-induced acute lung injury (ALI) remains incompletely illuminated. OBJECTIVE To explore the potential molecular mechanisms of Acacetin in alleviating ALI. MATERIALS & METHODS The network pharmacological approach was employed to screen the target genes and pathways of Acacetin. Lung injury was analyzed by Hematoxylin-Eosin (H&E) staining. Bronchoalveolar lavage fluid, serum and lung tissues were collected to detect the levels of proinflammatory cytokines and oxidative stress markers. Immunofluorescence and RT-qPCR experiments were used to observe the expression of CD45, COX2, Ly6G, and related-target proteins. In vitro, RAW264.7 macrophages were stimulated with LPS and treated with AMPK siRNA or an AMPK inhibitor Coumpound C to verify the role of AMPK/nuclear factor erythroid 2-related factor 2 (Nrf2)/high-mobility group box 1 (HMGB1) signaling in Acacetin-mediated alleviation of ALI. RESULTS Network data revealed that Acacetin could regulate HMGB1, AMPK, Nrf2, and IL-6. In vivo, Acacetin reversed pathological damage and the release of inflammatory factors, and alleviated oxidative stress and immune cell infiltration in ALI development. Acacetin remarkably upregulated the expression of AMPK and Nrf2, accompanied by HMGB1 downregulation. In vitro, inhibiting AMPK reversed the effects of Acacetin in LPS-treated RAW264.7, due to inactivation of AMPK/Nrf2/HMGB1 pathway. CONCLUSION The combination of network pharmacology and experimental studies revealed the role of Acacetin in improving ALI via the AMPK/Nrf2/HMGB1 signaling axis, which provided new insights into the treatment of ALI with Acacetin as a candidate drug.
Collapse
Affiliation(s)
- Zhisen Wang
- Department of Pharmacy, Zhejiang Hospital, Hangzhou 310013, China
| | - Zhihui Zhang
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Ting Yan
- Department of Endocrinology, Huai'an Cancer Hospital, Huai'an 223200, China
| | - Yuzhen Wang
- Department of Pharmacy, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Liucheng Li
- Department of Pharmacy, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
| | - Jingduo Li
- Department of Pathology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai 200080, China.
| | - Wencheng Zhou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China.
| |
Collapse
|
25
|
Imam M, Ji J, Zhang Z, Yan S. Targeting the initiator to activate both ferroptosis and cuproptosis for breast cancer treatment: progress and possibility for clinical application. Front Pharmacol 2025; 15:1493188. [PMID: 39867656 PMCID: PMC11757020 DOI: 10.3389/fphar.2024.1493188] [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: 09/08/2024] [Accepted: 11/12/2024] [Indexed: 01/28/2025] Open
Abstract
Breast cancer is the most commonly diagnosed cancer worldwide. Metal metabolism is pivotal for regulating cell fate and drug sensitivity in breast cancer. Iron and copper are essential metal ions critical for maintaining cellular function. The accumulation of iron and copper ions triggers distinct cell death pathways, known as ferroptosis and cuproptosis, respectively. Ferroptosis is characterized by iron-dependent lipid peroxidation, while cuproptosis involves copper-induced oxidative stress. They are increasingly recognized as promising targets for the development of anticancer drugs. Recently, compelling evidence demonstrated that the interplay between ferroptosis and cuproptosis plays a crucial role in regulating breast cancer progression. This review elucidates the converging pathways of ferroptosis and cuproptosis in breast cancer. Moreover, we examined the value of genes associated with ferroptosis and cuproptosis in the clinical diagnosis and treatment of breast cancer, mainly outlining the potential for a co-targeting approach. Lastly, we delve into the current challenges and limitations of this strategy. In general, this review offers an overview of the interaction between ferroptosis and cuproptosis in breast cancer, offering valuable perspectives for further research and clinical treatment.
Collapse
Affiliation(s)
| | | | | | - Shunchao Yan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
26
|
Chen K, Lu S, Shi K, Ali MH, Liu J, Yin F, Yin W. Hyperoside attenuates sepsis-induced acute lung injury by Nrf2 activation and ferroptosis inhibition. Int Immunopharmacol 2025; 145:113734. [PMID: 39657533 DOI: 10.1016/j.intimp.2024.113734] [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: 08/21/2024] [Revised: 11/11/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024]
Abstract
Sepsis-induced acute lung injury (ALI) is a life-threatening condition associated with high morbidity and mortality rates in intensive care units (ICUs). Emerging evidence from clinical studies suggests that compounds derived from traditional Chinese medicine (TCM) have shown promising therapeutic effects in treating sepsis-induced ALI. Hyperoside is a bioactive compound extracted from TCM. Prior studies reported that hyperoside exhibits potent anti-inflammatory, antioxidant, and organ-protective properties, however, the underlying mechanisms of its effects on ALI remain unclear. Hyperoside pretreatment significantly reduced inflammation, iron accumulation, and lipid peroxidation in the pulmonary tissues of ALI mice induced by CLP and in LPS-stimulated MLE-12 cells. In particular, hyperoside preferentially binds with Keap1 at Arg380 and Arg415, thereby inhibiting the ubiquitin-mediated degradation of nuclear Nrf2, promoting its translocation to the nucleus, and leading to upregulation of anti-ferroptosis gene expression. Moreover, the protective effects of hyperoside were significantly abrogated after Nrf2 expression was silenced or its activity was inhibited by chemical inhibitors, highlighting that Nrf2 is critically involved in the impact of hyperoside. This study confirms that hyperoside exhibits a therapeutically protective effect against sepsis-induced ALI by inhibiting ferroptosis through Nrf2-mediated signaling pathway. Hyperoside acts as an Nrf2 activator by preferentially binding to Arg380 and Arg415 of Keap1 and disrupting the Keap1/Nrf2 interaction.
Collapse
Affiliation(s)
- Kuida Chen
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Shipeng Lu
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Ke Shi
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China; Medical Research Center, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu 225001, China
| | - Mustafa Hussein Ali
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jian Liu
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Fangzhou Yin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210029, China.
| | - Wu Yin
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
27
|
Wang Z, Tian J, Wang L, Yan H, Feng S, Zhang Y. PGK1 Is Involved in the HIF-1 Signaling Pathway as a Hub Gene for Ferroptosis After Traumatic Brain Injury. Mol Neurobiol 2025; 62:233-245. [PMID: 38833128 DOI: 10.1007/s12035-024-04170-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 04/03/2024] [Indexed: 06/06/2024]
Abstract
The pathogenesis of ferroptosis in traumatic brain injury (TBI) is unclear; therefore, we aimed to identify key molecules associated with ferroptosis in TBI using bioinformatics analysis to determine its underlying mechanisms. GSE128543 dataset was downloaded from the Gene Expression Omnibus (GEO) database, and TBI-associated modules were obtained by weighted gene co-expression network analysis (WGCNA). We identified 60 differentially expressed genes (DEGs) by intersecting the modules with ferroptosis and glycolysis/gluconeogenesis gene libraries. The hypoxia-inducible factor-1 (HIF-1) signaling pathway was identified to be critical for ferroptosis post-TBI, and protein-protein interaction (PPI) network identified 20 hub genes, including phosphoglycerate kinase 1 (PGK1), ribosomal protein (RP) family, pyruvate kinase M1/2 (PKM), hypoxia-inducible factor 1α subunit (HIF-1α), and MYC genes. In this study, we further explored the role of PGK1, a gene involved in HIF-1 signaling pathway; however, its role and mechanism in TBI are still unclear. Moreover, we constructed a TBI mouse model and examined PGK1 and HIF-1α expression levels, and the results revealed their expressions increased after cortical injury in mice and they co-localized in the same cells. Furthermore, we examined the expressions of PGK1 in the cerebrospinal fluid of 20 clinical patients with different degrees of brain injuries within 48 h of surgery and examined the cognitive function of patients according to the Glasgow Coma Scale (GCS). The results revealed that PGK1 expression level was negatively correlated with the severity of the brain injury. These findings suggest that PGK1 may become a potential hub gene for ferroptosis via the HIF-1 signaling pathway, second to neurological injury after TBI, thereby affecting patient prognosis.
Collapse
Affiliation(s)
- Zhao Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Jinjie Tian
- Department of Neurosurgery, The Second Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Lei Wang
- Department of Emergency Center, The Second Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Hongyan Yan
- Department of Neurosurgery, The Second Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Sujuan Feng
- Department of Neurosurgery, The Second Affiliated Hospital of Nantong University, Nantong, 226001, China.
| | - Yi Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Nantong University, Nantong, 226001, China.
| |
Collapse
|
28
|
Jin S, Wang H, Zhang Z, Yan M. Targeting Ferroptosis: Small-molecule Inducers as Novel Anticancer Agents. Anticancer Agents Med Chem 2025; 25:517-532. [PMID: 39411969 DOI: 10.2174/0118715206342278241008081126] [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: 08/03/2024] [Revised: 09/12/2024] [Accepted: 09/23/2024] [Indexed: 05/14/2025]
Abstract
Ferroptosis, a distinct form of regulated cell death characterized by iron-dependent lipid peroxidation and reactive oxygen species (ROS) accumulation, is increasingly recognized for its role in cancer development and as a potential therapeutic target. This review consolidates insights into the molecular mechanisms underpinning ferroptosis and evaluates the therapeutic potential of small-molecule inducers, such as erastin, RSL3, sulfasalazine, and sorafenib, which selectively trigger ferroptosis in cancer cells. It highlights the distinct morphological and molecular signatures of ferroptosis, its complex interplay with iron, lipid, and amino acid metabolic pathways, and the resultant implications for cancer treatment strategies. Strategic manipulation of the ferroptosis pathway offers a groundbreaking approach to cancer treatment, potentially circumventing the resistance that cancers develop against traditional apoptosis-inducing agents. Furthermore, it also emphasizes the necessity of refining these small molecules for clinical application and exploring their synergistic potential when combined with current therapies to augment overall treatment efficacy and improve patient outcomes. Ferroptosis thus emerges as a promising avenue in the realm of cancer therapy. Moving forward, research endeavors should focus on a more nuanced understanding of the interconnections between ferroptosis and other cell death modalities. Additionally, comprehensive evaluations of the long-term safety and therapeutic indices of the involved compounds are imperative. Such investigations are poised to herald a transformative shift in the paradigm of oncology, paving the way for innovative and targeted interventions.
Collapse
Affiliation(s)
- Shihao Jin
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao Road, Jinan, 250000, China
| | - Huannan Wang
- School of Pharmacy, Jining Medical University, Rizhao, 276826, China
| | - Zhen Zhang
- School of Pharmacy, Jining Medical University, Rizhao, 276826, China
| | - Maocai Yan
- School of Pharmacy, Jining Medical University, Rizhao, 276826, China
| |
Collapse
|
29
|
Li H, Wang X, Liang X, Meng M, Zhang H, Li Z, Lin Y, Li J, Ma C. Verapamil inhibits ferroptosis in septic acute lung injury by blocking L-type calcium channels. Biochem Biophys Res Commun 2025; 744:151202. [PMID: 39708394 DOI: 10.1016/j.bbrc.2024.151202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS), result from pulmonary edema and alveolar-capillary barrier disruption due to inflammation, often triggered by conditions like sepsis. Sepsis-induced ALI (SALI) involves extensive damage to vascular endothelium and alveolar epithelium, leading to respiratory failure. Our study explores ferroptosis, an iron-dependent cell death pathway, and calcium dysregulation in SALI. Elevated cytosolic calcium early in ferroptosis exacerbates lipid peroxidation and cellular damage. We investigated verapamil, a calcium channel blocker, and found it reduces calcium influx, alleviates iron overload, and decreases oxidative stress, protecting against ferroptosis-induced apoptosis in lung cells. These insights suggest targeting ferroptosis pathways, including calcium and iron homeostasis, may offer new therapeutic strategies for SALI, potentially improving outcomes in ALI/ARDS.
Collapse
Affiliation(s)
- Hongru Li
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China.
| | - Xuan Wang
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China; Diagnostic Center of Infections, The Second Hospital of Hebei Medical University, Shijiazhuang, PR China.
| | - Xiangyang Liang
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China; School and Hospital of Stomatology, Hebei Medical University, 383 Zhongshan East Road, Shijiazhuang, Hebei, PR China.
| | - Meiqi Meng
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China.
| | - Haixia Zhang
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China.
| | - Zixin Li
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China.
| | - Yushan Lin
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China.
| | - Jihong Li
- Diagnostic Center of Infections, The Second Hospital of Hebei Medical University, Shijiazhuang, PR China.
| | - Cuiqing Ma
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China.
| |
Collapse
|
30
|
Wu T, Ji M, Li T, Luo L. The molecular and metabolic landscape of ferroptosis in respiratory diseases: Pharmacological aspects. J Pharm Anal 2025; 15:101050. [PMID: 40034685 PMCID: PMC11873008 DOI: 10.1016/j.jpha.2024.101050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 03/05/2025] Open
Abstract
Ferroptosis is a form of cell death that occurs when there is an excess of reactive oxygen species (ROS), lipid peroxidation, and iron accumulation. The precise regulation of metabolic pathways, including iron, lipid, and amino acid metabolism, is crucial for cell survival. This type of cell death, which is associated with oxidative stress, is controlled by a complex network of signaling molecules and pathways. It is also implicated in various respiratory diseases such as asthma, chronic obstructive pulmonary disease (COPD), acute lung injury (ALI), lung cancer, pulmonary fibrosis (PF), and the coronavirus disease 2019 (COVID-19). To combat drug resistance, it is important to identify appropriate biological markers and treatment targets, as well as intervene in respiratory disorders to either induce or prevent ferroptosis. The focus is on the role of ferroptosis in the development of respiratory diseases and the potential of targeting ferroptosis for prevention and treatment. The review also explores the interaction between immune cell ferroptosis and inflammatory mediators in respiratory diseases, aiming to provide more effective strategies for managing cellular ferroptosis and respiratory disorders.
Collapse
Affiliation(s)
- Tong Wu
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Miaorong Ji
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| |
Collapse
|
31
|
Lu Z, Tang Y, Chen H, Liu F, Liu M, Fu L, Wang X, Li M, Yu W, Sun Y. Identification and Functional Analysis of PANoptosis-Associated Genes in the Progression From Sepsis to ARDS. Immun Inflamm Dis 2025; 13:e70136. [PMID: 39854144 PMCID: PMC11760491 DOI: 10.1002/iid3.70136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 11/07/2024] [Accepted: 01/10/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Sepsis and acute respiratory distress syndrome (ARDS) are common inflammatory conditions in intensive care, with ARDS significantly increasing mortality in septic patients. PANoptosis, a newly discovered form of programmed cell death involving multiple cell death pathways, plays a critical role in inflammatory diseases. This study aims to elucidate the PANoptosis-related genes (PRGs) and their involvement in the progression of sepsis to ARDS. METHODS This study analyzed differentially expressed genes (DEGs) associated with PRGs to explore their role in the progression of immune disorders from sepsis to septic ARDS. A diagnostic prediction model was constructed based on key PRGs identified through bioinformatics analysis. Functional enrichment analyses were conducted to determine pathway involvement, and correlations with immune cells were assessed. Mendelian randomization analysis was applied to investigate potential causal links between specific PRGs and ARDS. Immunohistochemical analysis was used to evaluate PRG expression in lung tissue. RESULTS The prediction model effectively distinguished septic ARDS patients from those with sepsis. NDRG1 expression was elevated in ARDS, while DDX3X, PTPRC, and TNFSF8 were downregulated. NDRG1 showed a positive correlation with activated dendritic cells, whereas DDX3X, PTPRC, and TNFSF8 were positively associated with neutrophils and negatively correlated with CD56bright NK cells. Functional enrichment analysis highlighted spliceosome function, MAPK signaling, endocytosis, and antigen processing pathways as significantly associated with these PRGs. Mendelian randomization suggested a causal link between NDRG1 and ARDS, and immunohistochemical analysis revealed its predominant expression near vascular walls. In a mouse model of sepsis, suppression of NDRG1 alleviated lung injury. CONCLUSION PANoptosis may contribute to immune dysregulation in sepsis-associated ARDS. NDRG1 is identified as a potential therapeutic target, offering new avenues for mitigating ARDS progression and improving patient outcomes.
Collapse
Affiliation(s)
- Zhong‐Hua Lu
- The First Department of Critical Care MedicineThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Yan Tang
- Department of Rehabilitation MedicineThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Hu Chen
- The First Department of Critical Care MedicineThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Feng Liu
- Department of Critical Care MedicineThe First Affiliated Hospital of Chongqing Medical UniversityChongqing CityChina
| | - Mei Liu
- The First Department of Critical Care MedicineThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Lu Fu
- The First Department of Critical Care MedicineThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Xian‐Kai Wang
- The First Department of Critical Care MedicineThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Ming‐Juan Li
- The First Department of Critical Care MedicineThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Wei‐Li Yu
- The First Department of Critical Care MedicineThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Yun Sun
- The First Department of Critical Care MedicineThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| |
Collapse
|
32
|
Zhang H, Ouyang Z, Zhou T, Su F, Wang M. Novel anoikis-related diagnostic biomarkers for aortic dissection based on machine learning. Sci Rep 2024; 14:31314. [PMID: 39732886 PMCID: PMC11682115 DOI: 10.1038/s41598-024-82655-6] [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: 02/14/2023] [Accepted: 12/06/2024] [Indexed: 12/30/2024] Open
Abstract
Aortic dissection (AD) is one of the most dangerous diseases of the cardiovascular system, which is characterized by acute onset and poor prognosis, while the pathogenesis of AD is still unclear and may affect or even delay the diagnosis of AD. Anchorage-dependent cell death (Anoikis) is a special mode of cell death, which is programmed cell death caused by normal cells after detachment from extracellular matrix (ECM) and has been widely studied in the field of oncology in recent years. In this study, we applied bioinformatics analysis, according to the results of research analysis and Gene Ontology (GO), as well as Kyoto Encyclopedia of Genes and Genomes (KEGG), finally found 3 anoikis-related genes (ARGs) based on machine learning. Among these, TP53 and TUBB3 were further verified by receiver operating characteristic (ROC), gene set enrichment analysis (GSEA), gene set variation analysis (GSVA)and other methods. We hypothesize ARGs may be involved in the pathogenesis of AD through pathways such as oxidative stress, inflammatory response, and ECM. Therefore, we conclude that these ARGs can be potential factors in determining the diagnosis of AD.
Collapse
Affiliation(s)
- Hanyi Zhang
- Department of Dermatology, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, Hunan, China
- Xiang Ya School of Medcine, Central South University, 172 Tongzipo Road, Changsha, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhen Ouyang
- Department of Dermatology, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Tianji Zhou
- Xiang Ya Nursing School, Central South University, 172 Tongzipo Road, Changsha, Hunan, China
| | - Feng Su
- Department of Dermatology, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, Hunan, China.
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
- Department of Department of Emergency Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410000, Hunan, China.
| | - Mi Wang
- Department of Dermatology, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, Hunan, China.
- Department of Mental Health Center, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
33
|
Qi L, Wang S, Guo T, Qi Z, Wu S, Gao D, Yan Z, Tan B, Yang A. Mechanism of Qingdai in Alleviating Acute Lung Injury by Inhibiting the JAK2/STAT3 Signaling Pathway. J Inflamm Res 2024; 17:11403-11417. [PMID: 39722733 PMCID: PMC11669285 DOI: 10.2147/jir.s498299] [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: 09/27/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024] Open
Abstract
Objective Qingdai (QD) is a traditional Chinese medicine (TCM) commonly used in clinical practice to treat acute lung injury/acute respiratory distress syndrome (ALI/ARDS). However, the mechanisms underlying the effects of QD remain not fully understood. This investigation demonstrated QD alleviated LPS-induced ALI in mice and exerted anti-inflammatory effects by inhibiting the JAK2/STAT3 signaling pathway. Methods The active compounds of QD were identified through UPLC-LTQ-Orbitrap-MS/MS. Network pharmacology predicted potential pharmacological targets and the signaling pathways contributed to the effectiveness of QD in treating ALI. Molecular docking assessed the binding of active components to critical targets. ALI mice triggered by Lipopolysaccharides (LPS) were used for transcriptomic analysis to assess alterations in pulmonary gene expression. The pathological changes of lung tissue were analyzed via HE staining. Proinflammatory cytokine levels in serum were measured using ELISA, and the mRNA expression was measured by RT-qPCR. Western blot analysis evaluated protein expression related to the JAK2/STAT3 signaling pathway. Additionally, RAW264.7 cells induced by LPS were treated with QD to measure proinflammatory cytokines and JAK2/STAT3 signaling pathway protein expression. Results Six major components of QD were identified. Network pharmacology predicted JAK2 and STAT3 as targets for QD in ALI treatment, with KEGG analysis highlighting the JAK/STAT signaling pathway. Transcriptomics confirmed the JAK/STAT signaling pathway in the therapeutic effects of QD. Molecular docking demonstrated high binding affinities of bisindigotin, isoindigo, and 6-(3-oxoindolin-2-ylidene)indolo[2,1-b]quinazolin-12-one (IQO) to JAK2 and STAT3. In vivo, QD reduced lung inflammation, downregulated proinflammatory cytokines, and inhibited JAK2/STAT3 signaling pathway. In vitro, QD mitigated LPS-triggered inflammatory responses in RAW264.7 macrophages by inhibiting the same pathway. Conclusion The therapeutic effects of QD in ALI might be mediated by the modulation of the JAK2/STAT3 signaling pathway, which may make it a valuable therapeutic strategy for ALI/ARDS.
Collapse
Affiliation(s)
- Lu Qi
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
- The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People’s Hospital, Yichang, 443003, People’s Republic of China
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Shun Wang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Tao Guo
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Zhuocao Qi
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Suwan Wu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Dan Gao
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Zhiqiang Yan
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Bo Tan
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Aidong Yang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| |
Collapse
|
34
|
Tan R, Ge C, Yan Y, Guo H, Han X, Zhu Q, Du Q. Deciphering ferroptosis in critical care: mechanisms, consequences, and therapeutic opportunities. Front Immunol 2024; 15:1511015. [PMID: 39737174 PMCID: PMC11682965 DOI: 10.3389/fimmu.2024.1511015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/03/2024] [Indexed: 01/01/2025] Open
Abstract
Ischemia-reperfusion injuries (IRI) across various organs and tissues, along with sepsis, significantly contribute to the progression of critical illnesses. These conditions disrupt the balance of inflammatory mediators and signaling pathways, resulting in impaired physiological functions in human tissues and organs. Ferroptosis, a distinct form of programmed cell death, plays a pivotal role in regulating tissue damage and modulating inflammatory responses, thereby influencing the onset and progression of severe illnesses. Recent studies highlight that pharmacological agents targeting ferroptosis-related proteins can effectively mitigate oxidative stress caused by IRI in multiple organs, alleviating associated symptoms. This manuscript delves into the mechanisms and signaling pathways underlying ferroptosis, its role in critical illnesses, and its therapeutic potential in mitigating disease progression. We aim to offer a novel perspective for advancing clinical treatments for critical illnesses.
Collapse
Affiliation(s)
- Ruimin Tan
- School of Clinical Medical, North China University of Science and Technology, Tangshan, Hebei, China
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Chen Ge
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Yating Yan
- School of Clinical Medical, North China University of Science and Technology, Tangshan, Hebei, China
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - He Guo
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
- School of Graduate, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xumin Han
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
- School of Graduate, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qiong Zhu
- Department of Orthopaedics, The People’s Hospital Of Shizhu, Chongqing, China
| | - Quansheng Du
- Critical Care Department, Hebei General Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
35
|
Huo G, Lin Y, Liu L, He Y, Qu Y, Liu Y, Zhu R, Wang B, Gong Q, Han Z, Yin H. Decoding ferroptosis: transforming orthopedic disease management. Front Pharmacol 2024; 15:1509172. [PMID: 39712490 PMCID: PMC11659002 DOI: 10.3389/fphar.2024.1509172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
As a mechanism of cell death, ferroptosis has gained popularity since 2012. The process is distinguished by iron toxicity and phospholipid accumulation, in contrast to autophagy, apoptosis, and other cell death mechanisms. It is implicated in the advancement of multiple diseases across the body. Researchers currently know that osteosarcoma, osteoporosis, and other orthopedic disorders are caused by NRF2, GPX4, and other ferroptosis star proteins. The effective relief of osteoarthritis symptoms from deterioration has been confirmed by clinical treatment with multiple ferroptosis inhibitors. At the same time, it should be reminded that the mechanisms involved in ferroptosis that regulate orthopedic diseases are not currently understood. In this manuscript, we present the discovery process of ferroptosis, the mechanisms involved in ferroptosis, and the role of ferroptosis in a variety of orthopedic diseases. We expect that this manuscript can provide a new perspective on clinical diagnosis and treatment of related diseases.
Collapse
Affiliation(s)
- Guanlin Huo
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yumeng Lin
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Lusheng Liu
- Department of Acupuncture and Moxibustion, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuqi He
- Department of Blood Transfusion, Lu’an People’s Hospital, The Affiliated Hospital of Anhui Medical University, Lu’an, China
| | - Yi Qu
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yang Liu
- Orthopaedic Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Renhe Zhu
- Department of Blood Transfusion, Lu’an People’s Hospital, The Affiliated Hospital of Anhui Medical University, Lu’an, China
| | - Bo Wang
- Department of Orthopaedics, The Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Qing Gong
- Orthopaedic Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Zhongyu Han
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Hongbing Yin
- Orthopedic Center, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
36
|
Wang Q, Yu ZH, Nie L, Wang FX, Mu G, Lu B. Assessing the impact of gut microbiota and metabolic products on acute lung injury following intestinal ischemia-reperfusion injury: harmful or helpful? Front Cell Infect Microbiol 2024; 14:1491639. [PMID: 39687547 PMCID: PMC11647003 DOI: 10.3389/fcimb.2024.1491639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Ischemia-reperfusion injury (IRI) is a common and clinically significant form of tissue damage encountered in medical practice. This pathological process has been thoroughly investigated across a variety of clinical settings, including, but not limited to, sepsis, organ transplantation, shock, myocardial infarction, cerebral ischemia, and stroke. Intestinal IRI, in particular, is increasingly recognized as a significant clinical entity due to marked changes in the gut microbiota and their metabolic products, often described as the body's "second genome." These changes in intestinal IRI lead to profound alterations in the gut microbiota and their metabolic outputs, impacting not only the pathology of intestinal IRI itself but also influencing the function of other organs through various mechanisms. Notable among these are brain, liver, and kidney injuries, with acute lung injury being especially significant. This review seeks to explore in depth the roles and mechanisms of the gut microbiota and their metabolic products in the progression of acute lung injury initiated by intestinal IRI, aiming to provide a theoretical basis and directions for future research into the treatment of related conditions.
Collapse
Affiliation(s)
- Qiong Wang
- Department of Anesthesiology, Zigong Fourth People’s Hospital, Zigong, Sichuan, China
| | - Zi-Hang Yu
- Department of Anesthesiology, Fushun County People’s Hospital, Zigong, Sichuan, China
| | - Liang Nie
- Department of Anesthesiology, Fushun County People’s Hospital, Zigong, Sichuan, China
| | - Fei-Xiang Wang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Guo Mu
- Department of Anesthesiology, Zigong Fourth People’s Hospital, Zigong, Sichuan, China
| | - Bin Lu
- Department of Anesthesiology, Zigong Fourth People’s Hospital, Zigong, Sichuan, China
| |
Collapse
|
37
|
Zhou Q, Meng Y, Le J, Sun Y, Dian Y, Yao L, Xiong Y, Zeng F, Chen X, Deng G. Ferroptosis: mechanisms and therapeutic targets. MedComm (Beijing) 2024; 5:e70010. [PMID: 39568772 PMCID: PMC11577302 DOI: 10.1002/mco2.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
Ferroptosis is a nonapoptotic form of cell death characterized by iron-dependent lipid peroxidation in membrane phospholipids. Since its identification in 2012, extensive research has unveiled its involvement in the pathophysiology of numerous diseases, including cancers, neurodegenerative disorders, organ injuries, infectious diseases, autoimmune conditions, metabolic disorders, and skin diseases. Oxidizable lipids, overload iron, and compromised antioxidant systems are known as critical prerequisites for driving overwhelming lipid peroxidation, ultimately leading to plasma membrane rupture and ferroptotic cell death. However, the precise regulatory networks governing ferroptosis and ferroptosis-targeted therapy in these diseases remain largely undefined, hindering the development of pharmacological agonists and antagonists. In this review, we first elucidate core mechanisms of ferroptosis and summarize its epigenetic modifications (e.g., histone modifications, DNA methylation, noncoding RNAs, and N6-methyladenosine modification) and nonepigenetic modifications (e.g., genetic mutations, transcriptional regulation, and posttranslational modifications). We then discuss the association between ferroptosis and disease pathogenesis and explore therapeutic approaches for targeting ferroptosis. We also introduce potential clinical monitoring strategies for ferroptosis. Finally, we put forward several unresolved issues in which progress is needed to better understand ferroptosis. We hope this review will offer promise for the clinical application of ferroptosis-targeted therapies in the context of human health and disease.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yu Meng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Jiayuan Le
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yating Dian
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Lei Yao
- Department of General Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yixiao Xiong
- Department of Dermatology Tongji Hospital Huazhong University of Science and Technology Wuhan Hubei China
| | - Furong Zeng
- Department of Oncology Xiangya Hospital Central South University Changsha Hunan Province China
| | - Xiang Chen
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Guangtong Deng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| |
Collapse
|
38
|
Chen X, Dumbuya JS, Du J, Xue L, Zeng Q. Bovine pulmonary surfactant alleviates inflammation and epithelial cell apoptosis in the early phase of lipopolysaccharide-induced acute lung injury in rats. Biotechnol Genet Eng Rev 2024; 40:4361-4379. [PMID: 37154048 DOI: 10.1080/02648725.2023.2210452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/01/2023] [Indexed: 05/10/2023]
Abstract
We investigate the impact of bovine pulmonary surfactant (PS) on LPS-induced ALI in vitro and in vivo to improve recognition and prevent mortality in sepsis-induced ALI. Primary alveolar type II (AT2) cells were treated with LPS alone or in combination with PS. Cell morphology observation, CCK-8 proliferation assay, flow cytometry apoptosis assay, and ELISA for inflammatory cytokine levels were performed at different time points after treatment. An LPS-induced ALI rat model was established and treated with vehicle or PS. Lung wet/dry weight ratio, histopathological changes, lung function parameters, and serum inflammatory cytokine levels were examined 6 h after PS treatment. Survival analysis by Kaplan-Meier method. RNA sequencing was conducted to identify LPS-induced differentially expressed genes in rat lungs. Proapoptotic gene expression in rat lungs was determined by Western blot. LPS significantly inhibited cell proliferation while promoting apoptosis of AT2 cells starting 2 h after treatment, accompanied by a significant increase in inflammatory cytokine production; PS reversed these effects. PS decreased the lung wet/dry ratio in septic rats, histological abnormalities, alterations in lung function parameters, and inflammatory cytokines production; while improving the overall survival of rats. LPS-induced differentially expressed genes were closely associated with apoptosis. PS attenuated LPS-induced upregulation of proapoptotic gene expression starting 2 h after treatment in AT2 cells while restoring lung ATPase activity in vivo. Bovine PS alleviates LPS-induced ALI in the early phase, possibly by suppressing inflammation and AT2 cell apoptosis, as a preemptive therapeutic agent for managing sepsis-induced ALI.
Collapse
Affiliation(s)
- Xinxin Chen
- Department of Pediatrics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - John Sieh Dumbuya
- Department of Pediatrics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiang Du
- Department of Pediatrics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lijun Xue
- Department of neonatology, Songgang People's Hospital, Shenzhen, China
| | - Qiyi Zeng
- Department of Pediatrics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
39
|
Liu YY, Bao DQ, Zhang ZS, Zhu Y, Liu LM, Li T. Radix Sanguisorbae Improves Intestinal Barrier in Septic Rats via HIF-1 α/HO-1/Fe 2+ Axis. Chin J Integr Med 2024; 30:1101-1112. [PMID: 38212494 DOI: 10.1007/s11655-023-3550-2] [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] [Accepted: 02/28/2023] [Indexed: 01/13/2024]
Abstract
OBJECTIVE To investigate whether Radix Sanguisorbae (RS, Diyu) could restore intestinal barrier function following sepsis using a cecal ligation and puncture (CLP)-induced septic rat model and lipopolysaccharide (LPS)-challenged IEC-6 cell model, respectively. METHODS Totally 224 rats were divided into 4 groups including a control, sham, CLP and RS group according to a random number table. The rats in the control group were administrated with Ringer's lactate solution (30 mL/kg) with additional dopamine [10 µ g/(kg·min)] and given intramuscular injections of cefuroxime sodium (10 mg/kg) 12 h following CLP. The rats in the RS group were administrated with RS (10 mg/kg) through tail vein 1 h before CLP and treated with RS (10 mg/kg) 12 h following CLP. The rats in the sham group were only performed abdominal surgery without CLP. The rats in the CLP group were performed with CLP without any treatment. The other steps were same as control group. The effects of RS on intestinal barrier function, mesenteric microvessels barrier function, multi-organ function indicators, inflammatory response and 72 h survival window following sepsis were observed. In vitro, the effects of RS on LPS-challenged IEC-6 cell viability, the expressions of zona occludens-1 (ZO-1) and ferroptosis index were evaluated by cell counting kit-8, immunofluorescence and Western blot analysis. Bioinformatic tools were applied to investigate the pharmacological network of RS in sepsis to predict the active compounds and potential protein targets and pathways. RESULTS The sepsis caused severe intestinal barrier dysfunction, multi-organ injury, lipid peroxidation accumulation, and ferroptosis in vivo. RS treatment significantly prolonged the survival time to 56 h and increased 72-h survival rate to 7/16 (43.75%). RS also improved intestinal barrier function and relieved intestinal inflammation. Moreover, RS significantly decreased lipid peroxidation and inhibited ferroptosis (P<0.05 or P<0.01). Administration of RS significantly worked better than Ringer's solution used alone. Using network pharmacology prediction, we found that ferroptosis and hypoxia inducible factor-1 (HIF-1 α) signaling pathways might be involved in RS effects on sepsis. Subsequent Western blot, ferrous iron measurements, and FerroOrange fluorescence of ferrous iron verified the network pharmacology predictions. CONCLUSION RS improved the intestinal barrier function and alleviated intestinal injury by inhibiting ferroptosis, which was related in part to HIF-1 α/heme oxygenase-1/Fe2+ axis.
Collapse
Affiliation(s)
- Yi-Yan Liu
- State Key Laboratory of Trauma, Burns and Combined Injuries, Department of War Wound Shock and Transfusion, Institute of Surgery Research, Chongqing, 400042, China
| | - Dai-Qin Bao
- Department of Anesthesiology, Army Medical Center of PLA, Chongqing, 400042, China
| | - Zi-Sen Zhang
- State Key Laboratory of Trauma, Burns and Combined Injuries, Department of War Wound Shock and Transfusion, Institute of Surgery Research, Chongqing, 400042, China
| | - Yu Zhu
- State Key Laboratory of Trauma, Burns and Combined Injuries, Department of War Wound Shock and Transfusion, Institute of Surgery Research, Chongqing, 400042, China
| | - Liang-Ming Liu
- State Key Laboratory of Trauma, Burns and Combined Injuries, Department of War Wound Shock and Transfusion, Institute of Surgery Research, Chongqing, 400042, China
| | - Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injuries, Department of War Wound Shock and Transfusion, Institute of Surgery Research, Chongqing, 400042, China.
| |
Collapse
|
40
|
Zhang YJ, Chen LY, Lin F, Zhang X, Xiang HF, Rao Q. ROS responsive nanozyme loaded with STING silencing for the treatment of sepsis-induced acute lung injury. Toxicol Appl Pharmacol 2024; 493:117155. [PMID: 39537108 DOI: 10.1016/j.taap.2024.117155] [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: 06/28/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Acute lung injury (ALI) is a common complication of sepsis and a leading cause of mortality in septic patients. Studies indicate that STING may play a crucial role in the pathogenesis of sepsis-induced ALI by interacting with the PARP-1/NLRP3 pathway. Therefore, targeting STING inhibition has potential as a novel therapeutic strategy for ALI. However, effective inhibition remains challenging due to the widespread expression of STING across various tissues. In this study, we developed a nanozyme-based drug delivery system, DSPE-TK-mPEG-MnO2@siSTING (abbreviated as DTmM@siSTING), using DSPE-TK-mPEG-MnO2 as the carrier, and characterized it via scanning electron microscopy, dynamic light scattering, nanoparticle size analysis, and gel electrophoresis. To evaluate the therapeutic effects of DTmM@siSTING, an in vitro ALI cell model and an in vivo ALI mouse model were established, assessing the nanozyme's impact on ROS levels, inflammatory responses, and the PARP-1/NLRP3 pathway in sepsis-induced ALI. Results demonstrated that DTmM@siSTING exhibited good physiological stability. In vitro, DTmM@siSTING significantly reduced ROS levels, myeloperoxidase activity, and expression of inflammatory cytokines, while also inhibiting PARP-1/NLRP3 pathway activation. In vivo experiments further revealed that DTmM@siSTING effectively delivered siSTING to the lungs, mitigating sepsis-induced ALI and associated inflammatory responses. Additionally, DTmM@siSTING displayed excellent biocompatibility. In summary, our findings suggest that DTmM@siSTING significantly enhances the therapeutic efficacy of siSTING, alleviating ALI by inhibiting ROS production, inflammatory responses, and activation of the PARP-1/NLRP3 pathway. This novel approach presents a promising therapeutic avenue for sepsis-induced ALI.
Collapse
Affiliation(s)
- Yin-Jin Zhang
- Blood Purification Center, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Ling-Yang Chen
- Blood Purification Center, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Feng Lin
- Department of Anesthesiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Xia Zhang
- Department of Anesthesiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Hai-Fei Xiang
- Department of Anesthesiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, China.
| | - Qing Rao
- Department of Anesthesiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, China.
| |
Collapse
|
41
|
Zhang S, Yang L, Duoji Z, Qiangba D, Hu X, Jiang Z, Hou D, Hu Z, Basang Z. DNA Methylation Changes and Phenotypic Adaptations Induced Repeated Extreme Altitude Exposure at 8848 Meters. Int J Mol Sci 2024; 25:12652. [PMID: 39684363 DOI: 10.3390/ijms252312652] [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: 07/15/2024] [Revised: 08/13/2024] [Accepted: 08/23/2024] [Indexed: 12/18/2024] Open
Abstract
Repeated extreme environmental training (RET) enhances adaptability and induces lasting methylation modifications. We recruited 64 participants from a high-altitude region (4700 m), including 32 volunteers with repeated high-altitude exposure, reaching up to 8848 m and as many as 11 exposures. By analyzing 741,489 CpG loci and 39 phenotypes, we identified significant changes in 13 CpG loci (R2 > 0.8, ACC > 0.75) and 15 phenotypes correlated with increasing RET exposures. The phenotypic Bayesian causal network and phenotypic-CpG interaction networks showed greater robustness (node correlation) with more RET exposures, particularly in systolic blood pressure (SP), platelet count (PLT), and neutrophil count (NEUT). Six CpG sites were validated as significantly associated with hypoxia exposure using the GEO public da-taset (AltitudeOmics). Furthermore, dividing the participants into two groups based on the number of RET exposures (n = 9 and 4) revealed six CpG sites significantly corre-lated with PLT and red cell distribution width-standard deviation (RDW.SD). Our findings suggest that increased RET exposures strengthen the interactions between phenotypes and CpG sites, indicating that critical extreme adaptive states may alter methylation patterns, co-evolving with phenotypes such as PLT, RDW.SD, and NEUT.
Collapse
Affiliation(s)
- Shixuan Zhang
- High Altitude Health Science Research Centre of Tibet University, Tibet University, 10 East Zangda Road, Lhasa 850000, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences & Human Phenome Institute, Fudan University, Shanghai 200438, China
| | - La Yang
- High Altitude Health Science Research Centre of Tibet University, Tibet University, 10 East Zangda Road, Lhasa 850000, China
| | - Zhuoma Duoji
- High Altitude Health Science Research Centre of Tibet University, Tibet University, 10 East Zangda Road, Lhasa 850000, China
| | - Danzeng Qiangba
- High Altitude Health Science Research Centre of Tibet University, Tibet University, 10 East Zangda Road, Lhasa 850000, China
| | - Xiaoxi Hu
- State Key Laboratory of Genetic Engineering, School of Life Sciences & Human Phenome Institute, Fudan University, Shanghai 200438, China
| | - Zeyu Jiang
- State Key Laboratory of Genetic Engineering, School of Life Sciences & Human Phenome Institute, Fudan University, Shanghai 200438, China
| | - Dandan Hou
- State Key Laboratory of Genetic Engineering, School of Life Sciences & Human Phenome Institute, Fudan University, Shanghai 200438, China
| | - Zixin Hu
- Artificial Intelligence Innovation and Incubation Institute of Fudan University, Shanghai 200438, China
| | - Zhuoma Basang
- High Altitude Health Science Research Centre of Tibet University, Tibet University, 10 East Zangda Road, Lhasa 850000, China
| |
Collapse
|
42
|
Fan W, Tang Y, Liu Y, Ran Y, Pan G, Song X, Mai L, Jiang X, Chen D, Song F, Li H. ROS-responsive nanoparticles for bioimaging and treating acute lung injury by releasing dexamethasone and improving alveolar macrophage homeostasis. J Nanobiotechnology 2024; 22:729. [PMID: 39578796 PMCID: PMC11585220 DOI: 10.1186/s12951-024-03015-0] [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: 07/12/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Acute lung injury (ALI) triggers the activation of pulmonary macrophages, which in turn produce excessive amounts of reactive oxygen species (ROS). RESULTS We synthesized ROS-responsive red light-emitting carbon dots (RCMNs) that target lung macrophages, possess bioimaging capabilities, and efficiently eliminate intracellular ROS, thereby demonstrating anti-inflammatory effects for treating acute lung injury (ALI). In an LPS-induced ALI mouse model, RCMNs showed bioimaging and therapeutic potential, reducing lung damage and inflammation by targeting ROS-damaged tissue. RCMNs also improved alveolar macrophage activity, decreased inflammatory cytokines (TNF-α and IL-6), and enhanced survival in endotoxic shock, indicating their therapeutic potential for ALI. RNA-seq analysis revealed that RCMNs modulate signaling pathways related to calcium, TNF, and Toll-like receptors, highlighting their role in regulating inflammation and immune responses. Mechanistically, RCMNs alleviate inflammation in ALI by enhancing mitochondrial function in lung macrophages, as evidenced by improved mitochondrial morphology and membrane potential. CONCLUSIONS This protective effect is mediated through the regulation of intracellular Ca2+ levels and mitochondrial respiratory chain complexes, suggesting RCMNs as a therapeutic strategy for mitochondrial dysfunction in ALI.
Collapse
Affiliation(s)
- Wenna Fan
- Institute of Intelligent Chinese Medicine, Chongqing University of Chinese Medicine, Chongqing, 402760, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Yongyao Tang
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Yamin Liu
- Department of Obstetrics and Gyecology, Women and Children's Hospital of Chongqing Medical University/Chongqing Health Center for Women and Children, Chongqing, 400016, China
| | - Ya Ran
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
- General Surgery, Qianjiang Districet Chinese Medicine Hospital, Qianjiang District, Chongqing, 409000, China
| | - Guangrui Pan
- General Surgery (Breast Surgery), The Affiliated Hospital of Southwest Medical University, 25 Taiping Rd, Jiangyang District, Luzhou, 646000, Sichuan, China
| | - Xin Song
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Li Mai
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Xue Jiang
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Dan Chen
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Fangzhou Song
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China.
| | - Haiyu Li
- Institute of Intelligent Chinese Medicine, Chongqing University of Chinese Medicine, Chongqing, 402760, China.
| |
Collapse
|
43
|
Chen Y, Jiang Z, Li X. New insights into crosstalk between Nrf2 pathway and ferroptosis in lung disease. Cell Death Dis 2024; 15:841. [PMID: 39557840 PMCID: PMC11574213 DOI: 10.1038/s41419-024-07224-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/20/2024]
Abstract
Ferroptosis is a distinctive process of cellular demise that is linked to amino acid metabolism, lipid oxidation, and iron oxidation. The ferroptosis cascade genes, which are closely associated with the onset of lung diseases, are among the regulatory targets of nuclear factor erythroid 2-related factor 2 (Nrf2). Although the regulation of ferroptosis is mostly mediated by Nrf2, the precise roles and underlying regulatory mechanisms of ferroptosis and Nrf2 in lung illness remain unclear. This review provides new insights from recent discoveries involving the modulation of Nrf2 and ferroptosis in a range of lung diseases. It also systematically describes regulatory mechanisms involving lipid peroxidation, intracellular antioxidant levels, ubiquitination of Nrf2, and expression of FSP1 and GPX4. Finally, it summarises active ingredients and drugs with potential for the treatment of lung diseases. With the overarching aim of expediting improvements in treatment, this review provides a reference for novel therapeutic mechanisms and offers suggestions for the development of new medications for a variety of lung disorders.
Collapse
Affiliation(s)
- Yonghu Chen
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, 133002, P. R. China
| | - Zhe Jiang
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, 133002, P. R. China.
| | - Xuezheng Li
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, 133002, P. R. China.
| |
Collapse
|
44
|
Shi J, Chen W, Tang J, Zhang C, Qi M, Zheng X, Wang J, Liu Q, Liu L, Chen X, Han Z. Huperzine A protected against ferroptosis via activating PI3K/Akt signaling in lipopolysaccharide induced acute lung injury. Eur J Pharmacol 2024; 983:177004. [PMID: 39278310 DOI: 10.1016/j.ejphar.2024.177004] [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: 11/19/2023] [Revised: 08/17/2024] [Accepted: 09/13/2024] [Indexed: 09/18/2024]
Abstract
Huperzine A (Hup A), an extract from Huperzia serrata, exerted its anti-inflammation and anti-oxidation effect to protect against neurodegenerative disorders and organ injury. Ferroptosis was indicated to involve in the development of acute lung injury (ALI) accompanying by lipid reactive oxygen species (ROS) overexpressed. However, there is little research focused on the protective effect of Hup A on ALI, and the underlying molecular mechanism remains elusive. This study aims to determine the therapeutic effect of Hup A on ALI in vivo and in vitro. Hup A attenuated lung injury and cellular damage in lipopolysaccharide-induced ALI (LPS-ALI) models, both in vivo and in vitro, accompanied by the upregulation of ferroptosis-associated proteins (SLC7A11 and GPX4). Furthermore, the pretreatment with Hup A decreased the abundance of inflammation factors (IL-6, TNF-α), MDA, lipid ROS, and Fe2+ in the LPS-ALI model, while it also promoted the secretion of SOD and GSH to antagonize peroxidation. Mechanistically, RNA sequencing and network pharmacological analysis synergistically revealed the PI3K/Akt signaling pathway as a potential target of Hup A. In vitro experiments demonstrated that Hup A effectively activated GPX4 through the PI3K/Akt signaling pathway, which was subsequently reversed by LY294002, an inhibitor of the PI3K/Akt signaling pathway. Consequently, our results revealed that Hup A inhibited ferroptosis in LPS-ALI by activating the PI3K-Akt signaling pathway which indicated the potential therapeutical effect of Hup A and further emphasized the pivotal role of ferroptosis in ALI.
Collapse
Affiliation(s)
- Jun Shi
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China; School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Wei Chen
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China; College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing, 100091, China
| | - Jiajia Tang
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China; School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Chunyang Zhang
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China; College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing, 100091, China
| | - Man Qi
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China; College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing, 100091, China
| | - Xin Zheng
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Jiaxin Wang
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Qi Liu
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Lu Liu
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China; School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Xuxin Chen
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China; School of Medicine, South China University of Technology, Guangzhou, 510006, China; College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing, 100091, China.
| | - Zhihai Han
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China; School of Medicine, South China University of Technology, Guangzhou, 510006, China; College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing, 100091, China.
| |
Collapse
|
45
|
Cheng Y, Zhang H, Guan B, Zhang Y, Qin C, Li D, Zhang J, Zhang B, Lin Y, Li F. CircCDR1as orchestrates the advancement of asthma triggered by PM 2.5 through the modulation of ferroptosis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 950:175328. [PMID: 39117210 DOI: 10.1016/j.scitotenv.2024.175328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 07/27/2024] [Accepted: 08/04/2024] [Indexed: 08/10/2024]
Abstract
Exposure to fine particulate matter (PM2.5) in the ambient environment augments susceptibility to respiratory ailments. Circular RNAs, a distinctive subclass of endogenous non-coding RNAs, have been acknowledged as pivotal regulators of pathological conditions. Ferroptosis, an innovative iron-dependent form of cellular demise, has emerged as a consequential participant in numerous maladies. Despite the established association between PM2.5 exposure and the exacerbation of asthma, scant investigations have probed into the implication of circRNAs and ferroptosis in PM2.5-induced asthma. Consequently, this inquiry sought to scrutinize the potential involvement of circCDR1as and ferroptosis in PM2.5-induced asthma. Through the formulation of a PM2.5 exposure model in asthmatic mice and an in vitro cellular model, it was discerned that PM2.5 induced ferroptosis, thereby intensifying asthma progression. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) revealed an upregulation of circCDR1as in the PM2.5-stimulated asthma cell model. Molecular biology assays demonstrated that diminished circCDR1as expression hindered the onset of ferroptosis in response to PM2.5 exposure. Notably, Ferrostatin-1 (Fer-1), an inhibitor of ferroptosis, manifested the ability to impede the advancement of asthma. Mechanistically, RNA pull-down and molecular biology experiments substantiated that circCDR1as selectively bound to insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2), thereby modulating the occurrence of ferroptosis. CircCDR1as emerged as a potential orchestrator of asthma progression by regulating ferroptosis under PM2.5 exposure. Additionally, PM2.5 exposure elicited activation of the Wnt/β-catenin signaling pathway, subsequently influencing the expression of C-myc and Cyclin D1, ultimately exacerbating asthma development. In summation, the interaction between circCDR1as and IGF2BP2 in regulating ferroptosis was identified as a critical facet in the progression of asthma under PM2.5 exposure. This investigation underscores the pivotal roles of circCDR1as and ferroptosis in PM2.5-induced asthma, offering a novel theoretical foundation for the therapeutic and preventive approaches to asthma.
Collapse
Affiliation(s)
- Yu Cheng
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China
| | - Haimin Zhang
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China
| | - Boyu Guan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Yong Zhang
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China
| | - Chuhao Qin
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China
| | - Dongsheng Li
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China
| | - Jiahui Zhang
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China
| | - Boyu Zhang
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China
| | - Yingwei Lin
- Department of Laboratory Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China.
| | - Fasheng Li
- College of Medical Laboratory, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
46
|
Dai Y, Chen J, Duan Q. Epigenetic mechanism of EZH2-mediated histone methylation modification in regulating ferroptosis of alveolar epithelial cells in sepsis-induced acute lung injury. Drug Dev Res 2024; 85:e22263. [PMID: 39344139 DOI: 10.1002/ddr.22263] [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: 04/28/2024] [Revised: 07/05/2024] [Accepted: 09/12/2024] [Indexed: 10/01/2024]
Abstract
Sepsis-induced acute lung injury (SI-ALI) leads to significant deaths in critically ill patients worldwide. This study explores the mechanism of EZH2 regulating ferroptosis of alveolar epithelial cells (AECs) in SI-ALI. In vitro cell model and in vivo mouse lung injury model of sepsis were established. EZH2 expression in lung tissues was intervened by sh-EZH2, followed by H&E staining observation of lung tissue pathological changes. EZH2, H3K27me3, USP10, GPX4, and ACSL4 expressions were determined by qRT-PCR or Western blot. ROS, GSH, and iron ion levels were detected using fluorescent labeling and reagent kits, respectively. ChIP analyzed the enrichment of EZH2 and H3K27me3 on USP10 promoter. The binding between USP10 and GPX4, and the ubiquitination level of GPX4 were detected using Co-IP. EZH2 was highly expressed in lung tissues of SI-ALI mice. EZH2 silencing alleviated ALI and ferroptosis of AECs; EZH2 increased the H3K27me3 level on USP10 promoter through histone methylation. USP10 stabilized GPX4 protein expression through ubiquitination; inhibition of USP10 partially reversed the inhibitory effect of EZH2 silencing on ferroptosis of AECs. In conclusion, EZH2 depresses USP10 expression by promoting histone H3K27me3 modification on USP10 promoter, thereby enhancing ubiquitination degradation of GPX4 and ultimately facilitating ferroptosis of AECs in sepsis.
Collapse
Affiliation(s)
- Ying Dai
- Department of General Pediatrics, Taizhou People's Hospital, Taizhou, 225300, China
| | - Jiebin Chen
- Department of General Pediatrics, Taizhou People's Hospital, Taizhou, 225300, China
| | - Qingning Duan
- Department of General Pediatrics, Taizhou People's Hospital, Taizhou, 225300, China
| |
Collapse
|
47
|
Li K, Wang XQ, Liao ZL, Liu JY, Feng BH, Ren YC, Dai NN, Yu K, Yu H, Chen HJ, Mei H, Qin S. Wedelolactone inhibits ferroptosis and alleviates hyperoxia-induced acute lung injury via the Nrf2/HO-1 signaling pathway. Toxicol Sci 2024; 202:25-35. [PMID: 39110510 DOI: 10.1093/toxsci/kfae099] [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] [Indexed: 10/29/2024] Open
Abstract
Hyperoxia-induced acute lung injury (HALI) is a complication of oxygen therapy. Ferroptosis is a vital factor in HALI. This paper was anticipated to investigate the underlying mechanism of wedelolactone (WED) on ferroptosis in HALI. The current study used hyperoxia to injure two models, one HALI mouse model and one MLE-12 cell injury model. We found that WED treatment attenuated HALI by decreasing the lung injury score and lung wet/dry (W/D) weight ratio and alleviating pathomorphological changes. Then, the inflammatory reaction and apoptosis in HALI mice and hyperoxia-mediated MLE-12 cells were inhibited by WED treatment. Moreover, WED alleviated ferroptosis with less iron accumulation and reversed expression alterations of ferroptosis markers, including MDA, GSH, GPX4, SLC7A11, FTH1, and TFR1 in hyperoxia-induced MLE-12 cells in vitro and in vivo. Nrf2-KO mice and Nrf2 inhibitor (ML385) decreased WED's ability to protect against apoptosis, inflammatory response, and ferroptosis in hyperoxia-induced MLE-12 cells. Collectively, our data highlighted the alleviatory role of WED in HALI by activating the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Kang Li
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Xiao-Qin Wang
- Department of Pediatric, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Zhen-Liang Liao
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jun-Ya Liu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Bang-Hai Feng
- Department of Critical Care Medicine, Zunyi Hospital of Traditional Chinese Medicine, Zunyi, Guizhou 563000, P.R. China
| | - Ying-Cong Ren
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Ni-Nan Dai
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Kun Yu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Hong Yu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Hua-Jun Chen
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Hong Mei
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Song Qin
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
48
|
Zhan M, Liu D, Yao L, Wang W, Zhang R, Xu Y, Wang Z, Yan Q, Fang Q, Du J, Chen L. Gas6/AXL Alleviates Hepatic Ischemia/Reperfusion Injury by Inhibiting Ferroptosis via the PI3K/AKT Pathway. Transplantation 2024; 108:e357-e369. [PMID: 38725107 PMCID: PMC11495534 DOI: 10.1097/tp.0000000000005036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/08/2024] [Accepted: 02/16/2024] [Indexed: 10/24/2024]
Abstract
BACKGROUND Hepatic ischemia/reperfusion (I/R) injury is a major cause of complications in clinical liver surgery. AXL receptor tyrosine kinase (AXL) is a member of the TAM receptor tyrosine kinase family (TYRO3, AXL, and MERTK). Our previous study has shown that AXL expression was markedly upregulated in liver transplantation patients. However, the underlying mechanism of AXL in hepatic I/R injury remains unclear. METHODS A mouse liver warm I/R model and a primary hepatocyte hypoxia/reoxygenation model were established to investigate the role of AXL activation and ferroptosis in hepatic I/R injury by pretreating with recombinant mouse growth arrest-specific protein 6 (AXL activator) or R428 (AXL inhibitor). Moreover, we used LY294002 (phosphatidylinositol 3-kinase [PI3K] inhibitor) to evaluate the relationship between the PI3K/AKT (the Ser and Thr kinase AKT) pathway and ferroptosis in hepatic I/R injury. RESULTS Hepatic I/R injury decreased phosphorylation AXL expression and enhanced ferroptosis in liver transplantation patients and hepatic I/R-subjected mice. AXL activation attenuated lipid peroxidation and ferroptosis in hepatic I/R injury in vivo and in vitro. Inhibition of AXL activation exacerbated liver pathological damage and liver dysfunction, as well as iron accumulation and lipid peroxidation in hepatic I/R injury. Mechanistically, activated growth arrest-specific protein 6/AXL and its downstream PI3K/AKT signaling pathway inhibited ferroptosis during hepatic I/R injury. CONCLUSIONS AXL activation protects against hepatic I/R injury by preventing ferroptosis through the PI3K/AKT pathway. This study is the first investigation on the AXL receptor and ferroptosis, and activating AXL to mitigate ferroptosis may be an innovative therapeutic strategy to combat hepatic I/R injury.
Collapse
Affiliation(s)
- Mengting Zhan
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Deng Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Lei Yao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Weizhi Wang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Ruixin Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Yaru Xu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Zhen Wang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Anesthesiology, Ningbo First Hospital, Ningbo, China
| | - Qi Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Qi Fang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Jian Du
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Infectious Disease Research Center, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Lijian Chen
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| |
Collapse
|
49
|
Deng M, Chen S, Wu J, Su L, Xu Z, Jiang C, Sheng L, Yang X, Zeng L, Wang J, Dai W. Exploring the anti-inflammatory and immune regulatory effects of Taohe Chengqi decoction in sepsis-induced lung injury. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118404. [PMID: 38824977 DOI: 10.1016/j.jep.2024.118404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sepsis presents complex pathophysiological challenges. Taohe Chengqi Decoction (THCQ), a traditional Chinese medicine, offers potential in managing sepsis-related complications, though its exact mechanisms are not fully understood. AIM OF THE STUDY This research aimed to assess the therapeutic efficacy and underlying mechanisms of THCQ on sepsis-induced lung injury. MATERIALS AND METHODS The study began with validating THCQ's anti-inflammatory effects through in vitro and in vivo experiments. Network pharmacology was employed for mechanistic exploration, incorporating GO, KEGG, and PPI analyses of targets. Hub gene-immune cell correlations were assessed using CIBERSORT, with further scrutiny at clinical and single-cell levels. Molecular docking explored THCQ's drug-gene interactions, culminating in qPCR and WB validations of hub gene expressions in sepsis and post-THCQ treatment scenarios. RESULTS THCQ demonstrated efficacy in modulating inflammatory responses in sepsis, identified through network pharmacology. Key genes like MAPK14, MAPK3, MMP9, STAT3, LYN, AKT1, PTPN11, and HSP90AA1 emerged as central targets. Molecular docking revealed interactions between these genes and THCQ components. qPCR results showed significant modulation of these genes, indicating THCQ's potential in reducing inflammation and regulating immune responses in sepsis. CONCLUSION This study sheds light on THCQ's anti-inflammatory and immune regulatory mechanisms in sepsis, providing a foundation for further research and potential clinical application.
Collapse
Affiliation(s)
- Mingtao Deng
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China; Department of Medical Technology, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Siqi Chen
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China; Department of Medical Technology, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Jian Wu
- Department of Medical Technology, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Liling Su
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Zijin Xu
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Changrun Jiang
- Department of Critical Care Medicine, The First Affiliated Hospital of Jiangxi Medical College, No. 31 Qingfeng Road, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Lei Sheng
- Department of Critical Care Medicine, The First Affiliated Hospital of Jiangxi Medical College, No. 31 Qingfeng Road, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Xinyi Yang
- Department of Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yongwaizheng Street, Dong Lake District, Nanchang, Jiangxi Province, 330000, People's Republic of China
| | - Long Zeng
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Jingwei Wang
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China
| | - Wei Dai
- Shangrao Key Laboratory of Health Hazards and Bioprevention of Heavy Metals, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China; Department of Critical Care Medicine, The First Affiliated Hospital of Jiangxi Medical College, No. 31 Qingfeng Road, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China; Department of Clinical Medicine, Jiangxi Medical College, No. 399 Zhimin Avenue, Xinzhou District, Shangrao, Jiangxi Province, 334000, People's Republic of China.
| |
Collapse
|
50
|
Li Y, Shimizu H, Nakamura R, Lu Y, Sakamoto R, Omori E, Takahashi T, Morimatsu H. The protective effect of carbamazepine on acute lung injury induced by hemorrhagic shock and resuscitation in rats. PLoS One 2024; 19:e0309622. [PMID: 39441839 PMCID: PMC11498730 DOI: 10.1371/journal.pone.0309622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 08/14/2024] [Indexed: 10/25/2024] Open
Abstract
Hemorrhagic shock and resuscitation (HSR) enhances the risk of acute lung injury (ALI). This study investigated the protective effect of carbamazepine (CBZ) on HSR-induced ALI in rats. Male Sprague-Dawley rats were allocated into five distinct groups through randomization: control (SHAM), saline + HSR (HSR), CBZ + HSR (CBZ/HSR), dimethyl sulfoxide (DMSO) + HSR (DMSO/HSR), and CBZ + chloroquine (CQ) + HSR (CBZ/CQ/HSR). Subsequently, HSR models were established. To detect tissue damage, we measured lung histological changes, lung injury scores, and wet/dry weight ratios. We measured neutrophil counts as well as assessed the expression of inflammatory factors using RT-PCR to determine the inflammatory response. We detected autophagy-related proteins LC3II/LC3I, P62, Beclin-1, and Atg12-Atg5 using western blotting. Pretreatment with CBZ improved histopathological changes in the lungs and reduced lung injury scores. The CBZ pretreatment group exhibited significantly reduced lung wet/dry weight ratio, neutrophil aggregation and number, and inflammation factor (TNF-α and iNOS) expression. CBZ changed the expression levels of autophagy-related proteins (LC3II/LC3I, beclin-1, Atg12-Atg5, and P62), suggesting autophagy activation. However, after injecting CQ, an autophagy inhibitor, the beneficial effects of CBZ were reversed. Taken together, CBZ pretreatment improved HSR-induced ALI by suppressing inflammation, at least in part, through activating autophagy. Thus, our study offers a novel perspective for treating HSR-induced ALI.
Collapse
Affiliation(s)
- Yaqiang Li
- Department of Anesthesiology and Resuscitology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hiroko Shimizu
- Department of Anesthesiology and Resuscitology, Okayama University Medical School, Okayama, Japan
| | - Ryu Nakamura
- Department of Anesthesiology and Resuscitology, Okayama University Medical School, Okayama, Japan
| | - Yifu Lu
- Department of Anesthesiology and Resuscitology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Risa Sakamoto
- Department of Anesthesiology and Resuscitology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Emiko Omori
- Department of Anesthesiology and Resuscitology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | | | - Hiroshi Morimatsu
- Department of Anesthesiology and Resuscitology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|