1
|
Wang Y, Wang W, Zhang Y, Fleishman JS, Wang H. Targeting ferroptosis offers therapy choice in sepsis-associated acute lung injury. Eur J Med Chem 2025; 283:117152. [PMID: 39657462 DOI: 10.1016/j.ejmech.2024.117152] [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: 08/09/2024] [Revised: 10/06/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Sepsis-associated acute lung injury (SALI) is a common complication of sepsis, consisting of a dysfunctional host response to infection-mediated heterogenous complexes. SALI is reported in up to 50 % of patients with sepsis and causes poor outcomes. Despite high incidence, there is a lack of understanding in its pathogenesis and optimal treatment. A better understanding of the molecular mechanisms underlying SALI may help produce better therapeutics. The effects of altered cell-death mechanisms, such as non-apoptotic regulated cell death (RCD) (i.e., ferroptosis), on the development of SALI are beginning to be discovered, while targeting ferroptosis as a meaningful target in SALI is increasingly being recognized. Here, we outline how a susceptible lung alveoli may develop SALI. Then we discuss the general mechanisms underlying ferroptosis, and how it contributes to SALI. We then outline the chemical structures of the emerging agents or compounds that can protect against SALI by inhibiting ferroptosis, summarizing their potential pharmacological effects. Finally, we highlight key limitations and possible strategies to overcome them. This review suggests that a detailed mechanistic and biological understanding of ferroptosis can foster the development of pharmacological antagonists in the treatment of SALI.
Collapse
Affiliation(s)
- Yu Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Weixue Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Yi Zhang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| | - Hongquan Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China.
| |
Collapse
|
2
|
Zhu Z, Wang J, Cheng H, Zhao H, Liu C, Zhou X, Yang J. Combined Toxicity Assessment of Deoxynivalenol and Pb 2+ on HK-2 Cells Involved in Excessive ROS-Induced Ferroptosis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:2573-2584. [PMID: 39818813 DOI: 10.1021/acs.jafc.4c11077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
The cocontamination of food by several mycotoxins and heavy metals poses significant health risks, and their combined toxic effects remain poorly understood. Particularly, specific studies exploring their combined impact on ferroptosis remain limited. In this work, we investigated the combined toxic effects of a mycotoxin, called deoxynivalenol (DON), and a heavy metal, called plumbum (Pb), and explored the potential mechanisms of DON and Pb co-occurrence via excessive ROS-induced ferroptosis in HK-2 cells. It was found that combined toxicity of DON and Pb2+ showed a synergism at low concentrations and an antagonism at high concentrations. The increase of the ROS level and iron content as well as the change expression of four ferroptosis marker proteins were observed in DON and Pb2+ individual and combined groups. Furthermore, the addition of ferroptosis inhibitor Fer-1 could mitigate the imbalance of oxidative stress and ferroptosis. Our results suggest that the co-occurrence of DON and Pb2+ might pose a slight threat to the nephrotoxicity due to the interactions related to the excessive ROS-induced ferroptosis, which would provide valuable insights into their potential combined toxic impacts to human and animal health.
Collapse
Affiliation(s)
- Zuoyin Zhu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- Institute for Agro-food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, 1000 Jingqi Road, Shanghai 201403, China
| | - Jie Wang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- Institute for Agro-food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, 1000 Jingqi Road, Shanghai 201403, China
| | - Haisheng Cheng
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- Institute for Agro-food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, 1000 Jingqi Road, Shanghai 201403, China
| | - Hanke Zhao
- Institute for Agro-food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, 1000 Jingqi Road, Shanghai 201403, China
| | - Chengbin Liu
- Institute for Agro-food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, 1000 Jingqi Road, Shanghai 201403, China
| | - Xinli Zhou
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Junhua Yang
- Institute for Agro-food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, 1000 Jingqi Road, Shanghai 201403, China
| |
Collapse
|
3
|
Yuan Y, Wang Y, Yan Y, Kim E, Bai J, Zhao Y, Ma Q, Gu W, Song H. FBLN1 regulates ferroptosis in acute respiratory distress syndrome by reducing free ferrous iron by inhibiting the TGF-β/Smad pathway. PLoS One 2024; 19:e0314750. [PMID: 39671383 PMCID: PMC11643259 DOI: 10.1371/journal.pone.0314750] [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: 08/14/2024] [Accepted: 11/17/2024] [Indexed: 12/15/2024] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) / acute lung injury (ALI) is a serious medical disease characterized by pulmonary dysfunction and inflammation. This study aims to determine the main molecular modules linked to ARDS and investigate the role of Fibulin-1 (FBLN1) in regulating ferroptosis in ARDS. METHODS Weighted Gene Co-expression Network Analysis (WGCNA) was employed on the GSE263867 dataset to find key modules associated with ALI. Differentially expressed genes (DEGs) and protein-protein interaction (PPI) networks were analyzed. MLE-12 cells were treated with lipopolysaccharide (LPS) to induce ferroptosis. In vitro studies were conducted to investigate the effects of FBLN1 and Transforming Growth Factor Beta 1 (TGF-β) overexpression on cell viability, oxidative stress markers, and ferroptosis-related proteins. RESULTS WGCNA identified the turquoise module as significantly negatively correlated with ARDS. Five key overlapping genes (GRIA1, OGN, COL14A1, FBLN1, and COL6A3) were significantly downregulated in ARDS samples. LPS treatment induced ferroptosis in MLE-12 cells, indicated by increased malondialdehyde (MDA), lipid reactive oxygen species (ROS), and ferrous iron (Fe2⁺) levels, and decreased cell viability and glutathione (GSH) levels. FBLN1 overexpression partially reversed these effects. Additionally, FBLN1 inhibited the TGF-β/Smad signaling pathway, as shown by decreased TGF-β and p-Smad protein levels. TGF-β overexpression exacerbated LPS-induced oxidative stress and ferroptosis, reducing cell viability and GSH levels. FBLN1 overexpression counteracted this effect, suggesting antagonistic roles for FBLN1 and TGF-β in regulating ferroptosis. CONCLUSION This study highlights FBLN1 as a critical regulator of ferroptosis in ARDS. Targeting the TGF-β/Smad pathway to modulate FBLN1 expression offers a potential therapeutic strategy to alleviate oxidative stress and mitigate pulmonary injury in inflammatory lung diseases.
Collapse
Affiliation(s)
- Yaping Yuan
- Department of Pulmonary and Critical Care Medicine, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Youbo Wang
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yufeng Yan
- Department of Neurosurgery, Jinshan Hospital, Fudan University, Zhujing Town, Jinshan District, Shanghai, China
| | - Edward Kim
- Department of Immunology, DICAT National Biomedical Computation Centre, Vancouver, BC, Canada
| | - Jin Bai
- Department of Immunology, DICAT National Biomedical Computation Centre, Vancouver, BC, Canada
- CRT Medical Union, Time International, Beijing, China
| | - Yang Zhao
- Department of Immunology, DICAT National Biomedical Computation Centre, Vancouver, BC, Canada
- CRT Medical Union, Time International, Beijing, China
| | - Qinyun Ma
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenchao Gu
- Department of Pulmonary and Critical Care Medicine, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Haihan Song
- Department of Pulmonary and Critical Care Medicine, Shanghai Pudong New Area People’s Hospital, Shanghai, China
- Department of Immunology, DICAT National Biomedical Computation Centre, Vancouver, BC, Canada
- Central Lab, Shanghai Key Laboratory of Pathogenic Fungi Medical Testing, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| |
Collapse
|
4
|
Guo K, Yin Y, Zheng L, Wu Z, Rao X, Zhu W, Zhou B, Liu L, Liu D. Integration of microbiomics, metabolomics, and transcriptomics reveals the therapeutic mechanism underlying Fuzheng-Qushi decoction for the treatment of lipopolysaccharide-induced lung injury in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118584. [PMID: 39019418 DOI: 10.1016/j.jep.2024.118584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fuzheng-Qushi decoction (FZQS) is a practical Chinese herbal formula for relieving cough and fever. Therefore, the action and specific molecular mechanism of FZQS in the treatment of lung injury with cough and fever as the main symptoms need to be further investigated. AIMS OF THE STUDY To elucidate the protective effects of FZQS against lung injury in mice and reveal its potential targets and key biological pathways for the treatment of lung injury based on transcriptomics, microbiomics, and untargeted metabolomics analyses. MATERIALS AND METHODS Lipopolysaccharide (LPS) was used to induce a mouse model of lung injury, followed by the administration of FZQS. ELISA was used to detect IL-1β, IL-6, IL-17A, IL-4, IL-10, and TNF-α, in mouse lung tissues. Macrophage polarization and neutrophil activation were measured by flow cytometry. RNA sequencing (RNA-seq) was applied to screen for differentially expressed genes (DEGs) in lung tissues. RT-qPCR and Western blot assays were utilized to validate key DEGs and target proteins in lung tissues. 16S rRNA sequencing was employed to characterize the gut microbiota of mice. Metabolites in the gut were analyzed using untargeted metabolomics. RESULTS FZQS treatment significantly ameliorated lung histopathological damage, decreased pro-inflammatory cytokine levels, and increased anti-inflammatory cytokine levels. M1 macrophage levels in the peripheral blood decreased, M2 macrophage levels increased, and activated neutrophils were inhibited in mice with LPS-induced lung injury. Importantly, transcriptomic analysis showed that FZQS downregulated macrophage and neutrophil activation and migration and adhesion pathways by reversing 51 DEGs, which was further confirmed by RT-qPCR and Western blot analysis. In addition, FZQS modulated the dysbiosis of the gut microbiota by reversing the abundance of Corynebacterium, Facklamia, Staphylococcus, Paenalcaligenes, Lachnoclostridium, norank_f_Muribaculaceae, and unclassified_f_Lachnospiraceae. Meanwhile, metabolomics analysis revealed that FZQS significantly regulated tryptophan metabolism by reducing the levels of 3-Indoleacetonitrile and 5-Hydroxykynurenine. CONCLUSION FZQS effectively ameliorated LPS-induced lung injury by inhibiting the activation, migration, and adhesion of macrophages and neutrophils and modulating gut microbiota and its metabolites.
Collapse
Affiliation(s)
- Kaien Guo
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Yuting Yin
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Linxin Zheng
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Zenan Wu
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Xiaoyong Rao
- National Engineering Center for Manufacturing Technology of Solid Preparations of Traditional Chinese Medicine Manufacturing Technology, Nanchang, 330004, Jiangxi Province, China
| | - Weifeng Zhu
- Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Bugao Zhou
- Department of Research, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China.
| | - Liangji Liu
- Affiliated Hospital, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China.
| | - Duanyong Liu
- Formula-pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China; School of Nursing, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China.
| |
Collapse
|
5
|
Liu C, Xu J, Fan J, Liu C, Xie W, Kong H. DPP-4 exacerbates LPS-induced endothelial cells inflammation via integrin-α5β1/FAK/AKT signaling. Exp Cell Res 2024; 435:113909. [PMID: 38184221 DOI: 10.1016/j.yexcr.2023.113909] [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: 11/21/2023] [Revised: 12/18/2023] [Accepted: 12/30/2023] [Indexed: 01/08/2024]
Abstract
Endothelial dysfunction plays a pivotal role in the pathogenesis of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). Dipeptidyl peptidase IV (DPP-4), a cell surface glycoprotein, has been implicated in endothelial inflammation and barrier dysfunction. In this study, the role of DPP-4 on lipopolysaccharide (LPS)-induced pulmonary microvascular endothelial cells (HPMECs) dysfunction and the underlying mechanism were investigated by siRNA-mediated knockdown of DPP-4. Our results indicated that LPS (1 μg/ml) challenge resulted in either the production and releasing of DPP-4, as well as the secretion of IL-6 and IL-8 in HPMECs. DPP-4 knockdown inhibited chemokine releasing and monolayer hyper-permeability in LPS challenged HPMECs. When cocultured with human polymorphonuclear neutrophils (PMNs), DPP4 knockdown suppressed LPS-induced neutrophil-endothelial adhesion, PMN chemotaxis and trans-endothelial migration. Western blotting showed that DPP-4 knockdown attenuated LPS-induced activation of TLR4/NF-κB pathway. Immunoprecipitation and liquid chromatography-tandem mass spectrometry revealed that DPP-4 mediated LPS-induced endothelial inflammation by interacting with integrin-α5β1. Moreover, exogenous soluble DPP-4 treatment sufficiently activated integrin-α5β1 downstream FAK/AKT/NF-κB signaling, thereafter inducing ICAM-1 upregulation in HPMECs. Collectively, our results suggest that endothelia synthesis and release DPP-4 under the stress of endotoxin, which interact with integrin-α5β1 complex in an autocrine or paracrine manner to exacerbate endothelial inflammation and enhance endothelial cell permeability. Therefore, blocking DDP-4 could be a potential therapeutic strategy to prevent endothelial dysfunction in ALI/ARDS.
Collapse
Affiliation(s)
- Chang Liu
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Jian Xu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University, School of Medicine, Shanghai, 200433, China
| | - Jiahao Fan
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Chenyang Liu
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Weiping Xie
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| | - Hui Kong
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|