1
|
Ren J, Lei G, Dong A, Cao S, Han X, Li H. Therapeutic potential of ADSC-derived exosomes in acute lung injury by regulating macrophage polarization through IRF7/NLRP3 signaling. Int Immunopharmacol 2025; 156:114658. [PMID: 40252464 DOI: 10.1016/j.intimp.2025.114658] [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: 02/10/2025] [Revised: 04/02/2025] [Accepted: 04/08/2025] [Indexed: 04/21/2025]
Abstract
Alveolar macrophages (AMs) play a critical role in regulating pulmonary immunity and inflammation. Acute lung injury (ALI), frequently initiated by sepsis-induced systemic inflammation and cytokine storms, leads to heightened lung permeability and respiratory failure. Adipose-derived stem cell exosomes (ADSC-Exos) have shown promise as therapeutic agents due to their immunomodulatory properties. This study assesses the effectiveness of ADSC-Exos in mitigating ALI by modulating macrophage (mø) polarization and suppressing pyroptosis. In vivo, an LPS-induced ALI mouse model demonstrated that ADSC-Exos attenuated lung tissue inflammation and damage, as verified by histological staining, ELISA, and immunofluorescence. In vitro, LPS-stimulated MH-S cells treated with ADSC-Exos showed a decrease in M1 (iNOS, CD86) and an increase in M2 (CD206, Arg-1) markers, as evidenced by Western blotting (WB) and flow cytometry. Mechanistically, RNA sequencing pinpointed IRF7 as a key upstream regulator of pyroptosis. ADSC-Exos inhibited the NLRP3 inflammasome and pyroptosis, fostering a shift from pro-inflammatory M1 to anti-inflammatory M2 mø phenotypes. Overexpression of IRF7 negated these effects, undermining the protective role of ADSC-Exos. Notably, inhibition of exosome secretion with GW4869 nullified these immunomodulatory effects, underscoring the vital role of ADSC-Exos. This study underscores the therapeutic potential of ADSC-Exos in restoring alveolar mø homeostasis, modulating immune responses, and alleviating lung inflammatory injury in ALI. These findings suggest ADSC-Exos as a feasible strategy for treating sepsis-induced pulmonary complications.
Collapse
Affiliation(s)
- Jingyi Ren
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Guanhong Lei
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ajing Dong
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shuyan Cao
- Department of Orthopaedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiao Han
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Haibo Li
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
2
|
Yun J, Zhang Y, Zhang H, Xiao F, Wang Y, Xu P, Qu L. Anti-inflammatory properties of Pleione bulbocodioides extract through STING/ NF-κB pathway inhibition. Mol Immunol 2025; 183:83-92. [PMID: 40345080 DOI: 10.1016/j.molimm.2025.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Accepted: 04/27/2025] [Indexed: 05/11/2025]
Abstract
The members of Pleione (Orchidaceae) are popular worldwide for their ornamental appeal and medicinal properties. Pleione bulbocodioides, a CITES Appendix II-listed species, has been traditionally used in dermatological therapies but remains pharmacologically understudied. In this study, the in vitro anti-inflammatory effects of extracts from artificially cultivated P. bulbocodioides were investigated. We found the P. bulbocodioides extract (PE) significantly suppressed TNF-α/IFN-γ-induced expression of IL-6, IL-1β, CCL5, CCL8, CXCL8, CXCL3, and TMEM173 (STING) genes in both HaCaT cells and NHEKs. Transcriptomic analysis and Western blotting confirmed the inhibitory effects on STING/NF-κB signaling pathway of PE. And phytochemical characterization identified militarine and batatasin III as principal bioactive constituents responsible for STING/NF-κB pathway inhibition by PE. PE also demonstrated comparable anti-inflammatory efficacy in LPS-induced RAW 264.7 macrophages and SLS-irritated 3D reconstructed human epidermis. Thus, these findings indicate the potential of PE as a natural anti-inflammatory therapeutic or skincare ingredient for dermatological applications.
Collapse
Affiliation(s)
- Jinlei Yun
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Yunnan 650106, China
| | - Yindi Zhang
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Yunnan 650106, China
| | - Hongyan Zhang
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Yunnan 650106, China
| | - Fengkun Xiao
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Yunnan 650106, China
| | - Yichun Wang
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Yunnan 650106, China
| | - Peng Xu
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Yunnan 650106, China.
| | - Liping Qu
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Yunnan 650106, China; Yunnan Botanee Bio-technology Group Co., Ltd, Yunnan 650106, China; Botanee Research Institute, Shanghai Jiyan Biomedical Development Co., Ltd., Shanghai 201702, China.
| |
Collapse
|
3
|
Cheng S, Luo W, Zhang Z, Li J, Li X, Wang Y, Weng X, Dong Z. Shexiang Baoxin Pill alleviates atherosclerosis by inhibiting macrophage-mediated inflammation via suppressing KMT5A mediated Irf7 transcription. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119833. [PMID: 40252984 DOI: 10.1016/j.jep.2025.119833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/07/2025] [Accepted: 04/17/2025] [Indexed: 04/21/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shexiang Baoxin Pill (SBP) is a traditional compound formulation composed of seven Chinese medicinal ingredients. Although SBP has shown promising clinical outcomes in the treatment of cardiovascular diseases, its role and underlying mechanisms in alleviating atherosclerosis remain insufficiently studied. AIM OF THE STUDY This study aims to investigate the effects and mechanisms of SBP in epigenetic modulating macrophage inflammatory responses to mitigate atherosclerosis. MATERIALS AND METHODS ApoE-/- mice were treated with high fat diet (HFD) following varying concentrations of SBP. Oil Red O staining, hematoxylin-eosin (HE) staining, and ELISA were used to assess the anti-atherosclerotic and anti-inflammatory efficiency of SBP. Subsequently, RNA sequencing (RNA-seq), RT-PCR, Western blot (WB), immunofluorescence (IF) and chromatin immunoprecipitation (ChIP) were employed in bone marrow derived macrophages (BMDMs) to elucidate the epigenetic mechanisms of SBP in alleviating macrophage inflammatory responses. Lysine methyltransferase 5A (KMT5A) was overexpressed in vivo and in vitro for further validation. RESULTS SBP significantly attenuated atherosclerosis in HFD treated ApoE-/- mice by decreasing plaque areas, serum inflammation levels and macrophages infiltration in the aortic root and plaques. SBP treatment reduced BMDMs inflammatory responses following oxidized low-density lipoprotein (oxLDL) treatment. Mechanistically, SBP inhibited interferon regulatory factor 7 (IRF7) expression by reducing KMT5A-mediated mono-methylation of histone H4 lysine 20 (H4K20), thus decreasing the secretion of multiple pro-inflammatory cytokines, including interferon (IFN)-α, IFN-β, TNF-α. Overexpression of KMT5A abolished the anti-atherosclerotic and anti-inflammatory effects of SBP, further confirming that KMT5A/H4K20me/IRF7 axis is a key target for SBP exerting therapeutic effect. CONCLUSION SBP exerts anti-atherosclerotic effects by inhibiting macrophage inflammatory responses through downregulation of the H4K20 methylase KMT5A, thereby suppressing the transcription of Irf7. Our findings provide a novel epigenetic mechanism by which SBP alleviates atherosclerosis.
Collapse
Affiliation(s)
- Shuo Cheng
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wei Luo
- State Key Laboratory of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Beijing, 100029, China
| | - Zhonghua Zhang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jia Li
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiang Li
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yidan Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xinyu Weng
- State Key Laboratory of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China.
| | - Zheng Dong
- Department of Cardiology, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing Key Laboratory for Cardiovascular Information and Health Engineering Medicine, No. 321 Zhongshan Road, Nanjing, 210008, China; Nanjing Drum Towel Hospital, Clinical Collage, Nanjing University of Chinese medicine, China; State Key Laboratory of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China.
| |
Collapse
|
4
|
Zhao H, Zhang H, Shi J, Liu Y, Yu J, Yang Y, Weng J, Song Z, Zhou R, Min H, Yao J, Wang M, Zhang Z. Wuwei Shaji powder alleviates OVA-induced allergic asthma by protecting bronchial epithelial cells from ferroptosis via the S-sulfhydration of Keap1. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119649. [PMID: 40222689 DOI: 10.1016/j.jep.2025.119649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/09/2025] [Accepted: 03/16/2025] [Indexed: 04/15/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Asthma is a chronic inflammatory airway disease. Current treatments have limited efficacy and often cause severe side effects. Wuwei Shaji Powder (WSP), a traditional Mongolian remedy, is used for treating chronic pulmonary diseases, but its efficacy against asthma and underlying mechanisms are still unclear. AIM OF THE STUDY To investigate the therapeutic effect of WSP on asthma and elucidate its molecular mechanisms. MATERIALS AND METHODS An ovalbumin (OVA)-induced allergic asthma model was established in rats. Ferroptosis or apoptosis was induced in BEAS-2B cells using Erastin or CdCl2. Various techniques including histopathological staining, ELISA, Western blot, flow cytometry, and transmission electron microscopy were employed to assess WSP's effects and mechanisms. RESULTS WSP alleviated OVA-induced allergic asthma in rats without the immunosuppressive side effects observed with dexamethasone. WSP suppressed ferroptosis in bronchial epithelial cells both in vivo and in vitro. It reduced thiol- and sulfonic-based oxidative stress through Keap1 S-sulfhydration, disrupted the Keap1-Nrf2 interaction, and promoted Nrf2 nuclear translocation. Notably, we discovered that CdCl2 can induce ferroptosis in BEAS-2B cells, and WSP prevented both ferroptosis and apoptosis in these cells. CONCLUSION WSP alleviates OVA-induced allergic asthma by protecting bronchial epithelial cells from ferroptosis via S-sulfhydration of Keap1, providing new insights for its clinical application.
Collapse
Affiliation(s)
- Huimei Zhao
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Haiyan Zhang
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Jianyu Shi
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yanru Liu
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Jingao Yu
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yuangui Yang
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Jingyu Weng
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Zhongxing Song
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Rui Zhou
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Hong Min
- NMPA Key Laboratory for Testing Technology of Pharmaceutical Microbiology, Shaanxi Institute for Food and Drug Control, Xi'an, China
| | - Jian Yao
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Mei Wang
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China; Wangjing Hospital of China Academy of Traditional Chinese Medicine, Beijing, 100102, China.
| | - Zhen Zhang
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712046, China.
| |
Collapse
|
5
|
Ji H, Kuang G, Yang H, Liu H, Li Y, Hu S, Xiao A, You C, Sun H, Fan C, Sun G. Discrepancies between human and murine model cerebral aneurysms at single-cell resolution. Front Cell Dev Biol 2025; 13:1512938. [PMID: 40134579 PMCID: PMC11933115 DOI: 10.3389/fcell.2025.1512938] [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: 10/17/2024] [Accepted: 02/14/2025] [Indexed: 03/27/2025] Open
Abstract
Background The murine model of cerebral aneurysm (CA) serves as a prevalent tool for investigating the molecular underpinnings of CA. However, the extent to which the CA murine model aligns with that of human remains elusive. Methods The present study employed a comprehensive integration and exploration of the single-cell RNA-seq (scRNA-seq) datasets, along with multiple trajectory and gene regulatory network analyses, to investigate the cellular and molecular discrepancies between human and murine model CAs. Results The uniform manifold approximation and projection (umap) embedding exhibits that the primary discrepancies between human and murine model CAs reside in the cells of modifiable phenotype, encompassing vascular smooth muscle cell (vSMC), monocyte/macrophage, and neutrophil. The vSMCs from human CA tissue exhibit a fibroblast-like phenotype in comparison to that of murine model. Distinct patterns of neutrophil recruitment are observed in human and murine models, with the former characterized by neutrophil-derived CXCL8 and the latter by monocyte/macrophage-derived CCLs. In addition, macrophages originated from human unruptured CA express higher levels of M2 gene markers. Moreover, the inflammatory status of the CA tissue differs between humans and mouse models, with the former exhibiting a more acute and intense inflammation. Conclusion These findings demonstrate subtle but important disparities between human and murine model CAs, and may shed light upon an optimization of murine CA model.
Collapse
Affiliation(s)
- Hang Ji
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
- Department of Neurosurgery, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Guicheng Kuang
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - Hailan Yang
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - Haitao Liu
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - Yue Li
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - Shaoshan Hu
- Department of Neurosurgery, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Anqi Xiao
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - Chao You
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - Haogeng Sun
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - Chaofeng Fan
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - Guozhang Sun
- Department of Neurosurgery, Hei Longjiang Provincial People’s Hospital, Harbin, Hei Longjiang, China
| |
Collapse
|
6
|
Wu S, He Y, Li J, Zhuang H, Wang P, He X, Guo Y, Li Z, Shen H, Ye L, Lin F. TREM2 alleviates sepsis-induced acute lung injury by attenuating ferroptosis via the SHP1/STAT3 pathway. Free Radic Biol Med 2025; 229:111-126. [PMID: 39814108 DOI: 10.1016/j.freeradbiomed.2025.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
Sepsis-induced acute lung injury (ALI) is a complex and life-threatening condition characterized by excessive inflammatory responses, ferroptosis, and oxidative stress. A comprehensive investigation and effective therapeutic strategies are crucial for managing this condition. In this study, we established in vivo sepsis models using lipopolysaccharide (LPS) in wild-type (WT) mice and triggering receptor expressed on myeloid cells 2 (TREM2) knockout (TREM2-KO) mice to assess lung morphology, oxidative stress, and ferroptosis. In vitro, RAW264.7 cells with TREM2 overexpression (TREM2-OE) or knockdown (TREM2-SiRNA) were utilized to assess oxidative stress and ferroptosis. RNA sequencing of LPS-stimulated cells transfected with either vector or TREM2-OE revealed significant differences in inflammation- and ferroptosis-related pathways. LPS-induced lung injury and ferroptosis were exacerbated in TREM2-KO mice and TREM2-SiRNA cells but alleviated by the ferroptosis inhibitor ferrostatin-1 (Fer-1). Mechanistically, TREM2-KO led to SHP1 downregulation and STAT3-P upregulation, which were reversed by the SHP1 agonist SC-43. These findings highlight the role of TREM2 in the SHP1/STAT3 signaling pathway and its regulatory effects on ferroptosis. Our study demonstrates that TREM2, via the SHP1/STAT3 pathway, suppresses oxidative stress and ferroptosis, thereby significantly mitigating sepsis-induced ALI. These results underscore the pivotal role of TREM2 in modulating inflammatory responses and immunity, providing a theoretical foundation for developing therapeutic strategies.
Collapse
Affiliation(s)
- Siyi Wu
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China; Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Yuanjie He
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China; Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Jiemei Li
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China; Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Hanhong Zhuang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China; Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Peng Wang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China; Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Xiaojing He
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Youyuan Guo
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Zhiping Li
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Honglei Shen
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China; Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Liu Ye
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Fei Lin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China; Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
7
|
Meng T, Zhang Y, Wang H, Wu W, Peng W, Yue J, Huang C, Liu W, Liang C, Yang C, Chen J. Irf7 aggravates prostatitis by promoting Hif-1α-mediated glycolysis to facilitate M1 polarization. Cell Mol Life Sci 2025; 82:90. [PMID: 39985573 PMCID: PMC11846824 DOI: 10.1007/s00018-025-05608-w] [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: 10/17/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) is a common disorder associated with voiding symptoms and pain in the pelvic or perineal area. Macrophages, particularly the pro-inflammatory M1 subtype, are crucial initiating of CP/CPPS. Interferon regulatory factor 7 (Irf7) has been implicated in promoting M1 polarization, contributing to the onset and progression of autoimmunity. However, the role of Irf7 in the etiology and progression of CP/CPPS remains unclear. METHOD We established the experimental autoimmune prostatitis (EAP) mouse model by subcutaneous injection of prostate antigen combined with complete Freund's adjuvant. Six weeks after the first immunization, we analyzed the prostates, spleen, and blood to assess the degree of prostate inflammation, Irf7 expression levels, glycolysis, and M1 polarization to evaluate whether Irf7 could exacerbate the development of EAP by enhancing Hif-1α transcription, thereby increasing glycolysis and M1 polarization. Further investigations included sh-Irf7 intervention, Dimethyloxalylglycine (a Hif-1α agonist), and in vitro M1 polarization experiments. We also employed ChIP assays, dual-luciferase reporter assays, and q-PCR to explore if Irf7 could directly interact with the Hif-1α promoter in macrophages. RESULTS In the EAP mouse and cell models, elevated Irf7 expression was observed in inflamed tissues and cells. Reducing Irf7 expression decreased M1 cell glycolysis by inhibiting the nuclear translocation of Hif-1α, thus mitigating M1 cell polarization. Additionally, Irf7 was identified as a transcription factor that regulates Hif-1α transcription by interacting with its promoter in macrophages, confirmed through ChIP and dual-luciferase assays. Co-culturing macrophage cells with 3T3 fibroblasts with reduced Irf7 levels resulted in decreased fibrosis, and a significant reduction in prostate tissue fibrosis was noted in mice with Irf7 knockdown. CONCLUSION Our findings indicate that Irf7 can contribute to the development and progression of CP/CPPS by promoting glycolysis, which can enhance both M1 polarization as well as interstitial fibrosis in the prostate. This process was found to be mediated by the upregulation of Hif-1α transcription, presenting new potential therapeutic targets for managing CP/CPPS.
Collapse
Affiliation(s)
- Tong Meng
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Yi Zhang
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Huihui Wang
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Weikang Wu
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Wei Peng
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Jiabin Yue
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Cong Huang
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Wanqing Liu
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Chaozhao Liang
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China.
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China.
| | - Cheng Yang
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China.
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China.
| | - Jing Chen
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China.
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China.
| |
Collapse
|
8
|
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
|
9
|
Gan J, Zhang W, Pan F, Qiu Z, Chen X. TRIM11 modulates sepsis progression by promoting HOXB9 ubiquitination and inducing the NF-κB signaling pathway. Mol Biol Rep 2025; 52:194. [PMID: 39903348 DOI: 10.1007/s11033-024-10212-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/29/2024] [Indexed: 02/06/2025]
Abstract
INTRODUCTION The purpose of this investigation was to elucidate the functions of TRIM11 and HOXB9 in the pathogenesis of sepsis, focusing on their influence on inflammation, apoptosis, and the NF-κB signaling pathway. MATERIAL AND METHODS Through public databases, TRIM family genes related to sepsis were screened, and TRIM11 was evaluated as a sepsis biomarker through ROC analysis. The UbiBrowser database screened TRIM11 downstream genes and identified HOXB9 as an essential target. THP-1 cells were stimulated by Lipopolysaccharide (LPS) to induce inflammation and simulate sepsis. Flow cytometry, Enzyme-linked immunosorbent assay, and Western blot experiments were used to detect changes in cell apoptosis rate, apoptosis-related proteins, and inflammatory cytokines after TRIM11 and HOXB9 were silenced. Additionally, we investigated the ubiquitination interaction between TRIM11 and HOXB9 and their effects on the NF-κB signaling pathway. RESULTS Our findings demonstrated that sepsis patient samples had elevated levels of TRIM11 expression and had high clinical diagnostic value. Functional experiments showed that the knockdown of TRIM11 significantly alleviated LPS-induced THP-1 cell apoptosis and inflammation, while the knockdown of HOXB9 did the opposite. The simultaneous downregulation of TRIM11 and HOXB9 balanced these responses, suggesting they play a key role in regulating sepsis-associated inflammation and apoptosis. In addition, TRIM11 regulated the NF-κB signaling pathway by reversing HOXB9-induced activation through ubiquitination, suggesting a novel regulatory mechanism in the pathogenesis of sepsis. CONCLUSIONS Our findings highlight the interaction between TRIM11 and HOXB9 in regulating inflammation and apoptosis pathways, providing new insights into sepsis treatment.
Collapse
Affiliation(s)
- Jiaqi Gan
- Department of General Medicine, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai, 201199, China
| | - Wei Zhang
- Emergency Medical Department, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai, 201199, China
| | - Fei Pan
- Department of General Medicine, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai, 201199, China
| | - Zhiyun Qiu
- Emergency Medical Department, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai, 201199, China.
| | - Xiaobing Chen
- The First People's Hospital of Lianyungang Graduate Student Training Base, Jinzhou Medical University, No.6 East Zhenhua Road, Lianyungang, 222061, China.
| |
Collapse
|
10
|
Lv Y, Zhang L. IRF7 Activates LCN2 Transcription to Enhance LPS-Induced Acute Lung Injury by Inducing Macrophage Ferroptosis and M1 Polarization. Cell Biochem Biophys 2024:10.1007/s12013-024-01651-9. [PMID: 39738844 DOI: 10.1007/s12013-024-01651-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025]
Abstract
Acute lung injury (ALI), a severe pulmonary disorder that poses a significant threat to life, is closely associated with macrophage ferroptosis and polarization. Lipocalin 2 (LCN2) has been previously reported to be implicated in the pathogenesis of ALI. However, the specific role of LCN2 in macrophage ferroptosis and polarization remains undetermined. Lipopolysaccharide (LPS) was used to establish a mouse model of ALI and also to stimulate mouse RAW264.7 cells. H&E staining was used for histopathologic evaluation, and immunohistochemistry analysis was used to determine the 4-HNE-positive cells. The secretion levels of TNF-α, IL-6, and IL-1β were assessed by ELISA. Gene and protein expression assays were performed using quantitative PCR and immunoblotting. The levels of MDA, GSH, ROS, and lipid ROS were detected to evaluate the alteration in ferroptosis. CD86+ and CD206+ cells were quantified by flow cytometry. The relationship between LCN2 and interferon regulatory factor 7 (IRF7) was confirmed by chromatin immunoprecipitation (ChIP) and luciferase reporter assays. LCN2 expression was upregulated in the lungs of LPS-induced ALI mice and LPS-stimulated RAW264.7 cells. In LPS-induced ALI mice, the depletion of LCN2 alleviated lung injury and ferroptosis, and also inhibited inflammation and macrophage M1 polarization. In LPS-stimulated RAW264.7 cells, the depletion of LCN2 suppressed ferroptosis, inflammation, and M1 polarization. Mechanistically, IRF7 enhanced LCN2 transcription in RAW264.7 cells by binding to its promoter region. More importantly, the silencing of IRF7 inhibited ferroptosis and M1 polarization in LPS-stimulated RAW264.7 cells by downregulating LCN2. Taken together, the IRF7/LCN2 cascade enhances the ferroptosis and M1 polarization of LPS-stimulated macrophages, thereby exacerbating ALI. Anti-IRF7 and anti-LCN2 therapies might potentially be exploited for the prevention and treatment in ALI.
Collapse
Affiliation(s)
- Yali Lv
- Graduate School, Zhejiang Chinese Medical University, Hangzhou, China
- Emergency Intensive Care Unit (ICU), Jinyun County Second People's Hospital, Lishui, China
| | - Lefeng Zhang
- Department of Respiratory and Critical Care Medicine, Lishui Second People's Hospital, Lishui, China.
| |
Collapse
|
11
|
Zhang Z, He Y, Liu H, Liu Y, Wu T, Li R, Wang Y, Ma W. NLRP3 regulates ferroptosis via the JAK2/STAT3 pathway in asthma inflammation: Insights from in vivo and in vitro studies. Int Immunopharmacol 2024; 143:113416. [PMID: 39426227 DOI: 10.1016/j.intimp.2024.113416] [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: 07/09/2024] [Revised: 09/15/2024] [Accepted: 10/13/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Ferroptosis, an iron-dependent form of cell death, plays a pivotal role in the pathologic progression of asthma. Electroacupuncture (EA) has demonstrated considerable efficacy in mitigating asthma airway inflammation, although its underlying mechanisms remain partially elucidated. METHODS We investigated the regulatory effect of NLRP3 on ferroptosis using a lipopolysaccharide (LPS)-induced inflammation model in BEAS-2B cells, where NLRP3 expression was modulated with si-RNA and overexpression plasmids. The levels of inflammatory cytokines TNF-α, IL-1β, and IL-6 were quantified. We also assessed NLRP3 and JAK2/STAT3 pathway-related proteins, and evaluated lipid peroxidation, mitochondrial membrane potential (ΔΨm), and antioxidant system functionality. In vivo, we examined the impact of EA on ferroptosis and airway inflammation by modulating NLRP3 activation. Asthma inflammation severity was evaluated using H&E, Masson, and PAS staining, alongside ELISA. NLRP3 and JAK2/STAT3 pathway-related proteins, as well as ferroptosis indicators, were also analyzed. The mechanism by which NLRP3 activates ferroptosis was investigated through in vitro assays. RESULTS LPS exposure resulted in increased intracellular inflammatory cytokines, and activation of the NLRP3 and JAK2/STAT3 pathways, leading to enhanced lipid peroxidation, decreased ΔΨm, and disruption of antioxidant system balance, ultimately inducing ferroptosis. Si-NLRP3 countered the effects of LPS, whereas oe-NLRP3 exacerbated symptoms. In vivo studies revealed that EA reduced airway inflammation, inhibited NLRP3 activation, and decreased phosphorylation of JAK2/STAT3, effectively lowering ferroptosis-related indicators. Utilizing JAK2/STAT3 activators and inhibitors, we confirmed that NLRP3 mediates ferroptosis via the JAK2/STAT3 pathway. CONCLUSIONS EA alleviates HDM-induced asthma, primarily through the inhibition of NLRP3 activation, which modulates the JAK2/STAT3 pathway and mediates ferroptosis.
Collapse
Affiliation(s)
- Zhengze Zhang
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Yuewen He
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Hao Liu
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Yurui Liu
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Tong Wu
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Ruogen Li
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China
| | - Yong Wang
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China; State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China.
| | - Wuhua Ma
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China.
| |
Collapse
|
12
|
Hu W, Zhang X, Wu Z, Luo Y, Hu B, Zou X. Exploring and Validating the Mechanism of Ulinastatin in the Treatment of Sepsis-Associated Encephalopathy Based on Transcriptome Sequencing. J Inflamm Res 2024; 17:8753-8773. [PMID: 39564549 PMCID: PMC11573691 DOI: 10.2147/jir.s488400] [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: 08/30/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
Purpose Sepsis can induce sepsis-associated encephalopathy (SAE), with Ulinastatin (UTI) serving a critical anti-inflammatory role. This study aimed to identify the hub genes in an SAE mouse model following UTI intervention and investigate the underlying molecular mechanisms. Materials and Methods Through differential expression analysis to obtain differentially expressed genes (DEGs), ie, UTI vs CLP (DEGs1) and Con vs CLP (DEGs2). After taking the intersection of the genes with opposite differential trends in these two parts and immune-related genes (IRGs), DE-IRGs were obtained. Hub genes in the protein-protein interaction (PPI) network were then determined using six algorithms from the Cytohubba plugin in Cytoscape. Gene set enrichment analysis (GSEA) was employed to explore the functional relevance of these hub genes. Additionally, the immune microenvironment across the three groups was compared, and hub gene-related drugs were predicted using an online database. Finally, qRT-PCR was used to validate the expression of the hub genes in hippocampal tissue from CLP mice. Results RNA sequencing obtained 864 differentially expressed genes (DEGs) (CLP vs Con) and 279 DEGs (UTI vs CLP). Taking the intersection of DEGs with opposite expression trends yielded 165 DEGs. Six key genes (ICAM - 1, IRF7, IL - 1β, CCL2, IL - 6 and SOCS3) were screened by six algorithms. Immune infiltration analysis found that Treg cells were reversed after treatment with UTI in the diseased state. A total of 106 hub - gene - related drugs were predicted, among which BINDARIT - CCL2 and LIFITEGRAST - ICAM1 showed particularly high affinities. The qRT - PCR verification results were consistent with the sequencing results. Conclusion In conclusion, ICAM-1, IRF7, IL-1β, CCL2, IL-6, and SOCS3 were identified as potential therapeutic targets in SAE mice treated with UTI. This study offers theoretical support for UTI as a treatment option for SAE.
Collapse
Affiliation(s)
- Wen Hu
- Guizhou Medical University, Guiyang, Guizhou, 550004, People's Republic of China
| | - Xiaoyuan Zhang
- Guizhou Medical University, Guiyang, Guizhou, 550004, People's Republic of China
| | - Zhen Wu
- Guizhou Medical University, Guiyang, Guizhou, 550004, People's Republic of China
| | - Yushan Luo
- Guizhou Medical University, Guiyang, Guizhou, 550004, People's Republic of China
| | - Bailong Hu
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, People's Republic of China
| | - Xiaohua Zou
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, People's Republic of China
| |
Collapse
|
13
|
Cheng H, Wang X, Yao J, Yang C, Liu J. Mitophagy and Ferroptosis in Sepsis-Induced ALI/ARDS: Molecular Mechanisms, Interactions and Therapeutic Prospects of Medicinal Plants. J Inflamm Res 2024; 17:7819-7835. [PMID: 39494205 PMCID: PMC11531397 DOI: 10.2147/jir.s488655] [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: 07/26/2024] [Accepted: 10/17/2024] [Indexed: 11/05/2024] Open
Abstract
Sepsis is a common critical illness characterized by high mortality rates and a significant disease burden. In the context of sepsis-induced organ dysfunction, the lungs are among the initial organs affected, which may progress to acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). Recent studies have highlighted the crucial roles of mitophagy and ferroptosis in the development and progression of sepsis-induced ALI/ARDS. Identifying key convergence points in these processes may provide valuable insights for the treatment of this condition. In recent years, certain herbs and their bioactive compounds have demonstrated unique benefits in managing sepsis-induced ALI/ARDS by modulating mitophagy or ferroptosis. This review summary the mechanisms of mitophagy and ferroptosis, explores their interactions, and emphasizes their regulatory roles in the progression of sepsis-induced ALI/ARDS. Additionally, it offers a novel perspective on treatment strategies by summarizing various herbs and their bioactive compounds relevant to this condition.
Collapse
Affiliation(s)
- Huixin Cheng
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Xuehan Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Juyi Yao
- Traditional Chinese Medicine Hospital of Xinjiang Uygur Autonomous Region, Urumqi, People’s Republic of China
| | - Chunbo Yang
- Department of Critical Medicine Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi, People’s Republic of China
| | - Jian Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
- Department of Intensive Care Unit, Gansu Provincial Maternity and Child Health Hospital/Gansu Provincial General Hospital, Lan Zhou, Gansu Province, People’s Republic of China
| |
Collapse
|
14
|
Cheng Y, Zhu L, Xie S, Lu B, Du X, Ding G, Wang Y, Ma L, Li Q. Relationship between ferroptosis and mitophagy in acute lung injury: a mini-review. PeerJ 2024; 12:e18062. [PMID: 39282121 PMCID: PMC11397134 DOI: 10.7717/peerj.18062] [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: 03/12/2024] [Accepted: 08/19/2024] [Indexed: 09/18/2024] Open
Abstract
Acute lung injury (ALI) is one of the most deadly and prevalent diseases in the intensive care unit. Ferroptosis and mitophagy are pathological mechanisms of ALI. Ferroptosis aggravates ALI, whereas mitophagy regulates ALI. Ferroptosis and mitophagy are both closely related to reactive oxygen species (ROS). Mitophagy can regulate ferroptosis, but the specific relationship between ferroptosis and mitophagy is still unclear. This study summarizes previous research findings on ferroptosis and mitophagy, revealing their involvement in ALI. Examining the functions of mTOR and NLPR3 helps clarify the connection between ferroptosis and mitophagy in ALI, with the goal of establishing a theoretical foundation for potential therapeutic approaches in the future management of ALI.
Collapse
Affiliation(s)
- Yunhua Cheng
- The First School of Clinical Medicine of Gansu University of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu Province, China
- Department of Thoracic Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu Province, China
| | - Liling Zhu
- Department of Anesthesiology, Hunan Children's Hospital, Changsha, Hunan Province, China
| | - Shuangxiong Xie
- The First School of Clinical Medicine of Gansu University of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu Province, China
- Department of Thoracic Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu Province, China
| | - Binyuan Lu
- The First School of Clinical Medicine of Gansu University of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu Province, China
- Department of Thoracic Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu Province, China
| | - Xiaoyu Du
- Medical College of Northwest Minzu University, Northwest Minzu University, Lanzhou, Gansu Province, China
| | - Guanjiang Ding
- The First School of Clinical Medicine of Gansu University of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu Province, China
- Department of Thoracic Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu Province, China
| | - Yan Wang
- The First School of Clinical Medicine of Gansu University of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu Province, China
- Department of Thoracic Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu Province, China
| | - Linchong Ma
- Department of Thoracic Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu Province, China
| | - Qingxin Li
- Department of Thoracic Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu Province, China
| |
Collapse
|
15
|
Deng W, Zhong L, Ye S, Luo J, Ren G, Huang J, Zhuang X. Mir22hg facilitates ferritinophagy-mediated ferroptosis in sepsis by recruiting the m6A reader YTHDC1 and enhancing Angptl4 mRNA stability. J Bioenerg Biomembr 2024; 56:405-418. [PMID: 38842666 PMCID: PMC11217081 DOI: 10.1007/s10863-024-10022-1] [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: 03/07/2024] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Ferritinophagy-mediated ferroptosis plays a crucial role in fighting pathogen aggression. The long non-coding RNA Mir22hg is involved in the regulation of ferroptosis and aberrantly overexpression in lipopolysaccharide (LPS)-induced sepsis mice, but whether it regulates sepsis through ferritinophagy-mediated ferroptosis is unclear. METHODS Mir22hg was screened by bioinformatics analysis. Ferroptosis was assessed by assaying malondialdehyde (MDA), reactive oxygen species (ROS), and Fe2+ levels, glutathione (GSH) activity, as well as ferroptosis-related proteins GPX4 and SLC3A2 by using matched kits and performing western blot. Ferritinophagy was assessed by Lyso tracker staining and FerroOrange staining, immunofluorescence analysis of Ferritin and LC-3, and western blot analysis of LC-3II/I, p62, FTH1, and NCOA4. The bind of YTH domain containing 1 (YTHDC1) to Mir22hg or angiopoietin-like-4 (Angptl4) was verified by RNA pull-down and/or immunoprecipitation (RIP) assays. RESULTS Mir22hg silencing lightened ferroptosis and ferritinophagy in LPS-induced MLE-12 cells and sepsis mouse models, as presented by the downregulated MDA, ROS, Fe2+, NCOA4, and SLC3A2 levels, upregulated GPX4, GSH, and FTH1 levels, along with a decrease in autophagy. Mir22hg could bind to the m6A reader YTHDC1 without affecting its expression. Mechanistically, Mir22hg enhanced Angptl4 mRNA stability through recruiting the m6A reader YTHDC1. Furthermore, Angptl4 overexpression partly overturned Mir22hg inhibition-mediated effects on ferroptosis and ferritinophagy in LPS-induced MLE-12 cells. CONCLUSION Mir22hg contributed to in ferritinophagy-mediated ferroptosis in sepsis via recruiting the m6A reader YTHDC1 and strengthening Angptl4 mRNA stability, highlighting that Mir22hg may be a potential target for sepsis treatment based on ferroptosis.
Collapse
Affiliation(s)
- Wenlong Deng
- Emergency of Department, SSL Central Hospital of Dongguan City, No.1 Xianglong Road, Shilong Town, Dongguan, 523326, China.
| | - Liang Zhong
- Emergency of Department, SSL Central Hospital of Dongguan City, No.1 Xianglong Road, Shilong Town, Dongguan, 523326, China
| | - Shupei Ye
- Emergency of Department, SSL Central Hospital of Dongguan City, No.1 Xianglong Road, Shilong Town, Dongguan, 523326, China
| | - Jiajing Luo
- Emergency of Department, SSL Central Hospital of Dongguan City, No.1 Xianglong Road, Shilong Town, Dongguan, 523326, China
| | - Guobin Ren
- Emergency of Department, SSL Central Hospital of Dongguan City, No.1 Xianglong Road, Shilong Town, Dongguan, 523326, China
| | - Junhao Huang
- Emergency of Department, SSL Central Hospital of Dongguan City, No.1 Xianglong Road, Shilong Town, Dongguan, 523326, China
| | - Xiaolei Zhuang
- Emergency of Department, SSL Central Hospital of Dongguan City, No.1 Xianglong Road, Shilong Town, Dongguan, 523326, China
| |
Collapse
|
16
|
Li D, Deng Y, Wen G, Wang L, Shi X, Chen S, Chen R. Targeting BRD4 with PROTAC degrader ameliorates LPS-induced acute lung injury by inhibiting M1 alveolar macrophage polarization. Int Immunopharmacol 2024; 132:111991. [PMID: 38581996 DOI: 10.1016/j.intimp.2024.111991] [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: 12/12/2023] [Revised: 03/06/2024] [Accepted: 03/29/2024] [Indexed: 04/08/2024]
Abstract
OBJECTIVES Acute lung injury (ALI) is a highly inflammatory condition with the involvement of M1 alveolar macrophages (AMs) polarization, eventually leading to the development of non-cardiogenic edema in alveolar and interstitial regions, accompanied by persistent hypoxemia. Given the significant mortality rate associated with ALI, it is imperative to investigate the underlying mechanisms of this condition so as to identify potential therapeutic targets. The therapeutic effects of the inhibition of bromodomain containing protein 4 (BRD4), an epigenetic reader, has been proven with high efficacy in ameliorating various inflammatory diseases through mediating immune cell activation. However, little is known about the therapeutic potential of BRD4 degradation in acute lung injury. METHODS This study aimed to assess the protective efficacy of ARV-825, a novel BRD4-targeted proteolysis targeting chimera (PROTAC), against ALI through histopathological examination in lung tissues and biochemical analysis in bronchoalveolar lavage fluid (BALF). Additionally, the underlying mechanism by which BRD4 regulated M1 AMs was elucidated by using CUT & Tag assay. RESULTS In this study, we found the upregulation of BRD4 in a lipopolysaccharide (LPS)-induced ALI model. Furthermore, we observed that intraperitoneal administration of ARV-825, significantly alleviated LPS-induced pulmonary pathological changes and inflammatory responses. These effects were accompanied by the suppression of M1 AMs. In addition, our findings revealed that the administration of ARV-825 effectively suppressed M1 AMs by inhibiting the expression of IRF7, a crucial transcriptional factor involved in M1 macrophages. CONCLUSION Our study suggested that targeting BRD4 using ARV-825 is a potential therapeutic approach for ALI.
Collapse
Affiliation(s)
- Difei Li
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yao Deng
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Guanxi Wen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Lingwei Wang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xing Shi
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Shanze Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Rongchang Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
17
|
Peng Y, Yang Y, Li Y, Shi T, Xu N, Liu R, Luan Y, Yao Y, Yin C. Mitochondrial (mt)DNA-cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling promotes pyroptosis of macrophages via interferon regulatory factor (IRF)7/IRF3 activation to aggravate lung injury during severe acute pancreatitis. Cell Mol Biol Lett 2024; 29:61. [PMID: 38671352 PMCID: PMC11055249 DOI: 10.1186/s11658-024-00575-9] [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: 01/10/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Macrophage proinflammatory activation contributes to the pathology of severe acute pancreatitis (SAP) and, simultaneously, macrophage functional changes, and increased pyroptosis/necrosis can further exacerbate the cellular immune suppression during the process of SAP, where cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) plays an important role. However, the function and mechanism of cGAS-STING in SAP-induced lung injury (LI) remains unknown. METHODS Lipopolysaccharide (LPS) was combined with caerulein-induced SAP in wild type, cGAS -/- and sting -/- mice. Primary macrophages were extracted via bronchoalveolar lavage and peritoneal lavage. Ana-1 cells were pretreated with LPS and stimulated with nigericin sodium salt to induce pyroptosis in vitro. RESULTS SAP triggered NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome activation-mediated pyroptosis of alveolar and peritoneal macrophages in mouse model. Knockout of cGAS/STING could ameliorate NLRP3 activation and macrophage pyroptosis. In addition, mitochondrial (mt)DNA released from damaged mitochondria further induced macrophage STING activation in a cGAS- and dose-dependent manner. Upregulated STING signal can promote NLRP3 inflammasome-mediated macrophage pyroptosis and increase serum interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α levels and, thus, exacerbate SAP-associated LI (SAP-ALI). Downstream molecules of STING, IRF7, and IRF3 connect the mtDNA-cGAS-STING axis and the NLRP3-pyroptosis axis. CONCLUSIONS Negative regulation of any molecule in the mtDNA-cGAS-STING-IRF7/IRF3 pathway can affect the activation of NLRP3 inflammasomes, thereby reducing macrophage pyroptosis and improving SAP-ALI in mouse model.
Collapse
Affiliation(s)
- Yiqiu Peng
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, China
| | - Yuxi Yang
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, China
| | - Yingying Li
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, China
| | - Tingjuan Shi
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, China
| | - Ning Xu
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, China
| | - Ruixia Liu
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, China
| | - Yingyi Luan
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, China.
| | - Yongming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100048, China.
| | - Chenghong Yin
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No. 251 Yaojiayuan Road, Chaoyang District, Beijing, 100026, China.
| |
Collapse
|
18
|
Wu S, Tang W, Liu L, Wei K, Tang Y, Ma J, Li H, Ao Y. Obesity-induced downregulation of miR-192 exacerbates lipopolysaccharide-induced acute lung injury by promoting macrophage activation. Cell Mol Biol Lett 2024; 29:36. [PMID: 38486141 PMCID: PMC10938800 DOI: 10.1186/s11658-024-00558-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/29/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Macrophage activation may play a crucial role in the increased susceptibility of obese individuals to acute lung injury (ALI). Dysregulation of miRNA, which is involved in various inflammatory diseases, is often observed in obesity. This study aimed to investigate the role of miR-192 in lipopolysaccharide (LPS)-induced ALI in obese mice and its mechanism of dysregulation in obesity. METHODS Human lung tissues were obtained from obese patients (BMI ≥ 30.0 kg/m2) and control patients (BMI 18.5-24.9 kg/m2). An obese mouse model was established by feeding a high-fat diet (HFD), followed by intratracheal instillation of LPS to induce ALI. Pulmonary macrophages of obese mice were depleted through intratracheal instillation of clodronate liposomes. The expression of miR-192 was examined in lung tissues, primary alveolar macrophages (AMs), and the mouse alveolar macrophage cell line (MH-S) using RT-qPCR. m6A quantification and RIP assays helped determine the cause of miR-192 dysregulation. miR-192 agomir and antagomir were used to investigate its function in mice and MH-S cells. Bioinformatics and dual-luciferase reporter gene assays were used to explore the downstream targets of miR-192. RESULTS In obese mice, depletion of macrophages significantly alleviated lung tissue inflammation and injury, regardless of LPS challenge. miR-192 expression in lung tissues and alveolar macrophages was diminished during obesity and further decreased with LPS stimulation. Obesity-induced overexpression of FTO decreased the m6A modification of pri-miR-192, inhibiting the generation of miR-192. In vitro, inhibition of miR-192 enhanced LPS-induced polarization of M1 macrophages and activation of the AKT/ NF-κB inflammatory pathway, while overexpression of miR-192 suppressed these reactions. BIG1 was confirmed as a target gene of miR-192, and its overexpression offset the protective effects of miR-192. In vivo, when miR-192 was overexpressed in obese mice, the activation of pulmonary macrophages and the extent of lung injury were significantly improved upon LPS challenge. CONCLUSIONS Our study indicates that obesity-induced downregulation of miR-192 expression exacerbates LPS-induced ALI by promoting macrophage activation. Targeting macrophages and miR-192 may provide new therapeutic avenues for obesity-associated ALI.
Collapse
Affiliation(s)
- Siqi Wu
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, No 1. YouYi Road, Yuzhong District, Chongqing, 400016, China
| | - Wenjing Tang
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, No 1. YouYi Road, Yuzhong District, Chongqing, 400016, China
| | - Ling Liu
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, No 1. YouYi Road, Yuzhong District, Chongqing, 400016, China.
| | - Ke Wei
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, No 1. YouYi Road, Yuzhong District, Chongqing, 400016, China.
| | - Yin Tang
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, No 1. YouYi Road, Yuzhong District, Chongqing, 400016, China
| | - Jingyue Ma
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, No 1. YouYi Road, Yuzhong District, Chongqing, 400016, China
| | - Hongbin Li
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, No 1. YouYi Road, Yuzhong District, Chongqing, 400016, China
| | - Yichan Ao
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, No 1. YouYi Road, Yuzhong District, Chongqing, 400016, China
| |
Collapse
|
19
|
Lu X, Dai S, Li P, Zhou Y, Xu F. YBX-1 alleviates sepsis-stimulated lung epithelial cell injury. Allergol Immunopathol (Madr) 2024; 52:60-67. [PMID: 38459892 DOI: 10.15586/aei.v52i2.1068] [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: 12/21/2023] [Accepted: 01/29/2024] [Indexed: 03/11/2024]
Abstract
OBJECTIVE To explore the role of Y-box binding protein 1 (YBX-1) in the lipopolysaccharide (LPS)-stimulated inflammation and oxidative stress of BEAS-2B cell line and clarify the underlying mechanism. METHODS LPS-stimulated BEAS-2B cells were used as a cell model of sepsis-stimulated acute lung injury (ALI). Immunoblot and quantitative polymerase chain reaction assays were used to detect the expression of YBX-1 in LPS-stimulated BEAS-2B cells. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide, TdT-mediated dUTP nick end labeling, and immunoblot assays were conducted to determine the effects of YBX-1 on cell survival. JC-1 staining and adenosine triphosphate production were used to detect the effects of YBX-1 on mitochondrial function. Immunostaining and enzyme-linked immunosorbent serologic assay were performed to examine the effects of YBX-1 on the inflammation and oxidative stress of cells. Immunoblot assay was conducted to confirm the mechanism. RESULTS YBX-1 was lowly expressed in LPS-stimulated BEAS-2B cells and enhanced the survival of LPS-stimulated lung epithelial cells. In addition, YBX-1 improved mitochondrial function of LPS-stimulated BEAS-2B cells. YBX-1 inhibited the inflammation and oxidative stress of LPS-stimulated BEAS-2B cells. Mechanically, YBX-1 inhibited mitogen-activated protein kinase (MAPK) axis, thereby alleviating sepsis-stimulated ALI. CONCLUSION YBX-1 alleviated inflammation and oxidative stress of LPS-stimulated BEAS-2B cells via MAPK axis.
Collapse
Affiliation(s)
- Xin Lu
- Department of Emergency Medicine, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Shouqian Dai
- Department of Emergency Medicine, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Pengfei Li
- Department of Emergency Medicine, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Yuqian Zhou
- Department of Emergency Medicine, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Feng Xu
- Department of Emergency Medicine, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China;
| |
Collapse
|