1
|
Zhou P, Liu M, Lv T. Ferroptosis targeting offers a therapeutic target for septic cardiomyopathy. Tissue Cell 2025; 95:102930. [PMID: 40288080 DOI: 10.1016/j.tice.2025.102930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/05/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Sepsis-induced cardiac dysfunction, usually termed sepsis-induced cardiomyopathy or septic cardiomyopathy(SCM), is developed in approximately 70 % of the patients with sepsis, making it is a major concern for sepsis patients. However, the pathogenesis of SCM remain incompletely understood. Ferroptosis, a newly identified mechanism of regulated cell death, characterized by a decline in antioxidant capacity, iron accumulation, and lipid peroxidation(LPO), is involved in sepsis and SCM. Moreover, ferroptosis inhibitors confer a novel therapeutic regimen in SCM. In this Review, we first summarizes the core mechanism of ferroptosis, with an emphasis on how best to interpret ferroptosis leads to the genesis of SCM. We then highlights our focus on the emerging different types of therapeutic ferroptosis inhibitors and summarizes their pharmacological beneficial effect to treat SCM. This review highlights a novel potential therapeutic strategy for SCM by pharmacologically inhibiting ferroptosis.
Collapse
Affiliation(s)
- Pengsi Zhou
- Department of Cardiology, The Affiliated Hospital of Chifeng University, Chifeng 024005, China.
| | - Mengxue Liu
- Department of Cardiology, The Affiliated Hospital of Chifeng University, Chifeng 024005, China
| | - Tao Lv
- Department of Cardiology, The Affiliated Hospital of Chifeng University, Chifeng 024005, China.
| |
Collapse
|
2
|
Sun X, Teng R, Xu N, Sun Y, Zhang E, Chen X, Guo Q, Li S. PFOS exposure impairs porcine oocyte maturation and embryo development via mitochondria-dependent ferroptosis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 374:126185. [PMID: 40189092 DOI: 10.1016/j.envpol.2025.126185] [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: 01/03/2025] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025]
Abstract
Perfluorooctane sulfonate (PFOS) is a widely utilized chemical known for its exceptional environmental stability over extended periods, its significant potential to bioaccumulate in living organisms, and its considerable risks to both health and the environment. Several studies have suggested that PFOS may pose reproductive risks in mammals; however, the exact mechanisms driving these effects are not well understood. In this study, we explored the possible mechanisms by which PFOS toxicity affects the maturation of mammalian oocytes and the embryonic development employing porcine oocytes as a model system. SMART-seq results suggested that PFOS may affect oocyte maturation through mechanisms involving ferroptosis, autophagy, and alterations in membrane structure. Our results suggest that PFOS exposure adversely affects mitochondrial function and structure, thereby influencing peroxisome biogenesis and contributing to oxidative stress. Most importantly, we found that exposure to PFOS significantly elevated Fe2+ levels, an indicator associated with ferroptosis in oocytes. Furthermore, malondialdehyde (MDA) levels in the PFOS group were significantly higher than those in the control group. Additionally, the mRNA expression levels of PCBP1 and PCBP2, which are related to ferroptosis, as well as the expression level of P53, were significantly reduced in the PFOS group. Overall, exposure to PFOS in vitro results in mitochondrial damage in porcine oocytes, which induces lipid peroxidation and subsequently leads to the occurrence of ferroptosis.
Collapse
Affiliation(s)
- Xiaoqing Sun
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Ran Teng
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Ning Xu
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Yutong Sun
- Affiliated Middle School to Jilin University, Changchun, 130000, China
| | - Enbo Zhang
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Xingfu Chen
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Qing Guo
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Suo Li
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China.
| |
Collapse
|
3
|
Li Z, Bu Y, Wang C, Yu Y, Han L, Liu C, Chen G, Li C, Zhang Y, Cao H, Ma Z, Yue Z. Extracellular vesicle-packaged GBP2 from macrophages aggravates sepsis-induced acute lung injury by promoting ferroptosis in pulmonary vascular endothelial cells. Redox Biol 2025; 82:103614. [PMID: 40156957 PMCID: PMC11994402 DOI: 10.1016/j.redox.2025.103614] [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: 12/18/2024] [Revised: 03/13/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Macrophages play a critical role in the development of sepsis-induced acute lung injury (si-ALI), with extracellular vesicles (EVs) acting as crucial mediators. However, the effects and mechanisms of macrophage-derived EVs on si-ALI remain unclear. This study demonstrated that macrophage-derived EVs induce endothelial ferroptosis and barrier disruption during sepsis. Through proteomic sequencing and reanalysis of transcriptomic and single-cell sequencing data, guanylate-binding protein 2 (GBP2) was identified as a key EV molecule. Elevated GBP2 expression was observed in EVs and monocytes from the peripheral blood of sepsis patients, in LPS-stimulated THP-1 and RAW264.7 cells and their secreted EVs, and in macrophages within the lungs of CLP mice. Additionally, GBP2 expression in EVs showed a positive correlation with vascular barrier injury biomarkers, including ANGPT2, Syndecan-1, and sTM. Modulating GBP2 levels in macrophage-derived EVs affected EV-induced ferroptosis in endothelial cells. The mechanism by which GBP2 binds directly to OTUD5 and promotes GPX4 ubiquitination was elucidated using RNA interference, adeno-associated virus transfection, and endothelial-specific Gpx4 knockout mice. A high-throughput screening of small-molecule compounds targeting GBP2 was conducted. Molecular docking, molecular dynamics simulations, and cellular thermal shift assays further confirmed that Plantainoside D (PD) has a potent binding affinity for GBP2. PD treatment inhibited the interaction between GBP2 and OTUD5, leading to a reduction in GPX4 ubiquitination. Further research revealed that PD treatment enhanced the pulmonary protective effects of GBP2 inhibition. In conclusion, this study explored the role of EV-mediated signaling between macrophages and pulmonary vascular endothelial cells in si-ALI, highlighting the GBP2-OTUD5-GPX4 axis as a driver of endothelial ferroptosis and lung injury. Targeting this signaling axis presents a potential therapeutic strategy for si-ALI.
Collapse
Affiliation(s)
- Zhixi Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, 150001, PR China; The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, 150001, PR China; State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, 150001, PR China
| | - Yue Bu
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, 150001, PR China; Department of Pain Medicine, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China
| | - Cheng Wang
- Department of Environmental Hygiene, School of Public Health, Harbin Medical University, Harbin, 150081, PR China
| | - Yongjing Yu
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, 150001, PR China; The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, 150001, PR China; State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, 150001, PR China
| | - Lei Han
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, 150001, PR China
| | - Chang Liu
- The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, 150001, PR China; The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, 150001, PR China; State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, 150001, PR China; Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, 150081, PR China
| | - Guangmin Chen
- Department of Anesthesiology, First Affiliated Hospital of Harbin Medical University, 199 Dazhi Road, Harbin, 150001, PR China
| | - Chenglong Li
- Department of Anesthesiology, Fourth Affiliated Hospital of Harbin Medical University, 37 Yiyuan Road, Harbin, 150001, PR China
| | - Yan Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, 150001, PR China
| | - Hang Cao
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, 150001, PR China
| | - Zhaoxue Ma
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, 150001, PR China
| | - Ziyong Yue
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, PR China; The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, 150001, PR China.
| |
Collapse
|
4
|
Teng X, Wang Q, Ma J, Li D. Integrating bioinformatics and machine learning to discover sumoylation associated signatures in sepsis. Sci Rep 2025; 15:14398. [PMID: 40274894 PMCID: PMC12022290 DOI: 10.1038/s41598-025-96956-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 04/01/2025] [Indexed: 04/26/2025] Open
Abstract
Small Ubiquitin-like MOdifier-mediated modification (SUMOylation) is associated with sepsis; however, its molecular mechanism remains unclear. Herein, hub genes and regulatory mechanisms in sepsis was investigated. The GSE65682 and GSE95233 datasets were extracted from public databases. Differential analysis and Weighted Gene Co-expression Network Analysis (WGCNA) were conducted in GSE65682 to identify differentially expressed genes (DEGs) and key module genes. Candidate genes were derived by intersecting with SUMOylation-related genes (SUMO-RGs). The Least Absolute Shrinkage and Selection Operator (LASSO) and Support Vector Machine-Recursive Feature Elimination (SVM-RFE) were utilized to identify significant feature genes. The convergence of those genes was utilized for diagnostic assessment and expression validation. Hub genes were defined as those exhibiting an area under the curve (AUC) greater than 0.7, significant gene expression, and a consistent trend. Localization and functional analyses of hub genes were conducted to enhance the understanding of these genes. Immune analysis, regulatory network construction, and drug prediction were performed. Six hub genes were identified: RORA, L3MBTL2, PHC1, RPA1, CHD3, and RANGAP1. These genes possessed considerable diagnostic significance for sepsis and were also markedly downregulated in the condition. Hub genes were predominantly enriched in the ribosome pathway and exhibited a strong correlation with differential immune cells. Activated CD8 + T cells exhibited a positive correlation with RORA. Based on the predicted and established regulatory network, AC004687.1 was observed to modulate PHC1 expression via hsa-miR- 142 - 5p. A total of six hub genes (RORA, L3MBTL2, PHC1, RPA1, CHD3, and RANGAP1) associated with SUMOylation was identified in sepsis in the current study. The findings are likely to aid in the differentiation between control and disease states, offering substantiation for the diagnosis of sepsis.
Collapse
Affiliation(s)
- Xue Teng
- Department of Anesthesiology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China
- The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, Heilongjiang, China
| | - Qi Wang
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Jinling Ma
- Department of Intensive Care Medicine, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China
| | - Dongmei Li
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
- The Key Laboratory of Anesthesiology and Intensive Care Research of Heilongjiang Province, Harbin, Heilongjiang, China.
| |
Collapse
|
5
|
Wang TW, Zhou LL, Yuan J, Zhou WX, Wang HR, Yu TT, Zhai JC, Tang CB, Jiang W, Yu JQ, Zheng RQ, Yu HL, Shao J. Study of the relationship between iron metabolism disorders and sepsis-associated liver injury: A prospective observational study. World J Gastroenterol 2025; 31:104584. [PMID: 40248384 PMCID: PMC12001195 DOI: 10.3748/wjg.v31.i14.104584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/17/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Sepsis-associated liver injury (SALI) refers to secondary liver function impairment caused by sepsis, patients with SALI often have worse clinical outcomes. The early identification and assessment of the occurrence and progression of SALI are pressing issues that urgently need to be resolved. AIM To investigate the relationship between iron metabolism and SALI. METHODS In this prospective study, 139 patients were recruited, with 53 assigned to the SALI group. The relationships between SALI and various iron metabolism-related biomarkers were examined. These biomarkers included serum iron (SI), total iron-binding capacity (TIBC), serum ferritin, transferrin, and transferrin saturation. To identify independent risk factors for SALI, both univariate and multivariate logistic regression analyses were performed. Additionally, receiver operating characteristic curve analysis was utilized to assess the predictive value of these biomarkers for the occurrence of SALI. RESULTS There were no statistically significant differences in age, sex, body mass index, Sequential Organ Failure Assessment scores (excluding liver function), or APACHE II scores between the two groups of patients. Compared with the sepsis group, the SALI group presented significantly higher SI (P < 0.001), TIBC (P < 0.001), serum ferritin (P = 0.001), transferrin (P = 0.005), and transferrin saturation levels (P < 0.001). Multivariate logistic regression analysis revealed that SI (odds ratio = 1.24, 95% confidence interval: 1.11-1.40, P < 0.001) and TIBC levels (odds ratio = 1.13, 95% confidence interval: 1.05-1.21, P < 0.001) were independent predictors of SALI. Receiver operating characteristic curve analysis revealed that SI and TIBC had areas under the curve of 0.816 and 0.757, respectively, indicating moderate predictive accuracy for SALI. CONCLUSION Iron metabolism disorders are closely associated with the development of SALI, and SI and TIBC may serve as potential predictive biomarkers. The combined use of SI and TIBC has superior diagnostic efficacy for SALI. These findings provide valuable insights for the early identification and management of SALI among patients with sepsis.
Collapse
Affiliation(s)
- Tian-Wei Wang
- Department of Intensive Care Unit, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Lu-Lu Zhou
- Department of Intensive Care Unit, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Jing Yuan
- Department of Functional Examination, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Wen-Xin Zhou
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Hao-Ran Wang
- Department of Intensive Care Unit, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Ting-Ting Yu
- Department of Intensive Care Unit, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Ji-Chao Zhai
- Department of Intensive Care Unit, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Cheng-Bin Tang
- Department of Center for Cardiac Macrovascular Disease, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Wei Jiang
- Department of Intensive Care Unit, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Jiang-Quan Yu
- Department of Intensive Care Unit, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Rui-Qiang Zheng
- Department of Intensive Care Unit, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Hai-Long Yu
- Department of Neuro Intensive Care Unit, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
- Department of Neurology, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - Jun Shao
- Department of Center for Cardiac Macrovascular Disease, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| |
Collapse
|
6
|
Zhang X, Zhou Y, Li H, Chen M, Peng F, Li N. Integration of Transcriptomic and Single-Cell Data to Uncover Senescence- and Ferroptosis-Associated Biomarkers in Sepsis. Biomedicines 2025; 13:942. [PMID: 40299574 PMCID: PMC12025025 DOI: 10.3390/biomedicines13040942] [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: 03/07/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/01/2025] Open
Abstract
Background: Sepsis is a life-threatening condition characterized by organ dysfunction due to an imbalanced immune response to infection, with high mortality. Ferroptosis, an iron-dependent cell death process, and cellular senescence, which exacerbates inflammation, have recently been implicated in sepsis pathophysiology. Methods: Weighted gene co-expression network analysis (WGCNA) was used to identify ferroptosis- and senescence-related gene modules in sepsis. Differentially expressed genes (DEGs) were analyzed using public datasets (GSE57065, GSE65682, and GSE26378). Receiver operating characteristic (ROC) analysis was performed to evaluate their diagnostic potential, while single-cell RNA sequencing (scRNA-seq) was used to assess their immune-cell-specific expression. Molecular docking was conducted to predict drug interactions with key proteins. Results: Five key genes (CD82, MAPK14, NEDD4, TXN, and WIPI1) were significantly upregulated in sepsis patients and highly correlated with immune cell infiltration. MAPK14 and TXN exhibited strong diagnostic potential (AUC = 0.983, 0.978). Molecular docking suggested potential therapeutic interactions with diclofenac, flurbiprofen, and N-acetyl-L-cysteine. Conclusions: This study highlights ferroptosis and senescence as critical mechanisms in sepsis and identifies promising biomarkers for diagnosis and targeted therapy. Future studies should focus on clinical validation and precision medicine applications.
Collapse
Affiliation(s)
- Xiangqian Zhang
- Department of Blood Transfusion, Clinical Transfusion Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yiran Zhou
- Department of Blood Transfusion, Clinical Transfusion Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Hang Li
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, 07743 Jena, Germany
| | - Mengru Chen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Fang Peng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ning Li
- Department of Blood Transfusion, Clinical Transfusion Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
7
|
Zeng Z, Deng J, Wang G, Luo Z, Xiao W, Xie W, Liu J, Li K. Ferroptosis-related protein biomarkers for diagnosis, differential diagnosis, and short-term mortality in patients with sepsis in the intensive care unit. Front Immunol 2025; 16:1528986. [PMID: 40264754 PMCID: PMC12011590 DOI: 10.3389/fimmu.2025.1528986] [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: 11/21/2024] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
Background Sepsis is a disease with high mortality caused by a dysregulated response to infection. Ferroptosis is a newly discovered type of cell death. Ferroptosis-related genes are involved in the occurrence and development of sepsis. However, research on the diagnostic value of ferroptosis-related protein biomarkers in sepsis serum is limited. This study aims to explore the clinical value of Ferroptosis-related proteins in diagnosing sepsis and predicting mortality risk. Methods A single-center, prospective, observational study was conducted from January to December 2023, involving 170 sepsis patients, 49 non-septic ICU patients, and 50 healthy individuals. Upon ICU admission, biochemical parameters, GCS, SOFA, and APACHE II scores were recorded, and surplus serum was stored at -80°C for biomarker analysis via ELISA. Diagnostic efficacy was evaluated using ROC curve analysis. Results Baseline serum levels of ACSL4, GPX4, PTGS2, CL-11, IL-6, IL-8, PCT, and hs-CRP significantly differed among sepsis, non-septic, and healthy individuals (all p-value < 0.01). ACSL4, GPX4, PTGS2, IL-6, IL-8, PCT, and hs-CRP demonstrated high diagnostic and differential diagnostic performance (AUC: 0.6688 to 0.9945). IL-10 and TNF-α showed good diagnostic performance (AUC = 0.8955 and 0.7657, respectively). ACSL4 (AUC = 0.7127) was associated with predicting sepsis mortality. Serum levels of ACSL4, CL-11, and IL-6 above the cut-off value were associated with shorter survival times. ACSL4 levels were positively correlated with SOFA (Rho = 0.354, p-value < 0.0001), APACHE II (Rho = 0.317, p-value < 0.0001), and septic shock (Rho = 0.274, p-value = 0.003) scores but negatively correlated with the GCS score (Rho = -0.218, p-value = 0.018). GPX4 levels were positively correlated with SOFA (Rho = 0.204, p-value = 0.027) and APACHE II (Rho = 0.233, p-value = 0.011) scores. Conclusion ACSL4 and GPX4 have strong diagnostic and differential diagnostic value in sepsis, including the ability to predict 28-day mortality in sepsis patients, and may become new potential serum markers for the diagnostic and differential diagnostic of sepsis.
Collapse
Affiliation(s)
- Zhangrui Zeng
- Department of Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jie Deng
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Province Engineering Technology Research Center of Clinical Diseases Molecular Diagnosis, Luzhou, China
- Clinical Diseases Molecular Diagnosis Key Laboratory of LuZhou, Luzhou, China
| | - Gang Wang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Zixiang Luo
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Province Engineering Technology Research Center of Clinical Diseases Molecular Diagnosis, Luzhou, China
- Clinical Diseases Molecular Diagnosis Key Laboratory of LuZhou, Luzhou, China
| | - Weijia Xiao
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Province Engineering Technology Research Center of Clinical Diseases Molecular Diagnosis, Luzhou, China
- Clinical Diseases Molecular Diagnosis Key Laboratory of LuZhou, Luzhou, China
| | - Wenchao Xie
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Province Engineering Technology Research Center of Clinical Diseases Molecular Diagnosis, Luzhou, China
- Clinical Diseases Molecular Diagnosis Key Laboratory of LuZhou, Luzhou, China
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Province Engineering Technology Research Center of Clinical Diseases Molecular Diagnosis, Luzhou, China
- Clinical Diseases Molecular Diagnosis Key Laboratory of LuZhou, Luzhou, China
| | - Ke Li
- Department of Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
8
|
Yin Y, Mu F, Zhang L, Zhao J, Gong R, Yin Y, Zheng L, Du Y, Jin F, Wang J. Wedelolactone activates the PI3K/AKT/NRF2 and SLC7A11/GPX4 signalling pathways to alleviate oxidative stress and ferroptosis and improve sepsis-induced liver injury. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119557. [PMID: 40010556 DOI: 10.1016/j.jep.2025.119557] [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: 12/20/2024] [Revised: 02/11/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025]
Abstract
INTRODUCTION Sepsis-induced liver injury (SILI) is a severe complication of sepsis. Wedelolactone (WEL) can be used to treat liver diseases. However, its therapeutic mechanisms and efficacy in SILI remain unclear. To investigate the therapeutic effects of WEL on SILI and its potential mechanisms of action through in vitro and in vivo experiments. METHODS A SILI model based on lipopolysaccharide (LPS), and AML12 cells were treated with different concentrations of WEL, LY294002 and ML385. The SILI model was established by caecal ligation and puncture (CLP). C57BL/6 mice were administered WEL and biphenyl diester for seven consecutive days, and CLP was then performed 1 h later. Blood and liver tissue were collected 24 h later for subsequent analysis. HE staining, liver function index, oxidative stress index, JC-1 staining, transmission electron microscopy, immunofluorescence staining, Western blot, and inflammatory cytokines were used to detect oxidative stress and ferroptosis-related markers. RESULTS The in vivo experiments showed that WEL treatment reduced the pathological damage of the liver and decreased ALT and AST, MMP and ROS (the product of iron and lipid peroxidation) and inflammatory factors. WEL also decreased hepatocyte viability in vitro. Inhibition of NRF2 can lead to exacerbation of SILI. The expressions of P-PI3K and P-AKT were up-regulated while HO-1, GPX4, NRF2, and SLC7A11 were down-regulated in vitro and in vivo. CONCLUSIONS Ferroptosis and oxidative stress are pivotal in SILI. WEL mitigates SILI by inhibiting ferroptosis and oxidative stress, primarily through the PI3K/AKT/NRF2 and SLC7A11/GPX4 signalling pathways, thus suggesting a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yanping Yin
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China; College of Life Science, Northwest University, Xi'an, 710069, China
| | - Fei Mu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lulu Zhang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jinyi Zhao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Rui Gong
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yanli Yin
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lingling Zheng
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yang Du
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Fuxing Jin
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jingwen Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
9
|
Lu J, Wang J, Han K, Tao Y, Dong J, Pan X, Wen X. Identification and validation of m 6A RNA methylation and ferroptosis-related biomarkers in sepsis: transcriptome combined with single-cell RNA sequencing. Front Immunol 2025; 16:1543517. [PMID: 40124361 PMCID: PMC11925765 DOI: 10.3389/fimmu.2025.1543517] [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: 12/11/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
Background Sepsis, a systemic inflammatory response syndrome triggered by infection, is associated with high mortality rates and an increasing global incidence. While N 6-methyladenosine (m6A) RNA methylation and ferroptosis are implicated in inflammatory diseases, their specific genes and mechanisms in sepsis remain unclear. Methods Transcriptomic datasets of sepsis, along with m6A-related genes (m6A-RGs) and ferroptosis-related genes (FRGs), were sourced from public databases. Differentially expressed genes (DEGs) were identified between the sepsis and control groups, and m6A-RGs were analyzed through weighted gene co-expression network analysis (WGCNA) to uncover m6A module genes. These were then intersected with DEGs and FRGs to identify candidate genes. Biomarkers were identified using two machine learning methods, receiver operating characteristic (ROC) curves, and expression validation, followed by the development of a nomogram. Further in-depth analyses of the biomarkers were performed, including functional enrichment, immune infiltration, drug prediction, and molecular docking. Single-cell analysis was conducted to identify distinct cell clusters and evaluate biomarker expression at the single-cell level. Finally, reverse transcription-quantitative PCR (RT-qPCR) was employed to validate biomarker expression in clinical samples. Results DPP4 and TXN were identified as key biomarkers, showing higher expression in control and sepsis samples, respectively. The nomogram incorporating these biomarkers demonstrated strong diagnostic potential. Enrichment analysis highlighted their involvement in spliceosome function and antigen processing and presentation. Differential analysis of immune cell types revealed significant correlations between biomarkers and immune cells, such as macrophages and activated dendritic cells. Drug predictions identified gambogenic acid and valacyclovir as potential treatments, which were successfully docked with the biomarkers. Single-cell analysis revealed that the biomarkers were predominantly expressed in CD4+ memory cells, and CD16+ and CD14+ monocytes. The expression of DPP4 was further validated in clinical samples. Conclusions DPP4 and TXN were validated as biomarkers for sepsis, with insights into immune infiltration and therapeutic potential at the single-cell level, offering novel perspectives for sepsis treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaolan Wen
- Department of Emergency, People’s Hospital of Xinjiang Uygur Autonomous
Region, Urumqi, China
| |
Collapse
|
10
|
Xu J, Liang C, Yao S, Wang F. Melatonin Exerts Positive Effects on Sepsis Through Various Beneficial Mechanisms. Drug Des Devel Ther 2025; 19:1333-1345. [PMID: 40026332 PMCID: PMC11871935 DOI: 10.2147/dddt.s509735] [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: 12/02/2024] [Accepted: 02/12/2025] [Indexed: 03/05/2025] Open
Abstract
In recent years, our understanding of sepsis has greatly advanced. However, due to the complex pathological and physiological mechanisms of sepsis, the mechanisms of sepsis are currently not fully elucidated, and it is difficult to translate the research results into specific sepsis treatment methods. Melatonin possesses broad anti-inflammatory, antioxidant, and immune-regulatory properties, making it a promising therapeutic agent for sepsis. In recent years, further research has deepened our understanding of the potential mechanisms and application prospects of melatonin in sepsis. The mechanisms underlying the protective effects of melatonin in sepsis are multifaceted. In this review, based on a substantial body of clinical trials and animal research findings, we first highlighted the significance of melatonin as an important biomarker for disease progression and prognosis in sepsis. We also described the extensive regulatory mechanisms of melatonin in sepsis-induced organ damage. In addition to its broad anti-inflammatory, and anti-oxidant effects, melatonin exerts positive effects by regulating metabolic disorders, hemodynamics, cell autophagy, cellular ion channels, endothelial cell permeability, ferroptosis and other complex pathological mechanisms. Furthermore, as a safe exogenous supplement with low toxicity, melatonin demonstrates positive synergistic effects with other anti-sepsis agents. In the face of the urgent medical challenge of transforming the increasing knowledge of sepsis molecular mechanisms into therapeutic interventions to improve patient prognosis, melatonin seems to be a promising option.
Collapse
Affiliation(s)
- Jing Xu
- Department of Critical Care Medicine, Capital Medical University Electric Power Teaching Hospital/State Grid Beijing Electric Power Hospital, Beijing, People’s Republic of China
| | - Cui Liang
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Shanglong Yao
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Fuquan Wang
- Department of Pain Management, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| |
Collapse
|
11
|
Fan M, Chen M, Gao Y, Jiang H, Li Y, Zhu G, Chen S, Xu Y, Chen X. Construction of a novel gene signature linked to ferroptosis in pediatric sepsis. Front Cell Dev Biol 2025; 13:1488904. [PMID: 40070882 PMCID: PMC11893615 DOI: 10.3389/fcell.2025.1488904] [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: 08/30/2024] [Accepted: 01/20/2025] [Indexed: 03/14/2025] Open
Abstract
Introduction Pediatric sepsis is a complex and life-threatening condition characterized by organ failure due to an uncontrolled immune response to infection. Recent studies suggest that ferroptosis, a newly identified form of programmed cell death, may play a role in sepsis progression. However, the specific mechanisms of ferroptosis in pediatric sepsis remain unclear. Methods In this study, we analyzed microarray datasets from pediatric sepsis and healthy blood samples to identify ferroptosis-associated genes. A protein-protein interaction (PPI) network analysis and histological validation were performed to identify key genes. Additionally, immune infiltration analysis was conducted to explore the correlation between immune cells, immune checkpoint-related genes, and key genes. A competing endogenous RNA (ceRNA) network was constructed to investigate potential regulatory mechanisms involving long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and key ferroptosis-related genes. Results We identified 74 genes associated with ferroptosis in pediatric sepsis. Among them, five key genes (MAPK3, MAPK8, PPARG, PTEN, and STAT3) were confirmed through PPI network analysis and histological validation. Immune infiltration analysis revealed significant interactions between immune cells and key genes. The ceRNA network provided insights into the regulatory relationships between lncRNAs, miRNAs, and ferroptosis-related genes. Discussion These findings enhance our understanding of the role of ferroptosis in pediatric sepsis and highlight potential therapeutic targets for future research and clinical interventions.
Collapse
Affiliation(s)
- Mingyuan Fan
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Meiting Chen
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yongqi Gao
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Huilin Jiang
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yanling Li
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Gongxu Zhu
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shengkuan Chen
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yiming Xu
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xiaohui Chen
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
12
|
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
|
13
|
Sun M, Xu D, Liu D, Ran X, Li F, Wang J, Ge Y, Liu Y, Guo W, Liu J, Cao Y, Fu S. Stigmasterol from Prunella vulgaris L. Alleviates LPS-induced mammary gland injury by inhibiting inflammation and ferroptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 137:156362. [PMID: 39809030 DOI: 10.1016/j.phymed.2025.156362] [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: 09/11/2024] [Revised: 12/21/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025]
Abstract
BACKGROUND Dairy mastitis, a prevalent condition affecting dairy cattle, represents a significant challenge to both animal welfare and the quality of dairy products. However, current treatment options remain limited. Stigmasterol (ST) is a bioactive component of Prunella vulgaris L. (PV) with various pharmacological functions such as anti-inflammatory and anti-oxidation. At present, the specific effects and underlying mechanisms of PV and ST on dairy mastitis are still not fully understood. PURPOSE The aim of this research was to evaluate the pharmacological effects of PV and its active component ST on lipopolysaccharide (LPS) -stimulated bovine mammary epithelial cells (BMECs) and a mouse mastitis model, and to elucidate the possible mechanisms of action. METHODS UPLC-Q-TOF-MS/MS was employed to identify the constituents of PV. BMECs and mice were used to establish in vitro and in vivo models of mastitis. Western Blotting, RT-qPCR, immunofluorescence and other techniques were used to explore the effects of PV and ST on inflammatory factors, blood-milk barrier integrity, ferroptosis related indicators and their potential molecular mechanisms. RESULTS PV significantly attenuated the production of inflammatory mediators by LPS-stimulated BMECs. Subsequently, ST was found to be a potent anti-inflammatory agent in PV by inhibiting TLR4/NF-κB signaling pathway. This inhibition inhibits the myosin light chain (MLC)/MLC kinase signaling cascade and alleviates blood-milk barrier (BMB) disruption in BMECs. In addition, ferroptosis occurred in BMECs after LPS stimulation, and ST inhibited ferroptosis by stimulating Nrf2/GPX4 signaling pathway. Treatment of BMECs with the Nrf2 inhibitor ML385 significantly attenuated the therapeutic effect of ST. In vivo experiments further confirmed that both PV and ST attenuated LPS-induced breast tissue damage while reducing ferroptosis levels and restoring BMB. CONCLUSION ST from PV exhibits substantial anti-inflammatory properties and is a promising candidate for the treatment of dairy mastitis.
Collapse
Affiliation(s)
- Mingyang Sun
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Dianwen Xu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, Gansu, China
| | - Dianfeng Liu
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, China
| | - Xin Ran
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Feng Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jiaxin Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yusong Ge
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuhao Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Wenjin Guo
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Juxiong Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yu Cao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Shoupeng Fu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
14
|
Bai X, Liu Y, Liu J, Guo K, Guan H. ADSCs-derived exosomes suppress macrophage ferroptosis via the SIRT1/NRF2 signaling axis to alleviate acute lung injury in sepsis. Int Immunopharmacol 2025; 146:113914. [PMID: 39732105 DOI: 10.1016/j.intimp.2024.113914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/08/2024] [Accepted: 12/18/2024] [Indexed: 12/30/2024]
Abstract
Acute lung injury being one of the earliest and most severe complications during sepsis and macrophages play a key role in this process. To investigate the regulatory effects and potential mechanisms of adipose mesenchymal stem cell derived-exosomes (ADSC-exo) on macrophages and septic mice, ADSCs-exo was administrated to both LPS-induced macrophage and cecal ligation and puncture (CLP) induced sepsis mice. ADSCs-exo was confirmed to inhibit M1 polarization of macrophages and to reduce excessive inflammation. The use of ADSCs-exo in CLP mice and in LPS-induced macrophages relieved oxidative stress, cellular damage, and acute lung injury. During this process, ADSCs-exo increased the nuclear translocation of Nrf2, significantly upregulating the activation of the antioxidant pathway Nrf2/HO-1. Concurrently, they enhanced the expression of SIRT1 in macrophages. Further SIRT1 interference experiments demonstrated that ADSCs-exo mitigated macrophage inflammatory responses and LPS-induced ferroptosis by upregulating SIRT1. In the LPS-induced macrophage model, after SIRT1 was interfered with, ADSCs-exo failed to upregulate the Nrf2/HO-1 signaling pathway, leading to enhanced ferroptosis. Finally, in a CLP sepsis mouse model with myeloid-specific SIRT1 knockout, ADSCs-exo was observed to reduce lung tissue injury, oxidative stress damage, and ferroptosis. Still, these beneficial effects were reversed due to the myeloid-specific knockout of SIRT1, while co-administration of a ferroptosis inhibitor rescued this situation, alleviating lung injury and significantly reducing tissue levels of oxidative stress. In conclusion, this study elucidated a novel potential therapeutic mechanism wherein ADSCs-exo upregulates the levels of SIRT1 in macrophages through a non-delivery approach.
Collapse
Affiliation(s)
- Xiaozhi Bai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi 710032, China
| | - Yang Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi 710032, China
| | - Jiaqi Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi 710032, China
| | - Kai Guo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi 710032, China
| | - Hao Guan
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
15
|
Zhang H, Wang Y, Wang S, Xue X, Huang K, Xu D, Jiang L, Li S, Zhang Y. Tangeretin alleviates sepsis-induced acute lung injury by inhibiting ferroptosis of macrophage via Nrf2 signaling pathway. Chin Med 2025; 20:11. [PMID: 39815349 PMCID: PMC11734455 DOI: 10.1186/s13020-025-01063-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Sepsis-induced acute lung injury (ALI) is a severe clinical condition accompanied with high mortality. Tangeretin, which is widely found in citrus fruits, has been reported to exert antioxidant and anti-inflammatory properties. However, whether tangeretin protects against sepsis-induced ALI and the potential mechanisms remain unclear. METHODS We established an ALI model via intraperitoneally injected with 5 mg/kg lipopolysaccharides (LPS) for 12 h. Tangeretin was applied intraperitoneally 30 min before LPS treatment. Dexamethasone (Dex) was used as a positive control. Hematoxylin and eosin (HE) staining and protein content in bronchoalveolar lavage fluid (BALF) were determined to detect the degree of lung injury. RNA-seq was also applied to explore the effect of tangeretin on ALI. In vitro, RAW264.7 were treated with Nrf2 siRNA, the expression of ferroptosis-associated biomarkers, including glutathione peroxidase 4 (GPX4) and prostaglandin-endoperoxide synthase 2 (PTGS2) were assessed. Glutathione (GSH), malondialdehyde (MDA) levels, reactive oxygen species (ROS) and inflammatory factors were also determined both in vivo and in vitro. Furthermore, mice were treated with an Nrf2 inhibitor (ML385) to verify the mechanism of tangeretin in inhibiting sepsis-induced lung injury and ferroptosis. Data were analyzed using one way analysis of variance or two-tailed unpaired t tests. RESULTS Our study demonstrated that tangeretin significantly alleviated lung injury, reversed the LPS-induced reduction in GPX4 and GSH, and mitigates the elevation of PTGS2 and MDA levels. Tangeretin also reduced 4-HNE and iron levels. Besides, the levels of LPS-stimulated inflammatory factors IL-6, IL-1β and TNF-α were also decreased by tangeretin. RNA-seq and bioinformatics analysis demonstrated that tangeretin inhibited inflammatory response. Mechanistically, we identified that tangeretin inhibited the GPX4-dependent lipid peroxidation through activation of Nrf2. The silence of Nrf2 abolished the inhibitory effect of tangeretin on oxidative stress, inflammatory response and ferroptosis in RAW264.7 cells. Additionally, all the protective effects of tangeretin on ALI were abolished in Nrf2 inhibitor-treated mice. CONCLUSION We identified that ferroptosis as a critical mechanism contributing to sepsis-induced ALI. Tangeretin, a promising therapeutic candidate, effectively mitigates ALI through inhibiting ferroptosis via upregulating Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yan Wang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Shenghua Wang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Xiaomei Xue
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Kai Huang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Dunfeng Xu
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Siyuan Li
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| | - Yunqian Zhang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
16
|
Tao L, Xu J, Jiang L, Hu J, Tang Z. Investigation into the influence of mild hypothermia on regulating ferroptosis through the P53-SLC7A11/GPX4 signaling pathway in sepsis-induced acute lung injury. Intensive Care Med Exp 2025; 13:4. [PMID: 39812923 PMCID: PMC11735705 DOI: 10.1186/s40635-025-00713-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Sepsis-induced acute lung injury (S-ALI) significantly contributes to unfavorable clinical outcomes. Emerging evidence suggests a novel role for ferroptosis in the pathophysiology of ALI, though the precise mechanisms remain unclear. Mild hypothermia (32-34 °C) has been shown to inhibit inflammatory responses, reduce oxidative stress, and regulate metabolic processes. P53 has been reported to downregulate the transcriptional activity of solute carrier family 7 member 11 (SLC7A11), thereby limiting cystine uptake. This reduction in cystine availability compromises the activity of Glutathione peroxidase 4 (GPX4), a cystine-dependent enzyme, ultimately increasing cellular susceptibility to ferroptosis. However, it remains unclear whether mild hypothermia exerts protective effects through the P53-SLC7A11/GPX4 signaling pathway. This study investigates the influence of mild hypothermia on ferroptosis mediated by the P53-SLC7A11/GPX4 pathway in S-ALI. METHODS This study utilized both in vivo and in vitro models. In the vivo model, 64 Sprague-Dawley rats were employed, with 40 analyzed for survival outcomes. Sepsis was induced using the cecum ligation and puncture (CLP) method, after which rats were subjected to either normothermic (36-38 °C) or mild hypothermic (32-34 °C) conditions for a duration of 10 h. Twelve hours post-surgery, blood samples, bronchoalveolar lavage fluid, and lung tissue samples were harvested for histological analysis, measurement of inflammatory markers, wet/dry ratios, blood gas analysis, assessment of oxidative stress and ferroptosis, Western blotting, and RT-qPCR analysis. In the in vitro model, RLE-6TN cells were exposed to lipopolysaccharide (LPS) for 24 h under normothermic and mild hypothermic conditions. These cells were then evaluated for cell viability, inflammatory markers, oxidative stress levels, ferroptosis markers, as well as Western blot and RT-qPCR analyses. RESULTS CLP-induced sepsis led to elevated levels of inflammatory markers, increased lung injury scores, and heightened oxidative stress markers. These detrimental effects were significantly ameliorated by mild hypothermia. Furthermore, mild hypothermia reversed the modified expression of P53, SLC7A11, and GPX4 signaling molecules. Notably, mild hypothermia also improved the 5-day survival rate of CLP rats. CONCLUSION Mild hypothermia attenuates S-ALI and modulates ferroptosis through the P53-SLC7A11/GPX4 signaling pathway.
Collapse
Affiliation(s)
- Liujun Tao
- Intensive Care Unit, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Jie Xu
- Intensive Care Unit, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Liangyan Jiang
- Intensive Care Unit, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Juntao Hu
- Intensive Care Unit, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Zhanhong Tang
- Intensive Care Unit, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
17
|
Iba T, Helms J, Maier CL, Ferrer R, Levy JH. Mitochondrial dysfunction is a major cause of thromboinflammation and inflammatory cell death in critical illnesses. Inflamm Res 2025; 74:17. [PMID: 39806233 DOI: 10.1007/s00011-025-01994-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/02/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Mitochondria generate the adenosine triphosphate (ATP) necessary for eukaryotic cells, serving as their primary energy suppliers, and contribute to host defense by producing reactive oxygen species. In many critical illnesses, including sepsis, major trauma, and heatstroke, the vicious cycle between activated coagulation and inflammation results in tissue hypoxia-induced mitochondrial dysfunction, and impaired mitochondrial function contributes to thromboinflammation and cell death. METHODS A computer-based online search was performed using the PubMed and Web of Science databases for published articles concerning sepsis, trauma, critical illnesses, cell death, mitochondria, inflammation, coagulopathy, and organ dysfunction. RESULTS Mitochondrial outer membrane permeabilization triggers apoptosis by releasing cytochrome c and activating caspases. Apoptosis is a non-inflammatory programmed cell death but requires sufficient ATP supply. Therefore, conversion to inflammatory necrosis may occur due to a lack of ATP in critical illness. Severely damaged mitochondria release excess reactive oxygen species and injurious mitochondrial DNA, inducing cell death. Besides non-programmed necrosis, mitochondrial damage can trigger programmed inflammatory cell death, including necroptosis, pyroptosis, and ferroptosis. Additionally, a unique form of DNA-ejecting cell death, known as etosis, occurs in monocytes and granulocytes following external stimuli and mitochondrial damage. The type of cell death chosen remains uncertain but is known to depend on the cell type, the nature of the injury, and the degree of damage. CONCLUSIONS Mitochondria damage is the major contributor to the cell death mechanism that leads to organ damage in critical illnesses. Regulating and restoring mitochondrial function holds promise for developing new therapeutic approaches for mitigating critical diseases.
Collapse
Affiliation(s)
- Toshiaki Iba
- Department of Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Julie Helms
- Medical Intensive Care Unit - NHC, Strasbourg University (UNISTRA), Strasbourg University Hospital, INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, Strasbourg, France
| | - Cheryl L Maier
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Ricard Ferrer
- Intensive Care Department, Hospital Universitari Vall d'Hebron Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jerrold H Levy
- Department of Anesthesiology, Critical Care, and Surgery, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
18
|
de Man AM, Stoppe C, Koekkoek KW, Briassoulis G, Subasinghe LS, Cobilinschi C, Deane AM, Manzanares W, Grințescu I, Mirea L, Roshdy A, Cotoia A, Bear DE, Boraso S, Fraipont V, Christopher KB, Casaer MP, Gunst J, Pantet O, Elhadi M, Bolondi G, Forceville X, Angstwurm MW, Gurjar M, Biondi R, van Zanten AR, Berger MM. What do we know about micronutrients in critically ill patients? A narrative review. JPEN J Parenter Enteral Nutr 2025; 49:33-58. [PMID: 39555865 PMCID: PMC11717498 DOI: 10.1002/jpen.2700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 09/03/2024] [Accepted: 10/07/2024] [Indexed: 11/19/2024]
Abstract
Micronutrient (MN) status alterations (both depletion and deficiency) are associated with several complications and worse outcomes in critically ill patients. On the other side of the spectrum, improving MN status has been shown to be a potential co-adjuvant therapy. This review aims to collect existing data to better guide research in the critical care setting. This narrative review was conducted by the European Society of Intensive Care Medicine Feeding, Rehabilitation, Endocrinology, and Metabolism MN group. The primary objective was to identify studies focusing on individual MNs in critically ill patients, selecting the MNs that appear to be most relevant and most frequently investigated in the last decade: A, B1, B2, B3, B6, folate, C, D, E, copper, iron, selenium, zinc, and carnitine. Given the limited number of interventional studies for most MNs, observational studies were included. For each selected MN, the review summarizes the main form and functions, special needs and risk factors, optimal treatment strategies, pharmacological dosing, and clinical implications all specific to critically ill patients. A rigorous rebalancing of research strategies and priorities is needed to improve clinical practice. An important finding is that high-dose monotherapy of MNs is not recommended. Basal daily needs must be provided, with higher doses in diseases with known higher needs, and identified deficiencies treated. Finally, the review provides a list of ongoing trials on MNs in critically ill patients and identifies a priority list of future research topics.
Collapse
Affiliation(s)
- Angelique M.E. de Man
- Department of Intensive Care; Amsterdam Cardiovascular Sciences, Amsterdam UMClocation Vrije UniversiteitAmsterdamthe Netherlands
| | - Christian Stoppe
- University Hospital Wuerzburg, Department of Anaesthesiology, Intensive Care, Emergency, and Pain MedicineWuerzburgGermany
| | | | - George Briassoulis
- Postgraduate Program, Emergency and Intensive Care in Children Adolescents and Young Adults, School of MedicineUniversity of CreteHeraklionGreece
| | - Lilanthi S.D.P. Subasinghe
- Head of the Department ‐ Division of Intensive Care, University HospitalGeneral Sir John Kotelawala Defence UniversityColomboSri Lanka
| | - Cristian Cobilinschi
- Department of Anesthesiology and Intensive Care II“Carol Davila” University of Medicine and PharmacyBucharestRomania
- Department of Anesthesiology and Intensive Care I, Clinical Emergency Hospital of BucharestBucharestRomania
| | - Adam M. Deane
- Department of Critical Care, Melbourne Medical SchoolUniversity of MelbourneParkvilleVicAustralia
| | - William Manzanares
- Department of Critical Care, Hospital de Clínicas (University Hospital)Faculty of MedicineUdelaRMontevideoUruguay
| | - Ioana Grințescu
- Department of Anesthesiology and Intensive Care II“Carol Davila” University of Medicine and PharmacyBucharestRomania
- Department of Anesthesiology and Intensive Care I, Clinical Emergency Hospital of BucharestBucharestRomania
| | - Liliana Mirea
- Department of Anesthesiology and Intensive Care II“Carol Davila” University of Medicine and PharmacyBucharestRomania
- Department of Anesthesiology and Intensive Care I, Clinical Emergency Hospital of BucharestBucharestRomania
| | - Ashraf Roshdy
- Critical Care Medicine Department, Faculty of MedicineAlexandria UniversityAlexandriaEgypt
| | - Antonella Cotoia
- Department of Critical CareUniversity Hospital of FoggiaFoggiaItaly
| | - Danielle E. Bear
- Department of Nutritional Sciences, School of Life Course and Population SciencesKing's College LondonLondonUK
- Department of Nutrition and Dietetics and Department of Critical CareGuy's and St Thomas’ NHS Foundation TrustLondonUK
| | - Sabrina Boraso
- General and Neurosurgical Intensive Care Unit, Ospedale dell'AngeloMestre‐VeneziaItaly
| | | | - Kenneth B. Christopher
- Channing Division of Network Medicine, Brigham and Women's HospitalBostonUSA
- Division of Renal Medicine, Brigham and Women's HospitalBostonUSA
| | - Michael P. Casaer
- Department of Cellular and Molecular Medicine, Laboratory of Intensive Care MedicineKU LeuvenLeuvenBelgium
- Intensive Care MedicineUZ LeuvenBelgium
| | - Jan Gunst
- Department of Cellular and Molecular Medicine, Laboratory of Intensive Care MedicineKU LeuvenLeuvenBelgium
- Intensive Care MedicineUZ LeuvenBelgium
| | - Olivier Pantet
- Department of Intensive Care MedicineUniversity Hospital of LausanneLausanneSwitzerland
| | | | - Giuliano Bolondi
- Anesthesia and Intensive Care Unit, Ospedale BufaliniCesena (FC)Italy
| | - Xavier Forceville
- Inserm, CIC 1414 (Centre d′ Investigation Clinique de Rennes)Univ Rennes, CHU RennesRennesF‐35000France
| | | | - Mohan Gurjar
- Department of Critical Care MedicineSanjay Gandhi Post Graduate Institute of Medical SciencesIndia
| | | | - Arthur R.H. van Zanten
- Department of Intensive Care Medicine, Gelderse Vallei Hospital, Ede, the Netherlands; Wageningen University & Research, Division of Human Nutrition and HealthWageningenthe Netherlands
| | - Mette M. Berger
- Faculty of Biology and MedicineLausanne UniversityLausanneSwitzerland
| | | |
Collapse
|
19
|
Li M, Ren X, Lu F, Pang S, Ding L, Wang L, Xie S, Geng L, Xu J, Yang T. IDENTIFYING POTENTIAL KEY FERROPTOSIS-RELATED GENES AND THERAPEUTIC DRUGS IN SEPSIS-INDUCED ARDS BY BIOINFORMATICS AND EXPERIMENTAL VERIFICATION. Shock 2025; 63:141-154. [PMID: 39283066 DOI: 10.1097/shk.0000000000002478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2024]
Abstract
ABSTRACT Background: Acute respiratory distress syndrome (ARDS) is a serious pathological process with high mortality. Ferroptosis is pivotal in sepsis, whose regulatory mechanisms in sepsis-induced ARDS remains unknown. We aimed to determine key ferroptosis-related genes in septic ARDS and investigate therapeutic traditional Chinese medicine. Method: Sepsis-induced ARDS dataset obtained from Gene Expression Omnibus was analyzed to identify ferroptosis-related differentially expressed genes. Enrichment analysis and protein-protein interaction network construction were performed to identify hub genes. Immune cells infiltration was analyzed and competitive endogenous RNA network was constructed. The diagnostic value of hub genes in septic ARDS was analyzed and the occurrence of ferroptosis and the expression of hub genes were detected. Traditional Chinese medicine targeting hub genes was predicted via SymMap database and was verified. Results: Sixteen ferroptosis-related differentially expressed genes were obtained, among which the top four genes ( IL1B , TXN , MAPK3 , HSPB1 ) were selected as hub genes, which may be potential diagnostic markers of septic ARDS. Immunoassay showed that sepsis-induced ARDS and hub genes were closely related to immune cells. The competitive endogenous RNA network showed 26 microRNAs and 38 long noncoding RNA. Ferroptosis occurred and the expressions of IL1B , MAPK3 , and TXN were increased in septic ARDS mice and LPS-challenged human pulmonary alveolar epithelial cells. Sea buckthorn alleviated septic lung injury and affected hub genes expression. Conclusions: Ferroptosis-related genes of IL1B , MAPK 3, and TXN serve as potential diagnostic genes for sepsis-induced ARDS. Sea buckthorn may be therapeutic medication for ARDS. This study provides a new direction for septic ARDS treatment.
Collapse
Affiliation(s)
- Man Li
- Department of Anesthesiology, Tianjin Union Medical Center, Tianjin, China
| | - Xiaojing Ren
- Department of Anesthesiology, Tianjin Baodi Hospital, Tianjin, China
| | - Futai Lu
- Department of Anesthesiology, Tianjin Union Medical Center, Tianjin, China
| | - Shenyue Pang
- Department of Anesthesiology, Tianjin Union Medical Center, Tianjin, China
| | - Ling Ding
- Department of Anesthesiology, Tianjin Union Medical Center, Tianjin, China
| | - Lei Wang
- Department of Anesthesiology, Tianjin Union Medical Center, Tianjin, China
| | - Shuhua Xie
- Department of Anesthesiology, Tianjin Union Medical Center, Tianjin, China
| | - Licheng Geng
- Department of Anesthesiology, Tianjin Union Medical Center, Tianjin, China
| | - Jiangang Xu
- Department of Anesthesiology, Tianjin Union Medical Center, Tianjin, China
| | - Tao Yang
- Department of Anesthesiology, Tianjin Union Medical Center, Tianjin, China
| |
Collapse
|
20
|
Liu F, Wang Q, Ye H, Du Y, Wang M, Guo Y, He S. Identification of STAT3 and MYC as critical ferroptosis-related biomarkers in septic cardiomyopathy: a bioinformatics and experimental study. J Mol Med (Berl) 2025; 103:87-100. [PMID: 39557695 DOI: 10.1007/s00109-024-02502-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: 09/12/2024] [Revised: 10/27/2024] [Accepted: 11/06/2024] [Indexed: 11/20/2024]
Abstract
Ferroptosis is the well-known mechanism of septic cardiomyopathy (SCM). Bioinformatics analysis was employed to identify ferroptosis-related SCM differentially expressed genes (DEG). DEGs' functional enrichment was explored. Weighted gene co-expression network analysis (WGCNA) was employed to form gene clusters. The identified hub genes, signal transducer and activator of transcription 3 (STAT3) and myelocytomatosis (MYC) were further evaluated by generating receiver operator characteristic (ROC) curves and a nomogram prediction model. Additionally, survival rate, cardiac damage markers, and cardiac function and ferroptosis markers were evaluated in septic mouse model. STAT3 and MYC levels were measured in SCM heart tissue via immunohistochemical (IHC) staining, real-time polymerase chain reaction (qPCR) and western blot analysis. Analysis identified 225 DEGs and revealed 22 intersected genes. Of the 7 hub genes, STAT3 and MYC showed enrichment in septic heart tissue and a strong predicative ability based on AUC values. Cardiac damage, iron metabolism, and lipid peroxidation occurred in the SCM model. By experiments, STAT3 and MYC expression was increased in the SCM model. Impairment was reversed with a ferroptosis inhibitor, Fer-1. As conclusion, STAT3 and MYC are related with ferroptosis and may serve as potential SCM predictor indicators. KEY MESSAGES: Septic cardiomyopathy (SCM) often leads to high mortality in septic patients, and the diagnostic criteria still remains unclear. Ferroptosis as the pathogenic mechanism of SCM could help predict its progression and clinical outcomes. STAT3 and MYC are related with ferroptosis and may serve as potential SCM predictor biomarkers.
Collapse
Affiliation(s)
- Fangyu Liu
- Beijing University of Chinese Medicine, Beijing, China
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Qian Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Haoran Ye
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Capital Medical University, Beijing, China
| | - Yuan Du
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Capital Medical University, Beijing, China
| | - Mingjiao Wang
- Beijing University of Chinese Medicine, Beijing, China
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yuhong Guo
- Beijing University of Chinese Medicine, Beijing, China
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Capital Medical University, Beijing, China
| | - Shasha He
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China.
- Beijing Institute of Chinese Medicine, Beijing, China.
| |
Collapse
|
21
|
Hou Q, Dou Z, Zhu L, Li B. Shielding the Gut: Ghrelin and Ferrostatin-1's Protective Role Against Sepsis-Induced Intestinal Ferroptosis. Biomedicines 2024; 13:77. [PMID: 39857660 PMCID: PMC11761253 DOI: 10.3390/biomedicines13010077] [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: 11/29/2024] [Revised: 12/23/2024] [Accepted: 12/28/2024] [Indexed: 01/27/2025] Open
Abstract
Objective: This study investigates the therapeutic efficacy of ghrelin in alleviating sepsis-induced intestinal damage, focusing on its potential to inhibit ferroptosis and protect intestinal barrier integrity. Methods: This study evaluates the therapeutic efficacy of intraperitoneal ghrelin (80 μg/kg) and Ferrostatin-1 (5 mg/kg) using a cecal ligation and puncture (CLP) model in C57BL/6 mice to determine their potential in alleviating sepsis-induced intestinal damage. The investigation focuses on the impacts of ghrelin and Ferrostatin-1 on bacterial load, intestinal morphology, systemic inflammation, oxidative stress, and ferroptosis markers. Our comprehensive methodology encompasses histopathological evaluations, cytokine profiling, oxidative stress assays, and detailed analyses of ferroptosis indicators to thoroughly assess the interventions' efficacy. Results: Treatment with ghrelin significantly reduced bacterial proliferation, mitigated intestinal damage, and decreased systemic inflammation. Comparable outcomes were observed with Fer-1 treatment. Both interventions restored intestinal barrier functions, modulated inflammatory responses, and attenuated oxidative stress, indicating a suppression of the ferroptosis pathway. Conclusion: Ghrelin exhibits a protective role in sepsis-induced intestinal injury, likely through the inhibition of ferroptosis. This mechanism underscores ghrelin's therapeutic potential in sepsis management, suggesting avenues for further clinical exploration.
Collapse
Affiliation(s)
- Qiliang Hou
- Department of Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Department of Critical Care Medicine, The First Clinical Medical School, Lanzhou University, Lanzhou 730000, China
- National Key Laboratory of Critical Care Medicine, Lanzhou 730000, China
| | - Zhimin Dou
- Department of Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Department of Critical Care Medicine, The First Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Lei Zhu
- Department of Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Department of Critical Care Medicine, The First Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Bin Li
- Department of Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Department of Critical Care Medicine, The First Clinical Medical School, Lanzhou University, Lanzhou 730000, China
- National Key Laboratory of Critical Care Medicine, Lanzhou 730000, China
| |
Collapse
|
22
|
Xu F, Xie J, Mou W, Li D, Rui S, Lin L, Hu L, Yang G, Xie P, Tao Y, Yang F, Ma Y. The VDR/FFAR2 axis mitigates sepsis-induced lung injury by suppressing macrophage lipid peroxidation. Int Immunopharmacol 2024; 143:113328. [PMID: 39418731 DOI: 10.1016/j.intimp.2024.113328] [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: 06/09/2024] [Revised: 09/20/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
Sepsis-induced lung injury is a common critical condition in clinical practice, characterized by the accumulation of peroxides and inflammatory damage caused by excessive macrophage activation. Currently, effective treatments for sepsis-induced lung injury are lacking. Short-chain fatty acid receptor FFAR2 serves as an anti-inflammatory biomarker, but its role and mechanism in sepsis-induced lung injury remain unclear. To elucidate the influence and mechanism of FFAR2 on macrophage lipid peroxidation levels in sepsis-induced lung injury, this study conducted bioinformatics analysis and cellular experiments using the THP-1 macrophage cell line. By dual luciferase reporter and chromatin immunoprecipitation-quantitative PCR assays, it is confirmed that the transcription factor VDR upregulates FFAR2 expression in macrophages by binding to the promoter region -1695 ∼ 1525, thereby increasing the expression of iron death negative regulatory molecules and lowering macrophage lipid peroxidation levels. Moreover, both in vitro using THP-1 cells and bone marrow-derived macrophages (BMDMs) and in vivo using an LPS-induced septic mice model experiments revealed that activating the VDR/FFAR2 axis could reduce inflammation-induced macrophage lipid peroxide accumulation and alleviate lung injury in septic mice. This finding highlights the potential of FFAR2 as an immunotherapeutic target for mitigating sepsis-related lung injury.
Collapse
Affiliation(s)
- Fan Xu
- Chongqing Key Laboratory of Emergency Medicine, Chongqing Emergency Medical Center/Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400014, China
| | - Jia Xie
- Chongqing Key Laboratory of Emergency Medicine, Chongqing Emergency Medical Center/Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400014, China
| | - Weijiao Mou
- Chongqing Key Laboratory of Emergency Medicine, Chongqing Emergency Medical Center/Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400014, China
| | - Donglin Li
- Chongqing Key Laboratory of Emergency Medicine, Chongqing Emergency Medical Center/Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400014, China
| | - Shunli Rui
- Chongqing Key Laboratory of Emergency Medicine, Chongqing Emergency Medical Center/Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400014, China
| | - Ling Lin
- Chongqing Key Laboratory of Emergency Medicine, Chongqing Emergency Medical Center/Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400014, China
| | - Li Hu
- Chongqing Key Laboratory of Emergency Medicine, Chongqing Emergency Medical Center/Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400014, China
| | - Guo Yang
- Chongqing Key Laboratory of Emergency Medicine, Chongqing Emergency Medical Center/Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400014, China
| | - Puguang Xie
- Chongqing Key Laboratory of Emergency Medicine, Chongqing Emergency Medical Center/Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400014, China
| | - Yang Tao
- Department of Critical Care Medicine, Chongqing University Central Hospital, Chongqing 400014, China
| | - Fan Yang
- Chongqing Key Laboratory of Emergency Medicine, Chongqing Emergency Medical Center/Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400014, China.
| | - Yu Ma
- Chongqing Key Laboratory of Emergency Medicine, Chongqing Emergency Medical Center/Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400014, China; Department of Critical Care Medicine, Chongqing University Central Hospital, Chongqing 400014, China.
| |
Collapse
|
23
|
Zhang P, Liu W, Wang S, Wang Y, Han H. Ferroptosisand Its Role in the Treatment of Sepsis-Related Organ Injury: Mechanisms and Potential Therapeutic Approaches. Infect Drug Resist 2024; 17:5715-5727. [PMID: 39720615 PMCID: PMC11668052 DOI: 10.2147/idr.s496568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/12/2024] [Indexed: 12/26/2024] Open
Abstract
Sepsis is a complicated clinical disease caused by a defective host response to infection, leading to elevated morbidity and fatality globally. Sepsis patients have a significant risk of life-threatening organ damage, including hearts, brains, lungs, kidneys, and livers. Nevertheless, the molecular pathways driving organ injury in sepsis are not well known. Ferroptosis, a non-apoptotic cell death, occurs due to iron metabolism disturbance and lipid peroxide buildup. Multiple studies indicate that ferroptosis has a significant role in decreasing inflammation and lipid peroxidation during sepsis. Ferroptosis inhibitors and medications, aimed at the most studied ferroptosis process, including Xc-system, Nrf2/GPX4 axis, and NCOA4-FTH1-mediated ferritinophagy, alleviating sepsis effectively. However, few clinical trials demonstrated ferroptosis-targeted drugs's effectiveness in sepsis. Our study examines ferroptosis-targeted medicinal agents and their potential benefits for treating sepsis-associated organ impairment. This review indicates that ferroptosis suppression by pharmaceutical means may be a useful therapy for sepsis-associated organ injury.
Collapse
Affiliation(s)
- Pengyu Zhang
- The Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Wendi Liu
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Shu Wang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Yuan Wang
- Department of Histology and Embryology, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Han Han
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
24
|
Ding L, Zhang R, Du W, Wang Q, Pei D. The role of cGAS-STING signaling pathway in ferroptosis. J Adv Res 2024:S2090-1232(24)00606-4. [PMID: 39710299 DOI: 10.1016/j.jare.2024.12.028] [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: 10/14/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024] Open
Abstract
The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway has been identified as a crucial mechanism in antiviral defense and innate immunity pathway. Ferroptosis, characterized by iron dependence and lipid peroxidation, represents a specialized form of cell death. A burgeoning collection of studies has demonstrated that the cGAS-STING signaling pathway participates in the homeostatic regulation of the organism by modulating ferroptosis-associated enzyme activity or gene expression. Consequently, elucidating the specific roles of the STING signaling pathway and ferroptosis in vivo is vital for targeted disease intervention. This review systematically examines the interactions between the cGAS-STING signaling pathway and ferroptosis, highlighting their influence on disease progression in the contexts of inflammation, injury, and cancerous cell dynamics. Understanding these interactions may provide novel therapeutic strategies. The STING pathway has been implicated in the regulation of various cell death mechanisms, including apoptosis, pyroptosis, necroptosis, autophagy, and ferroptosis. Our focus primarily addresses the role and mechanism of the cGAS-STING signaling pathway and ferroptosis in diseases, limiting discussion of other cell death modalities and precluding a comprehensive overview of the pathway's additional functions.
Collapse
Affiliation(s)
- Lina Ding
- Department of Pathology, Xuzhou Medical University, Xuzhou, China.
| | - Ruicheng Zhang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| | - Wenqi Du
- Department of Human Anatomy, Xuzhou Medical University, Xuzhou, China.
| | - Qingling Wang
- Department of Pathology, Xuzhou Medical University, Xuzhou, China.
| | - Dongsheng Pei
- Department of Pathology, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
25
|
Guo Y, Chen H, Sun J, Zhang J, Yin Y. Maresin1 Inhibits Ferroptosis via the Nrf2/SLC7A11/GPX4 Pathway to Protect Against Sepsis-Induced Acute Liver Injury. J Inflamm Res 2024; 17:11041-11053. [PMID: 39691306 PMCID: PMC11651138 DOI: 10.2147/jir.s498775] [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: 10/07/2024] [Accepted: 11/29/2024] [Indexed: 12/19/2024] Open
Abstract
Purpose Maresin 1 (MaR1) is a specialized pro-resolving mediator with anti-inflammatory properties that promotes tissue repair. This study aims to investigate the molecular involvement of MaR1 in protecting against sepsis-induced acute liver injury (SI-ALI). Methods In vivo, a murine SI-ALI model was established using the cecal ligation and puncture (CLP) paradigm, providing a system in which the mechanistic functions of MaR1 could be tested. These analyses were supplemented through in vitro assays in which Alpha mouse liver 12 (AML12) hepatocytes and RAW264.7 macrophages were co-cultured in a Transwell system, with lipopolysaccharide (LPS) stimulation being used to establish a sepsis model. These cells were treated with MaR1 and/or nuclear factor erythroid 2-related factor 2 (Nrf2)inhibitor, while lentiviral transduction was used to knock down Nrf2 within AML12 cells. Hepatic pathological damage was assessed through hematoxylin and eosin staining. Lipid peroxidation-related analyses were conducted through the use of thiobarbituric acid, ferrous ions, glutathione, and appropriate fluorescent probes for reactive oxygen species detection. Liver enzymes and inflammatory mediators were quantified using appropriate Enzyme-Linked Immunosorbent Assays (ELISAs). Protein concentrations were evaluated via Western blot analysis. Results The presence of ferroptosis in SI-ALI. MaR1 was found to proficiently suppress ferroptosis in SI-ALI. Mechanistically, MaR1 enhanced Nrf2 expression in AML12 hepatocytes, while the Nrf2 inhibitor ML-385 or Nrf2 siRNA mitigated MaR1's regulatory influence on ferroptosis. Meanwhile, the expressions of the downstream genes solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase 4 (GPX4) diminished, suggesting that MaR1 has a protective function via activating the Nrf2/SLC7A11/GPX4 pathway to mitigate ferroptosis in septic liver injury. Conclusion The results indicate that MaR1 mitigates SI-ALI via stimulating the Nrf2/SLC7A11/GPX4 pathway to suppress ferroptosis. Moreover, it offers significant potential as a new agent for the prevention of SI-ALI.
Collapse
Affiliation(s)
- Yongjing Guo
- Department of Emergency and Critical Care, The Second Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| | - Huimin Chen
- Department of Pediatric Surgery, The Second Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| | - Jian Sun
- Department of Emergency and Critical Care, The Second Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| | - Jingxiao Zhang
- Department of Emergency and Critical Care, The Second Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| | - Yongjie Yin
- Department of Emergency and Critical Care, The Second Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| |
Collapse
|
26
|
Wang H, Su Z, Qian Y, Shi B, Li H, An W, Xiao Y, Qiu C, Guo Z, Zhong J, Wu X, Chen J, Wang Y, Zeng W, Zhan L, Wang J. Pentraxin-3 modulates hepatocyte ferroptosis and the innate immune response in LPS-induced liver injury. MOLECULAR BIOMEDICINE 2024; 5:68. [PMID: 39666228 PMCID: PMC11638432 DOI: 10.1186/s43556-024-00227-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024] Open
Abstract
The liver plays a crucial role in the immune response during endotoxemia and is one of the critical targets for sepsis-related injuries. As a secretory factor involved in inflammation, pentraxin-3 (PTX3) has been demonstrated to regulate hepatic homeostasis; however, the relationship between PTX3 and cell crosstalk between immune cells and hepatocytes in the liver remains incompletely understood. In this study, we revealed that, compared with WT mice, Ptx3-/- mice with lipopolysaccharide (LPS)-induced endotoxemia exhibited alleviated liver damage, with reduced serum alanine transaminase and aspartate transaminase levels and an improved survival rate. Mechanistically, RNA-Seq and western blot results revealed that Ptx3 knockdown in hepatocytes increased the expression of Tfrc and Ccl20; consequently, Ptx3 deficiency regulated LPS-induced hepatocyte ferroptosis via increased mitochondrial reactive oxygen species and Fe2+ and recruited more macrophages by CCL20/CCR6 axis to be involved in inflammation and the clearance of harmful substances. Moreover, western blot and immunofluorescence staining confirmed that the NF-κB signaling pathway was upregulated upon LPS treatment in Ptx3-knockdown macrophages, promoting phagocytosis and polarization toward M1 macrophages. Collectively, our findings show that the absence of Ptx3 can ameliorate sepsis-induced liver injury by regulating hepatocyte ferroptosis and promote the recruitment and polarization of M1 macrophages. These findings offer a key basis for the development of effective treatments for acute infections.
Collapse
Affiliation(s)
- Huitong Wang
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Zhaojie Su
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yunyun Qian
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Baojie Shi
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Hao Li
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Wenbin An
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yi Xiao
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Cheng Qiu
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Zhixiang Guo
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jianfa Zhong
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xia Wu
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jiajia Chen
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Ying Wang
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Wei Zeng
- Department of Gastroenterology, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Linghui Zhan
- Department of Critical Care Medicine, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, 361004, China.
| | - Jie Wang
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China.
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China.
| |
Collapse
|
27
|
Islam MM, Watanabe E, Salma U, Ozaki M, Irahara T, Tanabe S, Katsuki R, Oishi D, Takeyama N. Immunoadjuvant therapy in the regulation of cell death in sepsis: recent advances and future directions. Front Immunol 2024; 15:1493214. [PMID: 39720718 PMCID: PMC11666431 DOI: 10.3389/fimmu.2024.1493214] [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: 09/08/2024] [Accepted: 11/18/2024] [Indexed: 12/26/2024] Open
Abstract
Sepsis is characterized by a concomitant early pro-inflammatory response by immune cells to an infection, and an opposing anti-inflammatory response that results in protracted immunosuppression. The primary pathological event in sepsis is widespread programmed cell death, or cellular self-sacrifice, of innate and adaptive immune cells, leading to profound immunological suppression. This severe immune dysfunction hampers effective primary pathogen clearance, thereby increasing the risk of secondary opportunistic infections, latent viral reactivation, multiple organ dysfunction, and elevated mortality. The types of cell death include apoptosis (type I programmed cell death), autophagy (type II programmed cell death), NETosis (a program for formation of neutrophil extracellular traps (NETs)) and other programmed cell deaths like pyroptosis, ferroptosis, necroptosis, each contributing to immunosuppression in distinct ways during the later phases of sepsis. Extensive apoptosis of lymphocytes, such as CD4+, CD8+ T cells, and B cells, is strongly associated with immunosuppression. Apoptosis of dendritic cells further compromises T and B cell survival and can induce T cell anergy or promote regulatory Treg cell proliferation. Moreover, delayed apoptosis and impaired neutrophil function contribute to nosocomial infections and immune dysfunction in sepsis. Interestingly, aberrant NETosis and the subsequent depletion of mature neutrophils also trigger immunosuppression, and neutrophil pyroptosis can positively regulate NETosis. The interaction between programmed cell death 1 (PD-1) or programmed cell death 1 ligand (PD-L1) plays a key role in T cell modulation and neutrophil apoptosis in sepsis. The dendritic cell growth factor, Fms-like tyrosine kinase (FLTEL), increases DC numbers, enhances CD 28 expression, attenuates PD-L1, and improves survival in sepsis. Recently, immunoadjuvant therapies have attracted attention for their potential to restore host physiological immunity and homeostasis in patients with sepsis. This review focuses on several potential immunotherapeutic agents designed to bolster suppressed innate and adaptive immune responses in the management of sepsis.
Collapse
Affiliation(s)
- Md. Monirul Islam
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
- Department of Biochemistry and Biotechnology, University of Science and Technology Chittagong (USTC), Chattogram, Bangladesh
| | - Eizo Watanabe
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Umme Salma
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Masayuki Ozaki
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Takayuki Irahara
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Subaru Tanabe
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Ryusuke Katsuki
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Dai Oishi
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Naoshi Takeyama
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| |
Collapse
|
28
|
Zhou X, Wang H, Yan B, Nie X, Chen Q, Yang X, Lei M, Guo X, Ouyang C, Ren Z. Ferroptosis in Cardiovascular Diseases and Ferroptosis-Related Intervention Approaches. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07642-5. [PMID: 39641901 DOI: 10.1007/s10557-024-07642-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/08/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE Cardiovascular diseases (CVDs) are major public health problems that threaten the lives and health of individuals. The article has reviewed recent progresses about ferroptosis and ferroptosis-related intervention approaches for the treatment of CVDs and provided more references and strategies for targeting ferroptosis to prevent and treat CVDs. METHODS A comprehensive review was conducted using the literature researches. RESULTS AND DISCUSSION Many ferroptosis-targeted compounds and ferroptosis-related genes may be prospective targets for treating CVDs and our review provides a solid foundation for further studies about the detailed pathological mechanisms of CVDs. CONCLUSION There are challenges and limitations about the translation of ferroptosis-targeted potential therapies from experimental research to clinical practice. It warrants further exploration to pursure safer and more effective ferroptosis-targeted thereapeutic approaches for CVDs.
Collapse
Affiliation(s)
- Xianpeng Zhou
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Hao Wang
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Biao Yan
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Xinwen Nie
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Qingjie Chen
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Xiaosong Yang
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Min Lei
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Xiying Guo
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Changhan Ouyang
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Zhanhong Ren
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China.
| |
Collapse
|
29
|
Liu Y, Bao D, She H, Zhang Z, Shao S, Wu Z, Wu Y, Li Q, Wang L, Li T, Liu L. Role of Hippo/ACSL4 axis in ferroptosis-induced pericyte loss and vascular dysfunction in sepsis. Redox Biol 2024; 78:103353. [PMID: 39566164 PMCID: PMC11617880 DOI: 10.1016/j.redox.2024.103353] [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: 06/24/2024] [Revised: 08/27/2024] [Accepted: 09/11/2024] [Indexed: 11/22/2024] Open
Abstract
Sepsis is a critical condition characterized by a systemic inflammatory response to infection, often leading to severe vascular dysfunction and high mortality. One of the hallmarks of vascular dysfunction in sepsis is increased vascular permeability and the loss of pericytes, which are essential for maintaining vascular integrity. Despite the significance of pericyte loss in sepsis, the primary type of cell death responsible and the underlying molecular mechanisms remain incompletely understood. This study aims to elucidate these mechanisms by focusing on ferroptosis, a form of programmed cell death, and its regulation through the Hippo/ACSL4 axis. Our research confirmed significant pericyte loss in patients with sepsis. Through advanced single-cell analysis and proteomics, ferroptosis was identified as a key differentiating cell death type between sepsis and sham samples. Further metabolomics analysis revealed that Acyl-CoA Synthetase Long-Chain Family Member 4 (ACSL4) plays a pivotal role in the ferroptosis of pericytes during sepsis. In vitro experiments demonstrated that downregulation of ACSL4 effectively reduced lipopolysaccharide (LPS)-induced lipid peroxidation, restored pericyte viability, and improved endothelial permeability. In vivo studies with pericyte-specific ACSL4 knockout mice showed a marked decrease in pericyte loss and enhanced vascular barrier function following sepsis induction. To translate these findings into potential therapeutic strategies, we developed pericyte-targeting liposomes encapsulating ACSL4 shRNA adenovirus. These liposomes successfully restored pulmonary vascular barrier function and significantly reduced pericyte loss in septic conditions. The results of this study underscore the crucial role of ACSL4 in mediating ferroptosis in pericytes and highlight the therapeutic potential of targeting ACSL4 to mitigate vascular dysfunction in sepsis.
Collapse
Affiliation(s)
- Yiyan Liu
- Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Daiqin Bao
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Han She
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Zisen Zhang
- Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Shifeng Shao
- Department of Critical Care Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Zhengbin Wu
- Department of Critical Care Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yue Wu
- Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Qinghui Li
- Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Li Wang
- Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Tao Li
- Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Liangming Liu
- Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
30
|
Bertozzi G, Ferrara M, Calvano M, Pascale N, Di Fazio A. Oxidative/Nitrosative Stress and Brain Involvement in Sepsis: A Relationship Supported by Immunohistochemistry. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1949. [PMID: 39768830 PMCID: PMC11678000 DOI: 10.3390/medicina60121949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025]
Abstract
Background and Objectives: A large amount of recent evidence suggests that cellular inability to consume oxygen could play a notable part in promoting sepsis as a consequence of mitochondrial dysfunction and oxidative stress. The latter could, in fact, represent a fundamental stage in the evolution of the "natural history" of sepsis. Following a study previously conducted by the same working group on heart samples, the present research project aims to evaluate, through an immunohistochemical study, the existence and/or extent of oxidative stress in the brains of subjects who died due to sepsis and define, after reviewing the literature, its contribution to the septic process to support the use of medications aimed at correcting redox anomalies in the management of septic patients. Materials and Methods: 10 cases of subjects who died in healthcare facilities with ante-mortem clinical-laboratory signs that allowed the diagnosis of septic shock were selected as case studies, and 1 case of a subject who died immediately following a road traffic accident was used as a negative control. Samples of the cerebral cortex were then taken, fixed in formalin, and subjected to sections on which an immunohistochemical study was performed using anti-NOX-2, NT, iNOS, and 8-OHdG antibodies. Results: The results emerging from the present study demonstrate that despite a variable expressivity for the NT, iNOS, and NOX2 markers, the brain samples demonstrated univocal and high positivity for the 8-OHdG marker. Conclusions: This would allow us to hypothesize how, regardless of the mechanism of production of ROS and NOS (iNOS or NOX2 mediated) and the pathophysiological mechanisms that are triggered during sepsis, oxidative damage to DNA represents the event to which this whole process leads and, in fact, in the literature, is directly correlated to sepsis-dependent mortality. Neurons, conversely, appear to be more sensitive to oxidative stress because of a low number of protective or scavenger molecules (catalase, glutathione peroxidase, GSH, or vitamin E). Therefore, despite reduced production, the manifestation of the damage remains high. This evidence, together with that of the previous study, can only support the introduction of substances with an antioxidant function in the guidelines for the treatment of sepsis.
Collapse
Affiliation(s)
- Giuseppe Bertozzi
- SIC Medicina Legale, Via Potito Petrone, 85100 Potenza, Italy; (M.F.); (M.C.); (N.P.); (A.D.F.)
| | - Michela Ferrara
- SIC Medicina Legale, Via Potito Petrone, 85100 Potenza, Italy; (M.F.); (M.C.); (N.P.); (A.D.F.)
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00185 Rome, Italy
| | - Mariagrazia Calvano
- SIC Medicina Legale, Via Potito Petrone, 85100 Potenza, Italy; (M.F.); (M.C.); (N.P.); (A.D.F.)
| | - Natascha Pascale
- SIC Medicina Legale, Via Potito Petrone, 85100 Potenza, Italy; (M.F.); (M.C.); (N.P.); (A.D.F.)
| | - Aldo Di Fazio
- SIC Medicina Legale, Via Potito Petrone, 85100 Potenza, Italy; (M.F.); (M.C.); (N.P.); (A.D.F.)
| |
Collapse
|
31
|
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
|
32
|
Liu AB, Tan B, Yang P, Tian N, Li JK, Wang SC, Yang LS, Ma L, Zhang JF. The role of inflammatory response and metabolic reprogramming in sepsis-associated acute kidney injury: mechanistic insights and therapeutic potential. Front Immunol 2024; 15:1487576. [PMID: 39544947 PMCID: PMC11560457 DOI: 10.3389/fimmu.2024.1487576] [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: 08/28/2024] [Accepted: 10/11/2024] [Indexed: 11/17/2024] Open
Abstract
Sepsis represents a severe condition characterized by organ dysfunction resulting from a dysregulated host response to infection. Among the organs affected, the kidneys are particularly vulnerable, with significant functional impairment that markedly elevates mortality rates. Previous researches have highlighted that both inflammatory response dysregulation and metabolic reprogramming are crucial in the onset and progression of sepsis associated acute kidney injury (SA-AKI), making these processes potential targets for innovative therapies. This study aims to elucidate the pathophysiological mechanisms of renal injury in sepsis by perspective of inflammatory response dysregulation, with particular emphasis on pyroptosis, necroptosis, autophagy, and ferroptosis. Furthermore, it will incorporate insights into metabolic reprogramming to provide a detailed analysis of the mechanisms driving SA-AKI and explore potential targeted therapeutic strategies, providing solid theoretical framework for the development of targeted therapies for SA-AKI.
Collapse
Affiliation(s)
- An-Bu Liu
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Bin Tan
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Ping Yang
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Na Tian
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jin-Kui Li
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Si-Cong Wang
- Department of Emergency Medical, Yanchi County People’s Hospital, Wuzhong, Ningxia, China
| | - Li-Shan Yang
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Lei Ma
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jun-Fei Zhang
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
33
|
Yu Z, Qian YY. Aspirin use is associated with the reduced mortality risk in chronic obstructive pulmonary disease with sepsis: a retrospective study using the MIMIC-IV database. J Thorac Dis 2024; 16:6688-6698. [PMID: 39552902 PMCID: PMC11565322 DOI: 10.21037/jtd-24-952] [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: 06/12/2024] [Accepted: 08/23/2024] [Indexed: 11/19/2024]
Abstract
Background Sepsis has the characteristics of high morbidity and high mortality in intensive care unit (ICU) patients. Chronic obstructive pulmonary disease (COPD) is an important cause of death. Studies have shown the value of aspirin on COPD and sepsis, separately, but its role in the combined COPD and sepsis patients is unclear. This study aimed to analyze the association of aspirin use after ICU admission with the mortality risk in COPD patients with sepsis. Methods We conducted a retrospective study using the Medical Information Mart for Intensive Care (MIMIC)-IV database, enrolling 2,964 COPD patients with sepsis admitted to ICU. They were divided into aspirin users (n=1,642) and non-users (n=1,322). We evaluated the association of aspirin use with in-hospital and 28-day mortality using logistic regression, Kaplan-Meier survival analysis, and Cox proportional-hazards models. The role of aspirin dose and the association of aspirin use with 90-day and 1-year mortality were also assessed. Results Aspirin use was associated with lower in-hospital death (13.642% vs. 23.676%) and 28-day mortality (17.296% vs. 30.257%) (P<0.001). Adjusted models confirmed reduced mortality odds ratio (OR) with aspirin use: OR for in-hospital mortality was 0.574 [95% confidence interval (CI): 0.456, 0.721] and 28-day mortality was 0.539 (95% CI: 0.437, 0.665) in model 3. Survival analyses showed higher survival probabilities for aspirin users. Subgroup analyses supported consistent aspirin benefits across various clinical parameters. Additionally, aspirin users had lower 90-day (21.498% vs. 34.191%) and 1-year mortality (27.649% vs. 41.982%) (P<0.001). Conclusions Aspirin use is significantly related to in-hospital and 28-day mortality risk in COPD patients with sepsis. This highlighted the clinical relevance of aspirin in COPD patients with sepsis.
Collapse
Affiliation(s)
- Zhao Yu
- Emergency Department, Haining People’s Hospital, Haining, China
| | - Yuan-Yuan Qian
- Department of Respiratory and Critical Care, Haining People’s Hospital, Haining, China
| |
Collapse
|
34
|
Zhao Y, Zhou Z, Cui X, Yu Y, Yan P, Zhao W. Enhancing insight into ferroptosis mechanisms in sepsis: A genomic and pharmacological approach integrating single-cell sequencing and Mendelian randomization. Int Immunopharmacol 2024; 140:112910. [PMID: 39121604 DOI: 10.1016/j.intimp.2024.112910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
This research investigated the intricate relationship between ferroptosis and sepsis by utilizing advanced genomic and pharmacological methodologies. Specifically, we obtained expression quantitative trait loci (eQTLs) for 435 genes associated with ferroptosis from the eQTLGen Consortium and detected notable cis-eQTLs for 281 of these genes. Next, we conducted a detailed analysis to assess the impact of these eQTLs on susceptibility to sepsis using Mendelian randomization (MR) with data from a cohort of 10,154 sepsis patients and 452,764 controls sourced from the UK Biobank. MR analysis revealed 16 ferroptosis-related genes that exhibited significant associations with sepsis outcomes. To bolster the robustness of these findings, sensitivity analyses were performed to assess pleiotropy and heterogeneity, thus confirming the reliability of the causal inferences. Furthermore, single-cell RNA sequencing data from sepsis patients offered a detailed examination of gene expression profiles, demonstrating varying levels of ferroptosis marker expression across different cell types. Pathway enrichment analysis utilizing gene set enrichment analysis (GSEA) further revealed the key biological pathways involved in the progression of sepsis. Additionally, the use of computational molecular docking facilitated the prediction of interactions between identified genes and potential therapeutic compounds, highlighting novel drug targets. In conclusion, our integrated approach combining genomics and pharmacology offers valuable insights into the involvement of ferroptosis in sepsis, laying the groundwork for potential therapeutic strategies targeting this cell death pathway to enhance sepsis management.
Collapse
Affiliation(s)
- Yuanqi Zhao
- Department of Clinical Laboratory, School of Clinical Medicine, Dali University, Dali, China
| | - Zijian Zhou
- Department of Clinical Laboratory, School of Clinical Medicine, Dali University, Dali, China
| | - Xiuyu Cui
- Department of Clinical Laboratory, School of Clinical Medicine, Dali University, Dali, China
| | - Yiwei Yu
- Department of Clinical Laboratory, School of Clinical Medicine, Dali University, Dali, China
| | - Ping Yan
- Department of Gastroenterology, First Affiliated Hospital of Dali University, Dali, China.
| | - Weidong Zhao
- Department of Clinical Laboratory, School of Clinical Medicine, Dali University, Dali, China; Department of Clinical Laboratory, Second Infectious Disease Hospital of Yunnan Province, Dali, China; Immunology Discipline Team, School of Basic Medicine, Dali University, Dali, China.
| |
Collapse
|
35
|
Li X, Cui M, Xu L, Guo Q. Low miR-936-mediated upregulation of Pim-3 drives sorafenib resistance in liver cancer through ferroptosis inhibition by activating the ANKRD18A/Src/NRF2 pathway. Front Oncol 2024; 14:1483660. [PMID: 39507762 PMCID: PMC11540556 DOI: 10.3389/fonc.2024.1483660] [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: 08/20/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Objective Sorafenib, a multikinase inhibitor, is currently the standard treatment for advanced liver cancer. However, its application has become limited by the development of drug resistance. We intended to explore the mechanisms underlying the development of sorafenib resistance, therefore identifying an effective strategy to overcome sorafenib resistance remain challenges. Methods Here, the follow-up of liver cancer patients undergoing sorafenib therapy, as well as animal tumor challenge and treatment were performed. The sorafenib-resistant liver cancer cell lines Huh7/SOR and HepG2/SOR were also established. miRNA and mRNA microarray analyses, TargetScan prediction, dual luciferase reporter assay, RNA pull-down assay, co-mmunoprecipitation (Co-IP) and pull-down assays, a transcription factor-specific NRF2 assay, an iron detection assay, a lipid peroxidation quantification assay, a ROS measurement assay, and GSH/GSSG and GSH-px standard quantitative assays were used. Results We showed that upregulation of the provirus-integrating site for Moloney murine leukemia virus 3 (Pim-3) predicted poor response and unsatisfactory prognosis in sorafenib-treated liver cancer patients. Similarly, Pim-3 expression was positively associated with sorafenib resistance in liver cancer cells. Furthermore, microRNA-936 (miR-936) targeted the 3'-noncoding region (3'-UTR) of Pim-3 but exhibited lower expression in sorafenib-resistant liver cancer cells than in their parental cells. The high expression of Pim-3 mediated by miR-936 insufficiency activated the ANKRD18A/Src/NRF2 pathway which rearranged the expression of the indicated markers involved in iron distribution and lipid peroxidation homeostasis. MiR-936 overexpression and GV102-Pim-3-shRNA significantly attenuated the activity of the ANKRD18A/Src/NRF2 pathway to decrease the expression of Ankyrin repeat domain-containing protein 18A (ANKRD18A), Src, and Nuclear factor (erythroid-derived 2)-like 2 (NRF2), especially decreasing NRF2 nuclear retention and transcriptional activity. The transcriptional activity of NRF2 prompted cell ferroptosis because the transfection of miR-936 mimics, GV102-Pim-3-shRNA and GV102-NRF2-shRNA plasmid increased the expression of transferrin receptor 1 (TFR1) and divalent metal transporter 1 (DMT1) but decreased the expression of solute carrier family 7 member 11 (SLC7A11), glutathione peroxidase 4 (GPX4), quinone oxidoreductase 1 (NQO1), and heme oxygenase-1 (HO-1), thus facilitating the accumulation of intracellular Fe2+, lipid peroxides, and reactive oxygen species (ROS) but reducing the glutathione (GSH) level. Moreover, the elevated expression of Pim-3, resulting from the absence of miR-936 enhances sorafenib resistance in liver cancer by inhibiting cell ferroptosis. Conclusion Pim-3 can be regarded as a target in the treatment of sorafenib-resistant liver cancer.
Collapse
Affiliation(s)
| | | | | | - Qie Guo
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
36
|
Wang Y, Han Q, Liu L, Wang S, Li Y, Qian Z, Jiang Y, Yu Y. Natural hydrogen gas and engineered microalgae prevent acute lung injury in sepsis. Mater Today Bio 2024; 28:101247. [PMID: 39328786 PMCID: PMC11426111 DOI: 10.1016/j.mtbio.2024.101247] [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: 05/24/2024] [Revised: 08/25/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Background Hydrogen gas and microalgae both exist in the natural environment. We aimed to integrate hydrogen gas and biology nano microalgae together to expand the treatment options in sepsis. Methods Phosphoproteomics, metabolomics and proteomics data were obtained from mice undergoing cecum ligation and puncture (CLP) and inhalation of hydrogen gas. All omics analysis procedure were accordance with standards. Multi R packages were used in single cell and spatial transcriptomics analysis to identify primary cells expressing targeted genes, and the genes' co-expression relationships in sepsis related lung landscape. Then, network pharmacology method was used to identify candidate drugs. We used hydrophobic-force-driving self-assembly method to construct dihydroquercetin (DQ) nanoparticle. To cooperate with molecular hydrogen, ammonia borane (B) was added to DQ surface. Then, Chlorella vulgaris (C) was used as biological carrier to improve self-assembly nanoparticle. Vivo and vitro experiments were both conducted to evaluate anti-inflammation, anti-ferroptosis, anti-infection and organ protection capability. Results As a result, we identified Esam and Zo-1 were target phosphorylation proteins for molecular hydrogen treatment in lung. Ferroptosis and glutathione metabolism were two target pathways. Chlorella vulgaris improved the dispersion of DQB and reconstructed morphological features of DQB, formed DQB@C nano-system (size = 307.3 nm, zeta potential = -22mv), with well infection-responsive hydrogen release capability and biosafety. In addition, DQB@C was able to decrease oxidative stress and inflammation factors accumulation in lung cells. Through increasing expression level of Slc7a11/xCT and decreasing Cox2 level to participate with the regulation of ferroptosis. Also, DQB@C played lung and multi organ protection and anti-inflammation roles on CLP mice. Conclusion Our research proposed DQB@C as a novel biology nano-system with enormous potential on treatment for sepsis related acute lung injury to solve the limitation of hydrogen gas utilization in clinics.
Collapse
Affiliation(s)
- Yuanlin Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, 300052, Tianjin, China
- The Graduate School, Tianjin Medical University, 300070, Tianjin, China
| | - Qingqing Han
- The Graduate School, Tianjin Medical University, 300070, Tianjin, China
| | - Lingling Liu
- Department of Anesthesiology, Tianjin Medical University General Hospital, 300052, Tianjin, China
- Department of Anesthesiology, Tianjin Huanhu Hospital, Tianjin, 300350, China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Shuai Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, 300052, Tianjin, China
- The Graduate School, Tianjin Medical University, 300070, Tianjin, China
| | - Yongfa Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, 300052, Tianjin, China
- The Graduate School, Tianjin Medical University, 300070, Tianjin, China
| | - Zhanying Qian
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Yi Jiang
- Department of Anesthesiology, Tianjin Medical University General Hospital, 300052, Tianjin, China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, 300052, Tianjin, China
| |
Collapse
|
37
|
Xu Y, Qu X, Liang M, Huang D, Jin M, Sun L, Chen X, Liu F, Qiu Z. Focus on the role of calcium signaling in ferroptosis: a potential therapeutic strategy for sepsis-induced acute lung injury. Front Med (Lausanne) 2024; 11:1457882. [PMID: 39355841 PMCID: PMC11442327 DOI: 10.3389/fmed.2024.1457882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/05/2024] [Indexed: 10/03/2024] Open
Abstract
By engaging in redox processes, ferroptosis plays a crucial role in sepsis-induced acute lung injury (ALI). Although iron stimulates calcium signaling through the stimulation of redox-sensitive calcium pathways, the function of calcium signals in the physiological process of ferroptosis in septic ALI remains unidentified. Iron homeostasis disequilibrium in ferroptosis is frequently accompanied by aberrant calcium signaling. Intracellular calcium overflow can be a symptom of dysregulation of the cellular redox state, which is characterized by iron overload during the early phase of ferroptosis. This can lead to disruptions in calcium homeostasis and calcium signaling. The mechanisms controlling iron homeostasis and ferroptosis are reviewed here, along with their significance in sepsis-induced acute lung injury, and the potential role of calcium signaling in these processes is clarified. We propose that the development of septic acute lung injury is a combined process involving the bidirectional interaction between iron homeostasis and calcium signaling. Our goal is to raise awareness about the pathophysiology of sepsis-induced acute lung injury and investigate the relationship between these mechanisms and ferroptosis. We also aimed to develop calcium-antagonistic therapies that target ferroptosis in septic ALI and improve the quality of survival for patients suffering from acute lung injury.
Collapse
Affiliation(s)
- Yifei Xu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xintian Qu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Minghao Liang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Di Huang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Minyan Jin
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lili Sun
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xianhai Chen
- Department of Respiratory, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fen Liu
- Department of Respiratory, Shandong Institute of Respiratory Diseases, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Zhanjun Qiu
- Department of Respiratory, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
38
|
Ying-Hao P, Yu-Shan Y, Song-Yi C, Hua J, Peng Y, Xiao-Hu C. Time of day-dependent alterations of ferroptosis in LPS-induced myocardial injury via Bmal-1/AKT/ Nrf2 in rat and H9c2 cell. Heliyon 2024; 10:e37088. [PMID: 39296207 PMCID: PMC11407985 DOI: 10.1016/j.heliyon.2024.e37088] [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: 12/02/2023] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/21/2024] Open
Abstract
Background One of the most prevalent causes of death in sepsis is sepsis-induced cardiomyopathy (SICM). Circadian disruption is involved in the progress of sepsis. However, the molecular mechanism remains unclear. Methods Here, we built LPS-induced SICM in-vivo and in-vitro models. LPS was administrated at the particular Zeitgeber times (ZT), ZT4-ZT10-ZT16-ZT22 and ZT10-ZT22 in vivo and vitro experiments, respectively. Results In vivo experiment, injection of LPS at ZT10 induced higher infiltration of inflammatory cells and content of intracellular Fe2+, and lower level of Glutathione peroxidase 4 (GPX4) and cardiac function than other ZTs (P < 0.05), which indicated that myocardial ferroptosis in septic rat presented a time of day-dependent manner. Bmal-1 protein and mRNA levels of injection of LPS at ZT10 were lower than those at other three ZTs (P < 0.05). The ratios of pAKT/AKT at ZT4 and ZT10 LPS injection were lower than those at ZT16 and ZT22 (P < 0.05). Nrf2 protein levels at ZT10 LPS injection were lower than those at other three ZTs (P < 0.05). These results indicated that the circadian of Bmal-1 and its downstream AKT/Nrf2 pathway in rat heart were inhibited under SICM condition. Consistent with in-vivo experiment, we found LPS could significantly reduce the expressions of Bmal-1 protein and mRNA in H9c2 cell. Up-regulation of Bmal-1 could reduce the cell death, oxidative stress, ferroptosis and activation of AKT/Nrf2 pathway at both ZT10 and ZT22 LPS administration. Conversely, its down-regulation presented opposite effects. AKT siRNA could weaken the effect of Bmal-1 pcDNA. Conclusion Ferroptosis presented the time of day-dependent manners via Bmal-1/AKT/Nrf2 in vivo and vitro models of SICM.
Collapse
Affiliation(s)
- Pei Ying-Hao
- Department of Intensive Care Unit, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province, Nanjing, China
| | - Yang Yu-Shan
- Department of Cardiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province, Nanjing, China
- Department of Cardiology, the People's Hospital of Qingyang City, Gansu Province, China
| | - Cheng Song-Yi
- Department of Cardiology, Nanjing Hospital of Chinese Medicine affiliated to Nanjing university of Chinese medicine, Jiangsu Province, Nanjing, China
| | - Jiang Hua
- Department of Intensive Care Unit, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province, Nanjing, China
| | - Yu Peng
- Department of Cardiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province, Nanjing, China
| | - Chen Xiao-Hu
- Department of Cardiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province, Nanjing, China
| |
Collapse
|
39
|
Yang Z, Gao Y, Zhao L, Lv X, Du Y. Molecular mechanisms of Sepsis attacking the immune system and solid organs. Front Med (Lausanne) 2024; 11:1429370. [PMID: 39267971 PMCID: PMC11390691 DOI: 10.3389/fmed.2024.1429370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024] Open
Abstract
Remarkable progress has been achieved in sepsis treatment in recent times, the mortality rate of sepsis has experienced a gradual decline as a result of the prompt administration of antibiotics, fluid resuscitation, and the implementation of various therapies aimed at supporting multiple organ functions. However, there is still significant mortality and room for improvement. The mortality rate for septic patients, 22.5%, is still unacceptably high, accounting for 19.7% of all global deaths. Therefore, it is crucial to thoroughly comprehend the pathogenesis of sepsis in order to enhance clinical diagnosis and treatment methods. Here, we summarized classic mechanisms of sepsis progression, activation of signal pathways, mitochondrial quality control, imbalance of pro-and anti- inflammation response, diseminated intravascular coagulation (DIC), cell death, presented the latest research findings for each mechanism and identify potential therapeutic targets within each mechanism.
Collapse
Affiliation(s)
- Zhaoyun Yang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Yan Gao
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biomedical Sciences, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Lijing Zhao
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Xuejiao Lv
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Yanwei Du
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| |
Collapse
|
40
|
Liu Z, Ting Y, Li M, Li Y, Tan Y, Long Y. From immune dysregulation to organ dysfunction: understanding the enigma of Sepsis. Front Microbiol 2024; 15:1415274. [PMID: 39252831 PMCID: PMC11381394 DOI: 10.3389/fmicb.2024.1415274] [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: 04/10/2024] [Accepted: 08/05/2024] [Indexed: 09/11/2024] Open
Abstract
Sepsis is a syndrome precipitated by immune dysregulation in response to infection, and represents a pivotal factor in global mortality attributed to diseases. The recent consensus delineates sepsis as a perilous state of organ dysfunction arising from the host's maladaptive reaction to infection. It masks the complexity and breadth of the immune mechanisms involved in sepsis, which is characterized by simultaneous hyperinflammation and immunosuppression. Sepsis is highly correlated with the dysregulation of immune response, which is mainly mediated by various immune cells and their interactions. This syndrome can lead to a plethora of complications, encompassing systemic inflammatory response, metabolic disturbances, infectious shock, MODS, and DIC. Furthermore, more research studies have been conducted on sepsis in the past few years. The pathological characteristics of sepsis have been improved or treated by targeting signaling pathways like NF-B, JAK-STAT, PI3K-Akt, and p38-MAPK. Combined drug therapy is better than single drug therapy for sepsis. This article will review the latest progress in the pathogenesis and treatment of sepsis.
Collapse
Affiliation(s)
- Zhi Liu
- Department of Infectious Disease, Graduate Collaborative Training Base of Zhuzhou, Hengyang Medical School, University of South China, Hengyang, China
- Department of Infectious Disease, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Yuan Ting
- Department of Infectious Disease, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Miao Li
- Jishou University Zhuzhou Clinical College, Medical College, Jishou University, Zhuzhou, China
- Medical College, Jishou University, Xiangxi Tujia and Miao Autonomous Prefecture, Zhuzhou, China
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Yingzheng Tan
- Department of Infectious Disease, Graduate Collaborative Training Base of Zhuzhou, Hengyang Medical School, University of South China, Hengyang, China
- Department of Infectious Disease, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Yunzhu Long
- Department of Infectious Disease, Graduate Collaborative Training Base of Zhuzhou, Hengyang Medical School, University of South China, Hengyang, China
- Department of Infectious Disease, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| |
Collapse
|
41
|
Shen Y, He Y, Pan Y, Liu L, Liu Y, Jia J. Role and mechanisms of autophagy, ferroptosis, and pyroptosis in sepsis-induced acute lung injury. Front Pharmacol 2024; 15:1415145. [PMID: 39161900 PMCID: PMC11330786 DOI: 10.3389/fphar.2024.1415145] [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: 04/10/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Sepsis-induced acute lung injury (ALI) is a major cause of death among patients with sepsis in intensive care units. By analyzing a model of sepsis-induced ALI using lipopolysaccharide (LPS) and cecal ligation and puncture (CLP), treatment methods and strategies to protect against ALI were discussed, which could provide an experimental basis for the clinical treatment of sepsis-induced ALI. Recent studies have found that an imbalance in autophagy, ferroptosis, and pyroptosis is a key mechanism that triggers sepsis-induced ALI, and regulating these death mechanisms can improve lung injuries caused by LPS or CLP. This article summarized and reviewed the mechanisms and regulatory networks of autophagy, ferroptosis, and pyroptosis and their important roles in the process of LPS/CLP-induced ALI in sepsis, discusses the possible targeted drugs of the above mechanisms and their effects, describes their dilemma and prospects, and provides new perspectives for the future treatment of sepsis-induced ALI.
Collapse
Affiliation(s)
- Yao Shen
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Yingying He
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Ying Pan
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Li Liu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Yulin Liu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jing Jia
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
42
|
Lin S, Yan J, Wang W, Luo L. STAT3-Mediated Ferroptosis is Involved in Sepsis-Associated Acute Respiratory Distress Syndrome. Inflammation 2024; 47:1204-1219. [PMID: 38236387 DOI: 10.1007/s10753-024-01970-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/04/2024] [Accepted: 01/06/2024] [Indexed: 01/19/2024]
Abstract
Sepsis-induced acute respiratory distress syndrome (ARDS) poses a grave danger to life, resulting from sepsis-induced multi-organ failure. Although ferroptosis, a form of iron-dependent lipid peroxidative cell death, has been associated with sepsis-induced ARDS, the specific mechanisms are not fully understood. In this study, we utilized WGCNA, PPI, friends analysis, and six machine learning techniques (Lasso, SVM, RFB, XGBoost, AdaBoost, and LightGBM) to pinpoint STAT3 as a potential diagnostic marker. A significant increase in monocyte and neutrophil levels was observed in patients with sepsis-induced ARDS, as revealed by immune infiltration analyses, when compared to controls. Moreover, there was a positive correlation between STAT3 expression and the level of infiltration. Single-cell analysis uncovered a notable disparity in B-cell expression between sepsis and sepsis-induced ARDS. Furthermore, in vitro experiments using LPS-treated human bronchial epithelial cells (BEAS-2B) and THP1 cells demonstrated a significant increase in STAT3 phosphorylation expression. Additionally, the inhibition of STAT3 phosphorylation by Stattic effectively prevented LPS-induced ferroptosis in both BEAS-2B and THP1 cells. This indicates that the activation of STAT3 phosphorylation promotes ferroptosis in human bronchial epithelial cells in response to LPS. In summary, this research has discovered and confirmed STAT3 as a potential biomarker for the diagnosis and treatment of sepsis-induced ARDS.
Collapse
Affiliation(s)
- Shanshan Lin
- The Marine Biomedical Research Institute, The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Jiayu Yan
- The Marine Biomedical Research Institute, The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Wenjian Wang
- The Marine Biomedical Research Institute, The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute, The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China.
| |
Collapse
|
43
|
Chen YL, Wu JM, Chen KY, Wu MH, Yang PJ, Lee PC, Chen PD, Kuo TC, Yeh SL, Lin MT. Intravenous calcitriol administration improves the liver redox status and attenuates ferroptosis in mice with high-fat diet-induced obesity complicated with sepsis. Biomed Pharmacother 2024; 177:116926. [PMID: 38906016 DOI: 10.1016/j.biopha.2024.116926] [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: 04/22/2024] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 06/23/2024] Open
Abstract
Obesity aggravates ferroptosis, and vitamin D (VD) may inhibit ferroptosis. We hypothesized that weight reduction and/or calcitriol administration have benefits against the sepsis-induced liver redox imbalance and ferroptosis in obese mice. Mice were fed a high-fat diet for 11 weeks, then half of the mice continued to consume the diet, while the other half were transferred to a low-energy diet for 5 weeks. After feeding the respective diets for 16 weeks, sepsis was induced by cecal ligation and puncture (CLP). Septic mice were divided into four experimental groups: OS group, obese mice injected with saline; OD group, obese mice with calcitriol; WS group, weight-reduction mice with saline; and WD group, weight-reduction mice with calcitriol. Mice in the respective groups were euthanized at 12 or 24 h after CLP. Results showed that the OS group had the highest inflammatory mediators and lipid peroxide levels in the liver. Calcitriol treatment reduced iron content, enhanced the reduced glutathione/oxidized glutathione ratio, upregulated nuclear factor erythroid 2-related factor 2, ferroptosis-suppressing protein 1, and solute carrier family 7 member 11 expression levels. Also, mitochondrion-associated nicotinamide adenine dinucleotide phosphate oxidase 1, peroxisome proliferator-activated receptor-γ coactivator 1, hypoxia-inducible factor-1α, and heme oxidase-1 expression levels increased in the late phase of sepsis. These results were not noted in the WS group. These findings suggest that calcitriol treatment elicits a more-balanced glutathione redox status, alleviates liver ferroptosis, and enhances mitochondrial biogenesis-associated gene expressions. Weight reduction alone had minimal influences on liver ferroptosis and mitochondrial biogenesis in obese mice with sepsis.
Collapse
Affiliation(s)
- Ya-Ling Chen
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; Nutrition Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Jin-Ming Wu
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Kuen-Yuan Chen
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ming-Hsun Wu
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Po-Jen Yang
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Po-Chu Lee
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Po-Da Chen
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ting-Chun Kuo
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Sung-Ling Yeh
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ming-Tsan Lin
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| |
Collapse
|
44
|
Chen H, Xue H, Tang X, Wang C, Li X, Xie Y. IDENTIFICATION OF A NOVEL SEPSIS PROGNOSIS MODEL: BASED ON TRANSCRIPTOME AND PROTEOME ANALYSIS. Shock 2024; 62:217-226. [PMID: 38899838 DOI: 10.1097/shk.0000000000002388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
ABSTRACT Sepsis is a highly prevalent and deadly disease. Currently, there is a lack of ideal biomarker prognostis models for sepsis. We attempt to construct a model capable of predicting the prognosis of sepsis patients by integrating transcriptomic and proteomic data. Through analysis of proteomic and transcriptomic data, we identified 25 differentially expressed genes (DEGs). Single-factor Cox-Lasso regression analysis identified 16 DEGs (overall survival-DEGs) associated with patient prognosis. Through multifactor Cox-Lasso regression analysis, a prognostic model based on these 16 genes was constructed. Kaplan-Meier survival analysis and receiver operating characteristic curve analysis were used to further validate the high stability and good predictive ability of this prognostic model with internal and external data. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis of overall survival-DEGs and differentially expressed genes between high and low-risk groups based on the prognostic model revealed significant enrichment in immune-related pathways, particularly those associated with viral regulation.
Collapse
Affiliation(s)
- Haoran Chen
- Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Haoyue Xue
- Department of Emergency and Critical Care Medicine, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Xinyi Tang
- Department of Emergency and Critical Care Medicine, Lianyungang Clinical College of Xuzhou Medical University, Lianyungang, Jiangsu, China
| | - Chen Wang
- Department of Emergency and Critical Care Medicine, Lianyungang Clinical College of Xuzhou Medical University, Lianyungang, Jiangsu, China
| | - Xiaomin Li
- Department of Emergency and Critical Care Medicine, Lianyungang Clinical College of Xuzhou Medical University, Lianyungang, Jiangsu, China
| | | |
Collapse
|
45
|
Ferrer R, Iba T. Mitochondrial Damage in Sepsis. JUNTENDO IJI ZASSHI = JUNTENDO MEDICAL JOURNAL 2024; 70:269-272. [PMID: 39431175 PMCID: PMC11487371 DOI: 10.14789/jmj.jmj24-0016-p] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 06/10/2024] [Indexed: 10/22/2024]
Abstract
Mitochondria not only generate adenosine triphosphate (ATP) and act as the powerhouse of the cell but also contribute to host defense by producing reactive oxygen species. Therefore, mitochondrial damage in sepsis directly results in a shortage of energy currency and dysregulation of the immune system. Other than those, mitochondrial damage results in the release of highly dangerous mitochondrial DNA, facilitating acidosis by modulating the metabolism and inducing programmed cell death, thereby facilitating disease progression in sepsis. Various forms of cell death are induced by mitochondrial damage. Aponecrosis is a secondary conversion from apoptosis to necrosis. Although apoptosis is initially intended, it cannot be completed due to ATP depletion from mitochondrial damage, ultimately leading to inflammatory necrosis. Besides such accidental cell death, programmed inflammation-inducing cell deaths such as necroptosis, ferroptosis, and pyroptosis are induced by mitochondrial damage in sepsis. Based on these findings, the regulation of mitochondrial damage holds promise for the development of new therapeutic approaches for sepsis.
Collapse
|
46
|
Ma J, Fu L, Lu Z, Sun Y. Evaluating the Causal Effects of Circulating Proteome on the Risk of Sepsis and Related Outcomes. ACS OMEGA 2024; 9:23864-23872. [PMID: 38854583 PMCID: PMC11154893 DOI: 10.1021/acsomega.4c01934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/09/2024] [Accepted: 05/17/2024] [Indexed: 06/11/2024]
Abstract
The current investigation deployed Mendelian randomization (MR) to elucidate the causal relationship between circulating proteins and sepsis. A rigorous two-sample MR analysis evaluated the effect of plasma proteins on the sepsis susceptibility. To affirm the integrity of MR findings, a suite of supplementary analyses, including Bayesian colocalization, Steiger filtering, the assessment of protein-altering polymorphisms, and the correlation between expression quantitative trait loci and protein quantitative trait loci (pQTLs), was employed. The study further integrated the examination of protein-protein interactions and pathway enrichment, along with the identification of pharmacologically actionable targets, to advance our comprehension and outline potential sepsis therapies. Subsequent analyses leveraging cis-pQTLs within MR studies unveiled noteworthy relationships: 94 specific proteins exhibited significant links with sepsis-related 28 day mortality, while 96 distinct proteins correlated with survival outcomes in sepsis. Furthermore, incorporating both cis- and trans-pQTLs in MR investigations revealed more comprehensive findings, associating 201 unique proteins with sepsis-related 28 day mortality and 199 distinct proteins with survival outcomes in sepsis. Markedly, colocalization analyses confirmed that eight of these proteins exhibited prominent evidence for colocalization, emphasizing their potential criticality in sepsis pathophysiology. Further in silico analyses were conducted to delineate putative regulatory networks and to highlight prospective drug targets among these proteins. Employing the MR methodology has shed light on plasma proteins implicated in the etiopathogenesis of sepsis. This novel approach unveiled numerous biomarkers and targets, providing a scientific rationale for the development of new therapeutic strategies and prophylactic measures against sepsis.
Collapse
Affiliation(s)
- Jiawei Ma
- The
First Department of Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
- Department
of Critical Care Medicine, Wuxi No. 2 People’s
Hospital, Wuxi 214002, China
- Department
of Critical Care Medicine, Aheqi County
People’s Hospital, Xinjiang 843599, China
| | - Lu Fu
- The
First Department of Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Zhonghua Lu
- The
First Department of Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yun Sun
- The
First Department of Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| |
Collapse
|
47
|
Li FJ, Fu S, Ye H, Hu YH, Chen J, Privratsky JR, Yu W, Dong F, Reiter RJ, Dong M, Guo J, Ren J. Metallothionein Alleviates Glutathione Depletion-Induced Oxidative Cardiomyopathy through CISD1-Dependent Regulation of Ferroptosis in Murine Hearts. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:912-926. [PMID: 38417695 DOI: 10.1016/j.ajpath.2024.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/28/2024] [Accepted: 02/15/2024] [Indexed: 03/01/2024]
Abstract
This study was designed to discern the effect of heavy scavenger metallothionein on glutathione (GSH) deprivation-evoked cardiac anomalies and mechanisms involved with an emphasis on ferroptosis. Wild-type and cardiac metallothionein transgenic mice received GSH synthase inhibitor buthionine sulfoximine (BSO; 30 mmol/L in drinking water) for 14 days before assessment of myocardial morphology and function. BSO evoked cardiac remodeling and contractile anomalies, including cardiac hypertrophy, interstitial fibrosis, enlarged left ventricular chambers, deranged ejection fraction, fraction shortening, cardiomyocyte contractile capacity, intracellular Ca2+ handling, sarcoplasmic reticulum Ca2+ reuptake, loss of mitochondrial integrity (mitochondrial swelling, loss of aconitase activity), mitochondrial energy deficit, carbonyl damage, lipid peroxidation, ferroptosis, and apoptosis. Metallothionein itself did not affect myocardial morphology and function, although it mitigated BSO-provoked myocardial anomalies, loss of mitochondrial integrity and energy, and ferroptosis. Immunoblotting revealed down-regulated sarco(endo)plasmic reticulum Ca2+-ATPase 2a, glutathione peroxidase 4, ferroptosis-suppressing CDGSH iron-sulfur domain 1 (CISD1), and mitochondrial regulating glycogen synthase kinase-3β phosphorylation with elevated p53, myosin heavy chain-β isozyme, IκB phosphorylation, and solute carrier family 7 member 11 (SLC7A11) as well as unchanged SLC39A1, SLC1A5, and ferroptosis-suppressing protein 1 following BSO challenge, all of which, except glutamine transporter SLC7A11 and p53, were abrogated by metallothionein. Inhibition of CISD1 using pioglitazone nullified GSH-offered benefit against BSO-induced cardiomyocyte ferroptosis and contractile and intracellular Ca2+ derangement. Taken together, these findings support a regulatory modality for CISD1 in the impedance of ferroptosis in metallothionein-offered protection against GSH depletion-evoked cardiac aberration.
Collapse
Affiliation(s)
- Feng-Juan Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Shouzhi Fu
- Department of ICU/Emergency, Wuhan Third Hospital, Wuhan University, Wuhan, China
| | - Hua Ye
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Burns and Plastic and Wound Repair, Ganzhou People's Hospital, Ganzhou, China
| | - Yi-Han Hu
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Jianxin Chen
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Jamie R Privratsky
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina
| | - Wei Yu
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Feng Dong
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, Texas
| | - Maolong Dong
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Guo
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, China.
| | - Jun Ren
- National Clinical Research Center for Interventional Medicine, Shanghai, China; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
48
|
Zheng LY, Duan Y, He PY, Wu MY, Wei ST, Du XH, Yao RQ, Yao YM. Dysregulated dendritic cells in sepsis: functional impairment and regulated cell death. Cell Mol Biol Lett 2024; 29:81. [PMID: 38816685 PMCID: PMC11140885 DOI: 10.1186/s11658-024-00602-9] [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: 01/16/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Studies have indicated that immune dysfunction plays a central role in the pathogenesis of sepsis. Dendritic cells (DCs) play a crucial role in the emergence of immune dysfunction in sepsis. The major manifestations of DCs in the septic state are abnormal functions and depletion in numbers, which are linked to higher mortality and vulnerability to secondary infections in sepsis. Apoptosis is the most widely studied pathway of number reduction in DCs. In the past few years, there has been a surge in studies focusing on regulated cell death (RCD). This emerging field encompasses various forms of cell death, such as necroptosis, pyroptosis, ferroptosis, and autophagy-dependent cell death (ADCD). Regulation of DC's RCD can serve as a possible therapeutic focus for the treatment of sepsis. Throughout time, numerous tactics have been devised and effectively implemented to improve abnormal immune response during sepsis progression, including modifying the functions of DCs and inhibiting DC cell death. In this review, we provide an overview of the functional impairment and RCD of DCs in septic states. Also, we highlight recent advances in targeting DCs to regulate host immune response following septic challenge.
Collapse
Affiliation(s)
- Li-Yu Zheng
- Translational Medicine Research Center, Medical Innovation Research Division of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Yu Duan
- Department of Critical Care Medicine, Affiliated Chenzhou Hospital (the First People's Hospital of Chenzhou), Southern Medical University, Chenzhou, 423000, China
| | - Peng-Yi He
- Translational Medicine Research Center, Medical Innovation Research Division of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Meng-Yao Wu
- Translational Medicine Research Center, Medical Innovation Research Division of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Shu-Ting Wei
- Translational Medicine Research Center, Medical Innovation Research Division of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Xiao-Hui Du
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Ren-Qi Yao
- Translational Medicine Research Center, Medical Innovation Research Division of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Yong-Ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| |
Collapse
|
49
|
Sun J, Fleishman JS, Liu X, Wang H, Huo L. Targeting novel regulated cell death:Ferroptosis, pyroptosis, and autophagy in sepsis-associated encephalopathy. Biomed Pharmacother 2024; 174:116453. [PMID: 38513593 DOI: 10.1016/j.biopha.2024.116453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/04/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024] Open
Abstract
Sepsis-associated encephalopathy (SAE), a common neurological complication of sepsis, is a heterogenous complex clinical syndrome caused by the dysfunctional response of a host to infection. This dysfunctional response leads to excess mortality and morbidity worldwide. Despite clinical relevance with high incidence, there is a lack of understanding for its both its acute/chronic pathogenesis and therapeutic management. A better understanding of the molecular mechanisms behind SAE may provide tools to better enhance therapeutic efficacy. Mounting evidence indicates that some types of non-apoptotic regulated cell death (RCD), such as ferroptosis, pyroptosis, and autophagy, contribute to SAE. Targeting these types of RCD may provide meaningful targets for future treatments against SAE. This review summarizes the core mechanism by which non-apoptotic RCD leads to the pathogenesis of SAE. We focus on the emerging types of therapeutic compounds that can inhibit RCD and delineate their beneficial pharmacological effects against SAE. Within this review we suggest that pharmacological inhibition of non-apoptotic RCD may serve as a potential therapeutic strategy against SAE.
Collapse
Affiliation(s)
- Jingjing Sun
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 11004, China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Xueyan Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 11004, China
| | - Hongquan Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, 300060, China
| | - Liang Huo
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 11004, China.
| |
Collapse
|
50
|
Zhang W, Liu Y, Liao Y, Zhu C, Zou Z. GPX4, ferroptosis, and diseases. Biomed Pharmacother 2024; 174:116512. [PMID: 38574617 DOI: 10.1016/j.biopha.2024.116512] [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: 12/07/2023] [Revised: 03/03/2024] [Accepted: 03/27/2024] [Indexed: 04/06/2024] Open
Abstract
GPX4 (Glutathione peroxidase 4) serves as a crucial intracellular regulatory factor, participating in various physiological processes and playing a significant role in maintaining the redox homeostasis within the body. Ferroptosis, a form of iron-dependent non-apoptotic cell death, has gained considerable attention in recent years due to its involvement in multiple pathological processes. GPX4 is closely associated with ferroptosis and functions as the primary inhibitor of this process. Together, GPX4 and ferroptosis contribute to the pathophysiology of several diseases, including sepsis, nervous system diseases, ischemia reperfusion injury, cardiovascular diseases, and cancer. This review comprehensively explores the regulatory roles and impacts of GPX4 and ferroptosis in the development and progression of these diseases, with the aim of providing insights for identifying potential therapeutic strategies in the future.
Collapse
Affiliation(s)
- Wangzheqi Zhang
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yang Liu
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yan Liao
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Chenglong Zhu
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China.
| | - Zui Zou
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China.
| |
Collapse
|