1
|
Hu JJ, Deng F, Sun QS, Xiong QM, Min Y, Feng SY, Lin ZB, Chen PH, Hu Z, Wu L, Chen XF, Xie S, Liu WF, Li C, Liu KX. Time-restricted feeding protects against septic liver injury by reshaping gut microbiota and metabolite 3-hydroxybutyrate. Gut Microbes 2025; 17:2486515. [PMID: 40223164 PMCID: PMC12005432 DOI: 10.1080/19490976.2025.2486515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/13/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
Liver injury is an independent risk factor for multiple organ dysfunction and high mortality in patients with sepsis. However, the pathological mechanisms and therapeutic strategies for sepsis-associated liver injury have not been fully elucidated. Time-restricted feeding (TRF) is a promising dietary regime, but its role in septic liver injury remains unknown. Using 16S rRNA gene sequencing, Q200 targeted metabolomics, transcriptomics, germ-free mice, Hmgcs2/Lpin1 gene knockout mice, and Aml12 cells experiments, we revealed that TRF can mitigate septic liver injury by modulating the gut microbiota, particularly by increasing Lactobacillus murinus (L. murinus) abundance, which was significantly reduced in septic mice. Further study revealed that live L. murinus could markedly elevate serum levels of metabolite 3-hydroxybutyrate (3-HB) and alleviate sepsis-related injury, while the knockout of the key enzyme for 3-HB synthesis (3-hydroxy-3-methylglutaryl-CoA synthase 2, Hmgcs2) in the liver negated this protective effect. Additionally, serum 3-HB levels were significantly positively correlated with L. murinus abundance and negatively correlated with liver injury indicators in septic patients, demonstrating a strong predictive value for septic liver injury (AUC = 0.8429). Mechanistically, 3-HB significantly inhibited hepatocyte ferroptosis by activating the PI3K/AKT/mTOR/LPIN1 pathway, reducing ACSL4, MDA, LPO, and Fe2+ levels. This study demonstrates that TRF reduces septic liver injury by modulating gut microbiota to increase L. murinus, which elevates 3-HB to activate PI3K/AKT/mTOR/LPIN1 and inhibit hepatocyte ferroptosis. Overall, this study elucidates the protective mechanism of TRF against septic liver injury and identifies 3-HB as a potential therapeutic target and predictive biomarker, thereby providing new insights into the clinical management and diagnosis of septic liver injury.
Collapse
Affiliation(s)
- Jing-Juan Hu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Fan Deng
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Qi-Shun Sun
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Qing-Ming Xiong
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
- Department of Anesthesiology, The First People’s Hospital of Foshan, Foshan, China
| | - Yue Min
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Si-Yuan Feng
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Ze-Bin Lin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Peng-Han Chen
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Zhen Hu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Ling Wu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Xiao-Feng Chen
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Sun Xie
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Wei-Feng Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Cai Li
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Hou YC, Tseng SM, Kuo TC, Wu JM, Chen KY, Wu MH, Yang PJ, Lee PC, Chen PD, Yeh SL, Lin MT. The role of glutamine and leucine supplementation in liver metabolic reprogramming during sepsis. Life Sci 2025; 374:123708. [PMID: 40360090 DOI: 10.1016/j.lfs.2025.123708] [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: 10/29/2024] [Revised: 04/13/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025]
Abstract
AIMS Glutamine (Gln) and leucine (Leu) are amino acids known for modulating various biological functions. This study aimed to identify metabolism-related genes and their transcriptional pattern changes after Gln and/or Leu administration using next-generation sequencing technology in the liver during sepsis, a condition known to lead to liver metabolic reprogramming and damage. MATERIALS AND METHODS C57BL/6J mice were randomly assigned to a sham control group (C) and four septic groups subjected to cecal ligation and puncture (CLP). The septic groups were as follows: S group, sepsis control with saline injection after CLP; Gln group, injected with Gln after CLP; Leu group, injected with Leu after CLP; and GL group, injected with Gln plus Leu after CLP. All mice were sacrificed on day 4 after the operation, and liver samples were collected for further analysis. KEY FINDINGS Gln and/or Leu administration during sepsis significantly altered the hepatic transcriptome with different gene expression patterns. Notably, the G group had the highest number of gene changes among the amino acid-treated groups. Gln administration was associated with more pronounced downregulation of leukocyte inflammatory genes. Carbohydrate metabolic pathways were suppressed, but the oxidative phosphorylation pathway was enhanced by Gln administration, potentially improving metabolic reprogramming during sepsis. SIGNIFICANCE Gln and/or Leu treatment showed promise in alleviating sepsis-induced liver injury; however, only Gln administration alone demonstrated beneficial effects on hepatic macronutrient and energy metabolism during sepsis. These results highlight the potential therapeutic significance of specific amino acids on attenuating hepatic metabolic dysregulation and injury in septic insult.
Collapse
Affiliation(s)
- Yu-Chen Hou
- Master Program in Food Safety, College of Nutrition, Taipei Medical University, Taipei, Taiwan; School of Food Safety, College of Nutrition, Taipei Medical University, Taipei, Taiwan; Nutrition Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Shang-Ming Tseng
- Department of Traumatology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ting-Chun Kuo
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jin-Ming Wu
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kuen-Yuan Chen
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Hsun Wu
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Jen Yang
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Chu Lee
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Da Chen
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Sung-Ling Yeh
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Tsan Lin
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
3
|
Liu F, Tong R, Song H, Gong L, Ding X, Liu H, Li H, Liu L, Wang Y, Zhang R, Sun T. Metformin attenuated sepsis-related liver injury by modulating the DNA methylation and hydroxymethylation landscape. Int Immunopharmacol 2025; 157:114748. [PMID: 40339494 DOI: 10.1016/j.intimp.2025.114748] [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: 01/12/2025] [Revised: 03/21/2025] [Accepted: 04/24/2025] [Indexed: 05/10/2025]
Abstract
Sepsis-associated liver injury (SLI) is a common complication of sepsis, for which effective therapeutic strategies are lacking. DNA methylation and hydroxymethylation play crucial roles in the regulation of gene expression. This study investigated the effects of metformin on the DNA methylation landscape in a rat model of cecal ligation and puncture (CLP)-induced SLI. Reduced representation bisulfite sequencing (RRBS) and oxidative RRBS (ox-RRBS) were used to assess global DNA methylation and hydroxymethylation patterns in the liver tissues. The results showed that CLP-induced SLI was associated with global DNA hypomethylation and hyperhydroxymethylation, which were partially reversed by metformin treatment. The expression levels of DNA methyltransferases and ten-eleven translocation 2 (TET2) were elevated in the CLP group and were modulated by metformin. Functional enrichment analysis of differentially methylated and hydroxymethylated genes revealed their involvement in oxidative phosphorylation and metabolic pathways. Furthermore, integration of DNA methylation, hydroxymethylation, and transcriptome data identified two genes, A1cf and Atxn7l1, that exhibited increased methylation and decreased expression in CLP, which were reversed by metformin treatment. These findings provide novel insights into the epigenetic mechanisms underlying SLI and suggest that metformin exerts hepatoprotective effects by modulating DNA methylation and hydroxymethylation.
Collapse
Affiliation(s)
- Fengyu Liu
- Department of Critical Care Medicine, Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Zhengzhou 450052, China
| | - Ran Tong
- Department of Critical Care Medicine, Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Zhengzhou 450052, China
| | - Heng Song
- Department of Critical Care Medicine, Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Zhengzhou 450052, China
| | - Linmei Gong
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450052, China
| | - Xianfei Ding
- Department of Critical Care Medicine, Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Zhengzhou 450052, China
| | - Huan Liu
- Department of Critical Care Medicine, Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Zhengzhou 450052, China
| | - Hongyi Li
- Department of Critical Care Medicine, Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Zhengzhou 450052, China
| | - Liming Liu
- Department of Critical Care Medicine, Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Zhengzhou 450052, China
| | - Yuze Wang
- Department of Critical Care Medicine, Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Zhengzhou 450052, China
| | - Ruipeng Zhang
- The First Clinical Medical College of Zhengzhou University, Zhengzhou 450052, China
| | - Tongwen Sun
- Department of Critical Care Medicine, Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Henan Engineering Research Center for Critical Care Medicine, Henan Key Laboratory of Critical Care Medicine, Henan Key Laboratory of Sepsis in Health Commission, Zhengzhou Key Laboratory of Sepsis, Henan Sepsis Diagnosis and Treatment Center, Zhengzhou 450052, China.
| |
Collapse
|
4
|
Li H, Chen J, Guo H, Yang H, Liu J, Yuan H, Zhang J, Wang J, Liu S. Integrated transcriptomic and proteomic profiling reveals the anti-inflammatory mechanism of dihydroartemisinin in the treatment of acute liver injury by targeting CYBA and CYBB. Biochem Biophys Res Commun 2025; 764:151821. [PMID: 40250321 DOI: 10.1016/j.bbrc.2025.151821] [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: 01/24/2025] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
Acute liver injury (ALI) is a prevalent inflammatory disease with no currently available effective targeted therapies that characterized by high mortality and morbidity. Dihydroartemisinin (DHA), a derivative of the renowned antimalarial compound artemisinin, has garnered attention for its anti-inflammatory property. However, the precise anti-inflammatory mechanisms underlying its efficacy in treating ALI remain unclear. Notably, the excessive inflammatory cytokines secreted by macrophages represents a critical factor of liver damage. In our comprehensive study, transcriptome and proteomic analysis of M1 macrophages after DHA treatment was performed to unearth the potential anti-inflammatory targets for ALI treatment. Transcriptomics analysis indicated that DHA significantly mitigated inflammation, primarily by downregulating the expressions of CCL1, CCL2, CCL7, CCL13, and CXCL13. Concurrently, proteomics analysis identified six proteins, such as CYBA and CYBB, that were consistently downregulated in the DHA intervention groups compared to the M1 group. Intriguingly, a protein-protein interaction network analysis highlighted the close association of CYBA and CYBB with the aforementioned chemokine genes. Through meticulous screening, DHA curtailed the production of reactive oxygen species (ROS) by targeting CYBA and CYBB, subsequently suppressing the secretion of several chemokines and dampening the inflammatory response in M1 macrophages. More importantly, DHA not only reduced ROS and chemokine levels but also restored liver function by downregulating CYBA and CYBB to inhibit NF-κB pathway in ALI mice, demonstrating strong anti-inflammatory effects. In conclusion, our findings throw novel light into the underlying anti-inflammatory mechanism of DHA in ALI management, offering valuable insights for future clinical research and therapeutic strategies for inflammatory diseases.
Collapse
Affiliation(s)
- Honglian Li
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory of Drug Metabolism Research and Evaluation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jiayun Chen
- State Key Laboratory for Quality Ensuanse and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Huiyi Guo
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory of Drug Metabolism Research and Evaluation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hao Yang
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory of Drug Metabolism Research and Evaluation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jing Liu
- State Key Laboratory for Quality Ensuanse and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Haoxing Yuan
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory of Drug Metabolism Research and Evaluation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Junzhe Zhang
- State Key Laboratory for Quality Ensuanse and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jigang Wang
- State Key Laboratory for Quality Ensuanse and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory of Drug Metabolism Research and Evaluation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Southern Medical University, Guangzhou, 510515, China; MOE Innovation Center for Medical Basic Research on Inflammation and Immune Related Diseases, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
5
|
Parolini C. Sepsis and high-density lipoproteins: Pathophysiology and potential new therapeutic targets. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167761. [PMID: 40044061 DOI: 10.1016/j.bbadis.2025.167761] [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: 10/14/2024] [Revised: 01/19/2025] [Accepted: 02/25/2025] [Indexed: 03/10/2025]
Abstract
In 2020, sepsis has been defined a worldwide health major issue (World Health Organization). Lung, urinary tract and abdominal cavity are the preferred sites of sepsis-linked infection. Research has highlighted that the advancement of sepsis is not only related to the presence of inflammation or microbial or host pattern recognition. Clinicians and researchers now recognized that a severe immunosuppression is also a common feature found in patients with sepsis, increasing the susceptibility to secondary infections. Lipopolysaccharides (LPS) are expressed on the cell surface of Gram-negative, whereas Gram-positive bacteria express peptidoglycan (PGN) and lipoteichoic acid (LTA). The main mechanism by which LPS trigger host innate immune responses is binding to TLR4-MD2 (toll-like receptor4-myeloid differentiation factor 2), whereas, PGN and LTA are exogenous ligands of TLR2. Nucleotide-binding oligomerization domain (NOD)-like receptors are the most well-characterized cytosolic pattern recognition receptors, which bind microbial molecules, endogenous by-products and environmental triggers. It has been demonstrated that high-density lipoproteins (HDL), besides their major role in promoting cholesterol efflux, possess diverse pleiotropic properties, ranging from a modulation of the immune system to anti-inflammatory, anti-apoptotic, and anti-oxidant functions. In addition, HDL are able at i) binding LPS, preventing the activating of TLR4, and ii) inducing the expression of ATF3 (Activating transcription factor 3), a negative regulator of the TLR signalling pathways, contributing at justifying their capacity to hamper infection-based illnesses. Therefore, reconstituted HDL (rHDL), constituted by apolipoprotein A-I/apolipoprotein A-IMilano complexed with phospholipids, may be considered as a new therapeutic tool for the management of sepsis.
Collapse
Affiliation(s)
- Cinzia Parolini
- Department of Pharmacological and Biomolecular Sciences, "Rodolfo Paoletti", via Balzaretti 9 - Università degli Studi di Milano, 20133 Milano, Italy.
| |
Collapse
|
6
|
Lei J, Zhai J, Zhang Y, Qi J, Sun C. Supervised Machine Learning Models for Predicting Sepsis-Associated Liver Injury in Patients With Sepsis: Development and Validation Study Based on a Multicenter Cohort Study. J Med Internet Res 2025; 27:e66733. [PMID: 40418571 DOI: 10.2196/66733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 12/15/2024] [Accepted: 04/29/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Sepsis-associated liver injury (SALI) is a severe complication of sepsis that contributes to increased mortality and morbidity. Early identification of SALI can improve patient outcomes; however, sepsis heterogeneity makes timely diagnosis challenging. Traditional diagnostic tools are often limited, and machine learning techniques offer promising solutions for predicting adverse outcomes in patients with sepsis. OBJECTIVE This study aims to develop an explainable machine learning model, incorporating stacking techniques, to predict the occurrence of liver injury in patients with sepsis and provide decision support for early intervention and personalized treatment strategies. METHODS This retrospective multicenter cohort study adhered to the TRIPOD+AI (Transparent Reporting of a Multivariable Prediction Model for Individual Prognosis or Diagnosis, Extended for Artificial Intelligence) guidelines. Data from 8834 patients with sepsis in the Medical Information Mart for Intensive Care IV (MIMIC-IV) database were used for training and internal validation, while data from 4236 patients in the eICU-Collaborative Research Database (eICU-CRD) database were used for external validation. SALI was defined as an international normalized ratio >1.5 and total bilirubin >2 mg/dL within 1 week of intensive care unit admission. Nine machine learning models-decision tree, random forest (RF), extreme gradient boosting (XGBoost), light gradient boosting machine (LightGBM), support vector machine, elastic net, logistic regression, multilayer perceptron, and k-nearest neighbors-were trained. A stacking ensemble model, using LightGBM, XGBoost, and RF as base learners and Lasso regression as the meta-model, was optimized via 10-fold cross-validation. Hyperparameters were tuned using grid search and Bayesian optimization. Model performance was evaluated using accuracy, balanced accuracy, Brier score, detection prevalence, F1-score, Jaccard index, κ coefficient, Matthews correlation coefficient, negative predictive value, positive predictive value, precision, recall, area under the receiver operating characteristic curve (ROC-AUC), precision-recall AUC, and decision curve analysis. Shapley additive explanations (SHAP) values were used to quantify feature importance. RESULTS In the training set, LightGBM, XGBoost, and RF demonstrated the best performance among all models, with ROC-AUCs of 0.9977, 0.9311, and 0.9847, respectively. These models exhibited minimal variance in cross-validation, with tightly clustered ROC-AUC and precision-recall area under the curve distributions. In the internal validation set, LightGBM (ROC-AUC 0.8401) and XGBoost (ROC-AUC 0.8403) outperformed all other models, while RF achieved an ROC-AUC of 0.8193. In the external validation set, LightGBM (ROC-AUC 0.7077), XGBoost (ROC-AUC 0.7169), and RF (ROC-AUC 0.7081) maintained strong performance, although with slight decreases in ROC-AUC compared with the training set. The stacking model achieved ROC-AUCs of 0.995, 0.838, and 0.721 in the training, internal validation, and external validation sets, respectively. Key predictors-total bilirubin, lactate, prothrombin time, and mechanical ventilation status-were consistently identified across models, with SHAP analysis highlighting their significant contributions to the model's predictions. CONCLUSIONS The stacking ensemble model developed in this study yields accurate and robust predictions of SALI in patients with sepsis, demonstrating potential clinical utility for early intervention and personalized treatment strategies.
Collapse
Affiliation(s)
- Jingchao Lei
- Third Xiangya Hospital of Central South University, Changsha, China
| | - Jia Zhai
- Third Xiangya Hospital of Central South University, Changsha, China
| | - Yao Zhang
- Third Xiangya Hospital of Central South University, Changsha, China
| | - Jing Qi
- Third Xiangya Hospital of Central South University, Changsha, China
| | - Chuanzheng Sun
- Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
7
|
He Y, Chen X, Zhong J, Lin C, Situ J, Liu B, Yan Y, Gui S, Mao C, Xing S. Glucocorticoid reduces mortality in LPS-induced sepsis mouse model by inhibiting JAK1/STAT3-mediated inflammatory response and restoring tricarboxylic acid cycle. Life Sci 2025; 375:123744. [PMID: 40408938 DOI: 10.1016/j.lfs.2025.123744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/19/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025]
Abstract
AIMS Sepsis is characterized by a systemic inflammation disorder and multi-organ dysfunction caused by infection. Glucocorticoids (GCs), as anti-inflammatory properties drugs, are widely used for the management of patients with inflammatory diseases, including sepsis. However, the therapeutic effect and underlying mechanisms of GCs on sepsis remain incompletely understood. MATERIALS AND METHODS Mouse sepsis models were constructed using intraperitoneal injection of lipopolysaccharide (LPS) and treated with synthetic GC dexamethasone (DEX). Systemic inflammation and organ injury were evaluated by histological staining, immunophenotypic analysis, and detection of inflammatory cytokines. Inflammatory signaling pathways were identified by western blot (WB) and RT-qPCR. Finally, targeted metabolomics were conducted to examine metabolic changes. KEY FINDINGS Subcutaneous administration of DEX significantly alleviate LPS-induced acute organ injury and systemic inflammation, thereby reducing mortality in septic mice. DEX substantially reduced the amounts of intrahepatic neutrophils/macrophages, and inflammatory cytokines expressions in the livers of septic mice and in LPS-stimulated bone-marrow-derived macrophages (BMDMs). Mechanistically, DEX inhibit JAK1/STAT3 signaling pathway both in livers and BMDMs. Pharmacological inhibition of JAK1 or STAT3 via Upadacitinib or Stattic, respectively, partially mimic the anti-inflammatory function of GC, including the reduction of intrahepatic macrophages, proinflammatory cytokines production, and mortality in septic mice. Additionally, DEX could also promote the rewiring of tricarboxylic acid (TCA) cycle in the livers of septic mice. SIGNIFICANCE This study provides important insights into the molecular and metabolic mechanisms underlying GCs-mediated anti-inflammatory reactions.
Collapse
Affiliation(s)
- Yudan He
- Intensive Care Unit, The First Affiliated Hospital of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Infection Immunity and Inflammation, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Xinyue Chen
- Guangdong Provincial Key Laboratory of Infection Immunity and Inflammation, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jiayue Zhong
- Guangdong Provincial Key Laboratory of Infection Immunity and Inflammation, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Canfeng Lin
- Guangdong Provincial Key Laboratory of Infection Immunity and Inflammation, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jieying Situ
- Guangdong Provincial Key Laboratory of Infection Immunity and Inflammation, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Bing Liu
- Guangdong Provincial Key Laboratory of Infection Immunity and Inflammation, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Yiwen Yan
- Guangdong Provincial Key Laboratory of Infection Immunity and Inflammation, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Shuiqing Gui
- Intensive Care Unit, The First Affiliated Hospital of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Chengzhou Mao
- Guangdong Provincial Key Laboratory of Infection Immunity and Inflammation, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China.
| | - Shaojun Xing
- Intensive Care Unit, The First Affiliated Hospital of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Infection Immunity and Inflammation, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China; Marshall Laboratory of Biomedical Engineering, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
8
|
Wang Y, Zhang H, Miao C. Unraveling immunosenescence in sepsis: from cellular mechanisms to therapeutics. Cell Death Dis 2025; 16:393. [PMID: 40379629 DOI: 10.1038/s41419-025-07714-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/26/2025] [Accepted: 05/02/2025] [Indexed: 05/19/2025]
Abstract
Sepsis is a life-threatening multiple organ dysfunction resulting from a dysregulated host response to infection, and patients with sepsis always exhibit a state of immune disorder characterized by both overwhelming inflammation and immunosuppression. The aging of immune system, namely "immunosenescence", has been reported to be correlated with high morbidity and mortality in elderly patients with sepsis. Initially, immunosenescence was considered as a range of age-related alterations in the immune system. However, increasing evidence has proven that persistent inflammation or even a short-term inflammatory challenge during sepsis could trigger accelerated aging of immune cells, which might further exacerbate inflammatory cytokine storm and promote the shift towards immunosuppression. Thus, premature immunosenescence is found in young sepsis individuals, which further aggravates immune disorders and induces the progression of sepsis. Furthermore, in old sepsis patients, the synergistic effects of both sepsis and aging may cause immunosenescence-associated alterations more significantly, resulting in more severe immune dysfunction and a worse prognosis. Therefore, it is necessary to explore the potential therapeutic strategies targeting immunosenescence during sepsis.
Collapse
Affiliation(s)
- Yanghanzhao Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key laboratory of Perioperative Stress and Protection, Shanghai, China.
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key laboratory of Perioperative Stress and Protection, Shanghai, China.
- Department of Anesthesiology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Li S, Liu J, Wu J, Zheng X. Immunological Mechanisms and Effects of Bacterial Infections in Acute-on-Chronic Liver Failure. Cells 2025; 14:718. [PMID: 40422221 DOI: 10.3390/cells14100718] [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: 04/09/2025] [Revised: 05/09/2025] [Accepted: 05/10/2025] [Indexed: 05/28/2025] Open
Abstract
Acute-on-chronic liver failure (ACLF) is a severe clinical syndrome characterized by high morbidity and mortality rates. Bacterial infection is a frequent precipitating factor and complication in ACLF patients, significantly worsening patient outcomes. Elucidating the mechanisms underlying bacterial infections and their impact on ACLF pathophysiology is crucial for developing effective therapies to reduce infection rates and mortality. Current research highlights that immune suppression in ACLF increases susceptibility to bacterial infections, which in turn exacerbate immune dysfunction. However, a comprehensive review summarizing the emerging mechanisms underlying this immunosuppression is currently lacking. This review aims to provide an overview of the latest research, focusing on alterations in the immune responses of innate immune cells-including monocytes, macrophages, and neutrophils-as well as adaptive immune cells such as T and B lymphocytes during the onset and progression of bacterial infections in ACLF. In addition, recent advances in immunomodulatory therapies, including stem cell-based interventions, will also be discussed.
Collapse
Affiliation(s)
- Sumeng Li
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jing Liu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jun Wu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xin Zheng
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
10
|
Kuebart A, Kaltenbach N, Hoffmann C, Picker O, Herminghaus A. Effects of midazolam and dexmedetomidine on liver and intestinal mitochondrial respiration. Eur J Pharmacol 2025:177740. [PMID: 40381677 DOI: 10.1016/j.ejphar.2025.177740] [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: 11/12/2024] [Revised: 05/11/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND Mitochondrial dysfunction frequently occurs in critically ill patients and is thought to contribute to metabolic shutdown and hepatic or gastrointestinal failure. In addition to hypoxemia and inflammation, sedatives are considered potential contributors to further impairment of mitochondrial function. OBJECTIVE This study investigated the effect of the sedatives midazolam and dexmedetomidine on liver and colon mitochondrial function. Especially colon mitochondrial affection by both drugs was evaluated here for the first time. DESIGN Liver and colon tissue homogenates from healthy Wistar rats (n=60) were treated with therapeutical and supra-therapeutical doses of midazolam (0.25, 5, 50, 100, 500 μM) and dexmedetomidine (1, 5, 50, 100, 1000 nM). Samples were analyzed using the Clark electrode (Strathkelvin 782). Data analyses were performed via GraphPad Prism® 8 using the Kruskal-Wallis + Dunn's post-hoc test. p < 0.05 was considered significant. RESULTS In liver homogenates, treatment with 500 μM midazolam (48.6 ± 6.6 %) showed a significantly decreased respiratory control index (RCI) compared with the control (107.1 ± 14.8 %, p < 0.01) for complex II but without decreases in ADP:O ratio. Colon homogenate did not show significant alterations after midazolam treatment. Dexmedetomidine treatment did not affect colon or liver mitochondrial respiration in any dosage. CONCLUSIONS In clinical dosages on healthy organs, midazolam and dexmedetomidine did not show any effects on mitochondrial respiration in the colon and liver. However, in supra-therapeutical dosages, midazolam showed an organ-specific, dose-dependent, and complex-dependent effect on mitochondrial respiration by reducing the coupling of ETS and OXPHOS but without decreasing efficacy.
Collapse
Affiliation(s)
- Anne Kuebart
- Department of Anesthesiology, University Hospital Duesseldorf, Moorenstrasse 5, 40225 Duesseldorf, Germany.
| | - Nicola Kaltenbach
- Department of Anesthesiology, University Hospital Duesseldorf, Moorenstrasse 5, 40225 Duesseldorf, Germany.
| | - Christina Hoffmann
- Department of Anesthesiology, University Hospital Duesseldorf, Moorenstrasse 5, 40225 Duesseldorf, Germany.
| | - Olaf Picker
- Department of Anesthesiology, University Hospital Duesseldorf, Moorenstrasse 5, 40225 Duesseldorf, Germany.
| | - Anna Herminghaus
- Department of Anesthesiology, University Hospital Duesseldorf, Moorenstrasse 5, 40225 Duesseldorf, Germany.
| |
Collapse
|
11
|
Liu B, Tian X, Li L, Jiang N, Cheng M, Zhu J, Wu Z. SFTSV induces liver ferroptosis through m6A-related ferritinophagy. Autophagy 2025:1-14. [PMID: 40340535 DOI: 10.1080/15548627.2025.2503564] [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: 08/14/2024] [Revised: 04/15/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) is a widely prevalent infectious disease caused by severe fever with thrombocytopenia syndrome virus (SFTSV). SFTSV infection carries a high mortality rate and has emerged to be a public health concern. SFTSV infection could induce many classical cell death pathways. Ferroptosis, a novel iron-dependent form of regulated cell death, is shown to participate in various biological processes and is considered as a new therapeutic target. In the current study, we reported that SFTSV infection perturbed the classical redox cycle by downregulating the expression of GPX4, SLC7A11 and GSH, and increasing the level of reactive oxygen species (ROS) and malondialdehyde (MDA). Interestingly, we observed that the elevation of ATG5 mRNA m6A modification after SFTSV infection and mutation of the m6A-sites significantly rescued SFTSV infection-induced ferritinophagy. We further found that the NSs protein of SFTSV played a major role in driving the ferritinophagy. Finally, we found that ferroptosis inhibitor ferrostatin-1 prevented ferroptosis and suppressed SFTSV infection both in vitro and in vivo models. In summary, our study demonstrated that SFTSV infection could induce ferroptosis in liver, and m6A modified ATG5 mediated ferritinophagy to facilitate this process. Targeting ferroptosis may serve as a potential therapy for the treatment of SFTS.Abbreviations: ATG5: autophagy related 5; Baf-A1: bafilomycin A1; Fer-1: ferrostatin-1; Fe2+: ferrous iron; FTH1: ferritin heavy chain 1; GOT1/AST: glutamic-oxaloacetic transaminase 1; GPT/ALT: glutamic - pyruvic transaminase; GSH: glutathione; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MDA: malondialdehyde; NCOA4: nuclear receptor coactivator 4; ROS: reactive oxygen species; SFTSV: severe fever with thrombocytopenia virus; SQSTM1/p62: sequestosome 1.
Collapse
Affiliation(s)
- Bingxin Liu
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Xiaoyan Tian
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Linrun Li
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Na Jiang
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Min Cheng
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Jin Zhu
- The Department of Immunology, Huadong Medical Institute of Biotechnique, Nanjing, China
| | - Zhiwei Wu
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Q2 Science, Nanjing University, Nanjing, China
- School of Pharmacy, Dali University, Dali, Yunnan, China
| |
Collapse
|
12
|
Zhang B, He C, Su T, Qin Z, Cheng Q, Zhu Z, Chen M, Yu W. Association and dynamic change of fecal calprotectin with sepsis associated liver dysfunction in adults with sepsis. Sci Rep 2025; 15:16192. [PMID: 40346167 PMCID: PMC12064738 DOI: 10.1038/s41598-025-98904-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 04/15/2025] [Indexed: 05/11/2025] Open
Abstract
Fecal calprotectin (FC) is a biomarker of gut inflammation but its association with sepsis associated liver dysfunction (SALD) is unclear. This single-center, prospective cohort study investigated the relationship between FC and SALD in adults with sepsis. FC concentrations were measured on days 1 (FC1) and 3 (FC3). The difference between FC3 and FC1 (∆FC) was calculated. The relationship between FC and SALD was assessed using multivariate analysis. Receiver operating characteristic curves were used to assess the predictive value of SALD biomarkers. Ninety-five patients with sepsis (33 with SALD and 62 without SALD) were enrolled between January 2023 and March 2024. The median FC3 level and ∆FC were significantly higher in the SALD group than in the non-SALD group (both p < 0.001), whereas the median FC1 level did not differ significantly between groups. FC3 level and ∆FC were positively associated with SALD in the adjusted analysis (both p < 0.001) with areas under the curve of 0.784 and 0.848, respectively, and cut-off values of 930.88 μg/g and - 8.80 μg/g, respectively. In adults with sepsis, SALD occurrence was more strongly associated with elevated FC3 levels and ∆FC than with FC1 levels; therefore, monitoring FC3 levels and ∆FC could help detect SALD.
Collapse
Affiliation(s)
- Beiyuan Zhang
- Department of Critical Care Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China
| | - Chenzhe He
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, No. 321 Zhongshan Road, Nanjing, Jiangsu Province, China
| | - Ting Su
- Department of Critical Care Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China
| | - Zimeng Qin
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, No. 321 Zhongshan Road, Nanjing, Jiangsu Province, China
| | - Qi Cheng
- Department of Critical Care Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China
| | - Zhanghua Zhu
- Department of Critical Care Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China
| | - Ming Chen
- Department of Critical Care Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China.
| | - Wenkui Yu
- Department of Critical Care Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China.
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, No. 321 Zhongshan Road, Nanjing, Jiangsu Province, China.
| |
Collapse
|
13
|
Tsukiyama N, Tanaka Y, Yamane H, Tanimine N, Kuroda S, Tahara H, Ohira M, Ide K, Kobayashi T, Ohdan H. Impacts of high mobility group box protein 1 gene polymorphisms on morbidity and mortality after living donor liver transplantation. Transpl Immunol 2025; 90:102225. [PMID: 40157616 DOI: 10.1016/j.trim.2025.102225] [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: 11/18/2024] [Revised: 03/25/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
We investigated the effect of single-nucleotide polymorphisms (SNPs) in the high mobility group box 1 (HMGB1) gene on morbidity and mortality after liver transplantation (LT). Among 120 LT recipients and their living donors, the genotypes of HMGB1, and the SNPs rs2249825, rs1045411, rs1412125, and rs1360485 were determined. There were no significant associations between these four SNPs and the incidence of rejection or mortality. However, the incidence of early allograft dysfunction (EAD) (n = 43), which presents as functional insufficiency within 1 week of LT, was significantly higher in recipients with the GC + CC allele of rs2249825 (n = 17/34) than in those with the GG allele (n = 26/86) (p = 0.044). Although the impact of donor HMGB1 SNPs on the incidence of EAD was not statistically significant, recipients with the GC + CC allele of rs2249825 who received liver grafts from donors with the same genotype had the highest incidence of EAD (p = 0.052). In contrast, the donor TC + CC allele of rs1412125 was an independent risk factor for the development of sepsis (n = 33) in LT recipient (OR = 3.05, 95 % CI = 1.18-7.87, p = 0.021). Thus, the SNPs of the HMGB1 gene in either recipients or donors were not associated with mortality but influenced the incidence of EAD and sepsis, likely being a predictive biomarker for the risk of serious complications after LT.
Collapse
Affiliation(s)
- Naofumi Tsukiyama
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Yuka Tanaka
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan.
| | - Hiroaki Yamane
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Naoki Tanimine
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Shintaro Kuroda
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Hiroyuki Tahara
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Masahiro Ohira
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Kentaro Ide
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Tsuyoshi Kobayashi
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan.
| |
Collapse
|
14
|
Zhao X, Liu Y, Wang D, Li T, Xu Z, Li Z, Bai X, Wang Y. Role of GLP‑1 receptor agonists in sepsis and their therapeutic potential in sepsis‑induced muscle atrophy (Review). Int J Mol Med 2025; 55:74. [PMID: 40052580 PMCID: PMC11936484 DOI: 10.3892/ijmm.2025.5515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/17/2025] [Indexed: 03/27/2025] Open
Abstract
Sepsis‑induced myopathy (SIM) is a common complication in intensive care units, which is often associated with adverse outcomes, primarily manifested as skeletal muscle weakness and atrophy. Currently, the management of SIM focuses on prevention strategies, as effective therapeutic options remain elusive. Glucagon‑like peptide‑1 (GLP‑1) receptor agonists (GLP‑1RAs) have garnered attention as hypoglycemic and weight‑loss agents, with an increasing body of research focusing on the extrapancreatic effects of GLP‑1. In preclinical settings, GLP‑1RAs exert protective effects against sepsis‑related multiple organ dysfunction through anti‑inflammatory and antioxidant mechanisms. Based on the existing research, we hypothesized that GLP‑1RAs may serve potential protective roles in the repair and regeneration of skeletal muscle affected by sepsis. The present review aimed to explore the relationship between GLP‑1RAs and sepsis, as well as their impact on muscle atrophy‑related myopathy. Furthermore, the potential mechanisms and therapeutic benefits of GLP‑1RAs are discussed in the context of muscle atrophy induced by sepsis.
Collapse
Affiliation(s)
- Xuan Zhao
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yukun Liu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Dongfang Wang
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Tonghan Li
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhikai Xu
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhanfei Li
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiangjun Bai
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yuchang Wang
- Trauma Center, Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
15
|
Liu Z, Su M, Wan P, Zhen X, He L, Liang H. Identification of Natural Killer Cell-Related Genes as Potential Diagnostic Biomarkers for Pediatric Sepsis. APMIS 2025; 133:e70030. [PMID: 40411297 DOI: 10.1111/apm.70030] [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: 01/16/2024] [Revised: 04/16/2025] [Accepted: 05/06/2025] [Indexed: 05/26/2025]
Abstract
Pediatric sepsis is a prevalent severe condition with a high disability rate and mortality. The purpose of this study was to investigate how NK cell-related genes (NKGs) function in the diagnosis of pediatric sepsis. Differential analysis and Venn analysis were utilized to screen differentially expressed NKGs (DE-NKGs) in pediatric sepsis. The PPI network of DE-NKGs was generated using STRING. Candidate drugs are predicted using the CMAP database. miRNA and transcription factor (TF) targeting hub genes were screened using the Networkanalyst website. A total of 22 DE-NKGs were identified, which were mainly enriched in immune-related biological processes. Ten hub genes that were filtered out from DE-NKGs exhibited good diagnostic performance. Immune infiltration analysis revealed that macrophages and neutrophils had higher infiltration abundance in the pediatric sepsis group, whereas NK cells had higher infiltration abundance in the normal group. NKGs (LCK, FCER1G, PRF1, CD247, GZMB, CD2, CCL4, KLRD1, CCL5, and ITGB2) with diagnostic performance were successfully predicted, and some potential miRNAs, TFs, and candidate drugs were also predicted. In conclusion, this study threw light on a comprehensive understanding of the role of NK cells in pediatric sepsis.
Collapse
Affiliation(s)
- Zhihong Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning City, China
| | - Minghua Su
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning City, China
| | - Peiqi Wan
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning City, China
| | - Xiumei Zhen
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning City, China
| | - Lixia He
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning City, China
| | - Huan Liang
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning City, China
| |
Collapse
|
16
|
Li J, Xuan M, Yang L, Liu Y, Lou N, Fu L, Shi Q, Xue C. Comprehensive single-cell analysis deciphered the immunoregulatory mechanism of TPPU in alleviating sepsis-related acute liver injury. J Adv Res 2025; 71:457-470. [PMID: 39956402 DOI: 10.1016/j.jare.2025.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/21/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025] Open
Abstract
INTRODUCTION Sepsis-related acute liver injury involves complex immune dysfunctions. Epoxyeicosatrienoic acids (EETs), bioactive molecules derived from arachidonic acid (AA) via cytochrome P450 (CYP450) and rapidly hydrolyzed by soluble epoxide hydrolase (sEH), possess anti-inflammatory properties. Nevertheless, the impact of the sEH inhibitor TPPU on sepsis-related acute liver injury remains uncertain. OBJECTIVES This study utilized comprehensive single-cell analysis to investigate the immunoregulatory mechanism of TPPU in alleviating sepsis-related acute liver injury. METHODS Hepatic bulk RNA sequencing and proteomics analyses were employed to investigate the mechanisms underlying sepsis-related acute liver injury induced by cecal ligation and puncture in mice. Cytometry by time-of-flight and single-cell RNA sequencing were conducted to thoroughly examine the immunoregulatory role of TPPU at single-cell resolution. RESULTS Downregulation of AA metabolism and the CYP450 pathway was observed during sepsis-related acute liver injury, and TPPU treatment reduced inflammatory cytokine production and mitigated sepsis-related hepatic inflammatory injury. Comprehensive single-cell analysis revealed that TPPU promotes the expansion of anti-inflammatory CD206+CD73+ M2-like macrophages and PDL1-CD39-CCR2+ neutrophils, reprogramming liver neutrophils to an anti-inflammatory CAMP+NGP+CD177+ phenotype. Additionally, TPPU inhibits the CCL6-CCR1 signaling mediated by M2-like macrophages and CAMP+NGP+CD177+ neutrophils, altering intercellular communication within the septic liver immune microenvironment. CONCLUSION This study demonstrated TPPU's protective efficacy against sepsis-related acute liver injury, underscoring its vital role in modulating liver macrophages and neutrophils and enhancing prospects for personalized immunomodulatory therapy.
Collapse
Affiliation(s)
- Juan Li
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengjuan Xuan
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li Yang
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yingru Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Na Lou
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Leiya Fu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qingmiao Shi
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Chen Xue
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
17
|
Liu J, Hu J, Yao X, Xu M, Yuan A, Guo J, Wang C, Le Y, Yuan X, Lu D. CLICs Inhibitor IAA94 Alleviates Inflammation and Injury in Septic Liver by Preventing Pyroptosis in Macrophages. Inflammation 2025:10.1007/s10753-025-02304-6. [PMID: 40259192 DOI: 10.1007/s10753-025-02304-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/18/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025]
Abstract
Macrophage pyroptosis represents a pivotal mechanism underlying acute liver injury during sepsis. Chloride intracellular channel proteins (CLICs) have been linked to inflammatory reflexes, with IAA94 serving as an inhibitor of channel formation characteristic of CLICs. In a mouse model, IAA94 demonstrated efficacy in reducing pro-inflammatory cytokines in liver tissues, decreasing macrophage in the liver, inhibiting the development of the pro-fibrosis phenotype, and alleviating tissue injury. Additionally, IAA94 exhibited inhibitory effects on the activation of NLRP3 inflammasome, leading to the suppression of pyroptosis in J774A.1 cells and the liver. Additionally, IAA94 was observed to impede the interaction between NEK7 and NLRP3. Furthermore, it was observed that the conditioned medium of pyroptotic macrophages treated with IAA94 induced an attenuated inflammatory response in hepatocytes in comparison to that induced by the conditioned medium of pyroptotic macrophages. However, NLRP3 overexpression impeded the beneficial effects of IAA94. In conclusion, IAA94 has the capacity to impede NLRP3 inflammasome formation-mediated pyroptosis by blocking CLICs-mediated chloride efflux and the inhibition of NEK7-NLRP3 interactions, thereby establishing CLICs as a promising therapeutic target against liver inflammation.
Collapse
Affiliation(s)
- Jing Liu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jingwen Hu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xulei Yao
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Mengting Xu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Aini Yuan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jianan Guo
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Cui Wang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yifei Le
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xingyu Yuan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Dezhao Lu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
18
|
Wu CH, Hu S, Li D, Jiang XW, Ou-Yang H, Bi GF, Wang P, Liang FT, Zhou WH, Yang X, Fang JH, Bi HC. Pregnane X receptor alleviates sepsis-induced liver injury through activation of yes-associated protein in mice. Acta Pharmacol Sin 2025:10.1038/s41401-025-01552-4. [PMID: 40234620 DOI: 10.1038/s41401-025-01552-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/20/2025] [Indexed: 04/17/2025]
Abstract
The severity of sepsis is attributed to excessive inflammatory responses leading to liver injury. Pregnane X receptor (PXR), a nuclear receptor that controls xenobiotic and endobiotic metabolism, has been implicated in regulating inflammation and liver regeneration. This study aimed to investigate the role of PXR in sepsis-induced liver injury and the underlying mechanisms. Sepsis models were established in mice, the mice were administered the typical mouse PXR agonist PCN (100 mg·kg-1·d-1, i.p.) for 3 consecutive days in advance, then subjected to CLP operation or LPS administration 1 h after the last administration of PCN. The results showed that PCN pretreatment significantly increased the survival rate of septic mice, while the survival rate was reduced after the knockout of Pxr. In addition, PCN pretreatment effectively alleviated sepsis-induced liver injury. In Pxr knockout mice, liver injury was more severe, whereas the protective effects of PCN pretreatment were abolished. Mechanistically, PCN pretreatment significantly upregulated the expression of yes-associated protein (YAP) and its downstream targets and decreased the level of phosphorylated nuclear factor-κB (NF-κB). Moreover, liver-specific knockdown of Yap blocked the protective effects of PCN pretreatment against sepsis-induced liver injury and downregulated the phosphorylation level of NF-κB. In summary, this study demonstrated that PXR activation protects against sepsis-induced liver injury through activation of the YAP signaling pathway, providing a new strategy for the diagnosis and treatment of sepsis-induced liver injury.
Collapse
Affiliation(s)
- Cheng-Hua Wu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shuang Hu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Dan Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiao-Wen Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hui Ou-Yang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Guo-Fang Bi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Peng Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Feng-Ting Liang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wen-Hong Zhou
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiao Yang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jian-Hong Fang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hui-Chang Bi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
- The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen, 518000, China.
| |
Collapse
|
19
|
Qi F, Yi Z, Liu Y, Jia D, Zhao H, Jiang G, Gong J. CMTM4 promotes PD-L1-mediated macrophage apoptosis by enhancing STAT2 phosphorylation in sepsis. Exp Cell Res 2025; 447:114519. [PMID: 40122504 DOI: 10.1016/j.yexcr.2025.114519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Macrophage apoptosis is a key contributor to the elimination of immune cells and increased susceptibility during sepsis. CKLF like MARVEL transmembrane domain containing 4 (CMTM4) is a membrane protein with four transmembrane domains. It has recently been implicated in the regulation of immune cell biological functions. However, its role in regulating macrophage apoptosis during sepsis has not been extensively studied. METHODS Clinical samples were analyzed to determine CMTM4 expression levels and their correlation with clinical examination results. An in vitro model was developed using C57BL/6 mice and the THP-1 cell line. An immunofluorescence analysis was used to assess protein expression levels, apoptosis, and protein co-localization. Western blotting (WB) was used to measure protein expression levels, while flow cytometry was used to detect cell apoptosis. Transcriptomic sequencing was conducted to identify differentially expressed genes and to perform a functional enrichment analysis. Transcription factors were screened using databases. Chromatin immunoprecipitation, followed by quantitative PCR (ChIP-qPCR), was conducted to analyze protein-DNA interactions, and co-immunoprecipitation (Co-IP) was used to examine protein-protein interactions. RESULTS CMTM4 expression in macrophages was upregulated in sepsis. The inhibition of CMTM4 expression reduced macrophage apoptosis. PD-L1 was identified as a key molecule regulated by CMTM4 in macrophage apoptosis. CMTM4 regulates PD-L1 by promoting the phosphorylation of its transcription factor, STAT2, rather than directly binding to PD-L1. CONCLUSION In sepsis, CMTM4 facilitates PD-L1-dependent macrophage apoptosis by enhancing STAT2 phosphorylation. This discovery offers new insights for the diagnosis and treatment of sepsis.
Collapse
Affiliation(s)
- Feng Qi
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhujun Yi
- Department of Hepatobiliary Surgery, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Yan Liu
- Department of Hepatobiliary Surgery, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Degong Jia
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hui Zhao
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Gang Jiang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| | - Jianping Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
20
|
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
|
21
|
Brunsch V, Bergmann-Ewert W, Müller-Hilke B, Aleith J. Interleukin-6 overexpression and elevated granulocyte-to-lymphocyte ratio indicate hepatic stress in experimental group a Streptococcus sepsis. Med Microbiol Immunol 2025; 214:17. [PMID: 40178612 PMCID: PMC11968515 DOI: 10.1007/s00430-025-00826-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/14/2025] [Indexed: 04/05/2025]
Abstract
Group A Streptococcus (GAS) is a pathogen that is capable of colonizing various infection sites and can potentially elicit an inadequate immune response that will lead to sepsis. The processes underlying this misdirected immune reaction remain poorly understood, and reliable biomarkers for indicating impending organ failure during sepsis are still missing. The present study aims to identify parameters that can predict the onset of end-organ damage in the course of sepsis. To that extent, we investigated key aspects of the immune response in early-phase sepsis following infection of different tissues in a mouse model, using Brefeldin A to link cytokine production to specific cell types through multi-parameter flow cytometry. Subcutaneous and intravenous GAS infections resulted in clinical sepsis, which was paralleled by peripheral blood lymphopenia. Intravenous infection in particular was associated with a higher bacterial burden in the liver that strongly correlated with an increased granulocyte-to-lymphocyte ratio of the peripheral blood. Strikingly, IL-6 overexpression was more pronounced in intravenous infection and strongly correlated with hepatic stress, indicated by elevated bacterial loads in the liver. Collectively, our data highlight the potential utility of IL-6 in conjunction with an elevated granulocyte-to-lymphocyte ratio as promising early indicators of concomitant liver stress in sepsis.
Collapse
Affiliation(s)
- Valerie Brunsch
- Core Facility for Cell Sorting and Cell Analysis, Rostock University Medical Center, Rostock, Germany
| | - Wendy Bergmann-Ewert
- Core Facility for Cell Sorting and Cell Analysis, Rostock University Medical Center, Rostock, Germany
| | - Brigitte Müller-Hilke
- Core Facility for Cell Sorting and Cell Analysis, Rostock University Medical Center, Rostock, Germany
| | - Johann Aleith
- Core Facility for Cell Sorting and Cell Analysis, Rostock University Medical Center, Rostock, Germany.
| |
Collapse
|
22
|
Shao J, Wang T, Tang C, Yu J, Chen Y, Guo X, Wang H, Zhou L, Zhang G, Li Y, Yu H, Zheng R. The chemokine receptor type 5 inhibitor maraviroc alleviates sepsis-associated liver injury by regulating MAPK/NF-κB signaling. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3655-3666. [PMID: 39352530 PMCID: PMC11978715 DOI: 10.1007/s00210-024-03477-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/19/2024] [Indexed: 04/10/2025]
Abstract
Sepsis-related organ damage, as the most intractable problem in intensive care units (ICUs), receives a great deal of attention from healthcare professionals. Sepsis-associated liver injury (SALI) often leads to poor clinical outcomes due to its complex physiological mechanism. In previous studies, chemokine receptor 5 (CCR5) inhibitors were shown to exert unique anti-inflammatory effects. As the therapeutic effect of maraviroc (MVC) on SALI is still unclear, we aimed to explore whether MVC is effective in treating SALI. We established a model of SALI by cecal ligation and puncture (CLP) and intraperitoneally injected 20 mg/kg MVC 2 h after CLP. The results showed that MVC could significantly ameliorate liver injury after CLP. Furthermore, we demonstrated that MVC reduced inflammatory infiltration and apoptosis after SALI. In addition, we found that the function of MVC in reducing inflammation was obtained through the inhibition of the two inflammatory signaling pathways mentioned above. Finally, the JNK agonist AN was chosen for reverse research. As shown by the results, the therapeutic effects of MVC disappeared after AN treatment, indicating that MVC exerted anti-inflammatory and antiapoptotic effects through JNK. Our study revealed that MVC could reduce liver injury after SALI by inhibiting liver inflammation and hepatocyte apoptosis induced by CLP and that MVC exerted diminish inflammatory effects by inhibiting the NF-κB and MAPK signaling pathways.
Collapse
Affiliation(s)
- Jun Shao
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
- Department of Center for Cardiac Macrovascular Disease, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Tianwei Wang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
- Department of Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Chengbin Tang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
- Department of Center for Cardiac Macrovascular Disease, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Jiangquan Yu
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
- Department of Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Ying Chen
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
- Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, 225009, China
| | - Xin Guo
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
| | - Haoran Wang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
- Department of Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Lulu Zhou
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
- Department of Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Guozhong Zhang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
- Department of Center for Cardiac Macrovascular Disease, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Yuping Li
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China
- Department of Neuro Intensive Care Unit, Clinical Medical College of Yangzhou University, Yangzhou, 225001, China
| | - Hailong Yu
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China.
- Department of Neuro Intensive Care Unit, Clinical Medical College of Yangzhou University, Yangzhou, 225001, China.
- Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, 225009, China.
| | - Ruiqiang Zheng
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225009, China.
- Department of Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, 225001, China.
| |
Collapse
|
23
|
Ye L, Wang L, Kuang G, Zhang Z, Peng Q, He M, Fan J. IL-27 aggravates acute hepatic injury by promoting macrophage M1 polarization to induce Caspase-11 mediated Pyroptosis in vitro and in vivo. Cytokine 2025; 188:156881. [PMID: 39913960 DOI: 10.1016/j.cyto.2025.156881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 01/24/2025] [Accepted: 02/01/2025] [Indexed: 03/01/2025]
Abstract
OBJECTIVES Our aim was to explore the IL-27 effect in sepsis (SP)-related acute hepatic injury (AHI) as well as its possible mechanism. MATERIALS AND METHODS Herein, we utilized both wild-type (WT) and IL-27 receptor (WSX-1)-deficient (IL-27R-/-) mice alongside RAW264.7 cells. Our study established an SP-associated AHI model through the intraperitoneal injections of lipopolysaccharide (LPS) + D-galactosamine (D-G). For examining the IL-27 impact on AHI, mice serum and liver tissue samples were gathered. Inflammatory factor levels in the liver and serum were detected using ELISA and immunohistochemistry. Immunofluorescence and Western blot techniques were employed for the detection of protein expression associated with polarization and pyroptosis in the liver, including iNOS, ARG-1, caspase-11, RAGE, and GSDMD. To further verify the IL-27 effects on macrophage polarization and pyroptosis and explore possible mechanisms involved, we used LPS-triggered RAW264.7 macrophages to assess AMPK/SIRT1 expression after IL-27 intervention. This study utilized Compound C (CC) to block the AMPK/SIRT1 pathway. The inflammatory response level and protein expression related to macrophage polarization and pyroptosis were measured again to reveal IL-27 implication in AHI and determine whether its role is associated with the AMPK/SIRT1 pathway. RESULTS The results revealed that IL-27 exacerbated systemic inflammation and liver damage in AHI mice by promoting M1 macrophage polarization, thereby increasing pro-inflammatory phenotype macrophages (M1). This further exacerbated the inflammatory response and pyroptosis in vivo and in vitro. Additionally, IL-27 down-regulated p-AMPK and SIRT1 protein expression while overexpressing macrophage inflammatory mediators including IL-1β/6 and TNFα. Furthermore, IL-27 promoted increased RAGE and caspase-11 protein expression, aggravating macrophage pyroptosis. Employing CC to block the AMPK pathway further aggravated M1 macrophage polarization and pyroptosis in vitro and in vivo, ultimately worsening liver injury. CONCLUSIONS Here, IL-27 aggravates AHI by promoting macrophage M1 polarization to induce caspase-11-mediated pyroptosis in vitro and in vivo, which may be linked to the AMPK/SIRT1 signaling pathway.
Collapse
Affiliation(s)
- Lin Ye
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing City 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical Universit, Chongqing City, China
| | - Liuyang Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing City 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical Universit, Chongqing City, China
| | - Gang Kuang
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical Universit, Chongqing City, China; Department of Critical Care Medicine, Affiliated Dazu's Hospital of Chongqing Medical University, No. 1073, The second Ring South Road, Tangxiang Street, DaZu District, Chongqing City 402360, China
| | - Zhijiao Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing City 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical Universit, Chongqing City, China
| | - Qiaozhi Peng
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing City 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical Universit, Chongqing City, China
| | - Miao He
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Jing Fan
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing City 400016, China.
| |
Collapse
|
24
|
Xu X, Yang T, An J, Li B, Dou Z. Liver injury in sepsis: manifestations, mechanisms and emerging therapeutic strategies. Front Immunol 2025; 16:1575554. [PMID: 40226624 PMCID: PMC11985447 DOI: 10.3389/fimmu.2025.1575554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
Sepsis is defined as a condition related to infection that manifests with multiorgan dysfunction, representing a life-threatening state. Consequently, severe complications frequently occur, with liver injury being one of the most prevalent serious complications of sepsis. Liver dysfunction during sepsis serves as an independent predictor of mortality. This review provides a comprehensive overview of current research on sepsis-induced liver injury (SILI), encompassing the clinical manifestations, diagnostic criteria, pathogenesis and therapeutic strategies associated with this condition. SILI may manifest as hypoxic hepatitis due to ischemia and shock, cholestasis resulting from abnormal bile metabolism, or bile duct sclerosis. The pathophysiology of sepsis involves intricate interactions among the inflammatory response, oxidative stress, and cell death. All of these factors complicate treatment and represent potential targets for therapeutic intervention. Furthermore, this review addresses the limitations inherent in conventional therapies currently employed for managing SILI and emphasizes the potential of novel targeted strategies aimed at addressing the fundamental mechanisms underlying this condition.
Collapse
Affiliation(s)
- Xinqi Xu
- The First Hospital of Lanzhou University & The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Tingyu Yang
- The First Hospital of Lanzhou University & The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Jiapan An
- The First Hospital of Lanzhou University & The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Bin Li
- The First Hospital of Lanzhou University & The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Department of Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zhimin Dou
- The First Hospital of Lanzhou University & The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Department of Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
25
|
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
|
26
|
Shime N, Nakada TA, Yatabe T, Yamakawa K, Aoki Y, Inoue S, Iba T, Ogura H, Kawai Y, Kawaguchi A, Kawasaki T, Kondo Y, Sakuraya M, Taito S, Doi K, Hashimoto H, Hara Y, Fukuda T, Matsushima A, Egi M, Kushimoto S, Oami T, Kikutani K, Kotani Y, Aikawa G, Aoki M, Akatsuka M, Asai H, Abe T, Amemiya Y, Ishizawa R, Ishihara T, Ishimaru T, Itosu Y, Inoue H, Imahase H, Imura H, Iwasaki N, Ushio N, Uchida M, Uchi M, Umegaki T, Umemura Y, Endo A, Oi M, Ouchi A, Osawa I, Oshima Y, Ota K, Ohno T, Okada Y, Okano H, Ogawa Y, Kashiura M, Kasugai D, Kano KI, Kamidani R, Kawauchi A, Kawakami S, Kawakami D, Kawamura Y, Kandori K, Kishihara Y, Kimura S, Kubo K, Kuribara T, Koami H, Koba S, Sato T, Sato R, Sawada Y, Shida H, Shimada T, Shimizu M, Shimizu K, Shiraishi T, Shinkai T, Tampo A, Sugiura G, Sugimoto K, Sugimoto H, Suhara T, Sekino M, Sonota K, Taito M, Takahashi N, Takeshita J, Takeda C, Tatsuno J, Tanaka A, Tani M, Tanikawa A, Chen H, Tsuchida T, Tsutsumi Y, Tsunemitsu T, Deguchi R, Tetsuhara K, Terayama T, Togami Y, et alShime N, Nakada TA, Yatabe T, Yamakawa K, Aoki Y, Inoue S, Iba T, Ogura H, Kawai Y, Kawaguchi A, Kawasaki T, Kondo Y, Sakuraya M, Taito S, Doi K, Hashimoto H, Hara Y, Fukuda T, Matsushima A, Egi M, Kushimoto S, Oami T, Kikutani K, Kotani Y, Aikawa G, Aoki M, Akatsuka M, Asai H, Abe T, Amemiya Y, Ishizawa R, Ishihara T, Ishimaru T, Itosu Y, Inoue H, Imahase H, Imura H, Iwasaki N, Ushio N, Uchida M, Uchi M, Umegaki T, Umemura Y, Endo A, Oi M, Ouchi A, Osawa I, Oshima Y, Ota K, Ohno T, Okada Y, Okano H, Ogawa Y, Kashiura M, Kasugai D, Kano KI, Kamidani R, Kawauchi A, Kawakami S, Kawakami D, Kawamura Y, Kandori K, Kishihara Y, Kimura S, Kubo K, Kuribara T, Koami H, Koba S, Sato T, Sato R, Sawada Y, Shida H, Shimada T, Shimizu M, Shimizu K, Shiraishi T, Shinkai T, Tampo A, Sugiura G, Sugimoto K, Sugimoto H, Suhara T, Sekino M, Sonota K, Taito M, Takahashi N, Takeshita J, Takeda C, Tatsuno J, Tanaka A, Tani M, Tanikawa A, Chen H, Tsuchida T, Tsutsumi Y, Tsunemitsu T, Deguchi R, Tetsuhara K, Terayama T, Togami Y, Totoki T, Tomoda Y, Nakao S, Nagasawa H, Nakatani Y, Nakanishi N, Nishioka N, Nishikimi M, Noguchi S, Nonami S, Nomura O, Hashimoto K, Hatakeyama J, Hamai Y, Hikone M, Hisamune R, Hirose T, Fuke R, Fujii R, Fujie N, Fujinaga J, Fujinami Y, Fujiwara S, Funakoshi H, Homma K, Makino Y, Matsuura H, Matsuoka A, Matsuoka T, Matsumura Y, Mizuno A, Miyamoto S, Miyoshi Y, Murata S, Murata T, Yakushiji H, Yasuo S, Yamada K, Yamada H, Yamamoto R, Yamamoto R, Yumoto T, Yoshida Y, Yoshihiro S, Yoshimura S, Yoshimura J, Yonekura H, Wakabayashi Y, Wada T, Watanabe S, Ijiri A, Ugata K, Uda S, Onodera R, Takahashi M, Nakajima S, Honda J, Matsumoto T. The Japanese Clinical Practice Guidelines for Management of Sepsis and Septic Shock 2024. J Intensive Care 2025; 13:15. [PMID: 40087807 PMCID: PMC11907869 DOI: 10.1186/s40560-025-00776-0] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 01/21/2025] [Indexed: 03/17/2025] Open
Abstract
The 2024 revised edition of the Japanese Clinical Practice Guidelines for Management of Sepsis and Septic Shock (J-SSCG 2024) is published by the Japanese Society of Intensive Care Medicine and the Japanese Association for Acute Medicine. This is the fourth revision since the first edition was published in 2012. The purpose of the guidelines is to assist healthcare providers in making appropriate decisions in the treatment of sepsis and septic shock, leading to improved patient outcomes. We aimed to create guidelines that are easy to understand and use for physicians who recognize sepsis and provide initial management, specialized physicians who take over the treatment, and multidisciplinary healthcare providers, including nurses, physical therapists, clinical engineers, and pharmacists. The J-SSCG 2024 covers the following nine areas: diagnosis of sepsis and source control, antimicrobial therapy, initial resuscitation, blood purification, disseminated intravascular coagulation, adjunctive therapy, post-intensive care syndrome, patient and family care, and pediatrics. In these areas, we extracted 78 important clinical issues. The GRADE (Grading of Recommendations Assessment, Development and Evaluation) method was adopted for making recommendations, and the modified Delphi method was used to determine recommendations by voting from all committee members. As a result, 42 GRADE-based recommendations, 7 good practice statements, and 22 information-to-background questions were created as responses to clinical questions. We also described 12 future research questions.
Collapse
Affiliation(s)
- Nobuaki Shime
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Taka-Aki Nakada
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tomoaki Yatabe
- Emergency Department, Nishichita General Hospital, Tokai, Japan
| | - Kazuma Yamakawa
- Department of Emergency and Critical Care Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Yoshitaka Aoki
- Department of Anesthesiology and Intensive Care Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Shigeaki Inoue
- Department of Emergency and Critical Care Medicine, Wakayama Medical University, Wakayama, Japan
| | - Toshiaki Iba
- Department of Emergency and Disaster Medicine, Juntendo University, Tokyo, Japan
| | - Hiroshi Ogura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yusuke Kawai
- Department of Nursing, Fujita Health University Hospital, Toyoake, Japan
| | - Atsushi Kawaguchi
- Division of Pediatric Critical Care, Department of Pediatrics, School of Medicine, St. Marianna University, Kawasaki, Japan
| | - Tatsuya Kawasaki
- Department of Pediatric Critical Care, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Yutaka Kondo
- Department of Emergency and Critical Care Medicine, Juntendo University, Urayasu Hospital, Urayasu, Japan
| | - Masaaki Sakuraya
- Department of Emergency and Intensive Care Medicine, JA Hiroshima General Hospital, Hatsukaichi, Japan
| | - Shunsuke Taito
- Division of Rehabilitation, Department of Clinical Practice and Support, Hiroshima University Hospital, Hiroshima, Japan
| | - Kent Doi
- Department of Emergency and Critical Care Medicine, The University of Tokyo, Tokyo, Japan
| | - Hideki Hashimoto
- Department of Infectious Diseases, Hitachi Medical Education and Research Center University of Tsukuba Hospital, Hitachi, Japan
| | - Yoshitaka Hara
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| | - Tatsuma Fukuda
- Department of Emergency and Critical Care Medicine, Toranomon Hospital, Tokyo, Japan
| | - Asako Matsushima
- Department of Emergency and Critical Care, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Moritoki Egi
- Department of Anesthesia and Intensive Care, Kyoto University Hospital, Kyoto, Japan
| | - Shigeki Kushimoto
- Division of Emergency and Critical Care Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takehiko Oami
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Kazuya Kikutani
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Yuki Kotani
- Department of Intensive Care Medicine Kameda Medical Center, Kamogawa, Japan
| | - Gen Aikawa
- Department of Adult Health Nursing, College of Nursing, Ibaraki Christian University, Hitachi, Japan
| | - Makoto Aoki
- Division of Traumatology, National Defense Medical College Research Institute, Tokorozawa, Japan
| | - Masayuki Akatsuka
- Department of Intensive Care Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hideki Asai
- Department of Emergency and Critical Care Medicine, Nara Medical University, Nara, Japan
| | - Toshikazu Abe
- Department of Emergency and Critical Care Medicine, Tsukuba Memorial Hospital, Tsukuba, Japan
| | - Yu Amemiya
- Department of Emergency and Critical Care Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Ryo Ishizawa
- Department of Critical Care and Emergency Medicine, Tokyo Metropolitan Tama Medical Center, Tokyo, Japan
| | - Tadashi Ishihara
- Department of Emergency and Critical Care Medicine, Juntendo University, Urayasu Hospital, Urayasu, Japan
| | - Tadayoshi Ishimaru
- Department of Emergency Medicine, Chiba Kaihin Municipal Hospital, Chiba, Japan
| | - Yusuke Itosu
- Department of Anesthesiology, Hokkaido University Hospital, Sapporo, Japan
| | - Hiroyasu Inoue
- Division of Physical Therapy, Department of Rehabilitation, Showa University School of Nursing and Rehabilitation Sciences, Yokohama, Japan
| | - Hisashi Imahase
- Division of Intensive Care, Department of Anesthesiology and Intensive Care Medicine, Jichi Medical University School of Medicine, Shimotsuke, Japan
| | - Haruki Imura
- Department of Infectious Diseases, Rakuwakai Otowa Hospital, Kyoto, Japan
| | - Naoya Iwasaki
- Department of Anesthesiology and Intensive Care Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Noritaka Ushio
- Department of Emergency and Critical Care Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Masatoshi Uchida
- Department of Emergency and Critical Care Medicine, Dokkyo Medical University, Tochigi, Japan
| | - Michiko Uchi
- National Hospital Organization Ibarakihigashi National Hospital, Naka-Gun, Japan
| | - Takeshi Umegaki
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan
| | - Yutaka Umemura
- Division of Trauma and Surgical Critical Care, Osaka General Medical Center, Osaka, Japan
| | - Akira Endo
- Department of Acute Critical Care Medicine, Tsuchiura Kyodo General Hospital, Tsuchiura, Japan
| | - Marina Oi
- Department of Emergency and Critical Care Medicine, Kitasato University School of Medicine, Sagamihara, Japan
| | - Akira Ouchi
- Department of Adult Health Nursing, College of Nursing, Ibaraki Christian University, Hitachi, Japan
| | - Itsuki Osawa
- Department of Emergency and Critical Care Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Kohei Ota
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Takanori Ohno
- Department of Emergency and Crical Care Medicine, Shin-Yurigaoka General Hospital, Kawasaki, Japan
| | - Yohei Okada
- Department of Preventive Services, Kyoto University, Kyoto, Japan
| | - Hiromu Okano
- Department of Critical Care Medicine, St. Luke's International Hospital, Tokyo, Japan
| | - Yoshihito Ogawa
- Division of Trauma and Surgical Critical Care, Osaka General Medical Center, Osaka, Japan
| | - Masahiro Kashiura
- Department of Emergency and Critical Care Medicine, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Daisuke Kasugai
- Department of Emergency and Critical Care Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ken-Ichi Kano
- Department of Emergency Medicine, Fukui Prefectural Hospital, Fukui, Japan
| | - Ryo Kamidani
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Akira Kawauchi
- Department of Critical Care and Emergency Medicine, Japanese Red Cross Maebashi Hospital, Maebashi, Japan
| | - Sadatoshi Kawakami
- Department of Anesthesiology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Daisuke Kawakami
- Department of Intensive Care Medicine, Aso Iizuka Hospital, Iizuka, Japan
| | - Yusuke Kawamura
- Department of Rehabilitation, Showa General Hospital, Tokyo, Japan
| | - Kenji Kandori
- Department of Emergency and Critical Care Medicine, Japanese Red Cross Society Kyoto Daini Hospital , Kyoto, Japan
| | - Yuki Kishihara
- Department of Emergency and Critical Care Medicine, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Sho Kimura
- Department of Pediatric Critical Care Medicine, Tokyo Women's Medical University Yachiyo Medical Center, Yachiyo, Japan
| | - Kenji Kubo
- Department of Emergency Medicine, Japanese Red Cross Wakayama Medical Center, Wakayama, Japan
- Department of Infectious Diseases, Japanese Red Cross Wakayama Medical Center, Wakayama, Japan
| | - Tomoki Kuribara
- Department of Acute and Critical Care Nursing, School of Nursing, Sapporo City University, Sapporo, Japan
| | - Hiroyuki Koami
- Department of Emergency and Critical Care Medicine, Saga University, Saga, Japan
| | - Shigeru Koba
- Department of Critical Care Medicine, Nerima Hikarigaoka Hospital, Nerima, Japan
| | - Takehito Sato
- Department of Anesthesiology, Nagoya University Hospital, Nagoya, Japan
| | - Ren Sato
- Department of Nursing, Tokyo Medical University Hospital, Shinjuku, Japan
| | - Yusuke Sawada
- Department of Emergency Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Haruka Shida
- Data Science, Medical Division, AstraZeneca K.K, Osaka, Japan
| | - Tadanaga Shimada
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Motohiro Shimizu
- Department of Intensive Care Medicine, Ryokusen-Kai Yonemori Hospital, Kagoshima, Japan
| | | | | | - Toru Shinkai
- The Advanced Emergency and Critical Care Center, Mie University Hospital, Tsu, Japan
| | - Akihito Tampo
- Department of Emergency Medicine, Asahiakwa Medical University, Asahikawa, Japan
| | - Gaku Sugiura
- Department of Critical Care and Emergency Medicine, Japanese Red Cross Maebashi Hospital, Maebashi, Japan
| | - Kensuke Sugimoto
- Department of Anesthesiology and Intensive Care, Gunma University, Maebashi, Japan
| | - Hiroshi Sugimoto
- Department of Internal Medicine, National Hospital Organization Kinki-Chuo Chest Medical Center, Osaka, Japan
| | - Tomohiro Suhara
- Department of Anesthesiology, Keio University School of Medicine, Shinjuku, Japan
| | - Motohiro Sekino
- Department of Anesthesiology and Intensive Care Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kenji Sonota
- Department of Intensive Care Medicine, Miyagi Children's Hospital, Sendai, Japan
| | - Mahoko Taito
- Department of Nursing, Hiroshima University Hospital, Hiroshima, Japan
| | - Nozomi Takahashi
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jun Takeshita
- Department of Anesthesiology, Osaka Women's and Children's Hospital, Izumi, Japan
| | - Chikashi Takeda
- Department of Anesthesia and Intensive Care, Kyoto University Hospital, Kyoto, Japan
| | - Junko Tatsuno
- Department of Nursing, Kokura Memorial Hospital, Kitakyushu, Japan
| | - Aiko Tanaka
- Department of Intensive Care, University of Fukui Hospital, Fukui, Japan
| | - Masanori Tani
- Division of Critical Care Medicine, Saitama Children's Medical Center, Saitama, Japan
| | - Atsushi Tanikawa
- Division of Emergency and Critical Care Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hao Chen
- Department of Pulmonary, Yokohama City University Hospital, Yokohama, Japan
| | - Takumi Tsuchida
- Department of Anesthesiology, Hokkaido University Hospital, Sapporo, Japan
| | - Yusuke Tsutsumi
- Department of Emergency Medicine, National Hospital Organization Mito Medical Center, Ibaragi, Japan
| | | | - Ryo Deguchi
- Department of Traumatology and Critical Care Medicine, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Kenichi Tetsuhara
- Department of Critical Care Medicine, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Takero Terayama
- Department of Emergency Self-Defense, Forces Central Hospital, Tokyo, Japan
| | - Yuki Togami
- Department of Acute Medicine & Critical Care Medical Center, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Takaaki Totoki
- Department of Anesthesiology, Kyushu University Beppu Hospital, Beppu, Japan
| | - Yoshinori Tomoda
- Laboratory of Clinical Pharmacokinetics, Research and Education Center for Clinical Pharmacy, Kitasato University School of Pharmacy, Tokyo, Japan
| | - Shunichiro Nakao
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiroki Nagasawa
- Department of Acute Critical Care Medicine, Shizuoka Hospital Juntendo University, Shizuoka, Japan
| | | | - Nobuto Nakanishi
- Department of Disaster and Emergency Medicine, Kobe University, Kobe, Japan
| | - Norihiro Nishioka
- Department of Emergency and Crical Care Medicine, Shin-Yurigaoka General Hospital, Kawasaki, Japan
| | - Mitsuaki Nishikimi
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Satoko Noguchi
- Department of Anesthesiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Suguru Nonami
- Department of Emergency and Critical Care Medicine, Kyoto Katsura Hospital, Kyoto, Japan
| | - Osamu Nomura
- Medical Education Development Center, Gifu University, Gifu, Japan
| | - Katsuhiko Hashimoto
- Department of Emergency and Intensive Care Medicine, Fukushima Medical University, Fukushima, Japan
| | - Junji Hatakeyama
- Department of Emergency and Critical Care Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Yasutaka Hamai
- Department of Preventive Services, Kyoto University, Kyoto, Japan
| | - Mayu Hikone
- Department of Emergency Medicine, Tokyo Metropolitan Bokutoh Hospital, Tokyo, Japan
| | - Ryo Hisamune
- Department of Emergency and Critical Care Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Tomoya Hirose
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ryota Fuke
- Department of Internal Medicine, IMS Meirikai Sendai General Hospital, Sendai, Japan
| | - Ryo Fujii
- Emergency Department, Ageo Central General Hospital, Ageo, Japan
| | - Naoki Fujie
- Department of Pharmacy, Osaka Psychiatric Medical Center, Hirakata, Japan
| | - Jun Fujinaga
- Emergency and Critical Care Center, Kurashiki Central Hospital, Kurashiki, Japan
| | - Yoshihisa Fujinami
- Department of Emergency Medicine, Kakogawa Central City Hospital, Kakogawa, Japan
| | - Sho Fujiwara
- Department of Emergency Medicine, Tokyo Hikifune Hospital, Tokyo, Japan
- Department of Infectious Diseases, Tokyo Hikifune Hospital, Tokyo, Japan
| | - Hiraku Funakoshi
- Department of Emergency and Critical Care Medicine, Tokyobay Urayasu Ichikawa Medical Center, Urayasu, Japan
| | - Koichiro Homma
- Department of Emergency and Critical Care Medicine, Keio University School of Medicine, Shinjuku, Japan
| | - Yuto Makino
- Department of Preventive Services, Kyoto University, Kyoto, Japan
| | - Hiroshi Matsuura
- Osaka Prefectural Nakakawachi Emergency and Critical Care Center, Higashiosaka, Japan
| | - Ayaka Matsuoka
- Department of Emergency and Critical Care Medicine, Saga University, Saga, Japan
| | - Tadashi Matsuoka
- Department of Emergency and Critical Care Medicine, Keio University School of Medicine, Shinjuku, Japan
| | - Yosuke Matsumura
- Department of Intensive Care, Chiba Emergency and Psychiatric Medical Center, Chiba, Japan
| | - Akito Mizuno
- Department of Anesthesia and Intensive Care, Kyoto University Hospital, Kyoto, Japan
| | - Sohma Miyamoto
- Department of Emergency and Critical Care Medicine, St. Luke's International Hospital, Chuo-Ku, Japan
| | - Yukari Miyoshi
- Department of Emergency and Critical Care Medicine, Juntendo University, Urayasu Hospital, Urayasu, Japan
| | - Satoshi Murata
- Division of Emergency Medicine, Hyogo Prefectural Kobe Children's Hospital, Kobe, Japan
| | - Teppei Murata
- Department of Cardiology Miyazaki Prefectural, Nobeoka Hospital, Nobeoka, Japan
| | | | | | - Kohei Yamada
- Department of Traumatology and Critical Care Medicine, National Defense Medical College Hospital, Saitama, Japan
| | - Hiroyuki Yamada
- Department of Primary Care and Emergency Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryo Yamamoto
- Department of Emergency and Critical Care Medicine, Keio University School of Medicine, Shinjuku, Japan
| | - Ryohei Yamamoto
- Center for Innovative Research for Communities and Clinical Excellence (CIRC2LE), Fukushima Medical University, Fukushima, Japan
| | - Tetsuya Yumoto
- Department of Emergency, Critical Care and Disaster Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yuji Yoshida
- Department of Anesthesia and Intensive Care, Kyoto University Hospital, Kyoto, Japan
| | - Shodai Yoshihiro
- Department of Pharmaceutical Services, Hiroshima University Hospital, Hiroshima, Japan
| | - Satoshi Yoshimura
- Department of Emergency Medicine, Rakuwakai Otowa Hospital, Kyoto, Japan
| | - Jumpei Yoshimura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiroshi Yonekura
- Department of Anesthesiology and Pain Medicine, Fujita Health University Bantane Hospital, Nagoya, Japan
| | - Yuki Wakabayashi
- Department of Nursing, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Takeshi Wada
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Shinichi Watanabe
- Department of Physical Therapy, Faculty of Rehabilitation Gifu, University of Health Science, Gifu, Japan
| | - Atsuhiro Ijiri
- Department of Traumatology and Critical Care Medicine, National Defense Medical College Hospital, Saitama, Japan
| | - Kei Ugata
- Department of Intensive Care Medicine, Matsue Red Cross Hospital, Matsue, Japan
| | - Shuji Uda
- Department of Anesthesia and Intensive Care, Kyoto University Hospital, Kyoto, Japan
| | - Ryuta Onodera
- Department of Preventive Services, Kyoto University, Kyoto, Japan
| | - Masaki Takahashi
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Satoshi Nakajima
- Department of Emergency Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Junta Honda
- Department of Emergency and Critical Care Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tsuguhiro Matsumoto
- Department of Anesthesia and Intensive Care, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
27
|
He J, Huang Z, Zou R. Andrographolide ameliorates sepsis-induced acute liver injury by attenuating endoplasmic reticulum stress through the FKBP1A-mediated NOTCH1/AK2 pathway. Cell Biol Toxicol 2025; 41:56. [PMID: 40053226 PMCID: PMC11889056 DOI: 10.1007/s10565-025-10007-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/20/2025] [Indexed: 03/10/2025]
Abstract
Andrographolide (AP) has been shown to possess anti-inflammatory activities. In this study, the impact of AP in sepsis-induced acute liver injury (ALI) and the molecules involved were dissected. FKBP1A was predicted to be the sole target protein of AP that was also differentially expressed in the GSE166868 dataset. AP induced the protein expression of FKBP1A and suppressed that of NOTCH1 in a dose-dependent manner. AP ameliorated ALI in mice induced by D-galactosamine and LPS and inhibited LPS-induced liver parenchymal cell injury in vitro. By contrast, the protective effect of AP was significantly lost after the knockdown of FKBP1A. As a positive control, the therapeutic effect of dexamethasone on ALI may be related to NOTCH1, which was not related to FKBP1A. NOTCH1 promoted AK2 transcription in liver parenchymal cells, and FKBP1A inhibited endoplasmic reticulum (ER) stress by impairing NOTCH1/AK2 signaling. Restoration of NOTCH1 significantly reversed the hepatoprotective effect of AP in ALI mice and LPS-induced liver parenchymal cell injury by activating the ER stress pathway. Therefore, AP-promoted FKBP1A expression inhibits ALI progression by blocking the NOTCH1/AK2-mediated ER pathway.
Collapse
Affiliation(s)
- Jiaqi He
- Department of General Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, P.R. China
| | - Zepeng Huang
- Department of Hepatobiliary Surgery, Hainan Cancer Hospital, Haikou, 570000, Hainan, P.R. China
| | - Rui Zou
- Department of Hepatobiliary Surgery, Hainan Cancer Hospital, Haikou, 570000, Hainan, P.R. China.
- Hainan Clinical Research Center for Liver Disease and Liver Critical Illness, Haikou, 570000, Hainan, P.R. China.
| |
Collapse
|
28
|
Wang J, Hu X, Cao S, Zhao Y, Chen M, Hua T, Yang M. Aspirin is associated with improved 30-day mortality in patients with sepsis-associated liver injury: a retrospective cohort study based on MIMIC IV database. Front Pharmacol 2025; 16:1514392. [PMID: 40103585 PMCID: PMC11913821 DOI: 10.3389/fphar.2025.1514392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 02/14/2025] [Indexed: 03/20/2025] Open
Abstract
Background Sepsis-associated liver injury (SALI) is a common complication in sepsis patients, significantly affecting their prognosis. Previous studies have shown that aspirin can improve the prognosis of septic patients. However, there is currently a lack of clinical evidence supporting the use of aspirin in the treatment of SALI. Therefore, we conducted this study to explore the association between the use of aspirin and the prognosis of patients with SALI. Methods The patients in this study were obtained from the Medical Information Mart for Intensive Care IV (MIMIC-IV) database, version 3.0. The primary outcome was 30-day all-cause mortality. Baseline characteristics between the aspirin and non-aspirin groups were balanced using propensity score matching (PSM). The Kaplan-Meier survival curve and Cox regression analysis were used to investigate the association between aspirin use and the prognosis of patients with SALI. Results Of 657 SALI patients in this study, 447 (68%) patients had not used aspirin during hospitalization, whereas 210 (32%) had. After PSM, the 30-day mortality was 33.1% in the non-aspirin group and 21% in the aspirin group, indicating a significantly reduced mortality risk in the aspirin group (HR, 0.57; 95% CI, 0.37-0.90; P = 0.016). Similarly, the results of the multivariable Cox regression analysis and inverse probability weighting (IPW) analysis showed that, compared to the non-aspirin group, the aspirin group had a significantly lower 30-day mortality risk (Multivariable Cox regression analysis: HR, 0.69; 95% CI, 0.48-0.99; P = 0.047; IPW: HR, 0.62; 95% CI, 0.43-0.89; P = 0.010). Conclusion Aspirin can reduce 30-day mortality in SALI patients, regardless of the dose or timing of administration. However, careful assessment based on individual differences is essential to ensure the safety and effectiveness of aspirin use.
Collapse
Affiliation(s)
- Jianbao Wang
- The Second Department of Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Laboratory of Cardiopulmonary Resuscitation and Critical Care, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xuemei Hu
- The Second Department of Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Laboratory of Cardiopulmonary Resuscitation and Critical Care, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Susu Cao
- The Second Department of Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Laboratory of Cardiopulmonary Resuscitation and Critical Care, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yiwen Zhao
- Laboratory of Cardiopulmonary Resuscitation and Critical Care, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mengting Chen
- The Second Department of Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Laboratory of Cardiopulmonary Resuscitation and Critical Care, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Tianfeng Hua
- The Second Department of Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Laboratory of Cardiopulmonary Resuscitation and Critical Care, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Min Yang
- The Second Department of Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Laboratory of Cardiopulmonary Resuscitation and Critical Care, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
29
|
Zhu J, Jin Z, Wang J, Wu Z, Xu T, Tong G, Shen E, Fan J, Jiang C, Wang J, Li X, Cong W, Lin L. FGF21 ameliorates septic liver injury by restraining proinflammatory macrophages activation through the autophagy/HIF-1α axis. J Adv Res 2025; 69:477-494. [PMID: 38599281 PMCID: PMC11954821 DOI: 10.1016/j.jare.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/26/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024] Open
Abstract
INTRODUCTION Sepsis, a systemic immune syndrome caused by severe trauma or infection, poses a substantial threat to the health of patients worldwide. The progression of sepsis is heavily influenced by septic liver injury, which is triggered by infection and cytokine storms, and has a significant impact on the tolerance and prognosis of septic patients. The objective of our study is to elucidate the biological role and molecular mechanism of fibroblast growth factor 21 (FGF21) in the process of sepsis. OBJECTIVES This study was undertaken in an attempt to elucidate the function and molecular mechanism of FGF21 in therapy of sepsis. METHODS Serum concentrations of FGF21 were measured in sepsis patients and septic mice. Liver injury was compared between mice FGF21 knockout (KO) mice and wildtype (WT) mice. To assess the therapeutic potential, recombinant human FGF21 was administered to septic mice. Furthermore, the molecular mechanism of FGF21 was investigated in mice with myeloid-cell specific HIF-1α overexpression mice (LyzM-CreDIO-HIF-1α) and myeloid-cell specific Atg7 knockout mice (Atg7△mye). RESULTS Serum level of FGF21 was significantly increased in sepsis patients and septic mice. Through the use of recombinant human FGF21 (rhFGF21) and FGF21 KO mice, we found that FGF21 mitigated septic liver injury by inhibiting the initiation and propagation of inflammation. Treatment with rhFGF21 effectively suppressed the activation of proinflammatory macrophages by promoting macroautophagy/autophagy degradation of hypoxia-inducible factor-1α (HIF-1α). Importantly, the therapeutic effect of rhFGF21 against septic liver injury was nullified in LyzM-CreDIO-HIF-1α mice and Atg7△mye mice. CONCLUSIONS Our findings demonstrate that FGF21 considerably suppresses inflammation upon septic liver injury through the autophagy/ HIF-1α axis.
Collapse
Affiliation(s)
- Junjie Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Zhouxiang Jin
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Jie Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Zhaohang Wu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Tianpeng Xu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Gaozan Tong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China
| | - Enzhao Shen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China
| | - Junfu Fan
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Chunhui Jiang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Jiaqi Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Xiaokun Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China; Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Weitao Cong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China; Haihe Laboratory of Cell Ecosystem, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Li Lin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China.
| |
Collapse
|
30
|
Liu D, Sun S, Qiao H, Xin Q, Zhou S, Li L, Song N, Zhang L, Chen Q, Tian F, Mu X, Zhang S, Zhang J, Guo M, Wang H, Zhang XD, Zhang R. Ce 12V 6 Clusters with Multi-Enzymatic Activities for Sepsis Treatment. Adv Healthc Mater 2025; 14:e2401581. [PMID: 39129228 DOI: 10.1002/adhm.202401581] [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: 04/29/2024] [Revised: 07/22/2024] [Indexed: 08/13/2024]
Abstract
Artificial enzymes, especially nanozymes, have attracted wide attention due to their controlled catalytic activity, selectivity, and stability. The rising Cerium-based nanozymes exhibit unique SOD-like activity, and Vanadium-based nanozymes always hold excellent GPx-like activity. However, most inflammatory diseases involve polymerase biocatalytic processes that require multi-enzyme activities. The nanocomposite can fulfill multi-enzymatic activity simultaneously, but large nanoparticles (>10 nm) cannot be excreted rapidly, leading to biosafety challenges. Herein, atomically precise Ce12V6 clusters with a size of 2.19 nm are constructed. The Ce12V6 clusters show excellent glutathione peroxidase (GPx) -like activity with a significantly lower Michaelis-Menten constant (Km, 0.0125 mM versus 0.03 mM of natural counterpart) and good activities mimic superoxide dismutase (SOD) and peroxidase (POD). The Ce12V6 clusters exhibit the ability to scavenge the ROS including O2 ·- and H2O2 via the cascade reactions of multi-enzymatic activities. Further, the Ce12V6 clusters modulate the proinflammatory cytokines including tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β) and consequently rescue the multi-organ failure in the lipopolysaccharide (LPS)-induced sepsis mouse model. With excellent biocompatibility, the Ce12V6 clusters show promise in the treatment of sepsis.
Collapse
Affiliation(s)
- Di Liu
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Si Sun
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Huanhuan Qiao
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Qi Xin
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin Third Central Hospital, Tianjin, 300170, China
| | - Sufei Zhou
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Lingxia Li
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Nan Song
- Department of Physics, School of Science, Tianjin Chengjian University, Tianjin, 300384, 18, China
| | - Lijie Zhang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Qi Chen
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Fangzhen Tian
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Xiaoyu Mu
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Shaofang Zhang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Jing Zhang
- Department of Cardiology Tianjin Chest Hospital, Tianjin University, Tianjin, 300222, China
| | - Meili Guo
- Department of Physics, School of Science, Tianjin Chengjian University, Tianjin, 300384, 18, China
| | - Hao Wang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Xiao-Dong Zhang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin, 300350, China
| | - Ruiping Zhang
- The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
31
|
Song Y, Maged Abdulsalam Mohammed Ali AM, Yang W, Sun L. Clinical characteristics and prognosis of patients with early sepsis-related liver injury in Northeast China. J Intensive Care Med 2025; 40:253-262. [PMID: 39175409 DOI: 10.1177/08850666241277512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Background: Sepsis-associated liver injury (SALI) occurs in about a third of septic patients, and it is often a poor prognostic factor. However, there are few studies on early SALI and its impact on the clinical course of sepsis. Here we explored the clinical characteristics, risk factors, and prognosis of early SALI. Methods: Two hundred and one patients with confirmed sepsis were divided into those with and without early SALI (on admission) based on liver function. The clinical characteristics and prognosis were compared between groups and associated factors identified by multivariable regression analysis. Results: Sepsis-related liver injury was present in 18.9% of septic patients on admission. High aspartate transaminase (AST), high direct bilirubin, and low plasma thromboplastin antecedent (PTA, factor XI) were risk factors for sepsis with SALI: the area under the AST curve was 0.825, corresponding to a sensitivity of 0.67 and a specificity of 0.93 (cutoff 91.6 U/L), the area under the direct bilirubin curve was 0.86, corresponding to a sensitivity of 0.83 and a specificity of 0.71 (cutoff 8.35 μmol/L), and the area under the PTA curve was 0.678, corresponding to a sensitivity of 0.47 and a specificity of 0.93 (cutoff 54.0). Conclusion: Septic patients with early SALI have early-onset coagulation disorders that must be recognized to instigate early intervention and halt sepsis progression. Elevated AST, PTA, and direct bilirubin may be independent risk markers of sepsis-related liver injury, and extra clinical vigilance is required when these factors are noted in patients with sepsis.
Collapse
Affiliation(s)
- Yan Song
- Department of Emergency Medicine, First Hospital of Jilin University, Changchun, China
| | | | - Weiying Yang
- Department of Emergency Medicine, First Hospital of Jilin University, Changchun, China
| | - Lichao Sun
- Department of Emergency Medicine, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
32
|
Tang J, Li X, Li W, Cao C. The Protective Effect of Octanoic Acid on Sepsis: A Review. Nutr Rev 2025; 83:e1270-e1285. [PMID: 39101596 DOI: 10.1093/nutrit/nuae106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/06/2024] Open
Abstract
Sepsis, a systemic inflammation that occurs in response to a bacterial infection, is a significant medical challenge. Research conducted over the past decade has indicated strong associations among a patient's nutritional status, the composition of their gut microbiome, and the risk, severity, and prognosis of sepsis. Octanoic acid (OA) plays a vital role in combating sepsis and has a protective effect on both animal models and human patients. In this discussion, the potential protective mechanisms of OA in sepsis, focusing on its regulation of the inflammatory response, immune system, oxidative stress, gastrointestinal microbiome and barrier function, metabolic disorders and malnutrition, as well as organ dysfunction are explored. A comprehensive understanding of the mechanisms by which OA act may pave the way for new preventive and therapeutic approaches to sepsis.
Collapse
Affiliation(s)
- Jiabao Tang
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Xiaohua Li
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou 215004, China
- Department of Thyroid and Breast Surgery, Suzhou Wuzhong People's Hospital, Suzhou 215004, China
| | - Wei Li
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Chun Cao
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| |
Collapse
|
33
|
Fang L, Song Y, Chen J, Ding Y. The dual role of neutrophils in sepsis-associated liver injury. Front Immunol 2025; 16:1538282. [PMID: 40092997 PMCID: PMC11906405 DOI: 10.3389/fimmu.2025.1538282] [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] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Sepsis is often accompanied by liver injury and is associated with an increase in the number of circulating and hepatic neutrophils. In sepsis-associated liver injury, neutrophils exhibit phenotypic heterogeneity and perform both pro- and anti-inflammatory functions. Moreover, neutrophil dysfunction and neutrophil-associated immunosuppression are also involved in the pathogenesis of sepsis. Given the complex functionality of this cell type, the aim of this review was to describe the possible mechanistic role of neutrophils in sepsis-associated liver injury, with a brief introduction to neutrophil recruitment and subsequent discussion of the potential contributions of neutrophils to different subtypes of sepsis-associated liver injury.
Collapse
Affiliation(s)
- Lexin Fang
- Department of Intensive Care Unit, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Xinhua Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yu Song
- Department of Hepatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Xinhua Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jiangtao Chen
- Department of Intensive Care Unit, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Xinhua Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yueping Ding
- Department of Intensive Care Unit, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Xinhua Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| |
Collapse
|
34
|
You Y, Li Y, Ye L, Xu F, Fan J. Interleukins-27 Aggravates Liver Injury by Impairing the Antimicrobial Response of Macrophages via the Promotion of Mitochondrial Dysfunction in the Context of Sepsis. Mediators Inflamm 2025; 2025:6608718. [PMID: 40046257 PMCID: PMC11882325 DOI: 10.1155/mi/6608718] [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: 06/25/2024] [Accepted: 02/01/2025] [Indexed: 05/13/2025] Open
Abstract
Background and Aims: Plasma interleukin (IL)-27 is an important mediator of acute hepatic injury (AHI) associated with sepsis. Mitochondria contribute to the proper regulation of macrophage phagocytosis. In this study, we investigated the effect of IL-27 on mitochondrial function and the antimicrobial response of macrophages in sepsis-associated AHI. Methods: Wild-type (WT) and IL-27 receptor WSX-1 deficient (IL-27R-/-) mice underwent cecal ligation and puncture (CLP). The severity of hepatic injury, inflammatory cytokine levels, hepatic pyroptosis, and bacterial load in the liver and blood were assessed 24 h after CLP. In vitro, RAW264.7 cells and peritoneal macrophages were treated with lipopolysaccharide (LPS) and/or IL-27. The phagocytosis and killing functions of macrophages were detected. Mitochondrial function and mitophagy were detected using western blot, glutathione (GSH)/malondialdehyde (MDA) content measurement, fluorescence staining, and JC-1 staining in vivo and in vitro. After treatment with nicotinamide mononucleotide (NMN, NAD + precursor), a pharmacologic agent that improves mitochondrial function, the inflammatory response, hepatic injury, and hepatic pyroptosis were assessed. Results: IL-27R-/- mice exhibited a marked reduction in hepatic injury, pyroptosis (based on cleaved GSDMD and cleaved Caspases 1 protein levels), and systemic inflammation (based on serum IL-6, IL-10, and TNF-α levels) compared to WT mice following CLP. After CLP, mice lacking IL-27R displayed significantly higher bacterial clearance and greater local infection control. Subsequent studies demonstrated that IL-27 directly impaired the LPS-induced bacterial phagocytosis, killing capacity, and mitochondrial function of macrophages. Finally, enhanced mitochondrial function using NMN in vivo significantly alleviated pathological liver injury and inflammation. Conclusions: These findings indicated that IL-27 impairs the bacterial phagocytosis capacity of macrophages by aggravating mitochondrial dysfunction to aggravate AHI during sepsis.
Collapse
Affiliation(s)
- Yuehua You
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuyan Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Critical Care Medicine, The First People's Hospital of Chongqing High-tech Zone, Chongqing, China
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Ye
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Xu
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Fan
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
35
|
Bæk O, Muk T, Wu Z, Ye Y, Khakimov B, Casano AM, Gangadharan B, Bilic I, Brunse A, Sangild PT, Nguyen DN. Altered hepatic metabolism mediates sepsis preventive effects of reduced glucose supply in infected preterm newborns. eLife 2025; 13:RP97830. [PMID: 39992703 PMCID: PMC11850001 DOI: 10.7554/elife.97830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025] Open
Abstract
Preterm infants are susceptible to neonatal sepsis, a syndrome of pro-inflammatory activity, organ damage, and altered metabolism following infection. Given the unique metabolic challenges and poor glucose regulatory capacity of preterm infants, their glucose intake during infection may have a high impact on the degree of metabolism dysregulation and organ damage. Using a preterm pig model of neonatal sepsis, we previously showed that a drastic restriction in glucose supply during infection protects against sepsis via suppression of glycolysis-induced inflammation, but results in severe hypoglycemia. Now we explored clinically relevant options for reducing glucose intake to decrease sepsis risk, without causing hypoglycemia and further explore the involvement of the liver in these protective effects. We found that a reduced glucose regime during infection increased survival via reduced pro-inflammatory response, while maintaining normoglycemia. Mechanistically, this intervention enhanced hepatic oxidative phosphorylation and possibly gluconeogenesis, and dampened both circulating and hepatic inflammation. However, switching from a high to a reduced glucose supply after the debut of clinical symptoms did not prevent sepsis, suggesting metabolic conditions at the start of infection are key in driving the outcome. Finally, an early therapy with purified human inter-alpha inhibitor protein, a liver-derived anti-inflammatory protein, partially reversed the effects of low parenteral glucose provision, likely by inhibiting neutrophil functions that mediate pathogen clearance. Our findings suggest a clinically relevant regime of reduced glucose supply for infected preterm infants could prevent or delay the development of sepsis in vulnerable neonates.
Collapse
Affiliation(s)
- Ole Bæk
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of CopenhagenCopenhagenDenmark
- Department of Neonatology, RigshospitaletCopenhagenDenmark
| | - Tik Muk
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of CopenhagenCopenhagenDenmark
| | - Ziyuan Wu
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of CopenhagenCopenhagenDenmark
| | - Yongxin Ye
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of CopenhagenCopenhagenDenmark
- Department of Food Science, University of CopenhagenCopenhagenDenmark
| | - Bekzod Khakimov
- Department of Food Science, University of CopenhagenCopenhagenDenmark
| | - Alessandra Maria Casano
- Plasma-derived therapies, Baxalta Innovations GmbH, part of Takeda Pharmaceuticals LtdViennaAustria
| | - Bagirath Gangadharan
- Plasma-derived therapies, Baxalta Innovations GmbH, part of Takeda Pharmaceuticals LtdViennaAustria
| | - Ivan Bilic
- Plasma-derived therapies, Baxalta Innovations GmbH, part of Takeda Pharmaceuticals LtdViennaAustria
| | - Anders Brunse
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of CopenhagenCopenhagenDenmark
| | - Per Torp Sangild
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of CopenhagenCopenhagenDenmark
- Department of Neonatology, RigshospitaletCopenhagenDenmark
| | - Duc Ninh Nguyen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of CopenhagenCopenhagenDenmark
| |
Collapse
|
36
|
Guo Y, Guo W, Chen H, Sun J, Yin Y. Mechanisms of sepsis-induced acute liver injury: a comprehensive review. Front Cell Infect Microbiol 2025; 15:1504223. [PMID: 40061452 PMCID: PMC11885285 DOI: 10.3389/fcimb.2025.1504223] [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: 09/30/2024] [Accepted: 01/31/2025] [Indexed: 05/13/2025] Open
Abstract
Sepsis is a severe, often life-threatening form of organ dysfunction that arises from an inappropriately regulated host response to infectious pathogen exposure. As the largest gland in the body, the liver serves as a regulatory hub for metabolic, immune, and detoxification activity. It is also an early sepsis target organ such that hepatic dysfunction is observed in 34-46% of patients with sepsis. The precise mechanisms that give rise to sepsis-induced liver injury, however, remain incompletely understood. Based on the research conducted to date, dysregulated systemic inflammation, microbial translocation, microcirculatory abnormalities, cell death, metabolic dysfunction, and liver inflammation may all contribute to the liver damage that can arise in the context of septicemia. This review was developed to provide an overview summarizing the potential mechanisms underlying sepsis-induced liver injury, informing the selection of potential targets for therapeutic intervention and providing a framework for the alleviation of patient symptoms and the improvement of prognostic outcomes.
Collapse
Affiliation(s)
- Yongjing Guo
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun, China
| | - Wanxu Guo
- Department of Neonate, The Second Hospital of Jilin University, Changchun, China
| | - Huimin Chen
- Department of Neonate, The Second Hospital of Jilin University, Changchun, China
| | - Jian Sun
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun, China
| | - Yongjie Yin
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
37
|
Feng L, Cui J, Chen W, Zhu L, Li P, Zhou H, Sun Y, Yi W. Nrg4 Secreted by Brown Adipose Tissue Suppresses Ferroptosis of Sepsis-Induced Liver Injury. Inflammation 2025:10.1007/s10753-024-02230-z. [PMID: 39956880 DOI: 10.1007/s10753-024-02230-z] [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: 10/16/2024] [Revised: 12/07/2024] [Accepted: 12/23/2024] [Indexed: 02/18/2025]
Abstract
Sepsis is a leading cause of death, with the liver being particularly vulnerable to sepsis-related injuries. This damage significantly contributes to disease progression, underscoring the need for new treatments. Brown adipose tissue (BAT) secretes various cytokines, including neuregulin 4 (Nrg4), which plays a protective role in hepatic glucose and lipid metabolism. Ferroptosis, a key type of cell death in sepsis-induced liver injury, has recently gained attention. This study aimed to investigate how BAT-secreted cytokines alleviate liver ferroptosis in sepsis. Septic liver injury was induced in the control and BAT group using cecal ligation and puncture (CLP) and lipopolysaccharide injections. BAT removal worsened ferroptosis; in contrast, CL316243 activation reduced it. These findings suggest that Nrg4 secretion following BAT activation protects the liver during sepsis by inhibiting ferroptosis. Future therapies targeting BAT activation and Nrg4 could potentially mitigate sepsis-induced liver damage, offering new insights into treatment strategies.
Collapse
Affiliation(s)
- Linqi Feng
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jun Cui
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wenlong Chen
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Lei Zhu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Panpan Li
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Haitao Zhou
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yang Sun
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| | - Wei Yi
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
38
|
Zhang X, Zhang W, Zhang H, Liao X. Sepsis subphenotypes: bridging the gaps in sepsis treatment strategies. Front Immunol 2025; 16:1546474. [PMID: 40013154 PMCID: PMC11862915 DOI: 10.3389/fimmu.2025.1546474] [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: 12/17/2024] [Accepted: 01/20/2025] [Indexed: 02/28/2025] Open
Abstract
Sepsis, a heterogeneous illness produced by a dysregulated host response to infection, remains a severe mortality risk. Recent discoveries in sepsis research have stressed phenotyping as a feasible strategy for tackling heterogeneity and enhancing therapy precision. Sepsis phenotyping has moved from traditional stratifications based on severity and prognosis to dynamic, phenotype-driven therapeutic options. This review covers recent progress in connecting sepsis subgroups to personalized treatments, with a focus on phenotype-based therapeutic predictions and decision-support systems. Despite ongoing challenges, such as standardizing phenotyping frameworks and incorporating findings into clinical practice, this topic has enormous promise. By investigating phenotypic variation in therapy responses, we hope to uncover new biomarkers and phenotype-driven therapeutic solutions, laying the groundwork for more effective therapies and, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei Zhang
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huan Zhang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xuelian Liao
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Critical Care Medicine, West China Tianfu Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
39
|
Xiong L, Beyer D, Liu N, Lehmann T, Neugebauer S, Schaeuble S, Sommerfeld O, Ernst P, Svensson CM, Nietzsche S, Scholl S, Bruns T, Gaßler N, Gräler MH, Figge MT, Panagiotou G, Bauer M, Press AT. Targeting protein kinase C-α prolongs survival and restores liver function in sepsis: Evidence from preclinical models. Pharmacol Res 2025; 212:107581. [PMID: 39761839 DOI: 10.1016/j.phrs.2025.107581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 01/26/2025]
Abstract
Sepsis is a life-threatening organ failure resulting from a poorly regulated infection response. Organ dysfunction includes hepatic involvement, weakening the immune system due to excretory liver failure, and metabolic dysfunction, increasing the death risk. Although experimental studies correlated excretory liver functionality with immune performance and survival rates in sepsis, the proteins and pathways involved remain unclear. This study identified protein kinase C-α (PKCα) as a novel target for managing excretory liver function during sepsis. Using a preclinical murine sepsis model, we found that both PKCα knockout and the use of a PKCα-inhibitor midostaurin successfully restored liver function without hindering the host's response or ability to clear the pathogen, highlighting PKCα's vital role in excretory liver failure. In septic animals, both approaches significantly boosted survival rates. Midostaurin is the clinically approved active pharmaceutical ingredient in Rydapt, approved for the adjuvant treatment of FTL3-mutated AML. Here, it reduced plasma bile acids and related inflammation in those patients, opening a translational avenue for therapeutics in sepsis. Conclusively, our research underscores the significance of PKCα in controlling excretory liver function during inflammation. This suggests that targeting this protein could restore liver function without compromising the immune system, thereby decreasing sepsis mortality and supporting the recent paradigm that the liver is a hub for the host response to infection that might, in the future, result in novel host-directed therapies supporting the current state-of-the-art intensive care medicine in patients with sepsis-associated liver failure.
Collapse
Affiliation(s)
- Ling Xiong
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany
| | - Dustin Beyer
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany
| | - Na Liu
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany
| | - Tina Lehmann
- Jena University Hospital, Electron Microscopy Center, Friedrich-Schiller-University Jena, Jena, Germany
| | - Sophie Neugebauer
- Jena University Hospital, Institute of Clinical Chemistry and Laboratory Diagnostics, Friedrich-Schiller-University Jena, Jena, Germany
| | - Sascha Schaeuble
- Department of Microbiome Dynamics at Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (Leibniz-HKI), Jena, Germany
| | - Oliver Sommerfeld
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany
| | - Philipp Ernst
- Jena University Hospital, Clinic for Internal Medicine II, Department of Hematology and Internal Oncology, Friedrich-Schiller-University Jena, Jena, Germany
| | - Carl-Magnus Svensson
- Research Group Applied Systems Biology at Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (Leibniz-HKI), Jena, Germany
| | - Sandor Nietzsche
- Jena University Hospital, Electron Microscopy Center, Friedrich-Schiller-University Jena, Jena, Germany
| | - Sebastian Scholl
- Jena University Hospital, Clinic for Internal Medicine II, Department of Hematology and Internal Oncology, Friedrich-Schiller-University Jena, Jena, Germany
| | - Tony Bruns
- University Hospital RWTH Aachen, Department of Medicine III, Aachen, Germany
| | - Nikolaus Gaßler
- Jena University Hospital, Section of Pathology, Institute of Forensic Medicine, Friedrich-Schiller-University Jena, Jena, Germany
| | - Markus H Gräler
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany
| | - Marc Thilo Figge
- Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany; Research Group Applied Systems Biology at Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (Leibniz-HKI), Jena, Germany; Friedrich-Schiller-University Jena, Institute of Microbiology, Faculty of Biological Sciences, Jena, Germany; Friedrich-Schiller-University Jena, Cluster of Excellence Balance of the Microverse, Jena, Germany
| | - Gianni Panagiotou
- Department of Microbiome Dynamics at Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (Leibniz-HKI), Jena, Germany; Friedrich-Schiller-University Jena, Institute of Microbiology, Faculty of Biological Sciences, Jena, Germany; Friedrich-Schiller-University Jena, Cluster of Excellence Balance of the Microverse, Jena, Germany
| | - Michael Bauer
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany
| | - Adrian T Press
- Jena University Hospital, Department of Anesthesiology and Intensive Care Medicine, Friedrich-Schiller-University Jena, Jena, Germany; Jena University Hospital, Center for Sepsis Control and Care, Friedrich-Schiller-University Jena, Jena, Germany; Friedrich-Schiller-University Jena, Faculty of Medicine, Jena, Germany.
| |
Collapse
|
40
|
Wang J, Tao X, Liu Z, Yan Y, Cheng P, Liu B, Du H, Niu B. Noncoding RNAs in sepsis-associated acute liver injury: Roles, mechanisms, and therapeutic applications. Pharmacol Res 2025; 212:107596. [PMID: 39800175 DOI: 10.1016/j.phrs.2025.107596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/11/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Sepsis is a life-threatening syndrome characterized by organ dysfunction caused by a dysregulated host response to infection. Sepsis-associated acute liver injury (SA-ALI) is a frequent and serious complication of sepsis that considerably impacts both short-term and long-term survival outcomes. In intensive care units (ICUs), the mortality rate of patients with SA-ALI remains high, mostly due to the absence of effective early diagnostic markers and suitable therapeutic strategies. Recent studies have demonstrated the importance of non-coding RNAs (ncRNAs) in the development and progression of SA-ALI. This review focuses on the critical roles of ncRNAs, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), in regulating "cytokine storms", oxidative stress, mitochondrial dysfunction, and programmed cell death in SA-ALI, and summarizes the current state and limitations of existing studies on lncRNAs and circRNAs in SA-ALI. By integrating advancements in high-throughput sequencing technologies, this review provides novel insights into the dual potential of ncRNAs as diagnostic biomarkers and therapeutic targets, offers new ideas for SA-ALI diagnosis and treatment research and highlights potential challenges in clinical translation.
Collapse
Affiliation(s)
- Jialian Wang
- Department of Intensive Care Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400016, China
| | - Xingyu Tao
- Department of Intensive Care Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400016, China
| | - Zhengyang Liu
- Department of Nephrology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing 400016, China
| | - Yuan Yan
- Department of Intensive Care Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400016, China
| | - Peifeng Cheng
- Department of Intensive Care Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400016, China
| | - Bin Liu
- Department of Intensive Care Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400016, China
| | - Huimin Du
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Bailin Niu
- Department of Intensive Care Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400016, China.
| |
Collapse
|
41
|
Wang B, Wu X, Cheng J, Ye J, Zhu H, Liu X. Regulatory role of S1P and its receptors in sepsis-induced liver injury. Front Immunol 2025; 16:1489015. [PMID: 39935473 PMCID: PMC11811114 DOI: 10.3389/fimmu.2025.1489015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 01/13/2025] [Indexed: 02/13/2025] Open
Abstract
As an immune and metabolic organ, the liver affects the progression and prognosis of sepsis. Despite the severe adverse effects of sepsis liver injury on the body, treatment options remain limited. Sphingosine-1-phosphate (S1P) is a widely distributed lipid signaling molecule that binds to five sphingosine-1-phosphate receptors (S1PR) to regulate downstream signaling pathways involved in the pathophysiological processes of sepsis, including endothelial permeability, cytokine release, and vascular tone. This review summarizes current research on the role of S1P in normal liver biology and describes the mechanisms by which changes in S1P/S1PR affect the development of liver-related diseases. At the same time, the pathological processes underlying liver injury, as evidenced by clinical manifestations during sepsis, were comprehensively reviewed. This paper focused on the mechanistic pathways through which S1P and its receptors modulate immunity, bile acid metabolism, and liver-intestinal circulation in septic liver injury. Finally, the relationships between S1P and its receptors with liver inflammation and metabolism and the use of related drugs for the treatment of liver injury were examined. By elucidating the role of S1P and its receptor in the pathogenesis of sepsis liver injury, this review established a molecular targeting framework, providing novel insights into clinical and drug development.
Collapse
Affiliation(s)
- Bin Wang
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xiaoyu Wu
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jiangfeng Cheng
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Junming Ye
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Clinical College, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Hongquan Zhu
- Department of Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Critical Care Medicine, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xiaofeng Liu
- Clinical College, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Emergency, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
42
|
Nie K, Kadier A, Jia S, Liu D, Tong N, Tuerxun M, Aisha A, Peng H, Zhuang Y, Chen Y, Jiang J, Sun M, Fang Y, Chen H. Comparative study of transcriptomic alterations in sepsis-induced acute liver injury: Deciphering the role of alternative splicing in mouse models. Int Immunopharmacol 2025; 146:113878. [PMID: 39719759 DOI: 10.1016/j.intimp.2024.113878] [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: 04/02/2024] [Revised: 12/09/2024] [Accepted: 12/15/2024] [Indexed: 12/26/2024]
Abstract
BACKGROUND Sepsis represents a critical health crisis often leading to the failure of multiple organs, with the liver playing a pivotal role in controlling inflammation and defending against systemic infections. The exacerbation of liver damage can escalate sepsis severity, underscoring the necessity to delve into the molecular mechanisms underlying sepsis-induced acute liver injury (ALI). The role of alternative splicing (AS), a complex post-transcriptional mechanism, has been occasionally noted in relation to sepsis across different investigations. OBJECTIVE This research aimed to provide an extensive analysis of gene expression and alternative splicing variants in sepsis-induced ALI using mouse models, thus broadening the understanding of gene-level modulations during sepsis and pinpointing potential therapeutic targets. METHODS We employed mouse models of ALI induced via both cecal ligation and puncture (CLP) and lipopolysaccharides (LPS). An extensive evaluation was conducted to identify variances in gene expression and the occurrence of alternative splicing variants within the liver tissues of mice afflicted with sepsis. RESULTS The results of our study revealed significant alterations in the regulation of genes associated with RNA splicing and numerous pathways related to inflammation following exposure to CLP and LPS. We identified a total of 170 genes exhibiting both differential expression and splicing variations within the groups subjected to CLP and LPS interventions. Four key genes were specifically identified and validated, emphasizing their potential as treatment targets for ALI in sepsis. Among them, Nop58 was found to play a dual role in inflammation regulation, with intron retention linked to pro-inflammatory responses, while its full-length splicing variant exhibited anti-inflammatory properties. Furthermore, our data highlighted the potential role of specific splicing factors, such as Rbm3, Plrg1, and Snip1, in sepsis-induced liver abnormalities. CONCLUSION This study offers a comprehensive insight into the role of AS in sepsis-induced ALI, laying the groundwork for future therapeutic interventions. By demonstrating the functional relevance of specific splicing events, such as those involving Nop58, this work underscores the potential of targeting splicing mechanisms as innovative strategies to mitigate sepsis-induced liver injuries.
Collapse
Affiliation(s)
- Ke Nie
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China
| | - Aimaitijiang Kadier
- Department of Emergency, Kashi Prefecture Second People's Hospital, Kashi 844000, Xinjiang, China
| | - Shaohua Jia
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China
| | - Dan Liu
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China
| | - Nana Tong
- Department of Emergency, Kashi Prefecture Second People's Hospital, Kashi 844000, Xinjiang, China
| | - Muzaipaier Tuerxun
- Department of Emergency, Kashi Prefecture Second People's Hospital, Kashi 844000, Xinjiang, China
| | - Aisikaer Aisha
- Department of Emergency, Kashi Prefecture Second People's Hospital, Kashi 844000, Xinjiang, China
| | - Hu Peng
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China
| | - Yugang Zhuang
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China
| | - Yuanzhuo Chen
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China
| | - Jinxia Jiang
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China
| | - Mingming Sun
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China.
| | - Yi Fang
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China.
| | - Huaguo Chen
- Department of Emergency, Kashi Prefecture Second People's Hospital, Kashi 844000, Xinjiang, China; Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China.
| |
Collapse
|
43
|
Kou Y, Tian Y, Ha Y, Wang S, Sun X, Lv S, Luo B, Yang Y, Qin L. Comprehensive Sepsis Risk Prediction in Leukemia Using a Random Forest Model and Restricted Cubic Spline Analysis. J Inflamm Res 2025; 18:1013-1032. [PMID: 39867945 PMCID: PMC11766288 DOI: 10.2147/jir.s505813] [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: 11/12/2024] [Accepted: 01/12/2025] [Indexed: 01/28/2025] Open
Abstract
Background Sepsis is a severe complication in leukemia patients, contributing to high mortality rates. Identifying early predictors of sepsis is crucial for timely intervention. This study aimed to develop and validate a predictive model for sepsis risk in leukemia patients using machine learning techniques. Methods This retrospective study included 4310 leukemia patients admitted to the Affiliated Hospital of Guangdong Medical University from 2005 to 2024, using 70% for training and 30% for validation. Feature selection was performed using univariate logistic regression, LASSO, and the Boruta algorithm, followed by multivariate logistic regression analysis. Seven machine learning models were constructed and evaluated using receiver operating characteristic (ROC) curves and decision curve analysis (DCA). Shapley additive explanations (SHAP) were applied to interpret the results, and restricted cubic spline (RCS) regression explored the nonlinear relationships between variables and sepsis risk. Furthermore, we examined the interactions among predictors to better understand their potential interrelationships. Results The random forest (RF) model outperformed all others, achieving an AUC of 0.765 in the training cohort and 0.700 in the validation cohort. Key predictors of sepsis identified by SHAP analysis included C-reactive protein (CRP), procalcitonin (PCT), neutrophil count (Neut), lymphocyte count (Lymph), thrombin time (TT), red blood cell count (RBC), total bile acid (TBA), and systolic blood pressure (SBP). RCS analysis revealed significant non-linear associations between CPR, PCT, Neut, Lymph, TT, RBC and SBP with sepsis risk. Pairwise correlation analysis further revealed interactions among these variables. Conclusion The RF model exhibited robust predictive power for sepsis in leukemia patients, providing clinicians with a valuable tool for early risk assessment and the optimization of treatment strategies.
Collapse
Affiliation(s)
- Yanqi Kou
- Department of Hematology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, People’s Republic of China
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang City, Guangdong Province, People’s Republic of China
| | - Yuan Tian
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang City, Guangdong Province, People’s Republic of China
- Department of Pathology, Guangdong Medical University, Zhanjiang City, Guangdong Province, People’s Republic of China
| | - Yanping Ha
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang City, Guangdong Province, People’s Republic of China
- Department of Pathology, Guangdong Medical University, Zhanjiang City, Guangdong Province, People’s Republic of China
| | - Shijie Wang
- Department of Hematology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, People’s Republic of China
| | - Xiaobai Sun
- Department of Hematology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, People’s Republic of China
| | - Shuxin Lv
- Department of Hematology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, People’s Republic of China
| | - Botao Luo
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang City, Guangdong Province, People’s Republic of China
- Department of Pathology, Guangdong Medical University, Zhanjiang City, Guangdong Province, People’s Republic of China
| | - Yuping Yang
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang City, Guangdong Province, People’s Republic of China
| | - Ling Qin
- Department of Hematology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, People’s Republic of China
| |
Collapse
|
44
|
Wang J, Jiang Y, Yuan Y, Ma X, Li T, Lv Y, Zhang J, Chen L, Zhou J, Meng Y, Zhang B, Dong X, Ma L. Serum Exosomes miR-122-5P Induces Hepatic and Renal Injury in Septic Rats by Regulating TAK1/SIRT1 Pathway. Infect Drug Resist 2025; 18:185-197. [PMID: 39807206 PMCID: PMC11727329 DOI: 10.2147/idr.s499643] [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: 10/22/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025] Open
Abstract
Aim Sepsis is a potentially fatal condition characterized by organ failure resulting from an abnormal host response to infection, often leading to liver and kidney damage. Timely recognition and intervention of these dysfunctions have the potential to significantly reduce sepsis mortality rates. Recent studies have emphasized the critical role of serum exosomes and their miRNA content in mediating sepsis-induced organ dysfunction. The objective of this study is to elucidate the mechanism underlying the impact of miR-122-5p on sepsis-associated liver and kidney injury using inhibitors for miR-122-5p as well as GW4869, an inhibitor targeting exosome release. Materials and Methods Exosomes were isolated from serum samples of septic rats, sepsis patients, and control groups, while liver and kidney tissues were collected for subsequent analysis. The levels of miR-122-5p, inflammation indices, and organ damage were assessed using PCR, ELISA, and pathological identification techniques. Immunohistochemistry and Western blotting methods were employed to investigate the activation of inflammatory pathways. Furthermore, big data analysis was utilized to screen potential targets of miR-122-5p in vivo. Key Findings Serum exosomal levels of miR-122-5p were significantly elevated in septic patients as well as in LPS-induced septic rats. Inhibition of miR-122-5p reduced serum pro-inflammatory factors and ameliorated liver and kidney damage in septic rats. Mechanistically, miR-122-5p upregulated TAK1, downregulated SIRT1, and facilitated NF-κB activation. Conclusion Serum exosomal miR-122-5p promotes inflammation and induces liver/kidney injury in LPS-induced septic rats by modulating the TAK1/SIRT1/NF-κB pathway, highlighting potential therapeutic targets for sepsis management.
Collapse
Affiliation(s)
- Jiaqi Wang
- Department of Critical Care Medicine, Lanzhou University Second Hospital, Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| | - Yujing Jiang
- Department of Critical Care Medicine, Lanzhou University Second Hospital, Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| | - Yamin Yuan
- Department of Critical Care Medicine, Lanzhou University Second Hospital, Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| | - Xin Ma
- Department of Critical Care Medicine, Lanzhou University Second Hospital, Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| | - Tongqin Li
- Department of Critical Care Medicine, Lanzhou University Second Hospital, Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| | - YaTing Lv
- Department of Critical Care Medicine, Lanzhou University Second Hospital, Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| | - Jing Zhang
- Department of Critical Care Medicine, Lanzhou University Second Hospital, Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| | - Liao Chen
- Department of Critical Care Medicine, Lanzhou University Second Hospital, Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| | - Jinquan Zhou
- Department of Critical Care Medicine, Lanzhou University Second Hospital, Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| | - Yanfei Meng
- Department of Critical Care Medicine, Lanzhou University Second Hospital, Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| | - Bei Zhang
- Department of Critical Care Medicine, Lanzhou University Second Hospital, Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| | - Xiaorong Dong
- Department of Critical Care Medicine, Lanzhou University Second Hospital, Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| | - Li Ma
- Department of Critical Care Medicine, Lanzhou University Second Hospital, Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| |
Collapse
|
45
|
Xiao L, Ping Y, Sun S, Xu R, Zhou X, Wu H, Qi L. TMT-based quantitative proteomics unveils the protective mechanism of Polygonatum sibiricum polysaccharides on septic acute liver injury. J Proteomics 2025; 310:105331. [PMID: 39427987 DOI: 10.1016/j.jprot.2024.105331] [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: 05/23/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/22/2024]
Abstract
Polygonatum sibiricum polysaccharides (PSP) has been shown to possess multiple pharmacological functions. Our previous study found that PSP could protect against acute liver injury during sepsis via inhibiting inflammatory response. However, the underlying molecular mechanism by which PSP alleviates septic acute liver injury (SALI) remains unknown. Herein, TMT-based quantitative proteomics was utilized to explore the essential pathways and proteins involved in the protective effects of PSP on SALI. The results revealed that 632 and 176 differentially expressed proteins (DEPs) were identified in Model_vs_Control and PSP_vs_Model, respectively. GO annotation showed similar trends, suggesting that these DEPs were primarily involved in the cellular anatomical entity in Cellular Component, the cellular processe and the biological regulation in Biological Process, the binding and the catalytic activity in Molecular Function. Meanwhile, KEGG enrichment analysis implied that four common pathways, including the NF-κB signaling pathway, the IL-17 signaling pathway, the TNF signaling pathway and the Toll-like receptor signaling pathway, were closely associated with the pathogenesis of sepsis among the top 20 remarkably enriched pathways in Model_vs_Control_up and PSP_vs_Model_down. Moreover, the levels of several common DEPs, including TLR2, IKKi, JunB and CXCL9, were validated by WB, which was in line with the results of proteomics. Therefore, the protective effects of PSP on SALI might exert via blocking the above-mentioned inflammation pathways. Significance: PSP, recognized as a key component of Polygonatum sibiricum, exhibits a range of pharmacological functions. Our previous study found that PSP could protect against SALI, yet failing to clarify the mechanism of action. To reveal the underlying molecular mechanism involved in the protective effects of PSP on SALI, a TMT-based quantitative proteomic analysis was performed to detect and analyse the DEPs in liver tissue among the control group, the model group and the PSP group in this study. The results provide theoretical references for exploring the action mechanism of drugs and facilitate the comprehensive utilization of PSP. SIGNIFICANCE: PSP have been identified as the most crucial components of Polygonatum sibiricum with various pharmacological functions. Our previous study found that PSP could protect against SALI, but the mechanism of action remains unknown. To reveal the underlying molecular mechanism involved in the protective effects of PSP on SALI, a TMT-based quantitative proteomic analysis was performed to detect and analyse the DEPs in liver tissue among the control group, the model group and the PSP group in this study. The results provide theoretical references for exploring the action mechanism of drugs and facilitate the comprehensive utilization of PSP.
Collapse
Affiliation(s)
- Linxia Xiao
- School of Pharmacology, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Yinuo Ping
- School of Pharmacology, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Shangshang Sun
- School of Pharmacology, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Ran Xu
- School of Pharmacology, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Xinru Zhou
- School of Pharmacology, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Hongyan Wu
- School of Pharmacology, Jiangsu Vocational College of Medicine, Yancheng 224005, China; Institute of Biomedical Technology, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Liang Qi
- School of Pharmacology, Jiangsu Vocational College of Medicine, Yancheng 224005, China.
| |
Collapse
|
46
|
Van Nynatten LR, Patel MA, Daley M, Miller MR, Cepinskas G, Slessarev M, Russell JA, Fraser DD. Putative biomarkers of hepatic dysfunction in critically ill sepsis patients. Clin Exp Med 2025; 25:28. [PMID: 39751971 PMCID: PMC11698781 DOI: 10.1007/s10238-024-01545-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
Sepsis is a major cause of morbidity and mortality worldwide. Among the various types of end-organ damage associated with sepsis, hepatic injury is linked to significantly higher mortality rates compared to dysfunction in other organ systems. This study aimed to investigate potential biomarkers of hepatic injury in sepsis patients through a multi-center, case-control approach. We enrolled three matched cohorts: 37 sepsis patients with hepatic dysfunction (S-HD), 37 sepsis patients without hepatic dysfunction (S-CON), and 18 healthy controls (HC). We measured five proposed biomarkers of hepatic dysfunction-ARG1, MDH1, GSTα, 5-NT, and SDH-using multiplex immunoassays. These biomarkers were compared to traditional markers of hepatic dysfunction, including albumin, bilirubin, ALT, AST, and GGT, across the cohorts using both conventional statistical methods and machine learning techniques. The median age of participants was comparable across cohorts: S-HD (65.0 years, IQR 49.5-82.5), S-CON (65.0 years, IQR 48.0-81.5), and HC (62.5 years, IQR 53.0-65.0; P = 0.794). Patients with hepatic dysfunction (S-HD) exhibited higher illness severity scores compared to those without hepatic dysfunction (S-CON): MODS scores were median 7.0 (IQR 4.0-10.0) in S-HD versus median 4.0 (IQR 2.0-7.0) in S-CON (P = 0.005), and SOFA scores were median 7.0 (IQR 4.0-11.0) in S-HD versus median 3.0 (IQR 2.0-6.0) in S-CON (P < 0.001). Hemoglobin and platelet counts were lower, while creatinine levels were higher in S-HD compared to S-CON (P < 0.05). On ICU Day 1, bilirubin, ALT, AST, GGT, and INR were significantly elevated in S-HD relative to S-CON (P ≤ 0.001), and albumin levels were lower (P < 0.05). Additionally, ARG1, GSTα, 5-NT, and SDH were significantly higher in S-HD patients on ICU Day 1 compared to S-CON (P < 0.05). ARG1, MDH1, and SDH showed positive correlations with AST, ALT, and MODS (P < 0.01). From ICU Day 1 to Day 7, ARG1, GSTα, SDH, and AST levels significantly decreased in S-HD patients (P < 0.05), whereas MDH1 and 5-NT levels did not. Among the proposed biomarkers, GSTα and 5-NT did not correlate with traditional hepatic dysfunction markers but were significant in identifying S-HD patients (feature importance 0.131 and 0.097, respectively) in a random forest classification model. This comprehensive model demonstrated excellent performance in distinguishing sepsis patients with hepatic injury, with sensitivity 0.93, specificity 0.94, NPV 0.94, PPV 0.94, and AUC 0.94. The biomarkers ARG1, MDH1, GSTα, 5-NT, and SDH show promise as novel indicators of hepatic dysfunction associated with sepsis. This study provides a foundational basis for subsequent research aimed at characterizing and clinically validating these markers. Future investigations should focus on integrating these potential biomarkers into routine laboratory assessments for sepsis and related hepatic injury.
Collapse
Affiliation(s)
| | - Maitray A Patel
- Epidemiology and Biostatistics, Western University, London, ON, Canada
| | - Mark Daley
- Epidemiology and Biostatistics, Western University, London, ON, Canada
| | - Michael R Miller
- London Health Sciences Centre Research Institute, London, ON, Canada
| | - Gediminas Cepinskas
- London Health Sciences Centre Research Institute, London, ON, Canada
- Medical Biophysics, Western University, London, ON, Canada
- Anatomy and Cell Biology, Western University, London, ON, Canada
| | - Marat Slessarev
- Medicine, Western University, London, ON, Canada
- London Health Sciences Centre Research Institute, London, ON, Canada
| | - James A Russell
- University of British Columbia, St. Paul's Hospital, 1081 Burrard Street, Vancouver, BC, V6Z 1Y6, Canada.
| | - Douglas D Fraser
- Pediatrics, Western University, London, ON, Canada.
- London Health Sciences Centre Research Institute, London, ON, Canada.
- Physiology and Pharmacology, Western University, London, ON, Canada.
- Clinical Neurological Sciences, Western University, London, ON, Canada.
- London Health Sciences Centre, 800 Commissioners Road East, London, ON, N6A 5W9, Canada.
| |
Collapse
|
47
|
Daidouji Y, Suzuki S, Wang X, Fahreza RR, Nemoto E, Yamada S. Periodontal inflammation potentially inhibits hepatic cytochrome P450 expression and disrupts the omega-3 epoxidation pathway in a murine model. J Dent Sci 2025; 20:444-451. [PMID: 39873042 PMCID: PMC11763211 DOI: 10.1016/j.jds.2024.05.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 05/23/2024] [Indexed: 01/30/2025] Open
Affiliation(s)
- Yoshino Daidouji
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Shigeki Suzuki
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Xiuting Wang
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Rahmad Rifqi Fahreza
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Eiji Nemoto
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Satoru Yamada
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| |
Collapse
|
48
|
Lavarti R, Cai L, Alvarez‐Diaz T, Medina‐Rodriguez T, Bombin S, Raju RP. Senescence landscape in the liver following sepsis and senolytics as potential therapeutics. Aging Cell 2025; 24:e14354. [PMID: 39444093 PMCID: PMC11709100 DOI: 10.1111/acel.14354] [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: 04/04/2024] [Revised: 08/15/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024] Open
Abstract
Senescence, caused by cell-cycle arrest, is a hallmark of aging. Senescence has also been described in embryogenesis, wound healing, and acute injuries. Sepsis is characterized by a dysregulated host response to infection, leading to organ dysfunction and mortality. Most of the pathophysiology of human sepsis is recapitulated in the mouse model of polymicrobial sepsis, developed by cecal ligation and puncture (CLP). In this report, we demonstrate a rapid onset of cellular senescence in the liver of mice subjected to CLP-induced sepsis, characterized by the upregulation of p21, p53, and other senescence markers, including SA-βgal. Using RNAscope, confocal microscopy, and flow cytometry, we further confirm the emergence of p21-expressing senescence phenotype in the liver 24 h after sepsis induction. Senescence was observed in several cell types in the liver, including hepatocytes, endothelial cells, and macrophages. We determined the landscape of senescence phenotype in murine sepsis by single-cell sequencing, which further ascertained that this cell fate is not confined to any particular cell type but displays a heterogeneous distribution. Furthermore, we observed a significant reduction in mortality following sepsis when mice were treated with senolytics, a combination of dasatinib and quercetin, before the CLP surgery. Our experiments unequivocally demonstrated a rapid development of cellular senescence with sepsis and, for the first time, described the senescence landscape in the sepsis liver and the possible role of senescent cells in the worsening outcome following sepsis.
Collapse
Affiliation(s)
- Rupa Lavarti
- Department of Pharmacology and Toxicology, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| | - Lun Cai
- Department of Pharmacology and Toxicology, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| | - Tatiana Alvarez‐Diaz
- Department of Pharmacology and Toxicology, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| | - Thalia Medina‐Rodriguez
- Department of Pharmacology and Toxicology, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| | - Sergei Bombin
- Georgia Cancer Center, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| | - Raghavan Pillai Raju
- Department of Pharmacology and Toxicology, Medical College of GeorgiaAugusta UniversityAugustaGeorgiaUSA
| |
Collapse
|
49
|
Tang X, Ning J, Zhao Y, Feng S, Shao L, Liu T, Miao H, Zhang Y, Wang C. Intestine-derived fibroblast growth factor 19 alleviates lipopolysaccharide-induced liver injury by regulating bile acid homeostasis and directly improving oxidative stress. JOURNAL OF INTENSIVE MEDICINE 2025; 5:79-88. [PMID: 39872844 PMCID: PMC11763227 DOI: 10.1016/j.jointm.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 05/17/2024] [Accepted: 06/11/2024] [Indexed: 01/30/2025]
Abstract
Background Cholestasis plays a critical role in sepsis-associated liver injury (SALI). Intestine-derived fibroblast growth factor 19 (FGF19) is a key regulator for bile acid homeostasis. However, the roles and underlying mechanisms of FGF19 in SALI are still unclear. Methods We conducted a case-control study that included 58 pediatric patients aged from 1 month to 14-years-old diagnosed with sepsis at Shanghai Children's Hospital from January to December 2018 and 30 healthy individuals. The serum FGF19 levels of these patients with sepsis were analyzed and compared with those of healthy controls. Recombinant human FGF19 was intravenously injected in mice once a day for 7 days at a dose of 0.1 mg/kg body weight before lipopolysaccharide (LPS) treatment. Liver bile acid profiles and the gene expression involved in bile acid homeostasis were investigated in the mice groups. Metabolomic data were further integrated and analyzed using Ingenuity Pathways Analysis (IPA) software. In the in vitro analysis using HepG2 cells, the influence of FGF19 pretreatment on reactive oxygen species (ROS) production and mitochondrial dysfunction was analyzed. Compound C (CC), an inhibitor of AMP-activated protein kinase (AMPK) activation, was used to confirm the roles of AMPK activation in FGF19-mediated hepatoprotective effects. Results Serum FGF19 levels were significantly lower in children with sepsis than in healthy controls (115 pg/mL vs. 79 pg/mL, P=0.03). Pre-administration of recombinant human FGF19 alleviated LPS-induced acute liver injury (ALI) and improved LPS-induced cholestasis in mice. Moreover, FGF19 directly reversed LPS-induced intracellular ROS generation and LPS-decreased mitochondrial membrane potential in vitro and in vivo, resulting in hepatoprotection against LPS-induced apoptosis. More importantly, the inhibition of AMPK activity partially blocked the protective effects of FGF19 against LPS-induced oxidative stress and mitochondrial dysfunction. Conclusions Intestine-derived FGF19 alleviates LPS-induced ALI via improving bile acid homeostasis and directly suppressing ROS production via activating the AMPK signaling pathway.
Collapse
Affiliation(s)
- Xiaomeng Tang
- Department of Critical Care Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Laboratory of Critical Care Translational Medicine, Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingjing Ning
- Department of Critical Care Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Laboratory of Critical Care Translational Medicine, Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yilin Zhao
- Department of Critical Care Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Laboratory of Critical Care Translational Medicine, Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuyun Feng
- Department of Critical Care Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Laboratory of Critical Care Translational Medicine, Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lujing Shao
- Department of Critical Care Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Laboratory of Critical Care Translational Medicine, Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiantian Liu
- Department of Critical Care Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Laboratory of Critical Care Translational Medicine, Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huijie Miao
- Department of Critical Care Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Laboratory of Critical Care Translational Medicine, Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China
| | - Yucai Zhang
- Department of Critical Care Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Laboratory of Critical Care Translational Medicine, Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China
| | - Chunxia Wang
- Department of Critical Care Medicine, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Laboratory of Critical Care Translational Medicine, Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
50
|
Xu W, Hou H, Yang W, Tang W, Sun L. Immunologic role of macrophages in sepsis-induced acute liver injury. Int Immunopharmacol 2024; 143:113492. [PMID: 39471696 DOI: 10.1016/j.intimp.2024.113492] [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: 08/15/2024] [Revised: 10/11/2024] [Accepted: 10/21/2024] [Indexed: 11/01/2024]
Abstract
Sepsis-induced acute liver injury (SALI), a manifestation of sepsis multi-organ dysfunction syndrome, is associated with poor prognosis and high mortality. The diversity and plasticity of liver macrophage subpopulations explain their different functional responses in different liver diseases. Kupffer macrophages, liver capsular macrophages, and monocyte-derived macrophages are involved in pathogen recognition and clearance and in the regulation of inflammatory responses, exacerbating the progression of SALI through different pathways of pyroptosis, ferroptosis, and autophagy. Concurrently, they play an important role in maintaining hepatic homeostasis and in the injury and repair processes of SALI. Other macrophages are recruited to diseased tissues under pathological conditions and are polarized into various phenotypes (mainly M1 and M2 types) under the influence of signaling molecules, transcription factors, and metabolic reprogramming, thereby exerting different roles and functions. This review provides an overview of the immune role of macrophages in SALI and discusses the multiple roles of macrophages in liver injury and repair to provide a reference for future studies.
Collapse
Affiliation(s)
- Wanling Xu
- Department of Emergency, Jilin University First Hospital, 71 Xinmin Street, Changchun 130021, Jilin, China
| | - Hailong Hou
- Emergency Department, Meihekou Central Hospital, 2668 Aimin Street, Tonghua 135000, Jilin, China
| | - Weiying Yang
- Department of Emergency, Jilin University First Hospital, 71 Xinmin Street, Changchun 130021, Jilin, China
| | - Wenjing Tang
- Department of Emergency, Jilin University First Hospital, 71 Xinmin Street, Changchun 130021, Jilin, China
| | - Lichao Sun
- Department of Emergency, Jilin University First Hospital, 71 Xinmin Street, Changchun 130021, Jilin, China.
| |
Collapse
|