1
|
Zhang J, Tan B, Wu H, Han T, Fang D, Cai H, Hu B, Kang A. Scutellaria baicalensis Extracts Restrict Intestinal Epithelial Cell Ferroptosis by Regulating Lipid Peroxidation and GPX4/ACSL4 in Colitis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156708. [PMID: 40220415 DOI: 10.1016/j.phymed.2025.156708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/23/2025] [Accepted: 03/29/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Ferroptosis in colonic epithelial cells has been implicated in the development of ulcerative colitis (UC) and the accompanying gut leakage. Scutellaria baicalensis Georgi (Scu) is widely used herb medicine for alleviating UC. PURPOSE We aimed to clarify the therapeutic effect of Scu on UC by inhibiting intestinal epithelial cell ferroptosis and explore its regulatory mechanisms on lipid peroxidation and the GPX4/ACSL4 pathways. METHODS UPLC-Q-TOF/MS was employed to analyze chemicals in the herbal extract and the colonic exposure of prototypes in Scu-treated mice. Additionally, the main compounds were quantified using HPLC-UV. The ameliorative effects of Scu were comprehensively explored in a UC mouse model established by feeding with dextran sulfate sodium (DSS). HPLC-MS based metabolomic studies were conducted to identify the differential metabolites in colon tissues from Scu or vehicle treated UC mice. Network pharmacology was conducted for target prediction and potential pathway analysis. In conjunction with these bioinformatic analyses, we performed RT-qPCR, immunofluorescence, immunohistochemistry and immunoblotting to elucidate the regulatory mechanisms of Scu on ferroptosis-related pathways in both in vivo and in vitro models. RESULTS 78 chemical constituents in Scu were characterized, with 42 detected in the colonic tissues of Scu-treated mice. Scu could alleviate UC related symptoms in mice, including increased colon length and decreased pathological score. Furthermore, Scu inhibited pro-inflammatory cytokines and mediators, while improving gut barrier function by increasing the expression of ZO-1 and Occludin at both mRNA and protein levels. Based on metabolomic studies, a total of 71 differential metabolites exhibited a reversal trend following Scu administration. These findings, combined with results from network pharmacology, suggest that arachidonic acid (AA) metabolism and ferroptosis may serve as potential pathways for Scu intervention in UC. Further experiments indicated that the amelioratory actions of Scu on ferroptosis partially contributed to its modulation on lipid peroxidation and its regulatory influence on the GPX4/ASCL4 axis to ameliorate UC. When AA was administered at the same time as concurrently with Scu, the regulatory effects of Scu on ferroptosis, GPX4/ASCL4 axis, and its protective effects against UC were significantly reduced. Moreover, the inhibitory effect of Scu on ferroptosis was weakened when we knocked down GPX4 or overexpressed ACSL4 in vitro. CONCLUSION The ameliorative effect of Scu in UC is closely related to the regulation of lipid peroxidation and GPX4/ASCL4 mediated intestinal epithelial ferroptosis.
Collapse
Affiliation(s)
- Jingyan Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Bingyan Tan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hong Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tai Han
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Dan Fang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hong Cai
- Xuzhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Xuzhou 221009, China
| | - Bing Hu
- Xuzhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Xuzhou 221009, China.
| | - An Kang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
2
|
Su X, Sun Y, Dai A. New insights into pulmonary arterial hypertension: interaction between PANoptosis and perivascular inflammatory responses. Apoptosis 2025; 30:1097-1116. [PMID: 39979525 DOI: 10.1007/s10495-025-02086-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2025] [Indexed: 02/22/2025]
Abstract
Pulmonary arterial hypertension (PAH) is a heterogeneous disease characterized by various etiologies, with pulmonary vascular remodeling recognized as a main pathological change. Currently, it is widely accepted that vascular remodeling is closely associated with abnormal pulmonary vascular cell death and perivascular inflammation. The simultaneous activation of various pulmonary vascular cell death leads to immune cell adhesion and inflammatory mediator releases; And in turn, the inflammatory response may also trigger cell death and jointly promote the progression of vascular remodeling. Recently, PANoptosis has been identified as a phenomenon that describes the simultaneous activation and interaction of multiple forms of programmed cell death (PCD). Therefore, the relationship between PANoptosis and inflammation in PAH warrants further investigation. This review examines the mechanisms underlying apoptosis, necroptosis, pyroptosis, and inflammatory responses in PAH, with a focus on PANoptosis and its interactions with inflammation. And it aims to elucidate the significance of this emerging form of cell death and inflammation in the pathophysiology of PAH and to explore its potential as a therapeutic target.
Collapse
Affiliation(s)
- Xianli Su
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Yinhui Sun
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Aiguo Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China.
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China.
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410021, People's Republic of China.
| |
Collapse
|
3
|
Wu Q, Qi S, Kang Z, Bai X, Li Z, Cheng J, Dong X. PANoptosis in Sepsis: A Central Role and Emerging Therapeutic Target. J Inflamm Res 2025; 18:6245-6261. [PMID: 40386177 PMCID: PMC12085136 DOI: 10.2147/jir.s513367] [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: 12/20/2024] [Accepted: 05/04/2025] [Indexed: 05/20/2025] Open
Abstract
The pathogenesis of sepsis is intricately linked to regulated cell death. As a novel form of regulated cell death, PANoptosis plays a critical role in driving the inflammatory response, impairing immune cell function, and contributing to multi-organ dysfunction in sepsis. This review explores the molecular mechanisms underlying PANoptosis and its involvement in sepsis. By activating multiple pathways, PANoptosis promotes the release of inflammatory cytokines, triggering a cytokine storm that disrupts immune cell homeostasis and exacerbates organ damage. Emerging therapeutic strategies targeting PANoptosis, including chemotherapeutic agents and herbal remedies, are showing potential for clinical application. The concept of targeting PANoptosis offers a promising avenue for developing innovative treatments for sepsis.
Collapse
Affiliation(s)
- Qiqi Wu
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Siyuan Qi
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhaofeng Kang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xiangjun Bai
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhanfei Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jing Cheng
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xijie Dong
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
4
|
Milivojac T, Grabež M, Amidžić L, Prtina A, Krivokuća A, Malicevic U, Barudžija M, Matičić M, Uletilović S, Mandić-Kovačević N, Cvjetković T, Stojiljković MP, Gajić Bojić M, Mikov M, Gajanin R, Bolevich S, Petrović A, Škrbić R. Ursodeoxycholic and chenodeoxycholic bile acids alleviate endotoxininduced acute lung injury in rats by modulating aquaporin expression and pathways associated with apoptosis and inflammation. Front Pharmacol 2025; 16:1484292. [PMID: 40115259 PMCID: PMC11922783 DOI: 10.3389/fphar.2025.1484292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/06/2025] [Indexed: 03/23/2025] Open
Abstract
Introduction This study aimed to investigate the anti-inflammatory, antioxidant, and anti-apoptotic properties of ursodeoxycholic (UDCA) and chenodeoxycholic (CDCA) bile acids in a rat model of endotoxin (lipopolysaccharide, LPS)-induced acute lung injury (ALI). Methods The study included six groups of Wistar rats exposed to different pretreatments. The control and endotoxin groups were pretreated with propylene glycol, a solvent for bile acids, while the other groups received UDCA or CDCA for 10 days. On the 10th day, an endotoxin injection was given to evaluate the impact of these pretreatments. Lung tissue sections were analyzed by immunohistochemistry, targeting the pro-inflammatory marker nuclear factor kappa B (NF-κB), the anti-apoptotic marker B-cell lymphoma 2 (BCL-2), pro-apoptotic markers BCL-2-associated X protein (BAX) and caspase 3, as well as the aquaporins 1 and 5 (AQP1 and AQP5). Oxidative stress was assessed in bronchoalveolar lavage fluid (BALF). Results and discussion This study demonstrates that UDCA and CDCA can mitigate endotoxin-induced lung injury in rats. These effects are achieved through modulation of AQP1 and AQP5 expression, reduction of oxidative stress, regulation of apoptotic pathways (BAX, caspase 3, BCL-2), and attenuation of pro-inflammatory activity of NF-κB. Although the results indicate a significant association between the expression of these proteins and histopathological changes, the potential influence of additional factors cannot be excluded. These findings suggest that UDCA and CDCA provide lung protection by acting through complex mechanisms involving inflammatory, oxidative, and apoptotic pathways.
Collapse
Affiliation(s)
- Tatjana Milivojac
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Milkica Grabež
- Department of Hygiene, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Ljiljana Amidžić
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Alma Prtina
- Department of Pathophysiology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Aleksandra Krivokuća
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Pathophysiology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Ugljesa Malicevic
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Pathophysiology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Maja Barudžija
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Histology and Embryology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Milka Matičić
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Snežana Uletilović
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Medical Biochemistry and Chemistry, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Nebojša Mandić-Kovačević
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Pharmacy, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Tanja Cvjetković
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Medical Biochemistry and Chemistry, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Miloš P Stojiljković
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Milica Gajić Bojić
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Momir Mikov
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Radoslav Gajanin
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Sergey Bolevich
- Department of Pathologic Physiology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Aleksandar Petrović
- Department of Histology and Embryology, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Ranko Škrbić
- Centre for Biomedical Research, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
- Department of Pathologic Physiology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
5
|
Pan T, Wu F, Zhang J, Xiang B, Huang K, Chen Y, Jin X. The molecular structure of SHISA5 protein and its novel role in primary biliary cholangitis: From single-cell RNA sequencing to biomarkers. Int J Biol Macromol 2025; 296:139775. [PMID: 39800023 DOI: 10.1016/j.ijbiomac.2025.139775] [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: 11/20/2024] [Revised: 12/24/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
The study collected liver tissue samples from PBC patients and healthy controls and performed transcriptomic analysis of the cells in the samples using single-cell RNA sequencing. The expression characteristics of SHISA5 in PBC were revealed by comparing the difference of SHISA5 protein in the two groups of samples. The structure of SHISA5 protein was predicted and its possible biological function was analysed by bioinformatics method. The results showed that the expression of SHISA5 protein in liver tissue of PBC patients was significantly higher than that of healthy controls. Single-cell RNA sequencing data showed that SHISA5 was mainly expressed in hepatocytes and bile duct cells, and its expression level was positively correlated with the disease activity of PBC. Through structural prediction, we found that the SHISA5 protein molecule has a unique transmembrane domain and may be involved in cell signaling and intercellular interactions. Further functional analysis revealed that SHISA5 may participate in the pathological process of PBC by regulating the differentiation and function of bile duct cells.
Collapse
Affiliation(s)
- Tongtong Pan
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Faling Wu
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China; Department of Infectious Disease, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Jiarong Zhang
- Department of Infection Control, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Bingyu Xiang
- Department of Infectious Disease, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Kate Huang
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Yongping Chen
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China; Department of Infectious Disease, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China.
| | - Xiaoya Jin
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China; Department of Infectious Disease, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China; Department of Infection Control, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China.
| |
Collapse
|
6
|
Wang Y, Zhang X, Wang W, Zhang Y, Fleishman JS, Wang H. cGAS-STING targeting offers therapy choice in lung diseases. Biol Direct 2025; 20:20. [PMID: 39920718 PMCID: PMC11806777 DOI: 10.1186/s13062-025-00611-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 01/27/2025] [Indexed: 02/09/2025] Open
Abstract
Cyclic GMP/AMP (cGAMP) synthase (cGAS), along with the endoplasmic reticulum (ER)-associated stimulator of interferon genes (STING), are crucial elements of the type 1 interferon response. cGAS senses microbial DNA and self-DNA, labeling cGAS-STING as a crucial mechanism in autoimmunity, sterile inflammatory responses, and cellular senescence. However, chronic and aberrant activation of the cGAS-STING axis results in inflammatory and autoimmune diseases. cGAS-STING has emerged as a vital mechanism driving inflammation-related diseases, including lung diseases. Insights into the biology of the cGAS-STING pathway have enabled the discovery of small-molecule agents which have the potential to inhibit the cGAS-STING axis in lung diseases. In this review, we first outline the principal components of the cGAS-STING signaling cascade. Then, we discuss recent research that highlights general mechanisms by which cGAS-STING contributes to lung diseases. Then, we focus on summarizing a list of bioactive small-molecule compounds which inhibit the cGAS-STING pathway, reviewing their potential mechanisms.These review highlights a novel groundbreaking therapeutic possibilities through targeting cGAS-STING in lung diseases.
Collapse
Affiliation(s)
- Yu Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Xuan Zhang
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Weixue Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Yi Zhang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| | - Hongquan Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China.
| |
Collapse
|
7
|
Wang X, Zhang K, Zhang J, Xu G, Guo Z, Lu X, Liang C, Gu X, Huang L, Liu S, Wang L, Li J. Cordyceps militaris solid medium extract alleviates lipopolysaccharide-induced acute lung injury via regulating gut microbiota and metabolism. Front Immunol 2025; 15:1528222. [PMID: 39902053 PMCID: PMC11788161 DOI: 10.3389/fimmu.2024.1528222] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 12/30/2024] [Indexed: 02/05/2025] Open
Abstract
Acute lung injury (ALI) is a common respiratory disease, Cordycepin has been reported to reduce ALI, which is an effective component in Cordyceps militaris solid medium extract (CMME). Therefore, we aimed to explore the alleviating effect and mechanism of CMME on ALI. This study evaluated the effect of CMME on lipopolysaccharide (LPS)-induced ALI mice by analyzing intestinal flora and metabolomics to explore its potential mechanism. We assessed pulmonary changes, inflammation, oxidative stress, and macrophage and neutrophil activation levels, then we analyzed the gut microbiota through 16S rRNA and analyzed metabolomics profile by UPLC-QTOF/MS. The results showed that CMME treatment improved pulmonary injury, reduced inflammatory factors and oxidative stress levels, and decreased macrophage activation and neutrophil recruitment. The 16S rRNA results revealed that CMME significantly increased gut microbiota richness and diversity and reduced the abundance of Bacteroides compared with Mod group significantly. Metabolic analysis indicated that CMME reversed the levels of differential metabolites and may ameliorate lung injury through purine metabolism, nucleotide metabolism, and bile acid (BA) metabolism, and CMME did reverse the changes of BA metabolites in ALI mice, and BA metabolites were associated with inflammatory factors and intestinal flora. Therefore, CMME may improve lung injury by regulating intestinal flora and correcting metabolic disorders, providing new insights into its mechanism of action.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Lei Wang
- Traditional Chinese Veterinary Technology Innovation Center of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Jianxi Li
- Traditional Chinese Veterinary Technology Innovation Center of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| |
Collapse
|
8
|
Yang Z, Kao X, Zhang L, Huang N, Chen J, He M. Exploring the Anti-PANoptosis Mechanism of Dachaihu Decoction Against Sepsis-Induced Acute Lung Injury: Network Pharmacology, Bioinformatics, and Experimental Validation. Drug Des Devel Ther 2025; 19:349-368. [PMID: 39839500 PMCID: PMC11750123 DOI: 10.2147/dddt.s495225] [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: 10/17/2024] [Accepted: 12/31/2024] [Indexed: 01/23/2025] Open
Abstract
Background Dachaihu decoction (DCHD) is a common Chinese medicine formula against sepsis-induced acute lung injury (SALI). PANoptosis is a novel type of programmed cell death. Nevertheless, The mechanisms of DCHD against SALI via anti-PANoptosis remains unknown. Methods First, we identified the intersecting targets among DCHD, SALI, and PANoptosis using relevant databases and published literature. Then, protein-protein interaction (PPI) network, molecular docking, and functional enrichment analysis were conducted. In vivo, cecal ligation and puncture (CLP) was used to construct a sepsis mouse model, and the therapeutic effects of DCHD on SALI were evaluated using hematoxylin and eosin (H&E) staining, quantitative real-time PCR (qRT-PCR), and ELISA. Finally, qRT-PCR, immunofluorescence staining, and Western blotting were used to verify the effect of DCHD-containing serum (DCHD-DS) on LPS-induced RAW 264.7 macrophages in vitro. Results 82 intersecting targets were identified by mapping the targets of DCHD, SALI, and PANoptosis. Enrichment analysis showed that DCHD against SALI via anti-PANoptosis by modulating tumor necrosis factor (TNF), AGE-RAGE, phosphoinositide 3-kinase (PI3K)-AKT, and Toll-like receptor signaling pathways by targeting Casp3, cellular tumor antigen p53 (TP53), B-cell lymphoma 2 (Bcl2), toll-like receptor-4 (TLR4), STAT3, STAT1, RELA, NF-κB1, myeloid cell leukemia-1 (MCL1), JUN, IL-1β, HSP90AA1, Casp9, Casp8, and Bcl2l1. Molecular docking analysis revealed that the key components of DCHD have a high binding affinity to the core targets. In vivo, DCHD improved lung histopathological injury, reduced inflammatory factor expression, and alleviated oxidative stress injury in lung tissues. In vitro, DCHD-DS alleviated cell morphology changes, the release of pro-inflammatory factors, and p65 nucleus aggregation. Furthermore, we verified that DCHD-DS inhibited PANoptosis by downregulating the PI3K/AKT/NF-κB signalling pathway. Conclusion DCHD attenuates SALI by inhibiting PANoptosis via control of the PI3K/AKT/NF-κB pathway. Our study provides a solid foundation for investigating the mechanisms of DCHD and its clinical application in the treatment of SALI.
Collapse
Affiliation(s)
- Zhen Yang
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, Guangdong, People’s Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong, People’s Republic of China
| | - Xingyu Kao
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, Guangdong, People’s Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong, People’s Republic of China
| | - Lin Zhang
- Department of Cardiovascular, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, People’s Republic of China
| | - Na Huang
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, Guangdong, People’s Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong, People’s Republic of China
| | - Jingli Chen
- Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong, People’s Republic of China
| | - Mingfeng He
- Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong, People’s Republic of China
| |
Collapse
|
9
|
Zhang M, Shang L, Zhou F, Li J, Wang S, Lin Q, Cai Y, Yang S. Dachengqi decoction dispensing granule ameliorates LPS-induced acute lung injury by inhibiting PANoptosis in vivo and in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118699. [PMID: 39181290 DOI: 10.1016/j.jep.2024.118699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute lung injury (ALI) is a serious health-threatening syndrome of intense inflammatory response in the lungs, with progression leading to acute respiratory distress syndrome (ARDS). Dachengqi decoction dispensing granule (DDG) has a pulmonary protective role, but its potential modulatory mechanism to alleviate ALI needs further excavation. AIM OF THE STUDY This study aims to investigate the effect and potential mechanism of DDG on lipopolysaccharide (LPS)-induced ALI models in vivo and in vitro. MATERIALS AND METHODS LPS-treated Balb/c mice and BEAS-2B cells were used to construct in vivo and in vitro ALI models, respectively. Hematoxylin-eosin (HE), Wet weight/Dry weight (W/D) calculation of lung tissue, and total protein and Lactic dehydrogenase (LDH) assays in BALF were performed to assess the extent of lung tissue injury and pulmonary edema. Enzyme-linked immunosorbent assay (ELISA) was used to detect the levels of tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and interleukin-18 (IL-18) in BALF, serum, and cell supernatant. The qRT-PCR was used to detect inflammatory factors, Z-DNA binding protein 1 (ZBP1), and receptor-interacting protein kinase 1 (RIPK1) expression in lung tissues and BEAS-2B cells. Double immunofluorescence staining and co-immunoprecipitation were used to detect the relative expression and co-localization of ZBP1 and RIPK1. The effects of LPS and DDG on BEAS-2B cell activity were detected by Cell Counting Kit-8 (CCK-8). Western blot (WB) was performed to analyze the expression of PANoptosis-related proteins in lung tissues and BEAS-2B cells. RESULTS In vivo, DDG pretreatment could dose-dependently improve the pathological changes of lung tissue in ALI mice, and reduce the W/D ratio of lung, total protein concentration, and LDH content in BALF. In vitro, DDG reversed the inhibitory effect of LPS on BEAS-2B cell viability. Meanwhile, DDG significantly reduced the levels of inflammatory factors in vitro and in vivo. In addition, DDG could inhibit the expression levels of PANoptosis-related proteins, especially the upstream key regulatory molecules ZBP1 and RIPK1. CONCLUSION DDG could inhibit excessive inflammation and PANoptosis to alleviate LPS-induced ALI, thus possessing good anti-inflammatory and lung-protective effects. This study establishes a theoretical basis for the further development of DDG and provides a new prospect for ALI treatment by targeting PANoptosis.
Collapse
Affiliation(s)
- Mengqi Zhang
- Department of Clinical Nutrition, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1227 Jiefang Avenue, Wuhan City, 430022, Hubei Province, China
| | - Luorui Shang
- Department of Clinical Nutrition, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1227 Jiefang Avenue, Wuhan City, 430022, Hubei Province, China
| | - Fangyuan Zhou
- Department of Clinical Nutrition, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1227 Jiefang Avenue, Wuhan City, 430022, Hubei Province, China
| | - Jinxiao Li
- Department of Clinical Nutrition, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1227 Jiefang Avenue, Wuhan City, 430022, Hubei Province, China
| | - Shuhan Wang
- Department of Clinical Nutrition, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1227 Jiefang Avenue, Wuhan City, 430022, Hubei Province, China
| | - Qifeng Lin
- Department of Clinical Nutrition, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1227 Jiefang Avenue, Wuhan City, 430022, Hubei Province, China
| | - Yuju Cai
- Department of Clinical Nutrition, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1227 Jiefang Avenue, Wuhan City, 430022, Hubei Province, China
| | - Shenglan Yang
- Department of Clinical Nutrition, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1227 Jiefang Avenue, Wuhan City, 430022, Hubei Province, China.
| |
Collapse
|
10
|
Chen Y, Wu X, Jiang Z, Li X. KAE ameliorates LPS-mediated acute lung injury by inhibiting PANoptosis through the intracellular DNA-cGAS-STING axis. Front Pharmacol 2025; 15:1461931. [PMID: 39840115 PMCID: PMC11747328 DOI: 10.3389/fphar.2024.1461931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/19/2024] [Indexed: 01/23/2025] Open
Abstract
Background Acute lung injury (ALI) is a severe condition characterized by inflammation, tissue damage, and persistent activation of the cyclic GMP-AMP (cGAS)-stimulator of interferon genes (STING) pathway, which exacerbates the production of pro-inflammatory mediators and promotes the progression of ALI. Specific inhibition of this pathway has been shown to alleviate ALI symptoms. Kaempferol-3-O-α-L-(4″-E-p-coumaroyl)-rhamnoside (KAE), an active compound found in the flowers of Angelica acutiloba Kitagawa, exhibits anti-inflammatory and antioxidant properties. This study aimed to investigate the molecular mechanisms through which KAE regulates the cGAS-STING pathway in the context of ALI. Methods ALI was induced using LPS. Lung damage and anti-inflammatory/antioxidant effects were assessed by H&E staining, lung edema index, and SOD, MDA, and ELISA assays. NO release and mitochondrial membrane potential (MMP) were measured by JC-1 and Griess methods. The impact of KAE on the cGAS-STING pathway and PANoptosis was analyzed using flow cytometry, Western blot, and immunofluorescence. Results KAE significantly alleviated lipopolysaccharide-induced pulmonary injury by reducing inflammatory cell infiltration, alleviating pulmonary edema, enhancing antioxidant capacity, and decreasing levels of inflammatory cytokines in mouse lung tissues. In both in vitro and in vivo analyses, KAE downregulated the expression of key components of the cGAS-STING pathway, including cGAS, STING, p-TBK1, and nuclear factor-κB. KAE also reduced the assembly and activation of the PANoptosome, thereby attenuating apoptosis, necroptosis, and pyroptosis. Additionally, KAE inhibited cGAS activation by restoring the MMP, which reduced the release of cytosolic DNA. Conclusion KAE improve ALI by inhibiting the release of cytosolic DNA and suppressing cGAS-STING pathway activation, thereby protecting cells from PANoptosis. Our findings provide valuable insights for the development and application of novel therapeutic strategies for ALI.
Collapse
Affiliation(s)
| | | | | | - Xuezheng Li
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, China
| |
Collapse
|
11
|
Zhi H, Fu H, Zhang Y, Fan N, Zhao C, Li Y, Sun Y, Li Y. Progress of cGAS-STING signaling pathway-based modulation of immune response by traditional Chinese medicine in clinical diseases. Front Immunol 2024; 15:1510628. [PMID: 39737190 PMCID: PMC11683013 DOI: 10.3389/fimmu.2024.1510628] [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: 10/13/2024] [Accepted: 11/29/2024] [Indexed: 01/01/2025] Open
Abstract
The cGAS-STING signaling pathway is a critical component of the innate immune response, playing a significant role in various diseases. As a central element of this pathway, STING responds to both endogenous and exogenous DNA stimuli, triggering the production of interferons and pro-inflammatory cytokines to enhance immune defenses against tumors and pathogens. However, dysregulated activation of the STING pathway is implicated in the pathogenesis of multiple diseases, including autoinflammation, viral infections, and cancer. Traditional Chinese Medicines (TCMs), which have a long history of use, have been associated with positive effects in disease prevention and treatment. TCM formulations (e.g., Lingguizhugan Decoction, Yi-Shen-Xie-Zhuo formula) and active compounds (e.g., Glabridin, Ginsenoside Rd) can modulate the cGAS-STING signaling pathway, thereby influencing the progression of inflammatory, infectious, or oncological diseases. This review explores the mechanisms by which TCMs interact with the cGAS-STING pathway to regulate immunity, focusing on their roles in infectious diseases, malignancies, and autoimmune disorders.
Collapse
Affiliation(s)
- Hui Zhi
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hui Fu
- College of Integrated Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunxin Zhang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ni Fan
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chengcheng Zhao
- Experimental Teaching and Practical Training Center, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yunfei Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yujiao Sun
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingpeng Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
12
|
Cui Z, Li Y, Bi Y, Li W, Piao J, Ren X. PANoptosis: A new era for anti-cancer strategies. Life Sci 2024; 359:123241. [PMID: 39549938 DOI: 10.1016/j.lfs.2024.123241] [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: 07/29/2024] [Revised: 10/28/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024]
Abstract
Cancer cells possess an extraordinary ability to dodge cell death through various pathways, granting them a form of immortality-a key obstacle in oncotherapy. Thus, it's vital to unravel the intricate mechanisms behind newly discovered types of cell death that drive tumor suppression, going beyond apoptosis alone. The emergence of PANoptosis, a form of cell death intertwining necroptosis, pyroptosis, and apoptosis, offers a fresh perspective, integrating these pathways into one cohesive process. When cells detect damage signals, they assemble PANoptosome complexes that disrupt their balance, trigger immune responses, and lead to their eventual collapse. PANoptosis has been associated with multiple cellular pathways, including ferroptosis. Mitochondrial dysfunction also plays a critical role in sparking and advancing PANoptosis. In this review, we map out the molecular machinery and regulatory web controlling PANoptosis. We explore cutting-edge research and future trends in PANoptosis-centered tumor therapies, spotlighting promising innovations that could amplify cancer treatment effectiveness through harnessing this multifaceted cell death pathway. The development of nanomedicines and nanomaterials provides solutions to the therapeutic challenges of clinical drugs. Developing novel tumor nano-PANoptosis inducers by leveraging the advantages of nanomedicine is of research value. Traditional Chinese medicine (TCM) treatment is characterized by multiple targets, and it has distinct advantages in triggering PANoptosis through multiple pathways. Additionally, photodynamic Therapy (PDT) may offer new insights into promoting PANoptosis in tumor cells by increasing oxidative stress and reactive oxygen species levels. These will establish a solid theoretical groundwork for the development of integrated treatment methodologies.
Collapse
Affiliation(s)
- Ziheng Cui
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China
| | - Yuan Li
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China
| | - Yao Bi
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China
| | - Wenjing Li
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China; Department of Anesthesia, Affiliated Hospital of Yanbian University, Yanji, China
| | - Junjie Piao
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China
| | - Xiangshan Ren
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China; Central Laboratory, Yanbian University Hospital & Key Laboratory of Pathobiology, Yanbian University, State Ethnic Affairs Commission, Yanji, China.
| |
Collapse
|
13
|
Zhang YJ, Chen LY, Lin F, Zhang X, Xiang HF, Rao Q. ROS responsive nanozyme loaded with STING silencing for the treatment of sepsis-induced acute lung injury. Toxicol Appl Pharmacol 2024; 493:117155. [PMID: 39537108 DOI: 10.1016/j.taap.2024.117155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Acute lung injury (ALI) is a common complication of sepsis and a leading cause of mortality in septic patients. Studies indicate that STING may play a crucial role in the pathogenesis of sepsis-induced ALI by interacting with the PARP-1/NLRP3 pathway. Therefore, targeting STING inhibition has potential as a novel therapeutic strategy for ALI. However, effective inhibition remains challenging due to the widespread expression of STING across various tissues. In this study, we developed a nanozyme-based drug delivery system, DSPE-TK-mPEG-MnO2@siSTING (abbreviated as DTmM@siSTING), using DSPE-TK-mPEG-MnO2 as the carrier, and characterized it via scanning electron microscopy, dynamic light scattering, nanoparticle size analysis, and gel electrophoresis. To evaluate the therapeutic effects of DTmM@siSTING, an in vitro ALI cell model and an in vivo ALI mouse model were established, assessing the nanozyme's impact on ROS levels, inflammatory responses, and the PARP-1/NLRP3 pathway in sepsis-induced ALI. Results demonstrated that DTmM@siSTING exhibited good physiological stability. In vitro, DTmM@siSTING significantly reduced ROS levels, myeloperoxidase activity, and expression of inflammatory cytokines, while also inhibiting PARP-1/NLRP3 pathway activation. In vivo experiments further revealed that DTmM@siSTING effectively delivered siSTING to the lungs, mitigating sepsis-induced ALI and associated inflammatory responses. Additionally, DTmM@siSTING displayed excellent biocompatibility. In summary, our findings suggest that DTmM@siSTING significantly enhances the therapeutic efficacy of siSTING, alleviating ALI by inhibiting ROS production, inflammatory responses, and activation of the PARP-1/NLRP3 pathway. This novel approach presents a promising therapeutic avenue for sepsis-induced ALI.
Collapse
Affiliation(s)
- Yin-Jin Zhang
- Blood Purification Center, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Ling-Yang Chen
- Blood Purification Center, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Feng Lin
- Department of Anesthesiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Xia Zhang
- Department of Anesthesiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Hai-Fei Xiang
- Department of Anesthesiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, China.
| | - Qing Rao
- Department of Anesthesiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, China.
| |
Collapse
|
14
|
Li Y, Luo Y, Wang C, Xu L, Dai X, An Y, He L, Zeng D, Bai Y, Zhang H. VSV infection and LPS treatment alter serum bile acid profiles, bile acid biosynthesis, and bile acid receptors in mice. Microbiol Spectr 2024; 12:e0083624. [PMID: 39287458 PMCID: PMC11537081 DOI: 10.1128/spectrum.00836-24] [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] [Accepted: 08/13/2024] [Indexed: 09/19/2024] Open
Abstract
Pathogen infections remain a significant public health problem worldwide. Accumulating evidence regarding the crosstalk between bile acid (BA) metabolism and immune response reveals that BA metabolism regulates host immunity and microbial pathogenesis, making it an attractive target for disease prevention and infection control. However, the effect of infection on circulating BA profiles, the biosynthesis-related enzymes, and their receptors remains to be depicted. Here, we investigated the effect of viral (vesicular stomatitis virus, VSV) and bacterial (lipopolysaccharide, LPS) infections on BA metabolism and signaling. Infection models were successfully established by intraperitoneally injecting VSV and LPS, respectively. VSV and LPS injection significantly changed the circulating BA profiles, with highly increased levels of taurine-conjugated BAs and significant decreases in unconjugated BAs. Consistent with the decreased levels of circulating cholic acid (CA) and chenodeoxycholic acid (CDCA), the expression of BA biosynthesis-related rate-limiting enzymes (Cyp7a1, Cyp27a1, Cyp8b1, and Hsd3b7) were significantly reduced. Furthermore, hepatic and pulmonary BA receptors (BARs) expression varied in different infection models. LPS treatment had an extensive impact on tested hepatic and pulmonary BARs, resulting in the upregulation of TGR5, S1PR2, and VDR, while VSV infection only promoted VDR expression. Our study provides insights into the involvement of BA metabolism in the pathophysiology of infection, which may provide potential clues for targeting BA metabolism and BAR signaling to boost innate immunity and control infection. IMPORTANCE This study focuses on the crosstalk between bile acid (BA) metabolism and immune response in VSV infection and LPS treatment models and depicts the effect of infection on circulating BA profiles, the biosynthesis-related enzymes, and their receptors. These findings provide insights into the effect of infection on BA metabolism and signaling, adding a more comprehensive understanding to the relationship between infection, BA metabolism and immune responses.
Collapse
Affiliation(s)
- Yamei Li
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan, China
| | - Yan Luo
- Department of Pathology, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Chao Wang
- Department of Pathology, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Lei Xu
- Department of Pathology, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Xinhua Dai
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan, China
| | - Yunfei An
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan, China
| | - Lin He
- Department of Pathology, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Dongmei Zeng
- Department of Pathology, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Yangjuan Bai
- Department of Laboratory Medicine/Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan, China
| | - Hua Zhang
- Department of Pathology, General Hospital of Western Theater Command, Chengdu, Sichuan, China
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| |
Collapse
|
15
|
Wu L, Liu Y, Zhang Y, Xu R, Bi K, Li J, Wang J, Liu Y, Guo W, Wang Q, Chen Z. Identification of PANoptosis-related genes for idiopathic pulmonary fibrosis by machine learning and molecular subtype analysis. Sci Rep 2024; 14:24068. [PMID: 39402203 PMCID: PMC11473738 DOI: 10.1038/s41598-024-76263-7] [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: 03/27/2024] [Accepted: 10/11/2024] [Indexed: 10/17/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a severe interstitial lung disease characterized by a grim prognosis, in which various forms of cell death are significant contributors to its development. The objective of this study is to explore diagnostic biomarkers and molecular subtypes associated with PANoptosis in IPF, and to develop reliable diagnostic models based on PANoptosis-related mechanisms. The peripheral blood transcriptomic data of IPF from the Gene Expression Omnibus (GEO) database and PANoptosis-related genes from the GeneCards database were utilized to conduct differential gene expression analysis and weighted gene co-expression network analysis (WGCNA), identifying PANoptosis-related differentially expressed genes (PDEGs). We yielded 9 PDEGs and employed machine learning algorithms to identify 3 key diagnostic biomarkers for PANoptosis in IPF: MMP9, FCMR, NIBAN3. Consensus clustering algorithm was applied to recognize two PANoptosis-related subtypes. Cluster 1 exhibited higher abundance of adaptive immune response cells and significant enrichment in DNA damage and repair-related pathways. Cluster 2 exhibited greater prevalence of innate immune response cells and predominant enhancement in pathways related to lipid cholesterol metabolism and vascular remodeling. Diagnostic models were developed with the aid of clinical decision-making and a novel approach to the diagnosis and treatment for IPF.
Collapse
Affiliation(s)
- Li Wu
- Department of Anesthesiology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Yang Liu
- School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Yifan Zhang
- The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Rui Xu
- Department of Anesthesiology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Kaixin Bi
- The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Jing Li
- Department of Anesthesiology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Jia Wang
- Department of Anesthesiology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Yabing Liu
- Department of Anesthesiology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Wanjin Guo
- Department of Respiratory and Critical Care Medicine, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China.
| | - Qi Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China.
- School of Management, Shanxi Medical University, Taiyuan, China.
| | - Zhiqiang Chen
- Department of Galactophore, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, 030000, Taiyuan, China.
| |
Collapse
|
16
|
Feng R, Zhao J, Zhang Q, Zhu Z, Zhang J, Liu C, Zheng X, Wang F, Su J, Ma X, Mi X, Guo L, Yan X, Liu Y, Li H, Chen X, Deng Y, Wang G, Zhang Y, Liu X, Liu J. Generation of Anti-Mastitis Gene-Edited Dairy Goats with Enhancing Lysozyme Expression by Inflammatory Regulatory Sequence using ISDra2-TnpB System. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404408. [PMID: 39099401 PMCID: PMC11481229 DOI: 10.1002/advs.202404408] [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: 04/24/2024] [Revised: 06/29/2024] [Indexed: 08/06/2024]
Abstract
Gene-editing technology has become a transformative tool for the precise manipulation of biological genomes and holds great significance in the field of animal disease-resistant breeding. Mastitis, a prevalent disease in animal husbandry, imposes a substantial economic burden on the global dairy industry. In this study, a regulatory sequence gene editing breeding strategy for the successful creation of a gene-edited dairy (GED) goats with enhanced mastitis resistance using the ISDra2-TnpB system and dairy goats as the model animal is proposed. This included the targeted integration of an innate inflammatory regulatory sequence (IRS) into the promoter region of the lysozyme (LYZ) gene. Upon Escherichia Coli (E. coli) mammary gland infection, GED goats exhibited increased LYZ expression, showing robust anti-mastitis capabilities, mitigating PANoptosis activation, and alleviating blood-milk-barrier (BMB) damage. Notably, LYZ is highly expressed only in E. coli infection. This study marks the advent of anti-mastitis gene-edited animals with exogenous-free gene expression and demonstrates the feasibility of the gene-editing strategy proposed in this study. In addition, it provides a novel gene-editing blueprint for developing disease-resistant strains, focusing on disease specificity and biosafety while providing a research basis for the widespread application of the ISDra2-TnpB system.
Collapse
Affiliation(s)
- Rui Feng
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Jianglin Zhao
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Qian Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Zhenliang Zhu
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Junyu Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Chengyuan Liu
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Xiaoman Zheng
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Fan Wang
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Jie Su
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Xianghai Ma
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Xiaoyu Mi
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Lin Guo
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Xiaoxue Yan
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Yayi Liu
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Huijia Li
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Xu Chen
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Yi Deng
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Guoyan Wang
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Yong Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Xu Liu
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| | - Jun Liu
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureCollege of Veterinary MedicineNorthwest Agriculture & Forestry UniversityYanglingShaanxi712100China
| |
Collapse
|
17
|
Han Y, Qiu L, Wu H, Song Z, Ke P, Wu X. Focus on the cGAS-STING Signaling Pathway in Sepsis and Its Inflammatory Regulatory Effects. J Inflamm Res 2024; 17:3629-3639. [PMID: 38855170 PMCID: PMC11162626 DOI: 10.2147/jir.s465978] [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: 02/26/2024] [Accepted: 05/29/2024] [Indexed: 06/11/2024] Open
Abstract
Sepsis is a severe systemic inflammatory response commonly occurring in infectious diseases, caused by infection with virulent pathogens. In the pathogenesis of sepsis, the cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) synthase-stimulator of interferon genes (cGAS-STING) signaling pathway serves a crucial role as a fundamental immunoregulatory mechanism. This signaling pathway activates STING upon recognizing intracellular DNA damage and pathogen-derived DNA, subsequently inducing the production of numerous inflammatory mediators, including interferon and inflammatory cytokines, which in turn trigger an inflammatory response. The aim of this paper is to explore the activation mechanism of the cGAS-STING signaling pathway in sepsis and its impact on inflammatory regulation. By delving into the mechanism of action of the cGAS-STING signaling pathway in sepsis, we aim to identify new therapeutic strategies for the treatment and prevention of sepsis.
Collapse
Affiliation(s)
- Yupeng Han
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
- Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, Fujian, People’s Republic of China
| | - Liangcheng Qiu
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
- Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, Fujian, People’s Republic of China
| | - Haixing Wu
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
- Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, Fujian, People’s Republic of China
| | - Zhiwei Song
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
| | - Peng Ke
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
- Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, Fujian, People’s Republic of China
| | - Xiaodan Wu
- Department of Anesthesiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People’s Republic of China
- Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, Fujian, People’s Republic of China
| |
Collapse
|
18
|
Dong J, Liu W, Liu W, Wen Y, Liu Q, Wang H, Xiang G, Liu Y, Hao H. Acute lung injury: a view from the perspective of necroptosis. Inflamm Res 2024; 73:997-1018. [PMID: 38615296 DOI: 10.1007/s00011-024-01879-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/23/2024] [Accepted: 03/31/2024] [Indexed: 04/15/2024] Open
Abstract
BACKGROUND ALI/ARDS is a syndrome of acute onset characterized by progressive hypoxemia and noncardiogenic pulmonary edema as the primary clinical manifestations. Necroptosis is a form of programmed cell necrosis that is precisely regulated by molecular signals. This process is characterized by organelle swelling and membrane rupture, is highly immunogenic, involves extensive crosstalk with various cellular stress mechanisms, and is significantly implicated in the onset and progression of ALI/ARDS. METHODS The current body of literature on necroptosis and ALI/ARDS was thoroughly reviewed. Initially, an overview of the molecular mechanism of necroptosis was provided, followed by an examination of its interactions with apoptosis, pyroptosis, autophagy, ferroptosis, PANOptosis, and NETosis. Subsequently, the involvement of necroptosis in various stages of ALI/ARDS progression was delineated. Lastly, drugs targeting necroptosis, biomarkers, and current obstacles were presented. CONCLUSION Necroptosis plays an important role in the progression of ALI/ARDS. However, since ALI/ARDS is a clinical syndrome caused by a variety of mechanisms, we emphasize that while focusing on necroptosis, it may be more beneficial to treat ALI/ARDS by collaborating with other mechanisms.
Collapse
Affiliation(s)
- Jinyan Dong
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China
| | - Weihong Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China
| | - Wenli Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China
| | - Yuqi Wen
- Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China
| | - Qingkuo Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China
| | - Hongtao Wang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China
| | - Guohan Xiang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China
| | - Yang Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China.
| | - Hao Hao
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China.
| |
Collapse
|