1
|
Liu A, Gao XH, Mao Y, Zhang XQ, Xie WX, Su B, Li CY, Chen ZY. Esketamine mitigates lung injury in COPD rat models under mechanical ventilation: An RNA-sequencing and bioinformatics analysis of serum exosome miRNA profiles. Gene 2025; 962:149571. [PMID: 40389067 DOI: 10.1016/j.gene.2025.149571] [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: 05/07/2025] [Accepted: 05/12/2025] [Indexed: 05/21/2025]
Abstract
PURPOSE This study aims to investigate the protective effects of esketamine on lung injury in rat models of chronic obstructive pulmonary disease (COPD) under mechanical ventilation. The focus is on serum exosome miRNA profiles through RNA sequencing and bioinformatics analysis. METHODS Male Sprague-Dawley rats were categorized into four groups: Control (C), COPD model (M), COPD model + saline (MS), and COPD model + esketamine (MK). After inducing COPD and subjecting the animals to mechanical ventilation, lung structure, and respiratory function were evaluated. Serum exosomes were isolated for RNA sequencing to analyze differentially expressed miRNAs and their associated pathways. RESULTS Intervention with esketamine significantly improved lung histology, reduced inflammation, and enhanced respiratory mechanics in the MK group compared to the other COPD groups. Peak airway pressure, wet-to-dry weight ratio, and lung permeability index significantly improved. Analysis of serum exosome miRNA revealed the downregulation of several miRNAs, including rno-miR-143-3p, rno-miR-130a-3p, rno-miR-505-5p, rno-miR-122-5p, rno-miR-145-5p, and rno-miR-1b. These miRNAs were linked to target genes involved in critical pathways such as TGF-beta, PI3K-Akt, mTOR, and Ras signaling. Gene Ontology (GO) analysis indicated significant enrichment in biological processes related to signal transduction, cellular stress response, and intracellular trafficking. CONCLUSION Esketamine demonstrates significant potential to alleviate lung injury in COPD rat models subjected to mechanical ventilation, likely through modulating critical pathways related to inflammation, fibrosis, and cellular stress. Further research is needed to confirm these findings in clinical settings.
Collapse
Affiliation(s)
- Ang Liu
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University ,Quanzhou 362000,China; Department of Anesthesiology, Heze Municipal Hospital, Heze 274000, China
| | - Xiao-Hua Gao
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University ,Quanzhou 362000,China
| | - Yu Mao
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University ,Quanzhou 362000,China
| | - Xiao-Qi Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University ,Quanzhou 362000,China
| | - Wen-Xi Xie
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University ,Quanzhou 362000,China
| | - Bin Su
- Department of Anesthesiology, Heze Municipal Hospital, Heze 274000, China
| | - Chui-Yu Li
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University ,Quanzhou 362000,China.
| | - Zhi-Yuan Chen
- Department of Anesthesiology, the Second Affiliated Hospital of Fujian Medical University ,Quanzhou 362000,China.
| |
Collapse
|
2
|
Wang L, Guo H, Zhao W, Wang J, Cao X. Oxiracetam ameliorates neurological function after traumatic brain injury through competing endogenous RNA regulatory network. Psychopharmacology (Berl) 2025:10.1007/s00213-025-06797-9. [PMID: 40272502 DOI: 10.1007/s00213-025-06797-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
RATIONALE Oxiracetam (ORC) has been demonstrated to improve neurological function resulting from traumatic brain injury (TBI). OBJECTIVES This study aims to explore the precise molecular mechanism of ORC in the treatment of TBI. METHODS TBI rat model was established and treated with ORC. Modified Garcia score, rotarod test and HE staining were employed to evaluate the neuroprotective effects of ORC. Subsequently, RNA-seq was conducted on the hippocampus of sham, TBI and ORC rats to identify differential expression (DE) lncRNAs and mRNAs. Functional analysis of DE lncRNAs and mRNAs was performed. The real-time quantitative polymerase chain reaction (qRT-PCR) was used to determine the expression of DE lncRNAs and DE mRNAs. Western blot was performed to explore important pathway in ceRNA networks. RESULTS ORC has been demonstrated to effectively improve neurological function in TBI rats. A total of 10 ORC-treated DE lncRNAs and 61 DE mRNAs were obtained. A co-expression network comprising 79 lncRNA-mRNA pairs associated with the treatment of ORC was constructed. Furthermore, an lncRNA-miRNA-mRNA regulated ceRNA network was constructed, comprising 15 mRNAs, 41 miRNAs and 10 lncRNAs. Functional enrichment, qRT-PCR, and Western blot analysis showed that ORC improve neurological function of TBI rats by regulating multiple signaling pathways, including the JAK-STAT/PI3K-Akt pathway, as well as affecting the expression of key genes Prlr, Cdkn1a, and Cldn1. CONCLUSION Our study reveals the mechanism of ORC therapy in TBI rats, which mainly relies on the regulation of the JAK-STAT/PI3K-Akt pathway and the influence on the expression of key genes Prlr, Cdkn1a, and Cldn1.
Collapse
Affiliation(s)
- Liyi Wang
- Hospital Infection-Control Department, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China
| | - Han Guo
- Department of Oral Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China
| | - Weidong Zhao
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China
| | - Jiahao Wang
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China
| | - Xuhua Cao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Xinhua District, Shijiazhuang, Hebei Province, 050000, China.
| |
Collapse
|
3
|
Qin F, Tian L, Wang Z, Qiu X. Schisandrol A Mitigated Voriconazole-induced Liver Injury in Mice Through Regulation of Farnesoid X Receptor-mediated Bile Acid Metabolism. Chem Biodivers 2025:e202402769. [PMID: 40059080 DOI: 10.1002/cbdv.202402769] [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: 10/31/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 04/14/2025]
Abstract
Schisandrol A has been previously used to mitigate hepatotoxicity. However, the effects of schisandrol A on voriconazole-induced hepatic injury have not been investigated. In this study, we aimed to explore the effects of schisandrol A on voriconazole-induced hepatic injury in mice, as well as to elucidate the underlying mechanism. Mice were continuously treated with voriconazole with or without schisandrol A administration. Acetaminophen was used as a positive control to induce liver damage. Hematological, histological, and gene analyses were conducted, and the therapeutic target of schisandrol A was verified. Our results showed that voriconazole-induced hepatic injury was similar to that induced by acetaminophen. Importantly, schisandrol A alleviated voriconazole-induced hepatic steatosis, cell death, inflammation, and fibrosis. Schisandrol A reduced oxidative damage and inflammation in the liver. Furthermore, schisandrol A reduced total bile acid, cholesterol, and triglyceride levels in the liver and serum. The expression of farnesoid X receptor (FXR), small heterodimer partner, cytochrome P450 (CYP) 7A1, and sterol 12α-hydroxylase in the liver was altered by schisandrol A treatment. The binding of schisandrol A to FXR was verified by the cellular thermal shift assay and molecular docking. In conclusion, these findings suggested the favorable effects of schisandrol A against voriconazole-induced hepatotoxicity.
Collapse
Affiliation(s)
- Fangxu Qin
- Department of Clinical Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine, Zhongshan, China
| | - Lifeng Tian
- Department of Clinical Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine, Zhongshan, China
| | - Zhien Wang
- Department of Clinical Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine, Zhongshan, China
| | - Xiongquan Qiu
- Department of Clinical Pharmacy, Zhongshan Hospital of Traditional Chinese Medicine, Zhongshan, China
| |
Collapse
|
4
|
Liu X, Huang Q, Li W, Yu J, Yu J, Yang Y, Song H, Liu Y, Niu X, Li W. The inhibitory impact of Schisandrin on inflammation and oxidative stress alleviates LPS-induced acute kidney injury. Biotechnol Appl Biochem 2024; 71:1116-1128. [PMID: 38798098 DOI: 10.1002/bab.2602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/16/2024] [Accepted: 05/05/2024] [Indexed: 05/29/2024]
Abstract
Inflammation and oxidative stress (OS) are the major pathogenic characteristics of acute kidney injury (AKI). Studies have shown that Schisandrin (Sch) could regulate inflammatory disease. However, the function and mechanism of Sch in AKI progression are still unknown. Here, we investigated Sch's potential effects and mechanism on mice's renal damage and macrophages induced by lipopolysaccharide (LPS). Sch decreased LPS-induced inflammatory factor production while increasing the activity of related antioxidant enzymes in macrophages and mouse kidney tissues. Hematoxylin and eosin staining revealed that Sch may have the ability to profoundly inhibit inflammatory cell invasion and tissue damage caused by LPS in renal tissue. Furthermore, Western blot and immunohistochemical studies showed that Sch exerted its effects mainly through up-regulation of nuclear factor erythroid 2-related factor 2/heme oxygenase-1 and inhibition of Toll-like receptor 4‒mitogen-activated protein kinases/nuclear factor-kappa B pathways. Collectively, this study illustrates that Sch suppresses LPS-stimulated AKI by descending inflammation and OS, illuminating prospective AKI treatment options.
Collapse
Affiliation(s)
- Xinyao Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Qiuxia Huang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Wenqi Li
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P. R. China
| | - Jinjin Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Jiabao Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Yajie Yang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Huixin Song
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Yang Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, P. R. China
| |
Collapse
|
5
|
Liu X, Liu X, Wang H, Chen M, Zhang G, Ren D, Zhang N, Wei W. Quantitative proteomic analysis of circulating exosomes reveals the mechanism by which Triptolide protects against collagen-induced arthritis. Immun Inflamm Dis 2024; 12:e1322. [PMID: 38888462 PMCID: PMC11184934 DOI: 10.1002/iid3.1322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024] Open
Abstract
INTRODUCTION Triptolide (TP), a natural product derived from the herbal medicine Tripterygium wilfordii, exhibits potent immunosuppressive activity. However, the mechanisms underlying its effects in rheumatoid arthritis remain incompletely understood. METHODS Collagen-induced arthritis (CIA) model was induced in Sprague-Dawley rats by immunization with bovine type II collagen, and TP was administrated as treatment. The therapeutic effect of TP was evaluated based on paw swelling, histopathology, and serum levels of inflammatory factors. Exosomes isolated from rat serum were characterized by transmission electron microscopy, dynamic light scattering, and western blot analysis. Proteomic profiling of exosomes was analyzed by direct DIA quantitative proteomics analysis. Gene ontology and the Kyoto Encyclopedia of Genes and Genomes databases were employed for enrichment analysis related to molecular function, biological processes, and signaling pathways. Western blot analysis was used to analyze differentially expressed proteins. RESULTS TP treatment ameliorated arthritic phenotypes in CIA rats as evidenced by reduced arthritis score, paw swelling, pathological injury severity scores, and serum levels of inflammatory cytokines. The proteomic analysis revealed that TP treatment significantly inhibited complement and coagulation cascades, interleukin-17 signaling pathway, and cholesterol metabolism, which were reactivated in CIA rats. Importantly, lipocalin 2 (LCN2) and myeloperoxidase (MPO) levels were markedly upregulated in the CIA group but suppressed upon TP administration. Furthermore, in synovial tissues, LCN2 and MPO expression levels were also elevated in the CIA group but decreased following TP treatment. CONCLUSION Our findings demonstrate that TP alleviates CIA, possibly through modulation of exosomal LCN2 and MPO proteins.
Collapse
Affiliation(s)
- Xiuchan Liu
- Department of Infectious DiseasesTianjin HospitalTianjinChina
- Department of Rheumatology and ImmunologyTianjin Medical University General HospitalTianjinChina
| | - Xu Liu
- Department of Infectious DiseasesTianjin Medical University General Hospital, Binhai HospitalTianjinChina
| | - Hui Wang
- Department of Rheumatology and ImmunologyTianjin Medical University General HospitalTianjinChina
| | - Ming Chen
- Department of Rheumatology and ImmunologyTianjin Medical University General HospitalTianjinChina
| | - Geng Zhang
- Department of Infectious DiseasesTianjin Medical University Baodi Clinical CollegeTianjinChina
| | - Dongyun Ren
- Department of Infectious DiseasesTianjin HospitalTianjinChina
| | - Na Zhang
- Department of Rheumatology and ImmunologyTianjin Medical University General HospitalTianjinChina
| | - Wei Wei
- Department of Rheumatology and ImmunologyTianjin Medical University General HospitalTianjinChina
| |
Collapse
|
6
|
Chen H, Wang Z, Gong L, Chen J, Huang Y, Guo W, Zhang Q, Li Y, Bao G, Li D, Chen Y. Attenuation effect of a polysaccharide from large leaf yellow tea by activating autophagy. Int J Biol Macromol 2024; 265:130697. [PMID: 38490395 DOI: 10.1016/j.ijbiomac.2024.130697] [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: 09/06/2023] [Revised: 02/20/2024] [Accepted: 03/05/2024] [Indexed: 03/17/2024]
Abstract
Chemotherapy, the most common class of anticancer drugs, is considerably limited owing to its adverse side effects. In this study, we aimed to evaluate the protective effect and mechanism of action of large-leaf yellow tea polysaccharides (ULYTP-1, 1.29 × 104 Da) against chemotherapeutic 5-fluorouracil (5-Fu). Structural characterisation revealed that ULYTP-1 was a β-galactopyranouronic acid. Furthermore, ULYTP-1 promoted autolysosome formation, activating autophagy and reducing the oxidative stress and inflammation caused by 5-Fu. Our in vivo study of 4 T1 tumour-bearing mice revealed that ULYTP-1 also attenuated 5-Fu toxicity through modulation of the gut microbiota. Moreover, ULYTP-1 effectively protected immune organs and the liver from 5-Fu toxicity, while promoting its tumour-inhibitory properties. The current findings provide a new strategy for optimising chemotherapy regimens in the clinic.
Collapse
Affiliation(s)
- Hao Chen
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Zhuang Wang
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Lei Gong
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Jielin Chen
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Yuzhe Huang
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Wenqiang Guo
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Qiang Zhang
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Yong Li
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Guanhu Bao
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China.
| | - Daxiang Li
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, Anhui, China.
| | - Yan Chen
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China.
| |
Collapse
|
7
|
Chen S, Qin F, Yang Y, Zhao Y, Xiao S, Li W, Akihisa T, Jantrawut P, Ji J, Zhang J. Extraction, purification, structural characterization, and bioactivities of the genus Schisandra polysaccharides: A review. Int J Biol Macromol 2024; 262:130257. [PMID: 38423904 DOI: 10.1016/j.ijbiomac.2024.130257] [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: 11/06/2023] [Revised: 02/11/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024]
Abstract
The genus Schisandra, a member of the Magnoliaceae family, is a well-known tonic traditional Chinese medicine with a long history of traditional medicinal and functional food used in China. Polysaccharides are one of its main active constituents, which have a wide range of bioactivities, such as anti-inflammatory, anti-tumor, neuroprotection, anti-diabetes, hepatoprotection, immunomodulation, and anti-fatigue. In this paper, we review the extraction, isolation, purification, structural characterization, bioactivities, as well as structure-activity relationship of polysaccharides from the genus Schisandra. In conclusion, we hope that this review could provide reference for the subsequent research on structural, bioactivities, development and application of the genus Schisandra polysaccharides.
Collapse
Affiliation(s)
- Shujun Chen
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211112, China
| | - Fang Qin
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211112, China
| | - Ying Yang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211112, China
| | - Yu Zhao
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211112, China
| | - Shuyun Xiao
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211112, China
| | - Wei Li
- Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan
| | - Toshihiro Akihisa
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211112, China; Research Institute for Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Pensak Jantrawut
- Faculty of Pharmacy, Ching Mai University, Ching Mai, 50200, Thailand
| | - Jingyu Ji
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211112, China
| | - Jie Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211112, China.
| |
Collapse
|