1
|
Weng J, Weng S, Xu J, Liu D, Guo R, Shi B, Chen M. Tocilizumab alleviated lipopolysaccharide-induced acute lung injury by improving PI3K/AKT pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03786-9. [PMID: 39797985 DOI: 10.1007/s00210-025-03786-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
Acute lung injury (ALI) is a severe inflammatory condition of the respiratory system, associated with high morbidity and mortality. This study investigates the therapeutic potential of tocilizumab (TZ), an IL-6 receptor inhibitor, in mitigating lipopolysaccharide (LPS)-induced ALI by modulating the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) pathway. An ALI model was established using LPS induction. Lung damage was assessed by hematoxylin-eosin (H&E) staining, alongside measurements of respiratory function, including PaO2/Fio2 ratios, lung edema, airway resistance, and lung compliance. Western blotting analyzed the expression of phosphorylated PI3K and AKT (P-PI3K, P-AKT), while ELISA quantified levels of TNF-α, IL-1β, IL-6, and oxidative stress markers. Apoptosis was evaluated using the TUNEL assay, and key apoptotic proteins (Bcl-2, Bax, and caspase-3) were measured by Western blotting. The Cell Counting Kit-8 (CCK-8) assay was employed to determine cell viability. The LPS-induced model exhibited decreased P-PI3K and P-AKT levels, while TZ treatment significantly elevated these markers. TZ also reduced lung tissue damage, improved respiratory function, and decreased inflammation, oxidative stress, and apoptosis. However, co-administration with LY294002 (a PI3K inhibitor) blocked these benefits, reversing the protective effects of TZ. TZ alleviates lung injury and improves outcomes in LPS-induced ALI by enhancing the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Junting Weng
- Department of Clinical Medicine, Fujian Medical University, Fuzhou, 350000, China
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, No. 999 Dongzhen East Road, Licheng District, Putian, 351100, China
| | - Shuoyun Weng
- Wenzhou Medical University School of Optometry and Ophthalmology, Wenzhou, 325035, China
| | - Jitao Xu
- Department of Clinical Medicine, Fujian Medical University, Fuzhou, 350000, China
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, No. 999 Dongzhen East Road, Licheng District, Putian, 351100, China
| | - Danjuan Liu
- Department of Clinical Medicine, Fujian Medical University, Fuzhou, 350000, China
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, No. 999 Dongzhen East Road, Licheng District, Putian, 351100, China
| | - Rongjie Guo
- Department of Clinical Medicine, Fujian Medical University, Fuzhou, 350000, China
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, No. 999 Dongzhen East Road, Licheng District, Putian, 351100, China
| | - Bingbing Shi
- Department of Clinical Medicine, Fujian Medical University, Fuzhou, 350000, China
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, No. 999 Dongzhen East Road, Licheng District, Putian, 351100, China
| | - Min Chen
- Department of Clinical Medicine, Fujian Medical University, Fuzhou, 350000, China.
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, No. 999 Dongzhen East Road, Licheng District, Putian, 351100, China.
| |
Collapse
|
2
|
Sun Z, He W, Meng H, Li P, Qu J. Endoplasmic reticulum stress in acute lung injury and pulmonary fibrosis. FASEB J 2024; 38:e70232. [PMID: 39651914 DOI: 10.1096/fj.202401849rr] [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: 08/10/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/19/2024]
Abstract
Pulmonary fibrosis (PF) is a progressive and irreversible lung disease that leads to diminished lung function, respiratory failure, and ultimately death and typically has a poor prognosis, with an average survival time of 2 to 5 years. Related articles suggested that endoplasmic reticulum (ER) stress played a critical role in the occurrence and progression of PF. The ER is responsible for maintaining protein homeostasis. However, factors such as aging, hypoxia, oxidative stress, or inflammation can disrupt this balance, promoting the accumulation of misfolded proteins in the ER and triggering ER stress. To cope with this situation, cells activate the unfolded protein response (UPR). Since acute lung injury (ALI) is one of the key onset events of PF, in this review, we will discuss the role of ER stress in ALI and PF by activating multiple signaling pathways and molecular mechanisms that affect the function and behavior of different cell types, with a focus on epithelial cells, fibroblasts, and macrophages. Linking ER stress to these cell types may broaden our understanding of the mechanisms underlying lung fibrosis and help us target these cells through these mechanisms. The relationship between ER stress and PF is still evolving, and future research will explore new strategies to regulate UPR pathways, providing novel therapeutic targets.
Collapse
Affiliation(s)
- Zhiheng Sun
- College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
- State Key Laboratory of Cell Differentiation and Regulation, Xinxiang, Henan, China
| | - Wanyu He
- College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
- State Key Laboratory of Cell Differentiation and Regulation, Xinxiang, Henan, China
| | - Huiwen Meng
- College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang, Henan, China
- State Key Laboratory of Cell Differentiation and Regulation, Xinxiang, Henan, China
| | - Peizhi Li
- Department of Anesthesiology, Xinxiang First People's Hospital, The Affiliated People's Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Junxing Qu
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, Henan, China
- Xinxiang Key Laboratory for Tumor Drug Screening and Targeted Therapy, Xinxiang, Henan, China
| |
Collapse
|
3
|
Liu R, Zhang X, Yan J, Liu S, Li Y, Wu G, Gao J. Penehyclidine hydrochloride alleviates lung ischemia-reperfusion injury by inhibiting pyroptosis. BMC Pulm Med 2024; 24:207. [PMID: 38671448 PMCID: PMC11046774 DOI: 10.1186/s12890-024-03018-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
OBJECTIVE The aim of this research was to examine how penehyclidine hydrochloride (PHC) impacts the occurrence of pyroptosis in lung tissue cells within a rat model of lung ischemia-reperfusion injury. METHODS Twenty-four Sprague Dawley (SD) rats, weighing 250 g to 270 g, were randomly distributed into three distinct groups as outlined below: a sham operation group (S group), a control group (C group), and a test group (PHC group). Rats in the PHC group received a preliminary intravenous injection of PHC at a dose of 3 mg/kg. At the conclusion of the experiment, lung tissue and blood samples were collected and properly stored for subsequent analysis. The levels of malondialdehyde, superoxide dismutase, and myeloperoxidase in the lung tissue, as well as IL-18 and IL-1β in the blood serum, were assessed using an Elisa kit. Pyroptosis-related proteins, including Caspase1 p20, GSDMD-N, and NLRP3, were detected through the western blot method. Additionally, the dry-to-wet ratio (D/W) of the lung tissue and the findings from the blood gas analysis were also documented. RESULTS In contrast to the control group, the PHC group showed enhancements in oxygenation metrics, reductions in oxidative stress and inflammatory reactions, and a decrease in lung injury. Additionally, the PHC group exhibited lowered levels of pyroptosis-associated proteins, including the N-terminal segment of gasdermin D (GSDMD-N), caspase-1p20, and nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3). CONCLUSION Pre-administration of PHC has the potential to mitigate lung ischemia-reperfusion injuries by suppressing the pyroptosis of lung tissue cells, diminishing inflammatory reactions, and enhancing lung function. The primary mechanism behind anti-pyroptotic effect of PHC appears to involve the inhibition of oxidative stress.
Collapse
Affiliation(s)
- Rongfang Liu
- Department of Anesthesiology, the Second Hospital of Hebei Medical University, NO. 215 of HePing West Road, Xinhua District Shijiazhuang, 050000, Shijiazhuang, China
- Department of Anesthesiology, Affiliated Hospital of Hebei University, 071000, Baoding, China
| | - Xuguang Zhang
- Department of Thoracic surgery, Affiliated Hospital of Hebei University, 071000, Baoding, China
| | - Jing Yan
- Electron microscope room, Hebei Medical University, 050000, Shijiazhuang, China
| | - Shan Liu
- Department of Pathology, Affiliated Hospital of Hebei University, 071000, Baoding, China
| | - Yongle Li
- Department of Anesthesiology, Affiliated Hospital of Hebei University, 071000, Baoding, China
| | - Guangyi Wu
- Department of Anesthesiology, Affiliated Hospital of Hebei University, 071000, Baoding, China
| | - Jingui Gao
- Department of Anesthesiology, the Second Hospital of Hebei Medical University, NO. 215 of HePing West Road, Xinhua District Shijiazhuang, 050000, Shijiazhuang, China.
| |
Collapse
|
4
|
Gao FF, Chen DQ, Jiang YT, Han CF, Lin BY, Yang Z, Quan JH, Xiong YH, Chen XT. Functional roles of circular RNAs in lung injury. Front Pharmacol 2024; 15:1354806. [PMID: 38601461 PMCID: PMC11004487 DOI: 10.3389/fphar.2024.1354806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 01/16/2024] [Indexed: 04/12/2024] Open
Abstract
Lung injury leads to respiratory dysfunction, low quality of life, and even life-threatening conditions. Circular RNAs (circRNAs) are endogenous RNAs produced by selective RNA splicing. Studies have reported their involvement in the progression of lung injury. Understanding the roles of circRNAs in lung injury may aid in elucidating the underlying mechanisms and provide new therapeutic targets. Thus, in this review, we aimed to summarize and discuss the characteristics and biological functions of circRNAs, and their roles in lung injury from existing research, to provide a theoretical basis for the use of circRNAs as a diagnostic and therapeutic target for lung injury.
Collapse
Affiliation(s)
- Fei-Fei Gao
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Dian-Qing Chen
- Department of Hand and Foot Surgery, Armed Police Corps Hospital of Hebei, Shijiazhuang, Hebei, China
| | - Yue-Tong Jiang
- Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Cui-Fei Han
- Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Bi-Yun Lin
- Biotissue Repository, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Zhan Yang
- Biotissue Repository, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Juan-Hua Quan
- Laboratory of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Ying-Huan Xiong
- Biotissue Repository, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xin-Tian Chen
- Laboratory of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
5
|
Weng J, Liu D, Shi B, Chen M, Weng S, Guo R, Zhou X. Sivelestat sodium alleviated lipopolysaccharide-induced acute lung injury by improving endoplasmic reticulum stress. Gene 2023; 884:147702. [PMID: 37567453 DOI: 10.1016/j.gene.2023.147702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
Acute lung injury (ALI) is a common inflammatory respiratory disorder characterized by a high incidence and mortality rate. This study aimed to investigate the potential therapeutic effects of the neutrophil elastase inhibitor Sivelestat sodium (SIV) in improving endoplasmic reticulum stress (ERS) while treating lipopolysaccharide (LPS)-induced ALI. An ALI model was established using LPS induction. The effects of SIV on ALI were observed both in vivo and in vitro, along with its impact on ERS. Lung tissue damage was assessed using Hematoxylin-eosin (H&E) staining. Lung edema was measured by the lung wet/dry weight ratio. The expression levels of protein kinase R-like ER kinase (PERK), Phospho-protein kinase R-like ER kinase (p-PERK), activating transcription factor 4 (ATF4), eukaryotic translation initiation factor 2α (EIF2a), phosphorylated α subunit of eukaryotic initiation factor 2α (P-EIF2a), and C/EBP homologous protein (CHOP) were analyzed by Western blotting in vivo and in vitro. The levels of tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) in Lung tissue samples supernatants were measured by ELISA. Oxidative stress markers were measured by ELISA. Apoptosis was measured using the TUNEL assay. Apoptosis-associated proteins B-cell lymphoma-2 (Bcl-2)、Bcl2-associated × (Bax)、caspase-3 were evaluated through Western blotting in vivo and in vitro. The expression levels of ERS-related proteins, including p-PERK, ATF4, P-EIF2a, and CHOP, were significantly increased in the LPS-induced ALI model. However, SIV markedly reduced the expression levels of these proteins, suppressing the LPS-induced ERS response. Further investigations revealed that SIV exerted a protective effect on ALI by alleviating lung tissue damage and apoptosis, improving lung function, and reducing inflammation and oxidative stress levels. However, when SIV was co-administered with Tunicamycin (TUN), TUN blocked the beneficial effects of SIV on ERS and reversed the protective effects of SIV on ALI. In conclusion, SIV alleviated lung tissue damage and apoptosis, improving lung function, and reducing inflammation and oxidative stress in LPS-induced ALI by improving ERS.
Collapse
Affiliation(s)
- Junting Weng
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian 351100, China.
| | - Danjuan Liu
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian 351100, China.
| | - Bingbing Shi
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian 351100, China.
| | - Min Chen
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian 351100, China.
| | - Shuoyun Weng
- Wenzhou Medical University School of Optometry and Ophthalmology, China.
| | - Rongjie Guo
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian 351100, China.
| | - Xiaoping Zhou
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian 351100, China.
| |
Collapse
|
6
|
Liu Z, Meng Y, Miao Y, Yu L, Yu Q. Artesunate reduces sepsis-mediated acute lung injury in a SIRT1-dependent manner. BIOIMPACTS : BI 2023; 13:219-228. [PMID: 37431481 PMCID: PMC10329753 DOI: 10.34172/bi.2023.23585] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 05/18/2022] [Accepted: 05/18/2022] [Indexed: 07/12/2023]
Abstract
Introduction Sepsis-mediated acute lung injury (ALI) is a critical clinical condition. Artesunate (AS) is a sesquiterpene lactone endoperoxide that was discovered in Artemisia annua, which is a traditional Chinese herb. AS has a broad set of biological and pharmacological actions; however, its protective effect on lipopolysaccharide (LPS)-induced ALI remains unclear. Methods LPS-mediated ALI was induced in rats through bronchial LPS inhalation. Then NR8383 cells were treated with LPS to establish an in vitro model. Further, we administered different AS doses in vivo and in vitro. Results AS administration significantly decreased LPS-mediated pulmonary cell death and inhibited pulmonary neutrophil infiltration. Additionally, AS administration increased SIRT1 expression in pulmonary sections. Administration of a biological antagonist or shRNA-induced reduction of SIRT1 expression significantly inhibited the protective effect of AS against LPS-induced cellular injury, pulmonary dysfunction, neutrophil infiltration, and apoptosis. This demonstrates that enhanced SIRT1 expression is crucially involved in the observed protective effects. Conclusion Our findings could suggest the use of AS for treating lung disorders through a mechanism involving SIRT1 expression.
Collapse
Affiliation(s)
- Zhaohui Liu
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Yanli Meng
- Department of Gastroenterology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Yu Miao
- Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Lili Yu
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Qiannan Yu
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| |
Collapse
|
7
|
Yang J, Huang Q, Liao P, Zhang P, Sun S, Xu Q. Mechanism of miR-338-3p in sepsis-induced acute lung injury via indirectly modulating ATF4. Transpl Immunol 2023; 76:101681. [PMID: 35926799 DOI: 10.1016/j.trim.2022.101681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 01/31/2023]
Abstract
Sepsis is recognized as an inflammation-related syndrome in response to invading pathogens. Many patients suffer from sepsis including transplant recipients. Lipopolysaccharide (LPS) is known to trigger sepsis-related organ dysfunction. This study expounded on the possible effect of microRNA (miR)-338-3p in sepsis-induced acute lung injury (ALI). Firstly, human bronchial epithelial cell line 16HBE received LPS treatment to establish the cell models of sepsis-induced ALI. The expression patterns of miR-338-3p, long non-coding RNA OPA-interacting protein 5 antisense transcript 1 (lncRNA OIP5-AS1), and activating transcription factor 4 (ATF4) in 16HBE cells were examined. Afterwards, 16HBE cell viability, the apoptosis rate, and the levels of inflammation and lactate dehydrogenase (LDH) were determined to assess the degree of cell injury. We disclosed that LPS treatment triggered 16HBE cell injury, downregulated miR-338-3p, and upregulated OIP5-AS1 and ATF4. miR-338-3p overexpression repressed LPS-induced 16HBE cell injury. miR-338-3p diminished OIP5-AS1 stability via binding to OIP5-AS1 and downregulated OIP5-AS1 expression and OIP5-AS1 can enhance ATF4 mRNA stability and upregulate ATF4 mRNA level. The rescue experiments showed that ATF4 overexpression aggravated LPS-induced 16HBE cell injury. Overall, miR-338-3p overexpression decreased OIP5-AS1 expression and stability and further downregulated ATF4 mRNA level, thereby mitigating LPS-induced 16HBE cell injury.
Collapse
Affiliation(s)
- Jing Yang
- Department of Pediatric, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, Shandong, China.
| | - Qikun Huang
- Department of Pediatric, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, Shandong, China
| | - Peiyuan Liao
- Department of Pediatric, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, Shandong, China
| | - Pingli Zhang
- Department of Pediatric, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, Shandong, China
| | - Shijing Sun
- Department of Pediatric, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, Shandong, China
| | - Qianwen Xu
- Department of Pediatric, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, Shandong, China
| |
Collapse
|
8
|
Weng J, Liu D, Shi B, Chen M, Weng S, Guo R, Fu C. Penehyclidine hydrochloride protects against lipopolysaccharide-induced acute lung injury by promoting the PI3K/Akt pathway. Int J Immunopathol Pharmacol 2023; 37:3946320231192175. [PMID: 37500500 PMCID: PMC10655789 DOI: 10.1177/03946320231192175] [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: 04/11/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023] Open
Abstract
INTRODUCTION Acute lung injury (ALI) attracted attention among physicians because of its high mortality. We aimed to determine whether the phosphatidylinositol-3 kinase (PI3K)/protein kinase B (Akt) pathway is involved in the protective effects of penehyclidine hydrochloride (PHC) against lipopolysaccharide (LPS)-induced ALI. METHODS H&E staining was used to observed pathological changes in the lung tissues. ELISA was used to evaluate the concentration of inflammatory mediators in the bronchoalveolar lavage fluid (BALF). White-light microscopy was performed to observe the TUNEL-positive nuclei. The viability of NR8383 alveolar macrophages was determined by using CCK-8. The levels of MPO, MDA, SOD, and GSH-Px were analyzed using ELISA kits. Western blotting was used to evaluate the ERS-associated protein levels and the phosphorylation of PI3K and Akt. RESULTS PHC administration defended against LPS-induced histopathological deterioration and increased pulmonary edema and lung injury scores, while all of these beneficial effects were inhibited by LY. In addition, PHC administration mitigated oxidative stress as indicated by decreases in lung myeloperoxidase (MPO) and malondialdehyde (MDA) concentrations, and increases in glutathione peroxidase (GSH-Px) and superoxide dismutase (SOD) concentrations. It also alleviated LPS-induced inflammation. PHC administration attenuated apoptosis-associated protein levels, improved cell viability, and decreased the number of TdT-mediated dUTP Nick-End Labeling (TUNEL)-positive cells. Furthermore, PHC inhibited ERS-associated protein levels. Meanwhile, the protection of PHC against inflammation, oxidative stress, apoptosis, and ERS was inhibited by LY. Moreover, PHC administration increased PI3K and Akt phosphorylation, indicating that the upregulation of the PI3K/Akt pathway, while this pathway was inhibited by LY. CONCLUSION PHC significantly activates the PI3K/Akt pathway to ameliorate the extent of damage to pulmonary tissue, inflammation, oxidative stress, apoptosis, and ERS in LPS-induced ALI.
Collapse
Affiliation(s)
- Junting Weng
- Department of Critical Care Medicine, The Affiliated Hospital of Putian University, Putian, China
| | - Danjuan Liu
- Department of Critical Care Medicine, The Affiliated Hospital of Putian University, Putian, China
| | - Bingbing Shi
- Department of Critical Care Medicine, The Affiliated Hospital of Putian University, Putian, China
| | - Min Chen
- Department of Critical Care Medicine, The Affiliated Hospital of Putian University, Putian, China
| | - Shuoyun Weng
- School of Wenzhou Medical University, Wenzhou, China
| | - Rongjie Guo
- Department of Critical Care Medicine, The Affiliated Hospital of Putian University, Putian, China
| | - Chunjin Fu
- Department of Critical Care Medicine, The Affiliated Hospital of Putian University, Putian, China
| |
Collapse
|
9
|
Liu Z, Meng Y, Wei Q, Miao Y, Yu L, Li Y, Zhang B. The Protective Activity of Penehyclidine Hydrochloride against Renal Ischemia/Reperfusion-Mediated NLRP3 Inflammasome Activation is Induced by SIRT1. J INVEST SURG 2021; 35:1050-1061. [PMID: 34696682 DOI: 10.1080/08941939.2021.1995541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Background: The activation of alveolar macrophages (AMs) modulated via leucine-rich repeat (NLR) pyrin domain containing 3 (NLRP3) inflammasome activation is key to the progression of renal ischemia/reperfusion (rI/R)-mediated acute lung injury (ALI). Sirtuin-1 (SIRT1) can attenuate NLRP3 inflammasome activation during I/R stress and may be an important mechanism underlying ALI pathogenesis. Penehyclidine hydrochloride (PHC), an anticholinergic drug, exerts protective effects against rI/R-mediated ALI. This study aimed to decipher the effects of PHC on SIRT1 activation and the underlying mechanism of the protective activity of PHC against rI/R-mediated ALI.Materials and methods: We used an ALI rat model and the rat AMs cell line NR8383 to assess the degree of lung injury in vivo and in vitro.Results: The results show that PHC attenuates rI/R-mediated lung injury indices, myeloperoxidase, and apoptosis in vivo. It decreases the rI/R-mediated release of prostaglandin E2 and nitric oxide, mitochondrial reactive oxygen species production, and the activity of NADPH oxidase-4 in vitro. PHC ameliorates the rI/R-induced activation of the thioredoxin-interacting protein, caspase 1 (P10 unit), and NLRP3 inflammasome, along with reduced activation of interleukin-1β and interleukin-18 in vitro. We show that PHC alleviates the rI/R-induced reduction of SIRT1 and the depletion of SIRT1 eliminates the ameliorating activity of PHC on the NLRP3 inflammasome activation in vitro. Conclusions: In summary, the findings suggest that PHC ameliorates the rI/R-mediated ALI through the SIRT1-mediated NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Zhaohui Liu
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Yanli Meng
- Department of Gastroenterology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Qianjie Wei
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Yu Miao
- Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Lili Yu
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Yuqing Li
- Department of Anesthesiology, Botou Hospital, Cangzhou, Hebei, China
| | - Bing Zhang
- Department of Anesthesiology, Botou Hospital, Cangzhou, Hebei, China
| |
Collapse
|
10
|
Luo S, Ding X, Zhao S, Mou T, Li R, Cao X. Long non-coding RNA CHRF accelerates LPS-induced acute lung injury through microRNA-146a/Notch1 axis. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1299. [PMID: 34532436 PMCID: PMC8422153 DOI: 10.21037/atm-21-3064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/12/2021] [Indexed: 11/06/2022]
Abstract
Background The present study sought to investigate the regulatory role of the long non-coding RNA (lncRNA) cardiac hypertrophy-related factor (CHRF) in a mouse model of acute lung injury (ALI) and in primary mouse pulmonary microvascular endothelial cells (MPVECs) treated with lipopolysaccharide (LPS). Methods C57BL/6 mice were given adenovirus (Ad) sh-CHRF or negative control (NC) before undergoing cecal ligation and perforation. MPVECs transfected with Adsh-CHRF or NC were treated with LPS. Double luciferase assay was used to detect the binding of miR-146a to CHRF or Notch1. Subsequently, MPVECs were co-transfected with miR-146a inhibitor and sh-CHRF for 24 hours, and then treated with LPS. Results High expression of CHRF was detected in septic mice. Cecal ligation and perforation induced ALI and apoptosis in mice, whereas, CHRF knockout could inhibit ALI. The protein expression levels of TNF-α, IL-1β and IL-6 in the lung and bronchoalveolar lavage fluid of the CLP group were up-regulated, whereas the expression of IL-4 and IL-10 was down-regulated. CHRF inhibition reduced the production of proinflammatory cytokines in septic mice. The inhibitory effect of CHRF gene knockdown on lung inflammation and apoptosis was confirmed in the septic cell model. Mechanistic investigation showed that CHRF up-regulated the level of Notch1 by sponging miR-146a. Additionally, the low expression of miR-146a reversed the inhibitory effect of CHRF gene knockout on LPS-induced inflammatory response and apoptosis. Together, in vivo and in vitro results demonstrated that CHRF enhanced sepsis-induced ALI by targeting miR-146a and up-regulating Notch1. Conclusions CHRF can induce inflammation and apoptosis caused by sepsis by miR-146a/Notch1 axis. Therefore, it may serve as a potential drug target for treating sepsis-induced ALI.
Collapse
Affiliation(s)
- Shu Luo
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xuefeng Ding
- Department of Critical Care, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Shiqiao Zhao
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Tianyi Mou
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Ruixiu Li
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiaoping Cao
- Department of Emergency, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
11
|
Yuan M, Jing G, Kong Q, Ming T, Song X, Wu X. Tumor necrosis factor α‑induced protein 8‑like 2 contributes to penehyclidine hydrochloride pretreatment against lipopolysaccharide‑induced acute lung injury in a mouse model. Mol Med Rep 2021; 24:750. [PMID: 34468006 DOI: 10.3892/mmr.2021.12390] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/12/2021] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the effect of penehyclidine hydrochloride (PHC) pretreatment on mice with lipopolysaccharide (LPS)‑induced acute lung injury (ALI) and its possible underlying mechanisms. Mice were randomly separated into six groups: i) Sham group; ii) LPS group; iii) LPS + PHC group; iv) tumor necrosis factor a‑induced protein 8‑like protein 2 (TIPE2) group; v) LPS + TIPE2 group; and vi) LPS + TIPE2 + PHC group. The ALI model was induced using LPS through intratracheal injection. The mice received adenovirus gene to induce the overexpression of TIPE2. After mice were sacrificed, lung injury indices were assessed, and arterial blood, bronchoalveolar lavage fluid and lung tissues were collected for subsequent assays. Expression levels of related proteins were detected by using western blotting. It was found that compared with the sham group, the mice treated with LPS showed increased lung injury and dysfunctions of gas exchange. However, these trends were significantly ameliorated in the LPS + PHC group. Evaluation of protein expression in lung tissues showed that the increased expression of nuclear NF‑κB p65 and p‑c‑Jun N‑terminal kinase (JNK) induced by LPS were suppressed in the LPS + PHC group and the expression of TIPE2 was increased. The mice that received adenovirus gene to induce TIPE2 overexpression could also showed protective effects compared with the mice in the LPS group. However, the expression of TIPE2 decreased rather than increased in LPS group. In the mice pretreated with PHC, the expression of TIPE2 increased in mice with LPS‑induced ALI. To conclude, PHC pretreatment could inhibit the occurrence of inflammation and apoptosis in LPS‑induced ALI. This process may be related to the activation of TIPE2 and the inhibition of NF‑κB and JNK signaling pathway in the lungs of mice.
Collapse
Affiliation(s)
- Min Yuan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Guoqing Jing
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Qian Kong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Tingqian Ming
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xuemin Song
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
12
|
Activation of TREM-1 induces endoplasmic reticulum stress through IRE-1α/XBP-1s pathway in murine macrophages. Mol Immunol 2021; 135:294-303. [PMID: 33957479 DOI: 10.1016/j.molimm.2021.04.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 04/16/2021] [Accepted: 04/26/2021] [Indexed: 12/14/2022]
Abstract
Increasing evidence suggests that endoplasmic reticulum (ER) stress activates several pro-inflammatory signaling pathways in many diseases, including acute lung injury (ALI). We have reported that blocking triggering receptor expressed on myeloid cells 1 (TREM-1) protects against ALI by suppressing pulmonary inflammation in mice with ALI induced by lipopolysaccharides (LPS). However, the molecular mechanism underlying the TREM-1-induced pro-inflammatory microenvironment in macrophages remains unclearly. Herein, we aimed to determine whether TREM-1 regulates the inflammatory responses induced by LPS associated with ER stress activation. We found that the activation of TREM-1 by a monoclonal agonist antibody (anti-TREM-1) increased the mRNA and protein levels of IL-1β, TNF-α, and IL-6 in primary macrophages. Treatment of the anti-TREM-1 antibody increased the expression of ER stress markers (ATF6, PERK, IRE-1α, and XBP-1s) in primary macrophages. While pretreatment with 4-PBA, an inhibitor of ER stress, significantly inhibited the expression of ER stress markers and pro-inflammatory cytokines and reduced LDH release. Furthermore, inhibiting the activity of the IRE-1α/XBP-1s pathway by STF-083010 significantly mitigated the increased levels of IL-1β, TNF-α, and IL-6 in macrophages treated by the anti-TREM-1 antibody. XBP-1 silencing attenuated pro-inflammatory microenvironment evoked by activation of TREM-1. Besides, we found that blockade of TREM-1 with LR12 ameliorated ER stress induced by LPS in vitro and in vivo. In conclusion, we conclude that TREM-1 activation induces ER stress through the IRE-1α/XBP-1s pathway in macrophages, contributing to the pro-inflammatory microenvironment.
Collapse
|
13
|
Zhang E, Wang J, Chen Q, Wang Z, Li D, Jiang N, Ju X. Artesunate ameliorates sepsis-induced acute lung injury by activating the mTOR/AKT/PI3K axis. Gene 2020; 759:144969. [DOI: 10.1016/j.gene.2020.144969] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/07/2020] [Accepted: 07/17/2020] [Indexed: 12/25/2022]
|
14
|
Zou Z, Wang Q, Zhou M, Li W, Zheng Y, Li F, Zheng S, He Z. Protective effects of P2X7R antagonist in sepsis-induced acute lung injury in mice via regulation of circ_0001679 and circ_0001212 and downstream Pln, Cdh2, and Nprl3 expression. J Gene Med 2020; 22:e3261. [PMID: 32783373 DOI: 10.1002/jgm.3261] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Sepsis induces pulmonary P2X7 receptor (P2X7 R) expression and P2X7 R-knockout reduced lung inflammation in mice. The present study investigated the expression of circular RNA (circRNA) and mRNA in sepsis-induced acute lung injury (ALI) treated with a P2X7 R antagonist. METHODS Sepsis was induced by tracheal administration of lipopolysaccharide (LPS), and the mice were then divided into two groups: without [sepsis + dimethyl sulfoxide (DMSO)] or with P2X7 R antagonist treatment (sepsis + P2X7 A). Sham mice were administrated sterile normal saline. Serum levels of interleukin (IL)-1β and tumor necrosis factor (TNF)-α, pathological changes, cell apoptosis and P2X7 R expression in lung were assessed, followed by RNA sequencing (RNA-seq) and bioinformatics analyses. A quantitative reverse transcriptase-polymerase chain reaction (RT-qPCR) was used to validate circRNAs and mRNAs. RESULTS Compared to the sham group, LPS-induced sepsis produced obvious pathological changes in lung tissue, as well as increased apoptotic lung cells, serum TNF-α and IL-1β levels, and P2X7 R expression; P2X7 R antagonism significantly ameliorated these changes. RNA-seq identified many dysregulated circRNAs and mRNAs during sepsis, whereas this changed with P2X7 R antagonism. RT-qPCR confirmed that Mus musculus (mmu)_circ_0001679, mmu_circ_0001212, phospholamban (Pln), cadherin-2 (Cdh2) and nitrogen permease regulator 3-like (Nprl3) expression were significantly increased in the sepsis + DMSO group compared to that in the sham group but were decreased in the sepsis + P2X7 A group compared to that in the sepsis + DMSO group. The circRNA-microRNA-mRNA coexpression network indicated that mmu_circ_0001679 may regulate Nprl3 and that mmu_circ_0001212 may similarly regulate Pln, Cdh2 and Nprl3 as a competing endogenous RNA. CONCLUSIONS P2X7 R antagonism attenuates sepsis-induced ALI by inhibiting dysregulated expression of circRNA (circ_0001679, circ_0001212) and mRNA (Pln, Cdh2 and Nprl3).
Collapse
Affiliation(s)
- Zijun Zou
- Department of ICU, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Qin Wang
- Department of pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Minggen Zhou
- Department of ICU, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Weichao Li
- Department of ICU, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yikai Zheng
- Department of ICU, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Fanyi Li
- Department of ICU, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Shengcai Zheng
- Department of ICU, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhijie He
- Department of ICU, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
15
|
Liu Z, Li Y, Yu L, Chang Y, Yu J. Penehyclidine hydrochloride inhibits renal ischemia/reperfusion-induced acute lung injury by activating the Nrf2 pathway. Aging (Albany NY) 2020; 12:13400-13421. [PMID: 32652517 PMCID: PMC7377887 DOI: 10.18632/aging.103444] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/25/2020] [Indexed: 12/12/2022]
Abstract
The nuclear factor (NF)-κB and NOD-like receptor protein 3 (NLRP3) pathways promote inflammatory signaling that injures the kidneys, whereas the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway promotes anti-inflammatory signaling that inhibits oxidative damage. Penehyclidine hydrochloride (PHC) inhibits NF-κB and activates Nrf2 signaling. We investigated whether PHC induces communication between the Nrf2 and NF-κB/NLRP3 pathways, thereby protecting against renal ischemia/reperfusion (rI/R)-induced lung inflammation. Rat alveolar macrophages (NR8383 cells) were stimulated for 24 h with PHC with or without brusatol (a Nrf2 antagonist), after which they were treated for 4 h with tert-butyl hydroperoxide (10 mM). PHC Nrf2-dependently alleviated tert-butyl hydroperoxide-induced reactive oxygen species production in alveolar macrophages. Additionally, wild-type and Nrf2-/- rats were each divided into four groups: (1) sham, (2) PHC (1 mg/kg), (3) rI/R and (4) rI/R + PHC (1 mg/kg). PHC markedly induced the Nrf2 and adenosine monophosphate-activated protein kinase pathways and suppressed rI/R-induced NF-κB and NLRP3 activation in the lungs. Nrf2 deficiency diminished the ability of PHC to ameliorate rI/R-induced histopathological alterations and reactive oxygen species release in the lungs; however, PHC inhibited NLRP3 signaling Nrf2-dependently, while it inhibited NF-κB signaling Nrf2-independently. Our findings demonstrate the beneficial effects of PHC on rI/R-induced lung inflammation.
Collapse
Affiliation(s)
- Zhaohui Liu
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Yan Li
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Lili Yu
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Yulin Chang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Jingui Yu
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|