1
|
Ozkanlar S, Ozkanlar Y, Kara A, Dalkilinc E. Astaxanthin Alleviates Lung Injury by Regulating Oxidative Stress, Inflammatory Response, P2X7 Receptor, NF-κB, Bcl-2, and Caspase-3 in LPS-Induced Endotoxemia. ENVIRONMENTAL TOXICOLOGY 2025; 40:924-934. [PMID: 39873358 PMCID: PMC12069755 DOI: 10.1002/tox.24481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 10/07/2024] [Accepted: 01/16/2025] [Indexed: 01/30/2025]
Abstract
Sepsis remains the leading cause of multiple-organ injury due to endotoxemia. Astaxanthin (ASTA), widely used in marine aquaculture, has an extraordinary potential for antioxidant and anti-inflammatory activity. Purinergic receptor (e.g., P2X7R) activation is a powerful signaling in the modulation of inflammation. The effect of ASTA was investigated on the regulation of oxidative stress, inflammatory response, apoptotic mediators, and P2X7R expression in the lung injury during lipopolysaccharide (LPS)-induced endotoxemia. Twenty-four rats were blocked into four groups as Control, LPS, ASTA, and LPS + ASTA. LPS was administered by intraperitoneal injection and ASTA by gavage. Blood and lung samples were taken 6 h after the administrations. The methods were ELISA, western blotting, histopathology, and immunohistochemistry. Sepsis was confirmed by the elevations of IL-1β, IL-6, IL-10, and TNF-α levels in bloodstream. Lung injury was determined by histopathological changes. There were increased P2X7R expression, malondialdehyde (MDA), IL-1β, TNF-α, nuclear factor kappa B (NF-κB), and Caspase-3 and decreased B-cell lymphoma 2 (Bcl-2) and glutathione (GSH) in the septic lung tissue (p < 0.05). ASTA treatment improved MDA, GSH, IL-1β, TNF-α, P2X7R, NF-κB, Caspase-3, and Bcl-2 levels and reduced P2X7R immunoreactivity and histological abnormalities in the lung (p < 0.05). The production of pro-inflammatory cytokines, oxidative stress, P2X7R expression, and apoptotic mediators in the lung is associated with LPS-induced endotoxemia. The ASTA administration appears to regulate the expressions of P2X7R, NF-κB, Bcl-2, and Caspase-3 improving the antioxidative and anti-inflammatory response of the lung tissue in sepsis, in vivo.
Collapse
Affiliation(s)
- Seckin Ozkanlar
- Department of Biochemistry, Faculty of Veterinary MedicineAtaturk UniversityErzurumTurkey
| | - Yunusemre Ozkanlar
- Department of Internal Medicine, Faculty of Veterinary MedicineOndokuz Mayis UniversitySamsunTurkey
| | - Adem Kara
- Department of Genetics, Faculty of ScienceErzurum Technical UniversityErzurumTurkey
| | - Elif Dalkilinc
- Department of Biochemistry, Faculty of Veterinary MedicineAtaturk UniversityErzurumTurkey
| |
Collapse
|
2
|
Wang Y, Yuan M, Qin J, Chen X, Lei Z, Kong Q, Wang Q, Song X, Wu X. TIPE2 Alleviates Sepsis-induced Lung Injury By Inhibiting PANoptosis in Murine Alveolar Macrophages. Inflammation 2025:10.1007/s10753-025-02288-3. [PMID: 40299241 DOI: 10.1007/s10753-025-02288-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/20/2025] [Accepted: 03/05/2025] [Indexed: 04/30/2025]
Abstract
Sepsis-induced acute lung injury (ALI) is a life-threatening condition with high mortality rates, and its underlying mechanisms remain poorly understood. This study investigates the role of TNF-α-induced protein 8-like 2 (TIPE2) in modulating PANoptosis, an integrated form of programmed cell death that includes apoptosis, necroptosis, and pyroptosis, in the context of sepsis-induced lung injury. We utilized a cecal ligation and puncture (CLP) mouse model to examine the effects of TIPE2 knockout and overexpression on lung injury, inflammation, and cell death pathways. Our findings demonstrate that TIPE2 knockout exacerbates lung injury by promoting the abnormal activation of PANoptosis-related proteins, leading to increased inflammation and tissue damage. In contrast, overexpression of TIPE2 in macrophages significantly reduces these effects by inhibiting the ZBP1-dependent PANoptosis pathway via TRIF signaling. These results highlight the crucial role of TIPE2 in maintaining the balance between cell survival and death during sepsis and suggest that targeting TIPE2 could be a novel therapeutic strategy for treating sepsis-related lung injury.
Collapse
Affiliation(s)
- Yuxuan Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Min Yuan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jingxue Qin
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xue Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zihan Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qian Kong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qian Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xuemin Song
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
3
|
Han J, Miao Y, Song L, Zhou X, Liu Y, Wang L, Zhu K, Ma H, Ma Y, Li Q, Han D. Xuefu Zhuyu Decoction improves hyperlipidemia through the MAPK/NF-κB and MAPK/PPARα/CPT-1A signaling pathway. FASEB J 2025; 39:e70363. [PMID: 39878687 DOI: 10.1096/fj.202402688r] [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: 11/02/2024] [Revised: 12/29/2024] [Accepted: 01/21/2025] [Indexed: 01/31/2025]
Abstract
Xuefu Zhuyu Decoction (XZD) is widely used in the treatment of cardiovascular diseases. The purpose of this study was to explore the pharmacological effects and molecular mechanisms of XZD in improving hyperlipidemia and to provide a theoretical framework for clinical application. In this study, the signaling pathways regulated by XZD in improving hyperlipidemia were predicted by network pharmacology. Molecular docking was used to verify the affinity between the components in XZD and the target. Furthermore, a hyperlipidemic model in rats was constructed through feeding a high-fat diet. The effect of XZD on hyperlipidemia was verified by histopathological staining, Elisa, and western blot. The results found that the XZD improved dyslipidemia and inflammatory factor disorders, and inhibited liver function damage, pathological damage, and oxidative stress damage in hyperlipidemic rats. The findings from molecular docking and network pharmacology suggested that the mechanism of XZD improving hyperlipidemia may be closely related to the MAPK, NF-κB, and PPAR pathways. This study demonstrated that the XZD inhibited liver lipid metabolism disorder and inflammatory response by regulating the MAPK/NF-κB and MAPK/PPARα/CPT-1A pathway, significantly improved liver histopathological damage and oxidative stress injury, and played a protective role in hyperlipidemic rats.
Collapse
Affiliation(s)
- Jiajun Han
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin, China
| | - Yuyang Miao
- Department of Endocrinology and Metabolism, Jilin Province People's Hospital, Jilin, China
| | - Linze Song
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin, China
| | - Xianfeng Zhou
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin, China
| | - Yan Liu
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin, China
| | - Lin Wang
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin, China
| | - Kai Zhu
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin, China
| | - He Ma
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin, China
| | - Yan Ma
- Department of Endocrinology and Metabolism, Jilin Province People's Hospital, Jilin, China
| | - Qingjie Li
- Research Center of Traditional Chinese Medicine, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Dong Han
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin, China
| |
Collapse
|
4
|
Zhou B, Xue J, Wang J, Yu D, Zhou F, Duan JA, Niu Y, Wang H. Amygdalin alleviates LPS-induced acute lung injury in mice by targeting CD5L/iNOS pathway. Mol Immunol 2024; 176:22-29. [PMID: 39561489 DOI: 10.1016/j.molimm.2024.11.003] [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: 09/01/2024] [Revised: 11/02/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024]
Abstract
Amygdalins (AMY) from bitter almonds are distinguished by their anti-inflammatory, antibacterial and antioxidant properties, but their role in the treatment of acute lung injury (ALI) and their mechanisms need to be clarified. We sought to investigate whether AMY provides protection against lipopolysaccharide (LPS)-induced ALI in mice and explore the mechanisms of its protection. Results showed that AMY effectively alleviated LPS-induced ALI in a dose-dependent manner by reducing in vivo lung wet/dry ratio, lung/body weight ratio, and myeloperoxidase (MPO). In addition, AMY can significantly reduce lung histopathological injury, decreased bronchoalveolar lavage fluid (BALF) lymphocyte, neutrophil, and monocyte numbers, and decreased the secretion of inflammatory cytokines IL-6, IL-1β, and TNF-α. Through transcriptome sequencing, AMY was found to effectively reduce the mRNA level of CD5L in mice. In AAV-CD5L transfected mice, CD5L overexpression was found to block the protective effect of AMY in LPS-induced ALI mice. It was revealed that AMY inhibited NF-κB entry into the nucleus to reduce iNOS by targeting CD5L. Taken together, AMY can effectively reduce lung inflammation and alleviate ALI, and is a potential novel protective agent against LPS-induced ALI.
Collapse
Affiliation(s)
- Bo Zhou
- College of Pharmacy, Key Laboratory of Ningxia Minority Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Jiahui Xue
- College of Pharmacy, Key Laboratory of Ningxia Minority Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Jing Wang
- College of Pharmacy, Key Laboratory of Ningxia Minority Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Donghua Yu
- College of Pharmacy, Key Laboratory of Ningxia Minority Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Fangling Zhou
- College of Pharmacy, Key Laboratory of Ningxia Minority Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Jin-Ao Duan
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yang Niu
- College of Pharmacy, Key Laboratory of Ningxia Minority Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Hanqing Wang
- College of Pharmacy, Key Laboratory of Ningxia Minority Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia 750004, China; Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia 750004, China; Ningxia Regional Characteristic Traditional Chinese Medicine Collaborative Innovation Center Co-constructed by the Province and Ministry, Ningxia Engineering and Technology Research Center for Modernization of Regional Characteristic Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
5
|
Ming T, Liu H, Yuan M, Tian J, Fang Q, Liu Y, Kong Q, Wang Q, Song X, Xia Z, Wu X. The deubiquitinase OTUD1 deubiquitinates TIPE2 and plays a protective role in sepsis-induced lung injury by targeting TAK1-mediated MAPK and NF-κB signaling. Biochem Pharmacol 2024; 227:116418. [PMID: 38996928 DOI: 10.1016/j.bcp.2024.116418] [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: 04/16/2024] [Revised: 06/05/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
Ovarian tumor domain-containing protease 1 (OTUD1) is a critical negative regulator that promotes innate immune homeostasis and is extensively involved in the pathogenesis of sepsis. In this study, we performed a powerful integration of multiomics analysis and an experimental mechanistic investigation to elucidate the immunoregulatory role of OTUD1 in sepsis at the clinical, animal and cellular levels. Our study revealed the upregulation of OTUD1 expression and the related distinctive alterations observed via multiomics profiling in clinical and experimental sepsis. Importantly, in vivo and in vitro, OTUD1 was shown to negatively regulate inflammatory responses and play a protective role in sepsis-induced pathological lung injury by mechanistically inhibiting the activation of the transforming growth factor-beta-activated kinase 1 (TAK1)-mediated mitogen-activated protein kinase (MAPK) and nuclear factor kappa-B (NF-κB) signaling pathways in the present study. Subsequently, we probed the molecular mechanisms underlying OTUD1's regulation of NF-κB and MAPK pathways by pinpointing the target proteins that OTUD1 can deubiquitinate. Drawing upon prior research conducted in our laboratory, it has been demonstrated that tumor necrosis factor-α-induced protein 8-like 2 (TIPE2) performs a protective function in septic lung injury and septic encephalopathy by suppressing the NF-κB and MAPK pathways. Hence, we hypothesized that TIPE2 might be a target protein of OTUD1. Additional experiments, including Co-IP, immunofluorescence co-localization, and Western blotting, revealed that OTUD1 indeed has the ability to deubiquitinate TIPE2. In summary, OTUD1 holds potential as an immunoregulatory and inflammatory checkpoint agent, and could serve as a promising therapeutic target for sepsis-induced lung injury.
Collapse
Affiliation(s)
- Tingqian Ming
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Huifan Liu
- Department of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, PR China
| | - Min Yuan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Jingyuan Tian
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Qing Fang
- Department of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, PR China
| | - Yuping Liu
- Department of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, PR China
| | - Qian Kong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Qian Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Xuemin Song
- Department of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, PR China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China.
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China.
| |
Collapse
|
6
|
Li F, Yan W, Dong W, Chen Z, Chen Z. PNSC928, a plant-derived compound, specifically disrupts CtBP2-p300 interaction and reduces inflammation in mice with acute respiratory distress syndrome. Biol Direct 2024; 19:48. [PMID: 38902802 PMCID: PMC11191317 DOI: 10.1186/s13062-024-00491-0] [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] [Received: 01/29/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Prior research has highlighted the involvement of a transcriptional complex comprising C-terminal binding protein 2 (CtBP2), histone acetyltransferase p300, and nuclear factor kappa B (NF-κB) in the transactivation of proinflammatory cytokine genes, contributing to inflammation in mice with acute respiratory distress syndrome (ARDS). Nonetheless, it remains uncertain whether the therapeutic targeting of the CtBP2-p300-NF-κB complex holds potential for ARDS suppression. METHODS An ARDS mouse model was established using lipopolysaccharide (LPS) exposure. RNA-Sequencing (RNA-Seq) was performed on ARDS mice and LPS-treated cells with CtBP2, p300, and p65 knockdown. Small molecules inhibiting the CtBP2-p300 interaction were identified through AlphaScreen. Gene and protein expression levels were quantified using RT-qPCR and immunoblots. Tissue damage was assessed via histological staining. KEY FINDINGS We elucidated the specific role of the CtBP2-p300-NF-κB complex in proinflammatory gene regulation. RNA-seq analysis in LPS-challenged ARDS mice and LPS-treated CtBP2-knockdown (CtBP2KD), p300KD, and p65KD cells revealed its significant impact on proinflammatory genes with minimal effects on other NF-κB targets. Commercial inhibitors for CtBP2, p300, or NF-κB exhibited moderate cytotoxicity in vitro and in vivo, affecting both proinflammatory genes and other targets. We identified a potent inhibitor, PNSC928, for the CtBP2-p300 interaction using AlphaScreen. PNSC928 treatment hindered the assembly of the CtBP2-p300-NF-κB complex, substantially downregulating proinflammatory cytokine gene expression without observable cytotoxicity in normal cells. In vivo administration of PNSC928 significantly reduced CtBP2-driven proinflammatory gene expression in ARDS mice, alleviating inflammation and lung injury, ultimately improving ARDS prognosis. CONCLUSION Our results position PNSC928 as a promising therapeutic candidate to specifically target the CtBP2-p300 interaction and mitigate inflammation in ARDS management.
Collapse
Affiliation(s)
- Fan Li
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Wenqing Yan
- Department of Critical Care Medicine, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai, Shanghai, 200065, China
- Department of Emergency, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, China
- Department of Emergency, Jiangxi Provincial People's Hospital, No. 92, Aiguo Road, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Weihua Dong
- Department of Emergency, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, China
- Department of Emergency, Jiangxi Provincial People's Hospital, No. 92, Aiguo Road, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Zhiping Chen
- Department of Emergency, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, China.
- Department of Emergency, Jiangxi Provincial People's Hospital, No. 92, Aiguo Road, Donghu District, Nanchang, Jiangxi, 330006, China.
| | - Zhi Chen
- Department of Critical Care Medicine, Tongji Hospital, School of Medicine, Tongji University, No. 389 Xincun Road, Shanghai, Shanghai, 200065, China.
- Department of Emergency, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, China.
- Department of Emergency, Jiangxi Provincial People's Hospital, No. 92, Aiguo Road, Donghu District, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
7
|
Zheng Y, Hu R, Hu J, Feng L, Li S. Protective effects of butorphanol in oleic acid-endotoxin "two-hit" induced rat lung injury by suppression of inflammation and apoptosis. Sci Rep 2024; 14:14231. [PMID: 38902260 PMCID: PMC11190203 DOI: 10.1038/s41598-024-53483-5] [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: 05/23/2023] [Accepted: 01/31/2024] [Indexed: 06/22/2024] Open
Abstract
Butorphanol is widely used as an anesthetic drug, whether butorphanol could reduce organ injury and protecting lung tissue is unknown. This study explored the effects of butorphanol on ALI and investigated its underlying mechanisms. We established a "two-hit" rat model and "two-hit" cell model to prove our hypothesis. Rats were divided into four groups [control, "two-hit" (OA + LPS), "two-hit" + butorphanol (4 mg/kg and 8 mg/kg) (OA + LPS + B1 and OA + LPS + B2)]. RPMVE cells were divided into four groups [control, "two-hit" (OA + LPS), "two-hit" + butorphanol (4 μM and 8 μM) (OA + LPS + 4 μM and OA + LPS + 8 μM)]. Inflammatory injury was assessed by the histopathology and W/D ratio, inflammatory cytokines, and arterial blood gas analysis. Apoptosis was assessed by Western blotting and flow cytometry. The effect of NF-κB p65 was detected by ELISA. Butorphanol could relieve the "two-hit" induced lung injury, the expression of TNF, IL-1β, IL-6, and improve lung ventilation. In addition, butorphanol decreased Bax and cleaved caspase-3, increased an antiapoptotic protein (Bcl-2), and inhibited the "two-hit" cell apoptosis ratio. Moreover, butorphanol suppressed NF-κB p65 activity in rat lung injury. Our research showed that butorphanol may attenuate "two-hit"-induced lung injury by regulating the activity of NF-κB p65, which may supply more evidence for ALI treatment.
Collapse
Affiliation(s)
- Yanlei Zheng
- Department of Intensive Care Medicine, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, China
| | - Ronghua Hu
- Department of Intensive Care Medicine, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, China
| | - Jinrong Hu
- Department of Intensive Care Medicine, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, China
| | - Lina Feng
- Department of Intensive Care Medicine, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, China
| | - Shi Li
- Department of Intensive Care Medicine, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, China.
| |
Collapse
|
8
|
Yu PR, Tseng CY, Hsu CC, Chen JH, Lin HH. In vitro and in vivo protective potential of quercetin-3-glucuronide against lipopolysaccharide-induced pulmonary injury through dual activation of nuclear factor-erythroid 2 related factor 2 and autophagy. Arch Toxicol 2024; 98:1415-1436. [PMID: 38436694 DOI: 10.1007/s00204-024-03691-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/23/2024] [Indexed: 03/05/2024]
Abstract
In vitro and in vivo models of lipopolysaccharide (LPS)-induced pulmonary injury, quercetin-3-glucuronide (Q3G) has been previously revealed the lung-protective potential via downregulation of inflammation, pyroptotic, and apoptotic cell death. However, the upstream signals mediating anti-pulmonary injury of Q3G have not yet been clarified. It has been reported that concerted dual activation of nuclear factor-erythroid 2 related factor 2 (Nrf2) and autophagy may prove to be a better treatment strategy in pulmonary injury. In this study, the effect of Q3G on antioxidant and autophagy were further investigated. Noncytotoxic doses of Q3G abolished the LPS-caused cell injury, and reactive oxygen species (ROS) generation with inductions in Nrf2-antioxidant signaling. Moreover, Q3G treatment repressed Nrf2 ubiquitination, and enhanced the association of Keap1 and p62 in the LPS-treated cells. Q3G also showed potential in inducing autophagy, as demonstrated by formation of acidic vesicular organelles (AVOs) and upregulation of autophagy factors. Next, the autolysosomes formation and cell survival were decreased by Q3G under pre-treatment with a lysosome inhibitor, chloroquine (CQ). Furthermore, mechanistic assays indicated that anti-pulmonary injury effects of Q3G might be mediated via Nrf2 signaling, as confirmed by the transfection of Nrf2 siRNA. Finally, Q3G significantly alleviated the development of pulmonary injury in vivo, which may result from inhibiting the LPS-induced lung dysfunction and edema. These findings emphasize a toxicological perspective, providing new insights into the mechanisms of Q3G's protective effects on LPS-induced pulmonary injury and highlighting its role in dual activating Nrf2 and autophagy pathways.
Collapse
Affiliation(s)
- Pei-Rong Yu
- Department of Nutrition, Chung Shan Medical University, Taichung City, 40201, Taiwan
| | - Chiao-Yun Tseng
- Department of Nutrition, Chung Shan Medical University, Taichung City, 40201, Taiwan
| | - Cheng-Chin Hsu
- Department of Nutrition, Chung Shan Medical University, Taichung City, 40201, Taiwan
| | - Jing-Hsien Chen
- Department of Nutrition, Chung Shan Medical University, Taichung City, 40201, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan
| | - Hui-Hsuan Lin
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan.
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Road, Taichung City, 40201, Taiwan.
| |
Collapse
|
9
|
Liu D, Guo R, Shi B, Chen M, Weng S, Weng J. Fortunellin ameliorates LPS-induced acute lung injury, inflammation, and collagen deposition by restraining the TLR4/NF-κB/NLRP3 pathway. Immun Inflamm Dis 2024; 12:e1164. [PMID: 38501503 PMCID: PMC10949398 DOI: 10.1002/iid3.1164] [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: 05/17/2023] [Revised: 12/29/2023] [Accepted: 01/10/2024] [Indexed: 03/20/2024] Open
Abstract
OBJECTIVE Acute lung injury (ALI) is the prevalent respiratory disease of acute inflammation with high morbidity and mortality. Fortunellin has anti-inflammation property, but its role in ALI remains elusive. Thus, this study clarified the function of fortunellin on ALI pathogenesis. METHODS The ALI mouse model was established by lipopolysaccharide (LPS) induction, and lung tissue damage was evaluated utilizing hematoxylin-eosin (HE) staining. The edema of lung tissue was measured by the lung wet/dry (W/D) ratio. The lung capillary permeability was reflected by the protein content in bronchoalveolar lavage fluid (BALF). Inflammatory cell infiltration was measured by the evaluation of the content of myeloperoxidase (MPO), neutrophils, and leukocytes in BALF. Cell apoptosis was measured by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. The secretions of inflammatory cytokines were quantified using enzyme-linked immunosorbent assay (ELISA) assays. Lung tissue collagen deposition was evaluated by Masson staining. RESULTS Fortunellin attenuated LPS-induced lung tissue damage and reduced the W/D ratio, the content of MPO in lung tissue, the total protein contents in BALF, and the neutrophils and leukocytes number. Besides, fortunellin alleviated LPS-stimulated lung tissue apoptosis, inflammatory response, and collagen deposition. Furthermore, Fortunellin repressed the activity of the Toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB)/NLR Family Pyrin Domain Containing 3 (NLRP3) pathway in the LPS-stimulated ALI model and LPS-induced RAW264.7 cells. Moreover, fortunellin attenuated LPS-stimulated tissue injury, apoptosis, inflammation, and collagen deposition of the lung via restraining the TLR4/NF-κB/NLRP3 pathway. CONCLUSION Fortunellin attenuated LPS-stimulated ALI through repressing the TLR4/NF-κB/NLRP3 pathway. Fortunellin may be a valuable drug for ALI therapy.
Collapse
Affiliation(s)
- Danjuan Liu
- Department of Critical Care Medicinethe Affiliated Hospital of Putian UniversityPutianChina
| | - Rongjie Guo
- Department of Critical Care Medicinethe Affiliated Hospital of Putian UniversityPutianChina
| | - Bingbing Shi
- Department of Critical Care Medicinethe Affiliated Hospital of Putian UniversityPutianChina
| | - Min Chen
- Department of Critical Care Medicinethe Affiliated Hospital of Putian UniversityPutianChina
| | - Shuoyun Weng
- School of Ophthalmology & OptometryWenzhou Medical UniversityWenzhouChina
| | - Junting Weng
- Department of Critical Care Medicinethe Affiliated Hospital of Putian UniversityPutianChina
| |
Collapse
|
10
|
Wang Z, Li F, Aga EB, Liang X, He C, Yin L, Xu F, Li H, Tang H, Lv C. 'Pterocephalodes hookeri-Onosma hookeri' decoction protects against LPS-induced pulmonary inflammation via inhibiting TLR4/ NF-κB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116918. [PMID: 37453619 DOI: 10.1016/j.jep.2023.116918] [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: 05/12/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As the second-largest traditional medical system in China, Tibetan medicine has a long history and abundant resources. To promote the development of the Tibetan medicine industry, it is essential to study the pharmacological activities of Tibetan medicine based on its traditional usage methods. AIM OF THE STUDY Pneumonia has been a worldwide health problem with high morbidity and mortality rates, especially in the context of the COVID-19 epidemic. Given the unique advantages of traditional Tibetan medicine in treating pulmonary diseases, further research is warranted to develop potential anti-pneumonia drugs. MATERIALS AND METHODS In our study, the potential combined decoction from traditional Tibetan medicine was determined by the data mining method. The antioxidant activity in vitro, anti-inflammatory effects on the macrophage cell model, as well as the anti-pulmonary inflammation effects on the LPS-induced mice model, have been explored to investigate the potential anti-pneumonia role of the decoction. Additionally, we conducted network pharmacology analysis to identify the potential targets against pneumonia, which were further confirmed by western blot assays. RESULTS Following the combination therapy of Pterocephalodes hookeri (C.B.Clarke) V.Mayer & Ehrend. and Onosma hookeri var. longiflora (Duthie) A.V.Duthie ex Stapf ('P-O'), the clearance of DPPH radical and the total reducing power were all improved, as well as alleviated the toxicity. On the in vitro level, 'P-O' pre-treatment reduced the secretion of NO, TNF-α, IL-6, and IL-1β in LPS-stimulated RAW264.7 cells, while promoting the concentration of IL-10. Meanwhile, on the in vivo level, the 'P-O' pre-treating also could alleviate LPS-induced pulmonary inflammation by reducing the pulmonary edema and leakage of the lung microvascular, improving the pathological change of lung tissue and regulating the cytokines content in bronchoalveolar lavage fluid (BALF). Furthermore, network pharmacology analysis revealed that the mechanism of 'P-O' in treating pneumonia in a multi-component, multi-target, and multi-pathway network, with the TLR4/NF-κB signaling pathway playing a crucial role, as demonstrated by the western blot assay results. CONCLUSION In summary, the combination therapy of 'P-O' exhibited good antioxidant activity and anti-inflammatory activity in vitro, as well as a therapeutic effect against pulmonary inflammation in vivo. These findings provide evidence for the clinical application of 'P-O' and offer new approaches for treating pneumonia.
Collapse
Affiliation(s)
- Zhenyu Wang
- Natural Medicine Research Center, Department of Pharmacy, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Fanglong Li
- Natural Medicine Research Center, Department of Pharmacy, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Er-Bu Aga
- Medical College, Tibet University, Lasa, 850000, China.
| | - Xiaoxia Liang
- Natural Medicine Research Center, Department of Pharmacy, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Changliang He
- Natural Medicine Research Center, Department of Pharmacy, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Lizi Yin
- Natural Medicine Research Center, Department of Pharmacy, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Funeng Xu
- Natural Medicine Research Center, Department of Pharmacy, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Haohuan Li
- Natural Medicine Research Center, Department of Pharmacy, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Huaqiao Tang
- Natural Medicine Research Center, Department of Pharmacy, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Cheng Lv
- Natural Medicine Research Center, Department of Pharmacy, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
11
|
Li J, He X, Liu F, Zheng X, Jiang J. Tumor Necrosis Factor-α-Induced Protein-8-like 2 Transfected Adipose-Derived Stem Cells Regulated the Dysfunction of Monocrotaline Pyrrole-Induced Pulmonary Arterial Smooth Muscle Cells and Pulmonary Arterial Endothelial Cells. J Cardiovasc Pharmacol 2024; 83:73-85. [PMID: 38180455 DOI: 10.1097/fjc.0000000000001505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/12/2023] [Indexed: 01/06/2024]
Abstract
ABSTRACT Pulmonary arterial hypertension (PAH) is characterized by pulmonary arterial endothelial cell (PAEC) dysfunction and pulmonary arterial smooth muscle cell (PASMC) activation. For decades, the therapies for PAH based on stem cells have been shown to be effective. Meanwhile, tumor necrosis factor-α-induced protein-8-like 2 (TIPE2) promote the viability of human amniotic mesenchymal stem cells. Therefore, we aimed to explore the role of TIPE2 in adipose-derived stem cells (ADSCs) and the function of TIPE2-transfected ADSCs in the regulation of PAH. We first explored the role and underlying molecular mechanism of TIPE2 in viability and migration of ADSCs. Moreover, the ADSCs transfected with TIPE2 were cocultured with monocrotaline pyrrole (MCTP)-stimulated PASMCs or PAECs. The effects and mechanisms of TIPE2-transfected ADSCs on MCTP-induced PASMCs and PAECs were further investigated. The results showed that TIPE2 overexpression promoted viability and migration of ADSCs by activating the TLR4-ERK1/2 pathway. In addition, TIPE2-transfected ADSCs inhibited the abnormal proliferation and the impaired apoptosis of PASMCs via NF-κB signaling and promoted the conversion of PASMCs from synthetic to contractile. Meanwhile, TIPE2-transfected ADSCs reduced the apoptosis, endothelial-to-mesenchymal transition, and migration of PAECs via PI3K/AKT signaling after MCTP treatment. MCTP-induced oxidative stress and inflammation of PAECs were significantly decreased by TIPE2-transfected ADSCs. In rat model, TIPE2-ADSCs administration further decreased the monocrotaline-induced increase in the right ventricular systolic pressure and ratio of right ventricle weight/left ventricle and septa weight (L + S) and right ventricle weight/body weight compared with the ADSCs group. In conclusion, TIPE2-transfected ADSCs dramatically attenuated the PAH via inhibiting the dysfunction of PASMCs and PAECs.
Collapse
Affiliation(s)
- Jing Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; and
| | - Xin He
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; and
| | - Feng Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; and
| | - Xinglong Zheng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; and
| | - Jing Jiang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
12
|
Wu XY, Wang FY, Chen HX, Dong HL, Zhao ZQ, Si LF. Chronic heat stress induces lung injury in broiler chickens by disrupting the pulmonary blood-air barrier and activating TLRs/NF-κB signaling pathway. Poult Sci 2023; 102:103066. [PMID: 37769490 PMCID: PMC10539940 DOI: 10.1016/j.psj.2023.103066] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 10/02/2023] Open
Abstract
As an important respiratory organ, the lung is susceptible to damage during heat stress due to the accelerated breathing frequency caused by an increase in environmental temperature. This can affect the growth performance of animals and endanger their health. This study aimed to explore the mechanism of lung tissue damage caused by heat stress. Broilers were randomly divided into a control group (Control) and a heat stress group (HS). The HS group was exposed to 35°C heat stress for 12 h per d from 21-days old, and samples were taken from selected broilers at 28, 35, and 42-days old. The results showed a significant increase in lactate dehydrogenase (LDH) activity in the serum and myeloperoxidase (MPO) activity in the lungs of broiler chickens across all 3 age groups after heat stress (P < 0.01), while the total antioxidant capacity (T-AOC) was significantly enhanced at 35-days old (P < 0.01). Heat stress also led to significant increases in various proinflammatory factors in serum and expression levels of HSP60 and HSP70 in lung tissue. Histopathological results showed congestion and bleeding in lung blood vessels, shedding of pulmonary epithelial cells, and a large amount of inflammatory infiltration in the lungs after heat stress. The mRNA expression of TLRs/NF-κB-related genes showed an upward trend (P < 0.05) after heat stress, while the mRNA expression of MLCK, a gene related to pulmonary blood-air barrier, significantly increased after heat stress, and the expression levels of MLC, ZO-1, and occludin decreased in contrast. This change was also confirmed by Western blotting, indicating that the pulmonary blood-air barrier is damaged after heat stress. Heat stress can cause damage to the lung tissue of broiler chickens by disrupting the integrity of the blood-air barrier and increasing permeability. This effect is further augmented by the activation of TLRs/NF-κB signaling pathways leading to an intensified inflammatory response. As heat stress duration progresses, broiler chickens develop thermotolerance, which gradually mitigates the damaging effects induced by heat stress.
Collapse
Affiliation(s)
- Xing-Yue Wu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, PR China
| | - Fei-Yao Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, PR China
| | - Hao-Xiang Chen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, PR China
| | - Hui-Li Dong
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, PR China
| | - Zhan-Qin Zhao
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, PR China
| | - Li-Fang Si
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, PR China.
| |
Collapse
|
13
|
Yuan M, Jing G, Kong Q, Ming T, Zuo J, Wang Q, Feng Y, Liu W, Wu X, Xia Z. TIPE2 ameliorates neuroinflammation and cognitive impairment in sepsis-associated encephalopathy through regulating RhoA/ROCK2-NF-κB signaling pathway. Biochem Pharmacol 2023; 217:115816. [PMID: 37748665 DOI: 10.1016/j.bcp.2023.115816] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 09/27/2023]
Abstract
Sepsis-associated encephalopathy (SAE) is an acute brain dysfunction induced by systemic inflammation caused by sepsis and is one of the most common types of encephalopathy in intensive care units. Deteriorative neuroinflammation is closely related to the development of brain injury, which often transforms into common pathological manifestations in patients with severe sepsis. Therefore, taking necessary preventive and protective measures for potential brain injury and promptly reducing neuroinflammatory injury is necessary to improve the long-term prognoses of patients. Tumor necrosis factor-α-induced protein 8-like 2 (TIPE2) can play a significant protective role in septic lung injury, but studies on its expression and role in neurological diseases are rare. In the present study, we found that TIPE2 can expressed in microglia and ameliorate brain injury caused by SAE by suppressing neuroinflammation. The RhoA/ROCK2 pathway is the central coordinator of tissue injury response, and the activation of RhoA participates in the lipopolysaccharide-induced activation of the nuclear factor kappa B (NF-κB) signaling pathway. The activation of RhoA and phosphorylation of NF-κB was enhanced after TIPE2 deficiency. Importantly, TIPE2 negatively regulates inflammatory responses in vivo and in vitro and plays a protective role in SAE by inhibiting the activation of RhoA/ROCK2-NF-κB signaling pathways. The ultimate aim of our proposed project is to provide a theoretical basis for the development of a novel strategy for the early prevention and therapy of SAE.
Collapse
Affiliation(s)
- Min Yuan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Guoqing Jing
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qian Kong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Tingqian Ming
- Department of Anesthesiology, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Jing Zuo
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qian Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yong Feng
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Wanhong Liu
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
14
|
Liang H, Liu G, Fan Q, Nie Z, Xie S, Zhang R. Limonin, a novel AMPK activator, protects against LPS-induced acute lung injury. Int Immunopharmacol 2023; 122:110678. [PMID: 37481848 DOI: 10.1016/j.intimp.2023.110678] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/05/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
AMP-activated protein kinase (AMPK) activation plays crucial roles in the treatment of many oxidative stress- and inflammation-induced diseases, including acute lung injury (ALI). Limonin is a naturally occurring tetracyclic triterpenoid extracted from the plants of Rutaceae and Meliaceae. Limonin also serves as an AMPK activator with anti-inflammatory and anti-oxidation effects. However, the potential beneficial effects of limonin on ALI and the possible mechanisms have never been disclosed till now. Here, the effects of limonin on lipopolysaccharide (LPS)-induced ALI in C57 BL/6 mice, plus bone marrow-derived macrophages (BMDM) stimulated with LPS to induce in vitro ALI model were investigated. Limonin significantly improved pulmonary function and alleviated lung pathological injury in LPS-induced mice. Meanwhile, limonin also markedly decreased inflammation and oxidative stress in lung tissues from LPS-treated mice. In vitro experiments also unveiled that limonin could decrease inflammation and oxidative stress in LPS-induced BMDM in a concentration-dependent manner. Mechanically, limonin could promote the activation of AMPKα and upregulate the expression of nuclear factor erythroid 2-related factor 2 (NRF2) in lung tissues and BMDM. Pharmacological inhibition of AMPKα by Compound C or AMPKα knockout could abolish the pulmonary protection from limonin during ALI. In conclusion, limonin mediates the activation of AMPKα/NRF2 pathway, providing an attractive therapeutic target for ALI in the future.
Collapse
Affiliation(s)
- Hui Liang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Gaoli Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qinglu Fan
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhihao Nie
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Songping Xie
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Renquan Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
15
|
Gao J, Zhang H, Zhang F. Research progress of TIPE2 in immune-related diseases. Int Immunopharmacol 2023; 121:110514. [PMID: 37348234 DOI: 10.1016/j.intimp.2023.110514] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/02/2023] [Accepted: 06/13/2023] [Indexed: 06/24/2023]
Abstract
The tumor necrosis factor α-induced protein 8 (TNFAIP8) family, which consists of TNFAIP8 (TIPE), TNFAIP8L1 (TIPE1), TNFAIP8L2 (TIPE2) and TNFAIP8L3 (TIPE3), has recently emerged as a regulatory factor involved in immune response and tumorigenesis. Among its members, TIPE2 acts as a negative regulator of both innate and adaptive immunity, playing a crucial role in maintaining immune homeostasis by negatively regulating T cell receptor (TCR) and toll-like receptor (TLR) signal transduction. Immune homeostasis is an indispensable characteristic of the immune system, which prevents harmful inflammatory reactions and ensures the proper functioning of the body. A large number of studies have shown that abnormal TIPE2 expression exists in a variety of inflammation-related diseases such as asthma, colitis, and systemic lupus erythematosus, highlighting the importance of comprehending its function for the prevention and treatment of immune-related conditions. This review aims to provide an overview of the in vivo distribution and expression of TIPE2, its regulatory role in central and peripheral immune-related diseases, and the underlying mechanisms that govern its function in the inflammatory response. By delving into these aspects, a deeper understanding of the role and functionality of TIPE2 in inflammatory responses can be achieved.
Collapse
Affiliation(s)
- Jie Gao
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao 266000, China.
| | - Hanting Zhang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao 266000, China.
| | - Fang Zhang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao 266000, China.
| |
Collapse
|
16
|
Yang Z, Zhang F, Abdul M, Jiang J, Li Y, Li Y, Yin C, Xing Y, Liu S, Lu C. Tumor necrosis factor-α-induced protein 8-like 2 alleviates morphine antinociceptive tolerance through reduction of ROS-mediated apoptosis and MAPK/NF-κB signaling pathways. Neuropharmacology 2023:109667. [PMID: 37451333 DOI: 10.1016/j.neuropharm.2023.109667] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 07/09/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Chronic morphine tolerance is a repulsive barrier to the clinical treatment of pain. Whereas the underlying molecular mechanisms of morphine tolerance remain unknown. Here, we proposed that tumor necrosis factor-α-induced protein 8-like 2 (TIPE2) is an essential control point regarding the progression of chronic morphine tolerance. We found that TIPE2 levels in the lumbar spinal cord were significantly downregulated in the morphine tolerance mouse model. Specifically, decreased TIPE2 by morphine tolerance was primarily expressed in spinal neurons, while increased expression of spinal TIPE2 distinctly attenuated the chronic morphine antinociceptive tolerance and tolerance-associated hyperalgesia. We also observed that increased expression of spinal TIPE2 significantly reduced morphine tolerance-induced neuronal ROS production and apoptosis, along with the activation of MAPKs and NF-κB signaling pathways. Moreover, the increased TIPE2 expression inhibited neuronal activation and glial reactivity in the spinal dorsal horn after chronic morphine exposure. Additionally, TIPE2 overexpression in cultured SH-SY5Y cells significantly suppressed ROS production and apoptosis in response to morphine challenge. Therefore, we can conclude that the upregulation of spinal TIPE2 may attenuate the morphine antinociceptive tolerance via TIPE2-dependent downregulation of neuronal ROS, inhibition of neuronal apoptosis, suppression of MAPKs and NF-κB activation. TIPE2 may be a potential strategy for preventing morphine tolerance in the future studies and clinical settings. Schematic diagram for the proposed mechanisms of TIPE2 regulates morphine antinociceptive tolerance. TIPE2 may alleviate morphine antinociceptive tolerance by regulating MAPK/NF-κB signaling pathways and apoptosis, which might be associated with ROS production.
Collapse
Affiliation(s)
- Zhong Yang
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Feifei Zhang
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Mannan Abdul
- Department of Anesthesiology, Eye & ENT Hospital, Fudan University, Shanghai, China; School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jinhong Jiang
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yanqiang Li
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yeqi Li
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Cui Yin
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yanhong Xing
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Su Liu
- Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Chen Lu
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
17
|
Gao J, Wang N, Zong F, Dong J, Lin Y, Zhang H, Zhang F. TIPE2 regulates the response of BV2 cells to lipopolysaccharide by the crosstalk between PI3K/AKT signaling and microglia M1/M2 polarization. Int Immunopharmacol 2023; 120:110389. [PMID: 37245300 DOI: 10.1016/j.intimp.2023.110389] [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: 02/16/2023] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 05/30/2023]
Abstract
Tumor necrosis factor (TNF)-α-induced protein 8-like 2 (TIPE2) is a crucial negative regulator of both adaptive and innate immunity, which helps maintain the dynamic balance of the immune system by negatively regulating the signaling of T-cell receptors (TCR) and Toll-like receptors (TLR). In this study, we aimed to investigate the role and molecular mechanism of TIPE2 using a lipopolysaccharide (LPS)-induced inflammatory injury model in BV2 cells. Specifically, we constructed a BV2 cell line of TIPE2-overexpression or TIPE2-knockdown via lentiviral transfection. Our results demonstrated that overexpression of TIPE2 downregulated the expression of pro-inflammatory cytokines IL-1β and IL-6, which was reversed by knockdown of TIPE2 in the inflammation model of BV2 cells. In addition, overexpression of TIPE2 resulted in the conversion of BV2 cells to the M2 phenotype, while the knockdown of TIPE2 promoted the transformation of BV2 cells to the M1 phenotype. Notably, our co-culture experiments with neuronal cells SH-SY5Y showed that the overexpression of TIPE2 in inflammation-injured BV2 cells exhibited a protective effect on the neuronal cells. Finally, western blot analysis demonstrated that TIPE2 significantly reduced the expression of p-PI3K, p-AKT, p-p65, and p-IκBα in LPS treated BV2 cells, and inhibited the activation of NF-κB through the dephosphorylation of PI3K/AKT. These results suggest that TIPE2 plays an important role in mediating neuroinflammatory responses and may be involved in neuroprotection by modulating the phenotypic changes of BV2 cells and regulating the pro-inflammatory responses through the PI3K/AKT and NF-κB signaling pathways. In conclusion, our study provides new insights into the crucial role of TIPE2 in regulating neuroinflammatory responses and highlights its potential as a therapeutic target for neuroprotection.
Collapse
Affiliation(s)
- Jie Gao
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao 266000, China.
| | - Naidong Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Fangjiao Zong
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao 266000, China.
| | - Jiahao Dong
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao 266000, China.
| | - Yuanyuan Lin
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao 266000, China.
| | - Hanting Zhang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao 266000, China.
| | - Fang Zhang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao 266000, China.
| |
Collapse
|
18
|
Zeng J, Liu J, Huang JH, Fu SP, Wang XY, Xi C, Cui YR, Qu F. Aloperine alleviates lipopolysaccharide-induced acute lung injury by inhibiting NLRP3 inflammasome activation. Int Immunopharmacol 2023; 120:110142. [PMID: 37210910 DOI: 10.1016/j.intimp.2023.110142] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 05/23/2023]
Abstract
RATIONALE Excessive activation of the NLRP3 inflammasome is involved in the pathological progression of acute lung injury (ALI). Aloperine (Alo) has anti-inflammatory effects in many inflammatory disease models; however, its role in ALI remains elusive. In this study, we addressed the role of Alo in NLRP3 inflammasome activation in both ALI mice and LPS-treated RAW264.7 cells. METHODS The activation of the NLRP3 inflammasome in LPS-induced ALI lungs was investigated in C57BL/6 mice. Alo was administered in order to study its effect on NLRP3 inflammasome activation in ALI. RAW264.7 cells were used to evaluate the underlying mechanism of Alo in the activation of the NLRP3 inflammasome in vitro. RESULTS The activation of the NLRP3 inflammasome occurs in the lungs and RAW264.7 cells under LPS stress. Alo attenuated the pathological injury of lung tissue as well as downregulates the mRNA expression of NLRP3 and pro-caspase-1 in ALI mice and LPS-stressed RAW264.7 cells. The expression of NLRP3, pro-caspase-1, and caspase-1 p10 were also significantly suppressed by Alo in vivo and in vitro. Furthermore, Alo decreased IL-1β and IL-18 release in ALI mice and LPS-induced RAW264.7 cells. In addition, ML385, a Nrf2 inhibitor, weakened the activity of Alo, which inhibited the activation of the NLRP3 inflammasome in vitro. CONCLUSION Alo reduces NLRP3 inflammasome activation via the Nrf2 pathway in ALI mice.
Collapse
Affiliation(s)
- Jie Zeng
- Department of Physiology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China; Jiangxi Medical College, Shangrao, Jiangxi 334000, China
| | - Jie Liu
- Department of Physiology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Jun-Hao Huang
- Department of Pharmacology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | | | - Xin-Yi Wang
- Department of Physiology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Chao Xi
- Department of Pharmacology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Yan-Ru Cui
- Department of Physiology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China; Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China.
| | - Fei Qu
- Department of Pharmacology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China.
| |
Collapse
|
19
|
Yang H, Wu Q, Li J, Chen Q, Su L, He X, Li J, Qiu X. In Vivo Fate of CXCR2-Overexpressing Mesenchymal Stromal/Stem Cells in Pulmonary Diseases Monitored by Near-Infrared Region 2 Imaging. ACS APPLIED MATERIALS & INTERFACES 2023; 15:20742-20752. [PMID: 37071603 DOI: 10.1021/acsami.3c01741] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Lung-associated diseases pose a huge threat to human society. Mesenchymal stromal/stem cells (MSCs) hold great promise in the treatment of pulmonary diseases through cell transdifferentiation, paracrine factors, immune regulation, EV secretion, and drug loading. However, intravenous injection of MSCs often resulted in limited lesion tropism and apparent off-target accumulation. The IL-8-CXCR1/2 chemokine axis has been shown to be involved in progression of diseases including lung cancer and acute lung injury (ALI). Herein, we took advantage of this chemokine axis to enhance the homing of MSCs to cancerous and inflammation lesions. The in vivo distribution of MSCs was further monitored real-time by near-infrared region 2 (NIR-II) imaging owing to its outstanding performance in deep tissue imaging. Specifically, a new high-brightness D-A-D NIR-II dye, LJ-858, was synthesized and coprecipitated with a poly(d,l-lactic acid) polymer to form LJ-858 nanoparticles (NPs) with a relative quantum yield of 14.978%. LJ-858 NPs can efficiently label MSCs, and the NIR-II signal can be stable for 14 days without compromising the cell viability. Subcutaneous tracking of labeled MSCs showed no significant decline of NIR-II intensity within 24 h. The enhanced tropism of CXCR2-overexpressing MSCs to A549 tumor cells and the inflamed lung tissue was demonstrated through transwell models. The in vivo and ex vivo NIR-II imaging results further validated the significantly enhanced lesion retention of MSCCXCR2 in the lung cancer and ALI models. Taken together, this work reported a robust strategy to enhance the pulmonary disease tropism by the IL-8-CXCR1/2 chemokine axis. In addition, in vivo distribution of MSCs was successfully visualized by NIR-II imaging, which provides more insights into optimizing protocols for MSC-based therapies in the future.
Collapse
Affiliation(s)
- Huiying Yang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qingxia Wu
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jinwei Li
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qimingxing Chen
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Lili Su
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaoyan He
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jianfeng Li
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaoyan Qiu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
20
|
Yang X, Wu Y, Zhang M, Zhang L, Zhao T, Qian W, Zhu M, Wang X, Zhang Q, Sun J, Dong L. Piceatannol protects against age-related hearing loss by inhibiting cellular pyroptosis and inflammation through regulated Caspase11-GSDMD pathway. Biomed Pharmacother 2023; 163:114704. [PMID: 37100013 DOI: 10.1016/j.biopha.2023.114704] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/03/2023] [Accepted: 04/12/2023] [Indexed: 04/28/2023] Open
Abstract
Age-related hearing loss (ARHL) is a common issue associated with aging. One of the typical causes of hearing loss is the damage to inner ear hair cells. In addition, oxidative stress and inflammation contribute to ARHL. To avoid excessive inflammatory responses, non-classical scorch death pathway by cell membrane lipopolysaccharide (LPS) activates of caspase-11. Piceatannol (PCT) is also known for anti-tumor, antioxidant and anti-inflammatory effects; however, the protective effect of piceatannol (PCT) on ARHL is unclear. The aim of this study was to elucidate the mechanism underlying protective effect of PCT on ARHL-induced inner ear hair cell damage. In vivo experiments showed that PCT could protect mice from inflammatory aging-induced hearing loss as well as from inner hair cells (IHC) and spiral ganglion (SG) deficits. In addition, inflammatory vesicle inhibitor BAY11-7082 ameliorated ARHL, inhibited NLRP3 and reduced GSDMD expression. In in vitro experiments we used LPS and D-gal to simulate the aging inflammatory environment. The results showed that intracellular reactive oxygen species levels, expression of Caspase-11, NLRP3, and GSDMD were significantly increased, yet treatment with PCT or BAY11-7082 significantly improved HEI-OC-1 cell injury while reducing inflammation-associated protein expression as well as the occurrence of pyroptosis. In conclusion, these results suggest a protective role for PCT against ARHL, possibly through Caspase-11-GSDMD pathway. Our findings may provide a new target and theoretical basis for hearing loss treatment using PCT.
Collapse
Affiliation(s)
- Xu Yang
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology of Ministry of Education, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Yanlin Wu
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of USTC, Hefei 230001, China
| | - Menglian Zhang
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology of Ministry of Education, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Lingyu Zhang
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology of Ministry of Education, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Tianhao Zhao
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology of Ministry of Education, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Weiwei Qian
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology of Ministry of Education, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Mengmei Zhu
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology of Ministry of Education, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Xinya Wang
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology of Ministry of Education, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Qiannuo Zhang
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology of Ministry of Education, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Jiaqiang Sun
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of USTC, Hefei 230001, China.
| | - Liuyi Dong
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology of Ministry of Education, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
21
|
Wang Y, Deng F, Zhong X, Du Y, Fan X, Su H, Pan T. Dulaglutide provides protection against sepsis-induced lung injury in mice by inhibiting inflammation and apoptosis. Eur J Pharmacol 2023; 949:175730. [PMID: 37062504 DOI: 10.1016/j.ejphar.2023.175730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/04/2023] [Accepted: 04/14/2023] [Indexed: 04/18/2023]
Abstract
Sepsis is a dangerous condition with a high mortality rate. In addition to promoting insulin secretion in a glucose-dependent manner, glucagon-like peptide-1 (GLP-1) also exhibits anti-inflammatory properties. Dulaglutide is a glucagon-like peptide-1 receptor agonist (GLP-1 RA). In this study, we investigated the effects and mechanism of action of dulaglutide (Dul) in lipopolysaccharide (LPS) induced lung injury in mice with sepsis. In mice with LPS (15 mg/kg, ip, qd)-induced acute lung injury, the administration of dulaglutide (0.6 mg/kg, ip, qd) improved weight loss, reduced lung injury, reversed the increase in IL-1β, TNF-α, IL-6, CXCL1, CCL2 and CXCL2 expression in the lung, and reduced the infiltration of neutrophils and macrophages in the lung tissues. The decline in caspase-3, cleaved caspase-3, caspase-8, and Bcl-2/Bax expression and the increase in the number of TUNEL positive cells in the lung were reversed, suggesting that GLP-1RA could play a protective role in the lung by inhibiting inflammation and apoptosis. In addition, GLP-1RA could reduce the expression of P-STAT3 and NLRP3, suggesting that P-STAT3 and NLRP3 may be potential targets against lung injury in sepsis. Collectively, our data demonstrated that GLP-1RA exerts a protective effect against sepsis-induced lung injury through mechanisms related to the inhibition of inflammation, apoptosis, and STAT3 signaling.
Collapse
Affiliation(s)
- Yue Wang
- Department of Endocrinology, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Jingkai District, Hefei, 230061, Anhui Province, China; Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Jingkai District, Hefei 230061, Hefei, 230061, Anhui Province, China
| | - Fengyi Deng
- Department of Endocrinology, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Jingkai District, Hefei, 230061, Anhui Province, China; Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Jingkai District, Hefei 230061, Hefei, 230061, Anhui Province, China
| | - Xing Zhong
- Department of Endocrinology, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Jingkai District, Hefei, 230061, Anhui Province, China; Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Jingkai District, Hefei 230061, Hefei, 230061, Anhui Province, China
| | - Yijun Du
- Department of Endocrinology, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Jingkai District, Hefei, 230061, Anhui Province, China; Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Jingkai District, Hefei 230061, Hefei, 230061, Anhui Province, China
| | - Xingyu Fan
- Department of Endocrinology, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Jingkai District, Hefei, 230061, Anhui Province, China; Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Jingkai District, Hefei 230061, Hefei, 230061, Anhui Province, China
| | - Hong Su
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Shushan District, Hefei, 230031, Anhui Province, China
| | - Tianrong Pan
- Department of Endocrinology, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Jingkai District, Hefei, 230061, Anhui Province, China; Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Jingkai District, Hefei 230061, Hefei, 230061, Anhui Province, China.
| |
Collapse
|
22
|
Song J, Yang Q, Xiong H, Gu X, Chen M, Zhou C, Cai Y. TIPE3 protects mice from lipopolysaccharide-induced acute lung injury. Transpl Immunol 2023; 77:101799. [PMID: 36842565 DOI: 10.1016/j.trim.2023.101799] [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: 08/21/2022] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 02/28/2023]
Abstract
BACKGROUND Acute lung injury (ALI) is a severe inflammatory disease with high morbidity and mortality in patients and lung transplant recipients. Tumor necrosis factor-α-induced protein 8-like 3 (TIPE3) is one of the members of the TIPE family. While TIPE2 has been demonstrated to be protective against lipopolysaccharide (LPS)-induced ALI, the role of TIPE3 in ALI is currently unidentified. METHODS To examine the role of TIPE3 in ALI, we pretreated C57BL/6 mice with control or TIPE3-lentivirus in LPS-induced ALI models. The C57BL/6 mice were randomly divided into four groups: control group; ALI-induced group; ALI-induced group with control lentivirus; and ALI-induced group with TIPE3-lentivirus. Additionally, RAW 264.7 cells were used to validate the role and molecular mechanism of TIPE3 signaling in vitro. RESULTS An increased expression of TIPE3 reduced lung histopathological damage in ALI-affected mice. ALI-affected mice treated with TIPE3-lentivirus exhibited reduced lung microvascular permeability, myeloperoxidase (MPO) activity, neutrophil buildup, and inflammation response. Additionally, over-expression of TIPE3 significantly inhibited NF-κB activation and promoted the activation of Liver X receptors alpha (LXRα). In LPS-treated RAW264.7 cells, enforced TIPE3 expression produced anti-inflammatory effects, whereas the LXR inhibitor geranylgeranyl pyrophosphate (GGPP) reversed these effects. CONCLUSIONS TIPE3 protected against LPS-induced ALI by regulating the LXRα/NF-κB signaling pathway. These results suggest that TIPE3 might provide a novel insight into the prevention of ALI.
Collapse
Affiliation(s)
- Jie Song
- Department of Pediatrics, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Qiuping Yang
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Hui Xiong
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Xia Gu
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Mo Chen
- Department of Pediatrics, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Chuanxin Zhou
- Department of Pediatrics, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China.
| | - Yao Cai
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China.
| |
Collapse
|
23
|
Tian Y, Zhu CL, Li P, Li HR, Liu Q, Deng XM, Wang JF. Nicotinamide Mononucleotide Attenuates LPS-Induced Acute Lung Injury With Anti-Inflammatory, Anti-Oxidative and Anti-Apoptotic Effects. J Surg Res 2023; 283:9-18. [PMID: 36347171 DOI: 10.1016/j.jss.2022.09.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 08/23/2022] [Accepted: 09/18/2022] [Indexed: 11/07/2022]
Abstract
INTRODUCTION Nicotinamide mononucleotide (NMN) is a nucleotide that is commonly recognized for its role as an intermediate of nicotinamide adenine dinucleotide (NAD+) biosynthesis with multiple pharmacological effects. The purpose of this study was to evaluate the protective effect of nicotinamide mononucleotide (NMN) against lipopolysaccharide (LPS)-induced acute lung injury (ALI). METHODS We investigated the effect of NMN on ALI-induced inflammatory response, oxidative stress, and cell apoptosis. The ALI mouse model was performed by injecting LPS intratracheally at a dose of 10 mg/kg in 50 μL saline. Flow cytometry was used to detect neutrophil infiltration in bronchoalveolar lavage fluid (BALF), and ELISA was used to detect the contents of inflammatory cytokines TNF-α, IL-1β and IL-6 in BALF. Oxidative stress was evaluated by determining the superoxide dismutase (SOD) activity and malondialdehyde (MDA) content in lung tissue. ROS formation was analyzed by immunofluorescence. Western blotting was performed to detect apoptotic levels and p38MAPK/NF-κB phosphorylation levels in lung tissue. RESULTS In the ALI mouse model, NMN showed a significant therapeutic effect compared to the LPS group. NMN attenuated the pathological damage and cell apoptosis in lung tissue, decreased the levels of TNF-α, IL-1β, and IL-6 in BALF, and reduced the number of total cells and neutrophils in BALF. In addition, NMN attenuated the LPS-induced elevation of dry-to-wet ratio, MDA content, p38 MAPK and p65 NF-κB phosphorylation levels, and the SOD activity was increased by NMN treatment. CONCLUSIONS In conclusion, the present study showed that NMN exerted a protective effect on LPS-induced ALI with anti-inflammatory, antioxidative, and antiapoptotic effects.
Collapse
Affiliation(s)
- Ye Tian
- Department of Anesthesiology, The Sixth Medical Centre of General Hospital of PLA, Beijing, China
| | - Cheng-Long Zhu
- Changhai Hospital, Naval Medical University, Shanghai, China
| | - Peng Li
- Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hui-Ru Li
- Changhai Hospital, Naval Medical University, Shanghai, China
| | - Qiang Liu
- Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiao-Ming Deng
- Changhai Hospital, Naval Medical University, Shanghai, China.
| | - Jia-Feng Wang
- Changhai Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
24
|
Shan C, Xiong Y, Miao F, Liu T, Akhtar RW, Shah SAH, Gao H, Zhu E, Cheng Z. Hydroxytyrosol mitigates Mycoplasma gallisepticum-induced pulmonary injury through downregulation of the NF-κB/NLRP3/IL-1β signaling pathway in chicken. Poult Sci 2023; 102:102582. [PMID: 36940652 PMCID: PMC10033309 DOI: 10.1016/j.psj.2023.102582] [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: 01/05/2023] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/19/2023] Open
Abstract
In this study, the anti-inflammatory and antiapoptotic effects of hydroxytyrosol (HT) in Mycoplasma gallisepticum (MG)-infected chicken were investigated, and the underlying molecular mechanisms were explored. The results revealed severe ultrastructural pathological changes after MG infection in the lung tissue of chicken, including inflammatory cell infiltration, thickening of the lung chamber wall, visible cell swelling, mitochondrial cristae rupture, and ribosome shedding. MG possibly activated the nuclear factor κB (NF-κB)/nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3)/interleukin (IL)-1β signaling pathway in the lung. However, HT treatment significantly ameliorated MG-induced pathological damage of the lung. HT reduced the magnitude of pulmonary injury after MG infection by reducing apoptosis and releasing the proinflammatory factors. Compared with the MG-infected group, the HT-treated group exhibited significant inhibition of the expression of NF-κB/NLRP3/IL-1β signaling-pathway-related genes; for example, the expressions of NF-κB, NLRP3, caspase-1, IL-1β, IL-2, IL-6, IL-18, and TNF-α significantly decreased (P < 0.01 or <0.05). In conclusion, HT effectively inhibited MG-induced inflammatory response and apoptosis and protected the lung by blocking the activation of NF-κB/NLRP3/IL-1β signaling pathway and reducing the damage caused by MG infection in chicken. This study revealed that HT may be a suitable and effective anti-inflammatory drug against MG infection in chicken.
Collapse
Affiliation(s)
- Chunlan Shan
- College of Animal Science, Guizhou University, Guiyang 550000, PR China
| | - Yanling Xiong
- College of Animal Science, Guizhou University, Guiyang 550000, PR China
| | - Fujun Miao
- Yunnan Academy of Forestry and Grassland, Kunming 650204, PR China
| | - Ting Liu
- College of Animal Science, Guizhou University, Guiyang 550000, PR China
| | - Rana Waseem Akhtar
- Department of Animal Breeding and Genetics, Faculty of Veterinary and Animal Sciences, The Islamia University, Bahawalpur 63100, Pakistan
| | - Syed Aftab Hussain Shah
- Pakistan Scientific & Technological Information Center, Quaid-i-Azam University Campus, Islamabad 44000, Pakistan
| | - Hong Gao
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, PR China
| | - Erpeng Zhu
- College of Animal Science, Guizhou University, Guiyang 550000, PR China
| | - Zhentao Cheng
- College of Animal Science, Guizhou University, Guiyang 550000, PR China.
| |
Collapse
|
25
|
Li B, Wang Z, Yuan J, Liang D, Cheng Y, Wang Z. Knockdown of SHP2 attenuated LPS-induced ferroptosis via downregulating ACSL4 expression in acute lung injury. Allergol Immunopathol (Madr) 2023; 51:143-152. [PMID: 37169572 DOI: 10.15586/aei.v51i3.856] [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: 02/28/2023] [Accepted: 03/21/2023] [Indexed: 05/13/2023]
Abstract
BACKGROUND Acute lung injury (ALI) is a complex disease with a high mortality. Src homology 2 (SH2)-containing protein tyrosine phosphatase 2 (SHP2) is a protein tyrosine phosphatase that participates in pathogenesis of multiple diseases. Nevertheless, the role of SHP2 in ALI remains unknown. METHODS The in vivo and in vitro lipopolysaccharide (LPS)-induced ALI models were successfully established. The histopathological changes were evaluated by hematoxylin and eosin staining. The vascular permeability of lungs was assessed by Evans blue assay. The expression of ACSL4 and SHP2 was detected by western blot and qRT-PCR assay. The lactate dehydrogenase (LDH) activity, malondialdehyde (MDA), iron, and glutathione (GSH) levels were measured by commercial kits. RESULTS The SHP2 was upregulated in LPS-induced ALI mice and LPS-stimulated MLE-12 cells. In loss-of function experiment, the knockdown of SHP2 attenuated LPS-induced lung injury, microvessels damage, pulmonary edema, and increase of lung vascular permeability in vivo. Mechanically, shSHP2-rescued LPS induced increase in LDH activity, MDA, and iron levels, and decrease in GSH levels, as well as the accumulation of reactive oxygen species in vivo and in vitro, leading to an alleviation of LPS-induced ferroptosis. Notably, shSHP2 reduced the expression of Acyl-CoA synthetase long-chain 4 (ACSL4). In the rescued experiments, overexpression of ACSL4 abolished the shSHP2-induced reduction of LDH activity, MDA, and iron levels, and increase in GSH levels, thereby aggravating the LPS-induced ferroptosis. CONCLUSION These findings concluded that the knockdown of SHP2 attenuated LPS-induced ferroptosis via downregulation of ACSL4 expression in ALI, providing a novel sight for ALI treatment.
Collapse
Affiliation(s)
- Bin Li
- Department of Infectious Diseases, Linfen People's Hospital, Linfen, Shanxi, China
| | - Zhan Wang
- Research Division, National Health Commission of the People's Government of Linfen City, Linfen, Shanxi, China
| | - Jiayang Yuan
- Department of Infectious Diseases, Linfen People's Hospital, Linfen, Shanxi, China
| | - Dachuan Liang
- Department of Infectious Diseases, Linfen People's Hospital, Linfen, Shanxi, China
| | - Yanrong Cheng
- Intensive Care Unit, Linfen People's Hospital, Linfen, Shanxi, China
| | - Zheng Wang
- Intensive Care Unit, Linfen People's Hospital, Linfen, Shanxi, China;
| |
Collapse
|
26
|
Long noncoding RNA HOXA-AS2 ameliorates chronic intermittent hypoxia-induced lung inflammation by regulating miR-17-5p/tipe2 axis. Allergol Immunopathol (Madr) 2023; 51:36-44. [PMID: 36916086 DOI: 10.15586/aei.v51i2.701] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/27/2022] [Indexed: 03/08/2023]
Abstract
PURPOSE The purpose is to confirm whether long noncoding RNA HOXA-AS2 relieves chronic intermittent hypoxia (CIH)-induced lung inflammation. METHODS Male Sprague Dawley rats were used to establisha CIH rat model. Hematoxylin and Eosin staining was used on the lung tissue injury to determine the successful construction of CIH animal model. Arterial partial pressure of oxygen (PaO2) and carbon dioxide (PaCO2) were measured. HOXA-AS2 was overexpressed to evaluate its role in the progression and development of CIH. T cell differentiation and cytokine production were determined using flow cytometry. Cell apoptosis was determined using terminal deoxynucleotidyl transferase dUTP nick end labelling assay kit. The target of HOXA-AS2 and miR-17-5p was predicted by the Encyclopedia of RNA Interactomes (ENCORI) and confirmed using luciferase assay. RESULTS HOXA-AS2 was downregulated in CIH rat models. Lung tissue injury was observed in CIH rats, and the injury was attenuated by the overexpression of HOXA-AS2. PaO2 was reduced and PaCO2 was induced in CIH rats, which was reversed by the overexpression of HOXA-AS2. The overexpression of HOXA-AS2 inhibited CIH-induced cell apoptosis. It also reversed alterations in the levels of interferon gamma (IFNγ), interleukin (IL)-2, IL-6, IL-1β, tumor necrosis factor alpha (TNF-α), and transforming growth factor beta1 (TGF-β1) in rats caused by CIH. The overexpression of HOXA-AS2 prevented the induction in CD4+ IFN-γ+ T cells and reduction in CD4+TGF-β1+ T cells. The overexpression of HOXA-AS2 upregulated tumor necrosis factor-alpha-induced protein 8-like 2 (tipe2) key regulator through directly targeting miR-17-5p. Further experiments proved that tipe2 was the direct target of miR-17-5p. CONCLUSION This study manifested that HOXA-AS2 acted as an anti-inflammatory regulator and protected lung tissue injury from CIH in the rat model; this was mediated by upregulation of tipe2 through directly targeting miR-17-5p. HOXA-AS2 upregulated the expression of tipe2, providing new understanding and therapeutic target for CIH.
Collapse
|
27
|
Jian R, He X. TIPE2 knockdown exacerbates isoflurane-induced postoperative cognitive impairment in mice by inducing activation of STAT3 and NF-κB signaling pathways. Transl Neurosci 2023; 14:20220282. [PMID: 37069964 PMCID: PMC10105556 DOI: 10.1515/tnsci-2022-0282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 03/07/2023] [Accepted: 03/13/2023] [Indexed: 04/19/2023] Open
Abstract
Objective Anesthetic exposure causes learning and memory impairment, the mechanisms of which remain unknown. It has been reported that tumor necrosis factor-α-inducer protein 8-like 2 (TIPE2) is a newly discovered immune negative regulator that is essential for maintaining immune homeostasis. This study aimed to examine the role of TIPE2 in isoflurane-induced postoperative cognitive decline (POCD). Methods An AAV empty vector and AAV shTIPE2 vector for the knockdown of TIPE2 were injected into the dorsal hippocampus of mice. Mice were continuously exposed to 1.5% isoflurane followed by abdominal exploration. Behavioral tests including the open field test and fear conditioning test were performed on the third and fourth day post-operation. Apoptosis was detected by terminal deoxynucleotidyl-transferase-mediated dUTP nick end labeling staining. The kits were used to detect the activity of antioxidant enzymes. Inflammatory cytokine levels were detected by enzyme-linked immunosorbent assay. Signal transducer and activator of transcription 3 (STAT3) and nuclear factor-κB (NF-κB) signaling pathway activities were detected by western blotting. Results TIPE2 expression increased after isoflurane anesthesia and surgery. TIPE2 deficiency aggravated cognitive impairment in mice and further caused apoptosis and oxidative stress in hippocampal neurons. TIPE2 deficiency induced microglial activation and increased secretion of proinflammatory cytokines. In addition, TIPE2 deficiency promoted STAT3 and NF-κB signaling activation induced by isoflurane anesthesia and after surgery. Conclusion TIPE2 may play a neuroprotective role in POCD by regulating STAT3 and NF-κB pathways.
Collapse
Affiliation(s)
- Rui Jian
- Department of Rehabilitation Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xin He
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, No. 87, Xiangya Road, Kaifu District, Changsha, Hunan, 410008, China
| |
Collapse
|
28
|
Liu L, Yuan Y, Zhou Y, Yao L, Li J, Chen F. The transcription factor KLF14 attenuates LPS-induced acute lung injury by ameliorating apoptosis of alveolar epithelial cells in mice. Mol Immunol 2022; 152:67-77. [DOI: 10.1016/j.molimm.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 09/25/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
|
29
|
Liu X, Guan PY, Yu CT, Yang H, Shan AS, Feng XJ. Curcumin alleviated lipopolysaccharide-induced lung injury via regulating the Nrf2-ARE and NF-κB signaling pathways in ducks. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:6603-6611. [PMID: 35596659 DOI: 10.1002/jsfa.12027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/17/2022] [Accepted: 05/21/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Under the intensive modern poultry farming system, the lung of duck is one of the main target organs for various bacterial and viral infections. Curcumin is a kind of natural polyphenol compound for which various beneficial biological functions exist, including being an anti-inflammatory, antioxidant, and antiviral. The aim of this work was to investigate the mechanism of curcumin-alleviated lipopolysaccharide (LPS)-induced lung damage by the nuclear erythroid 2-related factor 2 (Nrf2)-antioxidant reaction element (ARE) and nuclear factor kappa B (NF-κB) signaling pathway in ducks. RESULTS In total, 450 one-day-old male specific pathogen-free ducks were randomly assigned into three dietary treatments: CON, basal diet; LPS, basal diet + LPS treatment; LPS + CUR, basal diet + LPS + 500 mg kg-1 of curcumin. At the end of the experiment (21 days), ducks in LPS treatment were challenged with 5 mg LPS per kilogram of body weight and the other two treatments were injected with the same dose of phosphate-buffered saline solution. The results showed that LPS caused acute inflammation, oxidation stress, and lung injury. Dietary addition of curcumin significantly relieved the oxidation stress and inflammation parameters. Moreover, the results showed that remission may be through the signaling pathways of both Nrf2-ARE and NF-κB. CONCLUSION In conclusion, dietary supplementation of 500 mg kg-1 of curcumin exhibited a lung-protective effect in ducks. This experiment broadens the mode of metabolism actions of curcumin in the target organs and provides an insight for the application of curcumin in waterfowl feed. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xiao Liu
- Institute of Animal Nutrition, College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Pei Yue Guan
- Institute of Animal Nutrition, College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Chun Ting Yu
- Institute of Animal Nutrition, College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Hao Yang
- Institute of Animal Nutrition, College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - An Shan Shan
- Institute of Animal Nutrition, College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Xing Jun Feng
- Institute of Animal Nutrition, College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| |
Collapse
|
30
|
Tan Y, Zou YF, Zhang HB, Liu X, Qian CY, Liu MW. The protective mechanism of salidroside modulating miR-199a-5p/TNFAIP8L2 on lipopolysaccharide-induced MLE-12 cells. Int J Immunopathol Pharmacol 2022; 36:3946320221132712. [PMID: 36214213 PMCID: PMC9551330 DOI: 10.1177/03946320221132712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVES Salidroside is used for treating inflammation-based diseases; however, its molecular mechanism is unclear. In this study, we determined the protective role of salidroside on the endotoxin-induced damage caused to the mouse alveolar epithelial type II (MLE-12) cells and its underlying mechanism. METHODS An in vitro model for acute lung injury was constructed by inducing the MLE-12 cells using lipopolysaccharide (lipopolysaccharides, 1 mg/L). Then, The MTT assay was conducted to assess the survival rate of the MLE-12 cells in the different groups. After the treatment, apoptosis of MLE-12 cells was determined, and the mRNA and protein expression of miR-199a-5p, HMGB1, NF-kB65, TNFAIP8L2, p-IkB-α, and TLR4 was estimated by Western Blotting and RT-PCR. ELISA was also used to measure the concentration of inflammatory cytokine molecules IL-1β, IL-6, TNF-α, and IL-18 in the cell-free supernatant. Lastly, cell morphology was examined using the AO/EB technique. RESULTS We showed that salidroside reduced the protein and gene expression of HMGB1, NF-kB65, miR-199a-5p, p-IkB-α, and TLR4, whereas it increased the gene and protein expression of TNFAIP8L2. Furthermore, it decreased the concentrations of cytokine molecules like IL-1β, IL-6, TNF-α, and IL-18 in the cell-free supernatant. MLE-12 also showed a lower apoptosis rate, higher survival rate, and better cell morphology. CONCLUSION Salidroside significantly inhibited the LPS-induced MLE-12 cell damage. Our results suggest that this could be by reducing miR-199a-5p and enhancing TNFAIP8L2 expression.
Collapse
Affiliation(s)
- Yang Tan
- Department of Emergency Medicine,
The First
Affiliated Hospital of Kunming Medical
University, Kunming, China
| | - Yong-fan Zou
- Department of Emergency Medicine,
The First
Affiliated Hospital of Kunming Medical
University, Kunming, China
| | - Huang-bo Zhang
- Trauma Center,
The First
Affiliated Hospital of Kunming Medical
University, Kunming, China
| | - Xu Liu
- Department of Infectious Diseases,
Yan-an Hospital
of Kunming City, Kunming, China
| | - Chuan-yun Qian
- Department of Emergency Medicine,
The First
Affiliated Hospital of Kunming Medical
University, Kunming, China
| | - Ming-Wei Liu
- Department of Emergency Medicine,
The First
Affiliated Hospital of Kunming Medical
University, Kunming, China
- Ming-Wei Liu, Department of Emergency
Medicine, The First Affiliated Hospital of Kunming Medical University, 295
Xichang Road, Wuhua District, Kunming 650032, China.
| |
Collapse
|
31
|
Zhang Q, Zeng M, Zhang B, Ren Y, Li S, Wang R, Hu Y, Fan R, Wang M, Yu X, Wu Z, Zheng X, Feng W. Salvianolactone acid A isolated from Salvia miltiorrhiza ameliorates lipopolysaccharide-induced acute lung injury in mice by regulating PPAR-γ. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 105:154386. [PMID: 35985183 DOI: 10.1016/j.phymed.2022.154386] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/28/2022] [Accepted: 08/08/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Severe inflammation of the lungs results from acute lung injury (ALI), a common life-threatening lung disease with a high mortality rate. The ligand-activated transcription factor peroxisome proliferator-activated receptor (PPAR) γ plays essential roles in diverse biological processes including inflammation, metabolism, development, and immune response. Salvianolactone acid A (SA) is a terpenoid derived from the herb Salvia miltiorrhiza. However, there is a scarcity of experimental evidence indicating whether the effect of SA on ALI occurs via PPAR-γ. METHODS SA (20 or 40 mg/kg, i.g., 1 time/day) was administered to mice for 3 d, followed by the induction of ALI by intranasal lipopolysaccharide (LPS, 10 mg/kg). The lung function and levels of inflammation, reactive oxygen species (ROS), immune cells, apoptosis, and PPAR-γ were examined. The antagonistic activity of GW9662 (GW, 1 µM, specific PPAR-γ blocker) and PPAR-γ transfection silencing against SA (10 μM) in BEAS-2B cells induced by LPS (10 μg/ml, 24 h) was also investigated to assess whether the observed effects caused by SA were mediated by PPAR-γ. RESULTS The results showed that lung histopathological injury, the B-line, the fluorescence intensity of live small animal, and the biomarkers in BALF or lung in the treatment of SA could regulate significantly. In addition, SA obviously decreased the levels of ROS and apoptosis in the primary lung cells, and MDA, increased the levels of GSH-Px and SOD. SA reduced levels of macrophages and neutrophils. Furthermore, SA reduced the protein levels of Keap-1, Cleaved-caspase-3, Cleaved-caspase-9, p-p65/p65, NLRP3, IL-1β, and upregulated the levels of p-Nrf2/Nrf2, HO-1, Bcl-2/Bax, PPAR-γ, p-AMPK/AMPK in lung tissue. In addition, silencing and inhibition of PPAR-γ effectively decreased the protective effects of SA in BEAS-2B cells induced by LPS, which might indicate that the active molecules of SA regulate ALI via mediation by PPAR-γ, which exhibited that the effect of SA related to PPAR-γ. CONCLUSIONS The anti-ALI effects of SA were partially mediated through PPAR-γ signaling. These data provide the molecular justification for the usage of SA in treating ALI and can assist in increasing the comprehensive utilization rate of Salvia miltiorrhiza.
Collapse
Affiliation(s)
- Qinqin Zhang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Mengnan Zeng
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan and Education Ministry of P.R., Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Beibei Zhang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Yingjie Ren
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Shujing Li
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Ru Wang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Yingbo Hu
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Ruyi Fan
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Mengya Wang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Xiao Yu
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Zhe Wu
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Xiaoke Zheng
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan and Education Ministry of P.R., Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Weisheng Feng
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan and Education Ministry of P.R., Henan University of Chinese Medicine, Zhengzhou 450046, China.
| |
Collapse
|
32
|
Zeng M, Zhang B, Ren Y, Wang S, Guo P, Liu M, Zhang Q, Jia J, Li J, Zheng X, Feng W. A sesquiterpene isolated from the stems and leaves of Dioscorea opposita thunb. Transforms the composition of immune cells through ERβ in a mouse model of LPS-induced lung injury. Heliyon 2022; 8:e10500. [PMID: 36105471 PMCID: PMC9465438 DOI: 10.1016/j.heliyon.2022.e10500] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/22/2022] [Accepted: 08/25/2022] [Indexed: 11/29/2022] Open
Abstract
Acute lung injury (ALI) is a common critical disease with a high mortality rate. Natural products have marked efficacy in the prevention and treatment of ALI, in addition, estrogen and its receptors are involved in the pathogenesis and development of lung injury. Our previous research shows that sesquiterpenes isolated from the stems and leaves of Dioscorea opposita Thunb. have anti-inflammatory and estrogenic-like activity. In the present study, sesquiterpene (A1) is a natural extract from the stems and leaves of Dioscorea opposita Thunb. with a view to determining whether A1 can improve lung function in a mouse model of LPS-induced ALI and exploring the involvement of the estrogen receptor β (ERβ) pathway. A1 (20 or 40 mg/kg, i. g., 2 times/day) was administered for 3 d, followed by the induction of ALI via an intratracheal LPS drip (5 mg/kg/2 h). The lung function and levels of inflammation, immune cells, apoptosis, and ERβ expression were examined. The antagonistic activity of specific ERβ blocker (THC, 1 μM) against A1 (20 μM) in co-cultured BEAS-2B cells and splenic lymphocytes induced with LPS (1 μg/mL, 24 h) was also investigated to assess whether the observed effects of A1 were mediated by ERβ. A1 improved lung function, regulated the immune system, and decreased inflammation and apoptosis. Moreover, A1 increased the expression of ERβ in LPS-induced mice, and antagonism of ERβ decreased the protective effects of A1 in a co-culture system. A1 had anti-ALI effects that might partially mediated through ERβ signaling. Our data provide molecular justification for the use of A1 in the treatment of ALI.
Collapse
Affiliation(s)
- Mengnan Zeng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Beibei Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Yingjie Ren
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Shengchao Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Pengli Guo
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Meng Liu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Qinqin Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Jufang Jia
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Jinyue Li
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, China
- Corresponding author.
| | - Weisheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, China
- Corresponding author.
| |
Collapse
|
33
|
Gu Z, Xin L, Yu H, Lu S, Wu J, Wang H, Huang D, Hu C. Cervical sympathetic trunk transection alleviates acute lung injury caused by intestinal obstruction via inhibition of phospholipase A 2 in rats. BMC Anesthesiol 2022; 22:270. [PMID: 35999492 PMCID: PMC9400334 DOI: 10.1186/s12871-022-01814-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Background Intestinal obstruction can result in inflammatory injury to distant organs, especially the lungs. Stellate ganglion block (SGB) provides sympathetic nervous homeostasis and inhibits the systemic inflammatory response. This study aimed to investigate whether SGB can alleviate acute lung injury by inhibiting phospholipase A2 expression in rats. Methods Thirty healthy male Sprague–Dawley rats were divided into three groups: C group (sham-operated); CLP group (cecal ligation and puncture with intestinal obstruction), and cervical sympathetic trunk transection (CSTT) group (transection of the cervical sympathetic trunk following CLP).Arterial blood samples were obtained to determine the ratio of partial arterial pressure of oxygen (PaO2) to fraction of oxygen in inspired air (FiO2). Venous blood samples were used to evaluate the serum concentrations of chemokines, tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-10 using enzyme-linked immunosorbent assays. Following euthanasia, the lungs were isolated to estimate the wet/dry lung weight (W/D) ratio, evaluate the pathological damage to lung tissues on microscopy, and determine secretory-type phospholipase A2 (sPLA2) expression using western blotting. Results Rats in the CLP group showed increased fatigue, decreased activity levels, and coarse, gray hair. The levels of chemokines, TNF-α, and IL-6 in the CLP and CSTT groups were higher than those in the C group. However, the levels were lower in the CSTT group than those in the CLP group. IL-10 levels in the CLP group were higher and lower than those in the C and CSTT groups, respectively. W/D ratios and PaO2/FiO2 in the CLP and CSTT groups were higher than those in the C group, whereas these ratios in the CSTT group were lower than those in the CLP group. No lung injury was noted in group C, and the lung injury scores were lower in the CSTT group than those in the CLP group. sPLA2 expression levels in the CLP group were higher than those in the C group, whereas these levels in the CSTT group were lower than those in the CLP group. Conclusions sPLA2 overexpression in the lungs may be a pathogenic factor in acute lung injury. CSTT alleviated acute lung injury by inhibiting sPLA2 expression. Supplementary Information The online version contains supplementary material available at 10.1186/s12871-022-01814-2.
Collapse
Affiliation(s)
- Zhengfeng Gu
- Department of Analgesia and Anesthesiology, Nanjing Medical University Affiliated Wuxi People's Hospital, Wuxi, China.
| | - Lian Xin
- Department of Analgesia and Anesthesiology, Nanjing Medical University Affiliated Wuxi People's Hospital, Wuxi, China
| | - Huizhi Yu
- Department of Analgesia and Anesthesiology, Nanjing Medical University Affiliated Wuxi People's Hospital, Wuxi, China
| | - Shunmei Lu
- Department of Analgesia and Anesthesiology, Nanjing Medical University Affiliated Wuxi People's Hospital, Wuxi, China
| | - Jinbo Wu
- Department of Analgesia and Anesthesiology, Nanjing Medical University Affiliated Wuxi People's Hospital, Wuxi, China
| | - Hui Wang
- Department of Analgesia and Anesthesiology, Nanjing Medical University Affiliated Wuxi People's Hospital, Wuxi, China
| | - Dongxiao Huang
- Department of Analgesia and Anesthesiology, Nanjing Medical University Affiliated Wuxi People's Hospital, Wuxi, China
| | - Chunxiao Hu
- Department of Analgesia and Anesthesiology, Nanjing Medical University Affiliated Wuxi People's Hospital, Wuxi, China
| |
Collapse
|
34
|
Jiang H, He J, Wang H, Zheng L, Wang X, Zhang H, Wu H, Shu Y. Gill Junction Injury and Microbial Disorders Induced by Microcystin-Leucine Arginine in Lithobates catesbeianus Tadpoles. Toxins (Basel) 2022; 14:toxins14070479. [PMID: 35878217 PMCID: PMC9322459 DOI: 10.3390/toxins14070479] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 02/01/2023] Open
Abstract
Microcystin-LR (MC-LR) is widely present in waters around the world, but its potential toxic effects and mechanisms on amphibian gills remain unknown. In the present study, tadpoles (Lithobates catesbeianus) were exposed to environmentally realistic concentrations of 0.5, 2 μg/L MC-LR, and 0 μg/L MC-LR (Control) for 30 days with the objective to unveil the impairment of gill health. The lysozyme was downregulated, while pattern recognition receptors and complement and adaptive immune processes were upregulated and the ability of gill supernatant to inhibit pathogenic bacteria decreased in the 0.5 and 2 μg/L MC-LR groups. The transcriptions of epithelial barrier components (e.g., CLDN1) were significantly decreased in MC-LR-exposed gills, while the gill content of lipopolysaccharide (LPS) endotoxins and the transcriptions of downstream responsive genes (e.g., TLR4 and NF-κB) were concurrently increased. In addition, the number of eosinophils and the expression of pro-inflammatory cytokines (e.g., IL-1β and TNF-α) were increased. These results imply that exposure of tadpoles to low environmentally concentrations of MC-LR leads to inflammation, increased permeability, and a reduced ability to inhibit pathogenic bacteria. The epithelial cells of inner gill filaments increased and transcriptions of hypoxic stress genes (e.g., HIF-1α, FLT1, and SERPINE1) were upregulated within the exposed group. As a consequence, exposure to MC-LR may lead to hypoxic stress. MC-LR exposure also drove gill microbiota to a dysbiosis. The relative abundance of Elizabethkingia was positively correlated with content of LPS and transcriptions of NF-κB and TNF-α. Overall, this study presents the first evidence about the pronounced impacts of MC-LR exposure on gills of amphibians, highlighting the susceptibility of early developing tadpoles to the environmental risks of MC-LR.
Collapse
Affiliation(s)
- Huiling Jiang
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-Founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China; (H.J.); (J.H.); (H.W.); (L.Z.); (X.W.); (H.Z.)
| | - Jun He
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-Founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China; (H.J.); (J.H.); (H.W.); (L.Z.); (X.W.); (H.Z.)
| | - Hui Wang
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-Founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China; (H.J.); (J.H.); (H.W.); (L.Z.); (X.W.); (H.Z.)
| | - Lingling Zheng
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-Founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China; (H.J.); (J.H.); (H.W.); (L.Z.); (X.W.); (H.Z.)
| | - Xiaoran Wang
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-Founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China; (H.J.); (J.H.); (H.W.); (L.Z.); (X.W.); (H.Z.)
| | - Huijuan Zhang
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-Founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China; (H.J.); (J.H.); (H.W.); (L.Z.); (X.W.); (H.Z.)
| | - Hailong Wu
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-Founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China; (H.J.); (J.H.); (H.W.); (L.Z.); (X.W.); (H.Z.)
- Correspondence: (H.W.); (Y.S.)
| | - Yilin Shu
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-Founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China; (H.J.); (J.H.); (H.W.); (L.Z.); (X.W.); (H.Z.)
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- Correspondence: (H.W.); (Y.S.)
| |
Collapse
|
35
|
Shen J, Ma X. miR‑374a‑5p alleviates sepsis‑induced acute lung injury by targeting ZEB1 via the p38 MAPK pathway. Exp Ther Med 2022; 24:564. [PMID: 35978929 PMCID: PMC9366279 DOI: 10.3892/etm.2022.11501] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 04/19/2022] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the effects of microRNA (miR)-374a-5p on sepsis-induced acute lung injury (ALI) and the associated mechanism. Lipopolysaccharide (LPS)-induced human pulmonary microvascular endothelial cells (HPMVECs) were used to construct the cellular model of sepsis. A luciferase reporter assay was performed to confirm the association between miR-374a-5p and zinc finger E-box binding homeobox 1 (ZEB1). Reverse transcription-quantitative polymerase chain reaction and western blot analysis were performed to assess the relative expression of miR-374a-5p, ZEB1 and apoptosis-related proteins. Cell viability and apoptosis were determined by Cell Counting Kit-8 assay and flow cytometry, respectively. Enzyme-linked immunosorbent assays were used to evaluate inflammatory cytokines. The results revealed that miR-374a-5p was downregulated in sepsis patients and LPS-treated HPMVECs. Upregulation of miR-374a-5p alleviated LPS-triggered cell injury in HPMVECs, as evidenced by restoration of cell viability, and inhibition of apoptosis and the production of proinflammatory cytokines. In addition, ZEB1 was revealed to be a downstream target of miR-374a-5p, and overexpression of ZEB1 could reverse the anti-apoptotic and anti-inflammatory effects of miR-374a-5p on an LPS-induced sepsis cell model. Moreover, miR-374a-5p-induced protective effects involved the p38 MAPK signaling pathway. Collectively, miR-374a-5p exerted a protective role in sepsis-induced ALI by regulating the ZEB1-mediated p38 MAPK signaling pathway, providing a potential target for the diagnosis and treatment of sepsis.
Collapse
Affiliation(s)
- Jia Shen
- Department of Intensive Care Unit, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750002, P.R. China
| | - Xiaojun Ma
- Department of Orthopedics, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| |
Collapse
|
36
|
Huang Z, Wang H, Long J, Lu Z, Chun C, Li X. Neutrophil Membrane-Coated Therapeutic Liposomes for Targeted Treatment in Acute Lung Injury. Int J Pharm 2022; 624:121971. [PMID: 35787461 PMCID: PMC9365401 DOI: 10.1016/j.ijpharm.2022.121971] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/22/2022] [Accepted: 06/29/2022] [Indexed: 10/26/2022]
Abstract
Acute lung injury (ALI) is one of the most common comorbidities associated with sepsis and can lead to acute respiratory distress syndrome. Intense inflammatory response due to excessive activation and uncontrolled infiltration of neutrophils are the central processes in the development of sepsis-induced ALI. In this study, a biomimetic nanoplatform that is a neutrophil membrane-coated liposome-loaded acidic fibroblast growth factor (aFGF@NMLs), which can selectively target the inflamed lung and effectively alleviate sepsis-induced ALI via inflammation suppression, was constructed. In vitro findings revealed that aFGF@NMLs has pro-inflammatory cytokine binding capabilities and can promote cellular uptake, substantially attenuate inflammatory responses, and enhance cellular antioxidant capacity. The in vivo results show that aFGF@NMLs can specifically accumulate in injured lungs in ALI mice after intravenous injection, thereby reducing the secretion of pro-inflammatory cytokines, inhibiting pulmonary cell apoptosis, and promoting lung function recovery. In conclusion, aFGF@NMLs demonstrated anti-inflammatory effects, mitigated the progression of ALI, and contributed to the disease prognosis. This research offers an innovative strategy and concept for the clinical treatment of diseases related to pulmonary inflammation.
Collapse
Affiliation(s)
- Zhiwei Huang
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Hengcai Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Juan Long
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of emergency and disaster medicine, Wenzhou 325035, China
| | - Zhongqiu Lu
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of emergency and disaster medicine, Wenzhou 325035, China
| | - Changju Chun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Xinze Li
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of emergency and disaster medicine, Wenzhou 325035, China.
| |
Collapse
|
37
|
Xiao J, Cheng P, Ma P, Wu Y, Feng F, Miao Y, Deng Q. Toxicological effects of traffic-related air pollution on the lungs: Evidence, biomarkers and intervention. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 238:113570. [PMID: 35512471 DOI: 10.1016/j.ecoenv.2022.113570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/13/2022] [Accepted: 04/25/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Numerous epidemiological studies have recently observed that exposure to traffic-related air pollution (TRAP) is associated with increased risk of various respiratory diseases. Major gaps in knowledge remain regarding the toxicological effects. OBJECTIVES We examined the toxicological effects of the gasoline exhaust particles (GEP), a paradigm of TRAP, in rats, with an objective to provide the evidence, obtain the biomarkers, and suggest effective intervention measure. METHODS We measured the airway hyperresponsiveness (AHR), inflammatory cells in the bronchoalveolar lavage (BAL) fluid, histological changes in the lung tissues, and the biomarkers so as to systematically examine the toxicological effects of GEPs at different dose levels (0.5, 2.5, 5 mg/kg BW). The intervention of vitamin E (VE), a natural antioxidant, on the toxicological effects was investigated. RESULTS The lung injury caused by GEP exposure was first indicated by the airway hyperresponsiveness (AHR). Compared with the control group, GEP exposure significantly increased the airway resistances and decreased the lung compliance; the higher the dose of GEP, the more serious the lung injury. Lung injury was also revealed by the increase of inflammatory cells, including the lymphocytes and neutrophils, in the BAL fluid. With the increase of GEP dose, histological changes in the lung tissues were further observed: inflammatory cell infiltration increased and alveolar wall thickened. The toxicology of GEP was demonstrated by the increase of the biomarkers of the oxidative stress, the pro-inflammatory cytokines and the apoptosis cytokine. However, administration of VE was found to be effective in restoring airway injury. CONCLUSION The toxicological effects of traffic-related air pollution (TRAP) on rat lungs are supported by evidence and biomarkers, and vitamin E intervention is feasible.
Collapse
Affiliation(s)
- Jiani Xiao
- XiangYa School of Public Health, Central South University, Changsha 410078, China
| | - Ping Cheng
- XiangYa School of Public Health, Central South University, Changsha 410078, China; Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, China
| | - Ping Ma
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Yang Wu
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Feifei Feng
- School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Yufeng Miao
- School of Energy Science and Engineering, Central South University, Changsha 430081, China.
| | - Qihong Deng
- School of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
38
|
Cui YR, Qu F, Zhong WJ, Yang HH, Zeng J, Huang JH, Liu J, Zhang MY, Zhou Y, Guan CX. Beneficial effects of aloperine on inflammation and oxidative stress by suppressing necroptosis in lipopolysaccharide-induced acute lung injury mouse model. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 100:154074. [PMID: 35397283 DOI: 10.1016/j.phymed.2022.154074] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 02/22/2022] [Accepted: 03/25/2022] [Indexed: 06/14/2023]
Abstract
RATIONALE Alveolar epithelial cell death, inflammation, and oxidative stress are typical features of acute lung injury (ALI). Aloperine (Alo), an alkaloid isolated from Sophora alopecuroides, has been reported to display various biological effects, such as anti-inflammatory, immunoregulatory, and anti-oxidant properties. In this study, we investigated the effects and mechanisms of Alo in treating a lipopolysaccharide (LPS)-induced ALI in a murine model. METHODS The effects of Alo in LPS-induced ALI were investigated in C57BL/6 mice. The RIPK1 inhibitor (Nec-1) and the RIPK3 inhibitor (GSK'872) were used to evaluate the relationship of necroptosis, NF-κB activation, and PDC subunits in LPS-treated mouse alveolar epithelial cells (MLE-12). Then the effects of Alo on necroptosis, inflammation, and oxidative stress of LPS-stimulated MLE-12 cells were evaluated. RESULTS Alo significantly attenuated histopathological lung injuries and reduced lung wet/dry ratio in LPS-induced ALI mice. Alo also remarkedly reduced total protein and neutrophils recruitment in bronchoalveolar lavage fluid of ALI mice. Meanwhile, Alo ameliorated the LPS-induced necroptosis in the lungs of ALI mice. The RIPK3 inhibitor GSK'872, but not the RIPK1 inhibitor Nec-1, reversed LPS-induced p65 phosphorylation and translocation to the nucleus in MLE-12 cells. GSK'872 also reversed the LPS-induced increase in ROS and binding of RIPK3 and PDC subunits in MLE-12 cells. Moreover, Alo down-regulated the levels of p-RIPK1, p-RIPK3, p-MLKL, p-p65, the translocation of p65 to the nucleus, and reduced the expression of IL-6 and IL-8 in LPS-stimulated MLE-12 cells. Alo also inhibited the binding of RIPK3 and PDC-E1α, PDC-E1β, PDC-E2, and PDC-E3 and the ROS production in LPS-treated MLE-12 cells. CONCLUSION The present study validated the beneficial effects of Alo on LPS-induced ALI , suggesting Alo may be a new drug candidate against ALI.
Collapse
Affiliation(s)
- Yan-Ru Cui
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China; Department of Physiology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Fei Qu
- Department of Pharmacology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Wen-Jing Zhong
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China
| | - Jie Zeng
- Department of Physiology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Jun-Hao Huang
- Department of Pharmacology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Jie Liu
- Department of Physiology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Ming-Yue Zhang
- Department of Pharmacology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China.
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|
39
|
Shi B, Hao Y, Li W, Dong H, Xu M, Gao P. TIPE2 May Target the Nrf2/HO-1 Pathway to Inhibit M1 Macrophage-Related Neutrophilic Inflammation in Asthma. Front Immunol 2022; 13:883885. [PMID: 35572500 PMCID: PMC9095941 DOI: 10.3389/fimmu.2022.883885] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/04/2022] [Indexed: 11/28/2022] Open
Abstract
Purpose Although recent studies have highlighted the link of TIPE2 and asthma airway inflammation, its roles and molecular mechanisms in different asthma inflammatory phenotypes remain largely unknown. We evaluated sputum TIPE2 expression level and its correlation with different asthma phenotypes. Additionally, we explored the roles and mechanism of TIPE2 in M1 polarization of macrophages. Methods A total of 102 asthma patients who underwent sputum induction were enrolled to evaluate the expression level of TIPE2 and its association with different asthma phenotypes. To explore the roles and mechanism of TIPE2 in M1 polarization of macrophages, THP-1 monocytes stimulated with phorbol-12-myristate-13-acetate, were used as a model of undifferentiated (M0) macrophages, and M0 macrophages were treated with lipopolysaccharide to induce M1 macrophages. Results The sputum TIPE2 level was significantly lower in patients with neutrophilic asthma (NA) and higher in patients with eosinophilic asthma (EA) compared with patients with paucigranulocytic asthma. The levels of IL-1β, TNF-α and IL-6 were highest in NA compared with other groups. TIPE2 levels in sputum negatively correlated with IL-1β and TNF-α levels but positively correlated with IL-4, IL-5, IL-13, and IL-10 levels (P < 0.05). In vitro, TIPE2 enhanced Nrf2/HO-1 pathway activation in macrophages and inhibited LPS-induced M1 macrophage differentiation and related cytokine release. Further analysis showed that the Nrf2 inhibitor ML385 weakened TIPE2-induced activation of the Nrf2/HO-1 pathway, as well as TIPE2-induced suppression in M1 polarization of macrophage and inflammatory cytokines secretion. Conclusions TIPE2 expression level was highly down-regulated in NA and was negatively correlated with inflammatory factors (IL-1β and TNF-α). Aberrant expression of TIPE2 may target the Nrf2/HO-1 pathway to inhibit M1 macrophage–related neutrophilic inflammation in asthma.
Collapse
Affiliation(s)
- Bingqing Shi
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Yuqiu Hao
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Wei Li
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Hongna Dong
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Mengting Xu
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Peng Gao
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
40
|
Regulation of inflammation and apoptosis by GPR43 via JNK/ELK1 in acute lung injury. Inflamm Res 2022; 71:603-614. [DOI: 10.1007/s00011-022-01556-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/08/2022] [Accepted: 02/18/2022] [Indexed: 11/05/2022] Open
|
41
|
Huang YD, Fang Y, Ma L, Feng PJ, Li WL, Zhou YQ, Qin YH, You ZJ, Dong L. Kindlin-2 Mediates Lipopolysaccharide-Induced Acute Lung Injury Partially via Pyroptosis in Mice. Inflammation 2022; 45:1199-1208. [PMID: 35133562 DOI: 10.1007/s10753-021-01613-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/16/2021] [Accepted: 12/13/2021] [Indexed: 11/26/2022]
Abstract
Acute lung injury (ALI) is characteristic of the wholesale destruction of the lung endothelial barrier, which results in protein-rich lung edema, influx of pro-inflammatory leukocytes, and intractable hypoxemia, contributing to high mortality. Kindlin-2 is involved in the process of tumor- and wound healing-associated inflammation. However, the effects of kindlin-2 on lipopolysaccharide (LPS)-induced ALI and its mechanisms remain unknown. In this study, we found that the concentration of kindlin-2 was elevated in the lungs of ALI mice. The specific deletion of kindlin-2 by kindlin-2 siRNA attenuated the severity of lung injury, which was demonstrated by the reduced number of pro-inflammatory cells in bronchoalveolar lavage fluid and lung wet/dry weight ratio, and ameliorated pathologic changes in the lungs of ALI mice. Furthermore, kindlin-2 siRNA decreased the mRNA levels of pro-inflammatory factors (IL-1β, IL-6, and TNF-α) and the protein levels of pyroptosis-related proteins. In vitro studies confirmed that LPS + ATP promoted the expressions of pro-inflammatory factors and pyroptosis-related proteins, which was prevented by kindlin-2 siRNA pretreatment in endothelial cells (ECs). In conclusion, inhibition of kindlin-2 developes protective effects against LPS-induced ALI and the cytotoxicity of ECs, which may depend on blocking pyroptosis.
Collapse
Affiliation(s)
- Yi-Dan Huang
- Department of Anesthesiology, Liuzhou Municipal People's Hospital, Liuzhou, 545006, Guangxi, China
| | - Yu Fang
- Medical Laboratory and Pathology Center, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410021, Hunan, China
| | - Li Ma
- Department of Anesthesiology, Liuzhou Municipal People's Hospital, Liuzhou, 545006, Guangxi, China
| | - Peng-Jiu Feng
- Department of Anesthesiology, Liuzhou Traditional Chinese Medicine Hospital, Liuzhou, 545001, Guangxi, China
| | - Wen-Long Li
- Department of Anesthesiology, Liuzhou Municipal People's Hospital, Liuzhou, 545006, Guangxi, China
| | - Yi-Qi Zhou
- Department of Anesthesiology, Liuzhou Municipal People's Hospital, Liuzhou, 545006, Guangxi, China
| | - Yuan-Hao Qin
- Department of Anesthesiology, Liuzhou Municipal People's Hospital, Liuzhou, 545006, Guangxi, China
| | - Zhi-Jian You
- Department of Anesthesiology, Liuzhou Municipal People's Hospital, Liuzhou, 545006, Guangxi, China.
| | - Liang Dong
- Department of Anesthesiology, Liuzhou Municipal People's Hospital, Liuzhou, 545006, Guangxi, China.
| |
Collapse
|
42
|
Etwebi Z, Goldsmith JR, Bou-Dargham M, Tian Y, Hood R, Spitofsky N, Li M, Sun H, Lou Y, Liu S, Lengner C, Chen YH. TIPE2 Promotes Tumor Initiation But Inhibits Tumor Progression in Murine Colitis-Associated Colon Cancer. Inflamm Bowel Dis 2021; 28:764-774. [PMID: 34894222 PMCID: PMC9074867 DOI: 10.1093/ibd/izab306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Colorectal cancer (CRC) is the third leading cause of cancer in the United States, and inflammatory bowel disease patients have an increased risk of developing CRC due to chronic intestinal inflammation with it being the cause of death in 10% to 15% of inflammatory bowel disease patients. TIPE2 (TNF-alpha-induced protein 8-like 2) is a phospholipid transporter that is highly expressed in immune cells and is an important regulator of immune cell function. METHODS The azoxymethane/dextran sulfate sodium murine model of colitis-associated colon cancer (CAC) was employed in Tipe2 -/- and wild-type mice, along with colonoid studies, to determine the role of TIPE2 in CAC. RESULTS Early on, loss of TIPE2 led to significantly less numbers of visible tumors, which was in line with its previously described role in myeloid-derived suppressor cells. However, as time went on, loss of TIPE2 promoted tumor progression, with larger tumors appearing in Tipe2 -/- mice. This was associated with increased interleukin-22/STAT3 phosphorylation signaling. Similar effects were also observed in primary colonoid cultures, together demonstrating that TIPE2 also directly regulated colonocytes in addition to immune cells. CONCLUSIONS This work demonstrates that TIPE2 has dual effects in CAC. In the colonocytes, it works as a tumor suppressor. However, in the immune system, TIPE2 may promote tumorigenesis through suppressor cells or inhibit it through IL-22 secretion. Going forward, this work suggests that targeting TIPE2 for CRC therapy requires cell- and pathway-specific approaches and serves as a cautionary tale for immunotherapy approaches in general in terms of colon cancer, as intestinal inflammation can both promote and inhibit cancer.
Collapse
Affiliation(s)
- Zienab Etwebi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason R Goldsmith
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Address correspondence to: Jason Rosenbaum Goldsmith, MD, PhD, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA ()
| | - Mayassa Bou-Dargham
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuhua Tian
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Ryan Hood
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nina Spitofsky
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Honghong Sun
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yunwei Lou
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, China
| | - Suxia Liu
- Institute of Immunology, Shandong University School of Medicine, Jinan, China
| | - Christopher Lengner
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Youhai H Chen
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Faculty of Pharmaceutical Sciences, CAS Shenzhen Institute of Advanced Technology, Shenzhen, China
| |
Collapse
|
43
|
The overexpression of Tipe2 in CRC cells suppresses survival while endogenous Tipe2 accelerates AOM/DSS induced-tumor initiation. Cell Death Dis 2021; 12:1001. [PMID: 34702807 PMCID: PMC8548391 DOI: 10.1038/s41419-021-04289-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 09/29/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022]
Abstract
Aging is a natural and progressive process characterized by an increased frequency of age-related diseases such as cancer. But its mechanism is unclear. TNFAIP8L2 (Tipe2) is an important negative regulator for homeostasis through inhibiting TLR and TCR signaling. Our work reveals that Tipe2 might have dual function by regulating senescence. One side, the overexpression of Tipe2 in CRC cells could induce typical senescent phenotype, especially exposure to oxidative stress. Tipe2 inhibits telomerase activity by regulating c-Myc and c-Est-2 binding to the hTERT promotor. Interestingly, Tipe2 KO mice treated with D-Gal showed a less serious inverse of CD4:CD8 ratio, a lower percentage of Treg compared to WT. Besides, Tipe2 KO mice were more tolerant to the initiation of AOM/DSS-induced CRC, accompanied by a lower level of Treg within IEL. Therefore, specific antibodies against CD25 effectively ameliorate tumorigenesis. These data suggest strongly that the overexpressed Tipe2 suppresses tumor cells proliferation and survival, but endogenous Tipe2 promotes the initiation of tumorigenesis when exposure to dangerous environment such as AOM/DSS-related inflammation.
Collapse
|
44
|
Li S, Lei Y, Lei J, Li H. All‑trans retinoic acid promotes macrophage phagocytosis and decreases inflammation via inhibiting CD14/TLR4 in acute lung injury. Mol Med Rep 2021; 24:868. [PMID: 34676874 PMCID: PMC8554390 DOI: 10.3892/mmr.2021.12508] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 02/18/2021] [Indexed: 12/22/2022] Open
Abstract
Acute lung injury (ALI) is a common clinical emergency and all-trans retinoic acid (ATRA) can alleviate organ injury. Therefore, the present study investigated the role of ATRA in ALI. Lipopolysaccharide (LPS)-induced ALI rats were treated with ATRA and the arterial partial pressure of oxygen (PaO2), lung wet/dry weight (W/D) ratio and protein content in the bronchial alveolar lavage fluid (BALF) were measured to evaluate the effect of ATRA on ALI rats. Alveolar macrophages were isolated from the BALF. The phagocytic function of macrophages was detected using the chicken erythrocyte phagocytosis method and flow cytometry. The viability of macrophages was measured using a Cell Counting Kit-8 assay, and apoptosis was analyzed using a TUNEL assay and flow cytometry. The expression levels of Toll-like receptor 4 (TLR4) and cluster of differentiation (CD)14 on the macrophage membrane were detected by immunofluorescence staining. The protein levels of TLR4, CD14, phosphorylated (p)-65, p65, p-IκBα and IκBα were analyzed using western blotting. The concentrations of IL-6, IL-1β and macrophage inflammatory protein-2 in the plasma of rats were detected by ELISA. Macrophages were treated with IAXO-102 (TLR4 inhibitor) to verify the involvement of CD14/TLR4 in the effect of ATRA on ALI. ATRA provided protection against LPS-induced ALI, as evidenced by the increased PaO2 and reduced lung W/D ratio and protein content in the BALF. ATRA enhanced macrophage phagocytosis and viability and reduced apoptosis and inflammation in ALI rats. Mechanically, ATRA inhibited CD14 and TLR4 expression and NF-κB pathway activation. ATRA enhanced macrophage phagocytosis and reduced inflammation by inhibiting the CD14/TLR4-NF-κB pathway in LPS-induced ALI. In summary, ATRA inactivated the NF-κB pathway by inhibiting the expression of CD14/TLR4 receptor in the alveolar macrophages of rats, thus enhancing the phagocytic function of macrophages in ALI rats, improving the activity of macrophages, inhibiting apoptosis, reducing the levels of inflammatory factors, and consequently playing a protective role in ALI model rats. This study may offer novel insights for the clinical management of ALI.
Collapse
Affiliation(s)
- Shuangxue Li
- Department of Respiratory and Critical Care Medicine, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| | - Yuansheng Lei
- Department of Neurology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jieyun Lei
- Department of Cardiology, Taiyuan Central Hospital, Taiyuan, Shanxi 030009, P.R. China
| | - Hui Li
- Department of Gynecology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| |
Collapse
|
45
|
Jiang W, Wang J, Xue W, Xin J, Shi C, Wen J, Feng X, Huang Y, Hu C. Caveolin-1 attenuates acetaminophen aggravated lipid accumulation in alcoholic fatty liver by activating mitophagy via the Pink-1/Parkin pathway. Eur J Pharmacol 2021; 908:174324. [PMID: 34246650 DOI: 10.1016/j.ejphar.2021.174324] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/28/2021] [Accepted: 07/07/2021] [Indexed: 12/25/2022]
Abstract
Alcoholic fatty liver (AFL) is a disease characterized by the abnormal structure and dysfunction of hepatocytes caused by long-term, excessive drinking. Acetaminophen (APAP) is a commonly used painkiller, but it can aggravate lipid deposition in the liver and cause liver injury when used in fatty liver disease. Here, we investigated the effect of caveolin-1 (CAV-1), an intracellular stent protein, on the pathogenesis of APAP aggravated lipid deposition in AFL mice. This study shows that lipid accumulation was more severe in APAP groups than in alcohol-treated mice. The CAV-1 stent-like domain (CSD, 82-101 amino acids of caveolin-1), used to upregulate CAV-1 expression, could reduce lipid accumulation and activate autophagy in AFL mice treated with APAP. The levels of CAV-1 and autophagy-related proteins (LC3-II/I and Beclin-1) had decreased, whereas SREBP-1c had increased in A/O (alcohol and oleic acid) and APAP-co-treated L02 cells. CAV-1 small interfering RNA and CAV1-overexpressing plasmid were separately transfected into A/O and APAP co-treated L02 cells. When CAV-1 was downregulated, the levels of Pink-1, Parkin, and autophagy-related proteins (LC3-II/I and Beclin-1) were decreased, whereas SREBP-1c was increased. The opposite trend was observed when CAV-1 was overexpressed. The results show that CAV-1 reduced lipid accumulation in L02 cells and activated Pink-1/Parkin-related mitophagy. This study highlights the positive role of CAV-1 in APAP-increased lipid accumulation under the AFL status and provides a new understanding of the function of CAV-1 in the liver through mitophagy associated with the Pink-1/Parkin pathway.
Collapse
Affiliation(s)
- Wei Jiang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Anhui Institute of Innovative Drugs, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Jiarong Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Anhui Institute of Innovative Drugs, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Weiju Xue
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Anhui Institute of Innovative Drugs, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Jiao Xin
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Anhui Institute of Innovative Drugs, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Congjian Shi
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Anhui Institute of Innovative Drugs, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Jiagen Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Anhui Institute of Innovative Drugs, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Xiaowen Feng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Anhui Institute of Innovative Drugs, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Yan Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Anhui Institute of Innovative Drugs, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Chengmu Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Anhui Institute of Innovative Drugs, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China.
| |
Collapse
|
46
|
A novel miRNA-762/NFIX pathway modulates LPS-induced acute lung injury. Int Immunopharmacol 2021; 100:108066. [PMID: 34492536 DOI: 10.1016/j.intimp.2021.108066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/03/2021] [Accepted: 08/09/2021] [Indexed: 11/23/2022]
Abstract
Severe acute lung injury (ALI) cause significant morbidity and mortality worldwide. MicroRNAs (miRNAs) are possible biomarkers and therapeutic targets for ALI. We aimed to explore the role of miR-762, a known oncogenic factor, in the pathogenesis of ALI. Levels of miR-762 in lung tissues of LPS-treated ALI mice and blood cells of patients with lung injury were measured. Injury of human lung epithelial cell line A549 was induced by LPS stimulation. A downstream target of miR-762, NFIX, was predicted using online tools. Their interactions were validated by luciferase reporter assay. Effects of targeted regulation of the miR-762/NFIX axis on cell proliferation, apoptosis, and inflammatory responses were tested in vitro in A549 cells in vivo with an ALI mouse model. We found that upregulation of miR-762 expression and downregulation of NFIX expression were associated with lung injury. Either miR-762 inhibition or NFIX overexpression in A549 lung cells significantly attenuated LPS-mediated impairment of cell proliferation and viability. Notably, increasing expressions of miR-762 inhibitor or NFIX in vivo via airway lentivirus infection alleviated the LPS-induced ALI in mice. Further, targeted downregulation of miR-762 expression or upregulation of NFIX expression in A549 cells markedly down-regulates NF-κB/IRF3 activation, and substantially reduces the production of inflammatory factors, including TNF-α, IL-6, and IL-8. This study reveals a novel role for the miR-762/NFIX pathway in ALI pathogenesis and sheds new light on targeting this pathway for diagnosis, prevention, and therapy.
Collapse
|
47
|
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
|
48
|
Yin J, Han B, Shen Y. RETRACTED: LncRNA NEAT1 inhibition upregulates miR-16-5p to restrain the progression of sepsis-induced lung injury via suppressing BRD4 in a mouse model. Int Immunopharmacol 2021; 97:107691. [PMID: 33962228 DOI: 10.1016/j.intimp.2021.107691] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/17/2021] [Accepted: 04/18/2021] [Indexed: 12/20/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern was raised about the reliability of the Western blot results in Figs. 5B and 6B, which appear to have the same eyebrow shaped phenotype as many other publications tabulated here (https://docs.google.com/spreadsheets/d/149EjFXVxpwkBXYJOnOHb6RhAqT4a2llhj9LM60MBffM/edit#gid=0 [docs.google.com]). The journal requested the corresponding author comment on these concerns and provide the raw data. However, the authors were not responsive to the request for comment. Since original data could not be provided, the overall validity of the results could not be confirmed. Therefore, the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Jianhong Yin
- Department of Emergency, the First People's Hospital of Yunnan Province, Kunming, 650032 Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032 Yunnan, China
| | - Bin Han
- Department of Emergency, the First People's Hospital of Yunnan Province, Kunming, 650032 Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032 Yunnan, China
| | - Yuan Shen
- Department of Emergency, the First People's Hospital of Yunnan Province, Kunming, 650032 Yunnan, China; The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032 Yunnan, China.
| |
Collapse
|
49
|
He X, Wu J, Tan T, Guo W, Xiong Z, Yang S, Feng Y, Wen Q. Quassinoids from Brucea javanica and attenuates lipopolysaccharide-induced acute lung injury by inhibiting PI3K/Akt/NF-κB pathways. Fitoterapia 2021; 153:104980. [PMID: 34186115 DOI: 10.1016/j.fitote.2021.104980] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 10/21/2022]
Abstract
Four new quassinoids (1-4) and twenty known analogues (5-24) were isolated from the seeds of Brucea javanica. All the compounds belong to tetracyclic quassinoids. The structures of the new compounds were elucidated by comprehensive spectroscopic analysis, including HRESIMS and 1D, 2D NMR. In in vitro bioassays, (5-9, 17-19 and 23) showed inhibitory activities for nitric oxide (NO) release in LPS-activated MH-S macrophages and IC50 values of 0.11-45.56 μM. Among them, bruceoside B significantly decreased LPS-induced NO, secretion of inflammatory factor cytokines tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and interleukin-1β (IL-1β). Western Blot was used to verify the expression of p-IκB-α, IκB-α, p-NF-κB, NF-κB, Bax, Bcl-2, Caspase-3, p-PI3K, PI3K, p-Akt, and Akt proteins in PI3K/Akt/NF-κB signal pathway. Bruceoside B inhibited the activity of Akt and its downstream pathways and reduced the activation of apoptotic. In vivo, it was found that bruceoside B had obvious therapeutic effect on LPS-induced acute lung injury (ALI) in mice, and the effect of tissue section was obvious. The regulatory signal pathway of bruceoside B on inflammation was consistent with the anti-inflammatory pathway in vitro. Therefore, the results implied that bruceoside B has a certain therapeutic effect on inflammation and has a certainly effect on acute lung injury.
Collapse
Affiliation(s)
- Xiao He
- Jiangxi University of Chinese Medicine, Nanchang 330006, Jiangxi, PR China; State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, 56 Yangming Road, Nanchang 330006, Jiangxi, China
| | - Jiahui Wu
- Jiangxi University of Chinese Medicine, Nanchang 330006, Jiangxi, PR China
| | - Ting Tan
- Jiangxi University of Chinese Medicine, Nanchang 330006, Jiangxi, PR China
| | - Wenjing Guo
- Jiangxi University of Chinese Medicine, Nanchang 330006, Jiangxi, PR China
| | - Ziwei Xiong
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, 56 Yangming Road, Nanchang 330006, Jiangxi, China
| | - Shilin Yang
- Jiangxi University of Chinese Medicine, Nanchang 330006, Jiangxi, PR China; State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, 56 Yangming Road, Nanchang 330006, Jiangxi, China
| | - Yulin Feng
- Jiangxi University of Chinese Medicine, Nanchang 330006, Jiangxi, PR China; State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, 56 Yangming Road, Nanchang 330006, Jiangxi, China.
| | - Quan Wen
- Jiangxi University of Chinese Medicine, Nanchang 330006, Jiangxi, PR China.
| |
Collapse
|
50
|
Up-regulating TIPE2 alleviates inflammatory pain by suppressing microglial activation-mediated inflammatory response via inhibiting Rac1/NF-κB pathway. Exp Cell Res 2021; 404:112631. [PMID: 33933441 DOI: 10.1016/j.yexcr.2021.112631] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 01/04/2023]
Abstract
TNF-α-inducible protein 8-like 2 (TIPE2) is a recently discovered regulator of inflammation that can maintain immune homeostasis, exerting a significant role in the development of inflammation-related diseases. Here, we aimed to explore the role and potential regulatory mechanism of TIPE2 in the progression of inflammatory pain. In the present study, a mouse BV2 microglia cell activation-mediated inflammatory model was developed with LPS induction, and a mouse inflammatory pain model was established with complete Freund's adjuvant (CFA) injection. In vitro, the TIPE2 expression was decreased in LPS-induced BV2 cells. Overexpression of TIPE2 mitigated LPS-medicated microglial activation via decreasing nitric oxide (NO) generation and the expression of microglia marker IBA-1. Notably, increasing TIPE2 expression alleviated microglial activation-triggered expression levels and releases of proinflammatory factors such as TNF-α, IL-1β, and IL-6. Mechanism analysis verified that overexpression of TIPE2 blunted Rac1-mediated activation of NF-κB pathway following LPS stimulation. More importantly, CFA injection reduced the expression of TIPE2 in a mouse inflammatory pain model and overexpression of TIPE2 alleviated CFA-mediated pain hypersensitivity and inflammatory response, and inactivated microglia cell in vivo. Furthermore, overexpression of TIPE2 decreased Rac1 expression and suppressed the activation of NF-κB pathway in spinal cord after CFA injection. In summary, the present study revealed that overexpression of TIPE2 mitigated inflammatory pain through suppressing microglial activation-induced inflammation by inactivating Rac1/NF-κB pathway. The study provides a novel theoretical foundation for the therapy of inflammatory pain.
Collapse
|