1
|
Li Y, Li G, Feng J, Li S, Liu N. Advances in Research on Marine Natural Products for Modulating the Inflammatory Microenvironment. Phytother Res 2025; 39:1238-1258. [PMID: 39844461 DOI: 10.1002/ptr.8418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 11/14/2024] [Accepted: 12/10/2024] [Indexed: 01/24/2025]
Abstract
In recent years, marine natural products (MNPs) have emerged as crucial sources of lead compounds for the advancement of anti-inflammatory drugs due to their abundant diversity, complexity, and distinctiveness. Inflammatory microenvironments (IMEs) are pervasive pathological features in the etiology of various chronic diseases, referring to the localized milieu or ecosystem where inflammatory responses occur, and they play a pivotal role in the onset and progression of inflammatory diseases. Uncontrolled IMEs can lead to dysregulation of inflammatory mediators within signaling pathways, thereby exerting detrimental effects on human health and even contributing to the development of inflammatory diseases such as cancer. Currently, inflammation treatment predominantly relies on chemical drugs. Nevertheless, these existing therapies are constrained by their numerous side effects and slow remission of symptoms. Consequently, there is an urgent need for the discovery and development of new drugs that exhibit minimal side effects while exerting potent anti-inflammatory effects. This article extensively explored the activities and mechanisms of MNPs (covering studies from 2010 to 2024) regulating key signaling pathways and inflammatory mediators in the IME, which establishes a theoretical basis for the further development of anti-inflammatory drugs.
Collapse
Affiliation(s)
- Yuru Li
- International Research Centre for Food and Health, College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Guangjie Li
- International Research Centre for Food and Health, College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Jingwen Feng
- International Research Centre for Food and Health, College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Songlin Li
- Research Centre of the Ministry of Agriculture and Rural Affairs on Environmental Ecology and Fish Nutrition, Shanghai Ocean University, Shanghai, China
- Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, Shanghai, China
| | - Ning Liu
- International Research Centre for Food and Health, College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
- Research Centre of the Ministry of Agriculture and Rural Affairs on Environmental Ecology and Fish Nutrition, Shanghai Ocean University, Shanghai, China
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, China
| |
Collapse
|
2
|
Holloman BL, Wilson K, Cannon A, Nagarkatti M, Nagarkatti PS. Indole-3-carbinol attenuates lipopolysaccharide-induced acute respiratory distress syndrome through activation of AhR: role of CCR2+ monocyte activation and recruitment in the regulation of CXCR2+ neutrophils in the lungs. Front Immunol 2024; 15:1330373. [PMID: 38596679 PMCID: PMC11002125 DOI: 10.3389/fimmu.2024.1330373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/27/2024] [Indexed: 04/11/2024] Open
Abstract
Introduction Indole-3-carbinol (I3C) is found in cruciferous vegetables and used as a dietary supplement. It is known to act as a ligand for aryl hydrocarbon receptor (AhR). In the current study, we investigated the role of AhR and the ability of I3C to attenuate LPS-induced Acute Respiratory Distress Syndrome (ARDS). Methods To that end, we induced ARDS in wild-type C57BL/6 mice, Ccr2gfp/gfp KI/KO mice (mice deficient in the CCR2 receptor), and LyZcreAhRfl/fl mice (mice deficient in the AhR on myeloid linage cells). Additionally, mice were treated with I3C (65 mg/kg) or vehicle to investigate its efficacy to treat ARDS. Results I3C decreased the neutrophils expressing CXCR2, a receptor associated with neutrophil recruitment in the lungs. In addition, LPS-exposed mice treated with I3C revealed downregulation of CCR2+ monocytes in the lungs and lowered CCL2 (MCP-1) protein levels in serum and bronchoalveolar lavage fluid. Loss of CCR2 on monocytes blocked the recruitment of CXCR2+ neutrophils and decreased the total number of immune cells in the lungs during ARDS. In addition, loss of the AhR on myeloid linage cells ablated I3C-mediated attenuation of CXCR2+ neutrophils and CCR2+ monocytes in the lungs from ARDS animals. Interestingly, scRNASeq showed that in macrophage/monocyte cell clusters of LPS-exposed mice, I3C reduced the expression of CXCL2 and CXCL3, which bind to CXCR2 and are involved in neutrophil recruitment to the disease site. Discussion These findings suggest that CCR2+ monocytes are involved in the migration and recruitment of CXCR2+ neutrophils during ARDS, and the AhR ligand, I3C, can suppress ARDS through the regulation of immune cell trafficking.
Collapse
Affiliation(s)
| | | | | | | | - Prakash S. Nagarkatti
- Nagarkatti Laboratory, University of South Carolina School of Medicine, Department of Pathology, Microbiology, and Immunology, Columbia, SC, United States
| |
Collapse
|
3
|
Yan J, Zhu J, Li X, Yang R, Xiao W, Huang C, Zheng C. Blocking LTB 4 signaling-mediated TAMs recruitment by Rhizoma Coptidis sensitizes lung cancer to immunotherapy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:154968. [PMID: 37531900 DOI: 10.1016/j.phymed.2023.154968] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 05/31/2023] [Accepted: 07/15/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND Immune checkpoint blockade (ICB) induces durable immune responses across a spectrum of advanced cancers and revolutionizes the oncology field. However, only a subset of patients achieves long-lasting clinical benefits. Tumor-associated macrophages (TAMs) usually secrete immunosuppressive cytokines and contribute to the failure of ICB therapy. Therefore, it is crucial to mechanically manipulate the abundance and function of TAMs in the tumor microenvironment (TME), which can offer a promising molecular basis to improve the clinical response efficacy of ICB in cancer patients. PURPOSE This study aims to investigate TAMs in the immunosuppressive microenvironment to identify new therapeutic targets, improve the ability to predict and guide responses to clinical immunotherapy, and develop new strategies for immunotherapy of lung tumors. METHODS Lewis lung carcinoma (LLC) xenograft-bearing mouse models were established to analyze the antitumor activity of Rhizoma Coptidis (RC) in vivo. A systems pharmacology strategy was used to predict the correlation between RC and M2 macrophages. The effect of RC on the abundance of M2 macrophages was analyzed by flow cytometry of murine samples. Western blot was performed to analyze the expression of Leukotriene A4 hydrolase (LTA4H) and LTB4 receptor 1 (BLT1) in harvested lung cancer tissues. The impact of blocking leukotriene B4 (LTB4) signaling by RC on the recruitment of M2 macrophages was assessed in vitro and in vivo. Transwell migration assays were conducted to clarify the inhibition of macrophage migration by blocking LTB4. Lta4h-/- mice were used to investigate the sensitivity of immunotherapy to lung cancer by blocking the LTB4 signaling. RESULTS Here, we report that RC, an herbal medicine from the family Ranunculaceae, suppresses the recruitment and immunosuppressive function of TAMs, which in turn sensitizes lung cancer to ICB therapy. Firstly, a systems pharmacology strategy was proposed to identify combinatorial drugs for ICB therapy with a systems biology perspective of drug-target-pathway-TME phenotype. We predicted and verified that RC significantly inhibits tumor growth and the infiltration of M2-TAMs into TME of LLC tumor-bearing mice. Then, RC inhibits the recruitment of macrophages to the tumor TME via blocking LTB4 signaling, and suppresses the expression of immunosuppressive factors (IL-10, TGF-β and VEGF). As a result, RC enables CD8+ T cells to retain their proliferative and infiltrative abilities within the TME. Ultimately, these events promote cytotoxic T-cell-mediated clearance of tumor cells, which is further enhanced by the addition of anti-PD-L1 therapy. Furthermore, we employed LTA4H deficient mice (Lta4h-/- mice) to evaluate the antitumor efficiency, the results showed that the efficacy of immunotherapy was enhanced due to the synergistic effect of LTB4 signaling blockage and ICB inhibition, leading to remarkable inhibition of tumor growth in a mouse model of lung adenocarcinoma. CONCLUSIONS Taken together, these findings suggest that RC enhances antitumor immunity, providing a rationale for combining RC with immunotherapies as a potential anti-cancer treatment strategy.
Collapse
Affiliation(s)
- Jiangna Yan
- College of Medicine, Yan'an University, Yan'an, Shaanxi 716000, PR China
| | - Jinglin Zhu
- College of Medicine, Yan'an University, Yan'an, Shaanxi 716000, PR China
| | - Xiaolan Li
- Department of Pathology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi 710038, PR China
| | - Ruijie Yang
- School of Life Sciences, East China Normal University, Shanghai 200241, PR China
| | - Wei Xiao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Parmaceutical Co. Ltd., Lianyungang, Jiangsu 222001, PR China
| | - Chao Huang
- School of Basic Medical Sciences, Institute of Molecular and Translational Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China.
| | - Chunli Zheng
- College of Medicine, Yan'an University, Yan'an, Shaanxi 716000, PR China.
| |
Collapse
|
4
|
Liu X, Tao Q, Shen Y, Liu X, Yang Y, Ma N, Li J. Aspirin eugenol ester ameliorates LPS-induced inflammatory responses in RAW264.7 cells and mice. Front Pharmacol 2023; 14:1220780. [PMID: 37705535 PMCID: PMC10495573 DOI: 10.3389/fphar.2023.1220780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/18/2023] [Indexed: 09/15/2023] Open
Abstract
Introduction: Inflammation is a defensive response of the body and the pathological basis of many diseases. However, excessive inflammation and chronic inflammation impair the homeostasis of the organism. Arachidonic acid (AA) has a close relationship with inflammation and is the main mediator of the pro-inflammatory response. Based on the prodrug principle, the new pharmaceutical compound aspirin eugenol ester (AEE) was designed and synthesized. However, the effects of AEE on key enzymes, metabolites and inflammatory signaling pathways in the AA metabolic network have not been reported. Methods: In this study, the anti-inflammation effects of AEE were first investigated in mice and RAW264.7 cells in LPS induced inflammation model. Then, the changes of the key enzymes and AA metabolites were explored by RT-PCR and targeted metabolomics. Moreover, the regulatory effects on NF-kB and MAPKS signaling pathways were explored by Western Blotting. Results: Results indicated that AEE significantly reduced the number of leukocyte and increased the lymphocyte percentage. AEE decreased the expression levels of IL-1β, IL-6, IL-8 and TNF-α both in vivo and in vitro. In the liver of mice, AEE downregulated the levels of AA, prostaglandin D2 (PGD2) and upregulated 12- hydroxyeicosatetraenoic acid (12-HETE). However, the changes of PGE2, PGF2α, 6-keto-prostaglandin F1α (6-KETO-PGF1α), 9-hydroxy-octadecenoic acid (9- HODE), 13-HODE, 15-HETE, docosahexaenoic acid (DHA) and thromboxane B2 (TXB2) were not significant. Additionally, it was found that AEE decreased the relative mRNA expression levels of p65 and p38 and the ratio of p-p65/p65. Discussion: It was concluded that AEE might inhibit the LPS-induced inflammatory response through the regulation of AA metabolism. This study provides the theoretical foundation for the development of AEE as a medicinal anti-inflammatory drug.
Collapse
Affiliation(s)
- Xu Liu
- Hebei Veterinary Biotechnology Innovation Center, College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, China
| | - Qi Tao
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Youming Shen
- Quality Inspection and Test Center for Fruit and Nursery Stocks, Ministry of Agriculture and Rural Affairs (Xingcheng), Research Institute of Pomology Chinese Academy of Agricultural Sciences, Xingcheng, Liaoning, China
| | - Xiwang Liu
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yajun Yang
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Ning Ma
- Hebei Veterinary Biotechnology Innovation Center, College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, China
| | - Jianyong Li
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| |
Collapse
|
5
|
Gong R, Luo H, Long G, Xu J, Huang C, Zhou X, Shang Y, Zhang D. Integrative proteomic profiling of lung tissues and blood in acute respiratory distress syndrome. Front Immunol 2023; 14:1158951. [PMID: 37197655 PMCID: PMC10184823 DOI: 10.3389/fimmu.2023.1158951] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 04/20/2023] [Indexed: 05/19/2023] Open
Abstract
Introduction Acute respiratory distress syndrome and acute lung injury (ARDS/ALI) still lack a recognized diagnostic test and pharmacologic treatments that target the underlying pathology. Methods To explore the sensitive non-invasive biomarkers associated with pathological changes in the lung of direct ARDS/ALI, we performed an integrative proteomic analysis of lung and blood samples from lipopolysaccharide (LPS)-induced ARDS mice and COVID-19-related ARDS patients. The common differentially expressed proteins (DEPs) were identified based on combined proteomic analysis of serum and lung samples in direct ARDS mice model. The clinical value of the common DEPs was validated in lung and plasma proteomics in cases of COVID-19-related ARDS. Results We identified 368 DEPs in serum and 504 in lung samples from LPS-induced ARDS mice. Gene ontology (GO) classification and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that these DEPs in lung tissues were primarily enriched in pathways, including IL-17 and B cell receptor signaling pathways, and the response to stimuli. In contrast, DEPs in the serum were mostly involved in metabolic pathways and cellular processes. Through network analysis of protein-protein interactions (PPI), we identified diverse clusters of DEPs in the lung and serum samples. We further identified 50 commonly upregulated and 10 commonly downregulated DEPs in the lung and serum samples. Internal validation with a parallel-reacted monitor (PRM) and external validation in the Gene Expression Omnibus (GEO) datasets further showed these confirmed DEPs. We then validated these proteins in the proteomics of patients with ARDS and identified six proteins (HP, LTA4H, S100A9, SAA1, SAA2, and SERPINA3) with good clinical diagnostic and prognostic value. Discussion These proteins can be viewed as sensitive and non-invasive biomarkers associated with lung pathological changes in the blood and could potentially serve as targets for the early detection and treatment of direct ARDS especially in hyperinflammatory subphenotype.
Collapse
Affiliation(s)
- Rui Gong
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, Anhui, China
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
| | - Hong Luo
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
| | - Gangyu Long
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
| | - Jiqian Xu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
| | - Chaolin Huang
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
| | - Xin Zhou
- SpecAlly Life Technology Co., Ltd, Wuhan, Hubei, China
| | - You Shang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, Anhui, China
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- *Correspondence: Dingyu Zhang, ; You Shang,
| | - Dingyu Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, Anhui, China
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, Hubei, China
- *Correspondence: Dingyu Zhang, ; You Shang,
| |
Collapse
|
6
|
Tim-3 Blockade Decreases the Apoptosis of CD8 + T Cells and Reduces the Severity of Sepsis in Mice. J Surg Res 2022; 279:8-16. [PMID: 35716447 DOI: 10.1016/j.jss.2022.05.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 04/05/2022] [Accepted: 05/21/2022] [Indexed: 11/23/2022]
Abstract
INTRODUCTION The T cell immunoglobulin and mucin domain 3 (Tim-3) mediated immunosuppressive pathway has been shown to play an essential role in the development of sepsis. However, the influence of Tim-3 blockade during sepsis and the possible effects on T cells' function remains largely unknown. Our study investigates the role of Tim-3 in cecal ligation and puncture (CLP)-induced sepsis in mice. METHODS Sepsis was induced in C57BL/6 male mice via CLP. The expression of Tim-3 in CD8+ T cells after CLP challenge was measured. A dose of 50 μg anti-Tim-3 antibodies was injected intraperitoneally 30 min after surgery. Postoperative survival, bacterial clearance in the blood and peritoneal lavage fluid, cytokine secretion in the blood, and lung and liver histology were evaluated. In addition, the apoptosis of immune cells in the spleen and thymus was examined, respectively. RESULTS Tim-3 expression was elevated in the splenic CD8+ T cells of septic mice. At the early stage of CLP-induced sepsis, blocking Tim-3 with anti-Tim-3 antibodies reduced the severity of sepsis. The anti-Tim-3 antibodies alleviated the morphology of lung and liver injuries in septic mice. The anti-Tim-3 antibodies also reduced the severity of the inflammatory responses and lymphocyte apoptosis in septic mice. CONCLUSIONS Anti-Tim-3 antibodies might be a potential therapeutic strategy for sepsis.
Collapse
|
7
|
Lv XC, Wu Q, Cao YJ, Lin YC, Guo WL, Rao PF, Zhang YY, Chen YT, Ai LZ, Ni L. Ganoderic acid A from Ganoderma lucidum protects against alcoholic liver injury through ameliorating the lipid metabolism and modulating the intestinal microbial composition. Food Funct 2022; 13:5820-5837. [PMID: 35543349 DOI: 10.1039/d1fo03219d] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Alcoholic liver injury is mainly caused by long-term excessive alcohol consumption and has become a global public threat to human health. It is well known that Ganoderma lucidum has excellent beneficial effects on liver function and lipid metabolism. The object of this study was to investigate the hepatoprotective effects of ganoderic acid A (GAA, one of the main triterpenoids in G. lucidum) against alcohol-induced liver injury and reveal the underlying mechanisms of its protective effects. The results showed that oral administration of GAA significantly inhibited the abnormal elevation of the liver index, serum total triglyceride (TG), cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in mice exposed to alcohol intake, and also significantly protected the liver against alcohol-induced excessive lipid accumulation and pathological changes. Besides, alcohol-induced oxidative stress in the liver was significantly ameliorated by the dietary intervention of GAA through decreasing the hepatic levels of lactate dehydrogenase (LDH) and malondialdehyde (MDA), and increasing hepatic activities of catalase (CAT), superoxide dismutase (SOD), alcohol dehydrogenase (ADH), aldehyde dehydrogenase (ALDH), and hepatic levels of glutathione (GSH). In addition, GAA intervention evidently ameliorated intestinal microbial disorder by markedly increasing the abundance of Muribaculaceae, Prevotellaceae, Jeotgalicoccus, Bilophila, Family_XIII_UCG_001, Aerococcus, Ruminococcaceae_UCG_005, Harryflintia, Christensenellaceae, Rumonpcpccaceae, Prevotelaceae_UCG_001, Clostridiales_vadinBB60_group, Parasutterella and Bifidobacterium, but decreasing the proportion of Lactobacillus, Burkholderia_Caballeroria_Paraburkholderia, Escherichia_Shigella and Erysipelatoclostridium. Furthermore, liver metabolomics based on UPLC-QTOF/MS demonstrated that oral administration of GAA had a significant regulatory effect on the composition of liver metabolites in mice exposed to alcohol intake, especially the levels of the biomarkers involved in the metabolic pathways of riboflavin metabolism, glycine, serine and threonine metabolism, pyruvate metabolism, glycolysis/gluconeogenesis, biosynthesis of unsaturated fatty acids, synthesis and degradation of ketone bodies, fructose and mannose metabolism. Moreover, dietary supplementation of GAA significantly regulated the hepatic mRNA levels of lipid metabolism and inflammatory response related genes. Conclusively, these findings demonstrate that GAA has beneficial effects on alleviating alcohol-induced liver injury and is expected to become a new functional food ingredient for the prevention of alcoholic liver injury.
Collapse
Affiliation(s)
- Xu-Cong Lv
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, Fujian 350108, China.
| | - Qi Wu
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, Fujian 350108, China.
| | - Ying-Jia Cao
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, Fujian 350108, China. .,National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, P. R. China
| | - Yi-Chen Lin
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, Fujian 350108, China. .,National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, P. R. China
| | - Wei-Ling Guo
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, Fujian 350108, China.
| | - Ping-Fan Rao
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, Fujian 350108, China.
| | - Yan-Yan Zhang
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, Fujian 350108, China. .,Department of Flavor Chemistry, Institute of Food Science and Biotechnology, University of Hohenheim, Stuttgart 70599, Germany
| | - You-Ting Chen
- Fujian Abdominal Surgery Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China.
| | - Lian-Zhong Ai
- School of Medical Instruments and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Li Ni
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, Fujian 350108, China.
| |
Collapse
|
8
|
LTB4 Promotes Acute Lung Injury via Upregulating the PLCε-1/TLR4/NF-κB Pathway in One-Lung Ventilation. DISEASE MARKERS 2022; 2022:1839341. [PMID: 35059042 PMCID: PMC8766192 DOI: 10.1155/2022/1839341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/23/2021] [Indexed: 11/21/2022]
Abstract
Background Mechanical ventilation (MV) can provoke acute lung injury (ALI) by increasing inflammation activation and disrupting the barrier in lung tissues even causing death. However, the inflammation-related molecules and pathways in MV-induced ALI remain largely unknown. Hence, the purposes of this study are to examine the role and mechanism of a novel inflammation-related molecule, leukotriene B4 (LTB4), in ALI. Methods The functions of LTB4 in one-lung ventilation (OLV) model were detected by the loss-of-function experiments. H&E staining was used to examine the pathologic changes of lung tissues. Functionally, PLCε-1 knockdown and Toll-like receptor 4 (TLR4)/NF-κB pathway inhibitor were used to detect the regulatory effects of LTB4 on the phospholipase Cε (PLCε-1)/TLR4/nuclear factor-kappa B (NF-κB) pathway. The levels of genes and proteins were determined by RT-qPCR and western blotting assay. The levels of inflammation cytokines and chemokines were measured by ELISA. Results Here, we found LTA4H, leukotriene B (4) receptor 1 (BLT1), LTB4, and PLCε-1 upregulated in OLV rats and associated with inflammatory activation and lung permeability changes of lung tissues. Inhibition of LTB4 alleviated the OLV-induced ALI by inhibiting inflammatory activation and lung permeability changes of lung tissues. For mechanism analyses, LTB4 promoted OLV-induced ALI by activating the PLCε-1/TLR4/NF-κB pathway. Conclusion LTB4 induced ALI in OLV rats by activating the PLCε-1/TLR4/NF-κB pathway. Our findings might supply a new potential therapeutic for OLV-induced ALI.
Collapse
|
9
|
Lin WC, Fessler MB. Regulatory mechanisms of neutrophil migration from the circulation to the airspace. Cell Mol Life Sci 2021; 78:4095-4124. [PMID: 33544156 PMCID: PMC7863617 DOI: 10.1007/s00018-021-03768-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/22/2020] [Accepted: 01/16/2021] [Indexed: 02/07/2023]
Abstract
The neutrophil, a short-lived effector leukocyte of the innate immune system best known for its proteases and other degradative cargo, has unique, reciprocal physiological interactions with the lung. During health, large numbers of ‘marginated’ neutrophils reside within the pulmonary vasculature, where they patrol the endothelial surface for pathogens and complete their life cycle. Upon respiratory infection, rapid and sustained recruitment of neutrophils through the endothelial barrier, across the extravascular pulmonary interstitium, and again through the respiratory epithelium into the airspace lumen, is required for pathogen killing. Overexuberant neutrophil trafficking to the lung, however, causes bystander tissue injury and underlies several acute and chronic lung diseases. Due in part to the unique architecture of the lung’s capillary network, the neutrophil follows a microanatomic passage into the distal airspace unlike that observed in other end-organs that it infiltrates. Several of the regulatory mechanisms underlying the stepwise recruitment of circulating neutrophils to the infected lung have been defined over the past few decades; however, fundamental questions remain. In this article, we provide an updated review and perspective on emerging roles for the neutrophil in lung biology, on the molecular mechanisms that control the trafficking of neutrophils to the lung, and on past and ongoing efforts to design therapeutics to intervene upon pulmonary neutrophilia in lung disease.
Collapse
Affiliation(s)
- Wan-Chi Lin
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, P.O. Box 12233, MD D2-01, Research Triangle Park, NC, 27709, USA
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, P.O. Box 12233, MD D2-01, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
10
|
Tiwari P, Nagatake T, Hirata S, Sawane K, Saika A, Shibata Y, Morimoto S, Honda T, Adachi J, Abe Y, Isoyama J, Tomonaga T, Kiyono H, Kabashima K, Kunisawa J. Dietary coconut oil ameliorates skin contact hypersensitivity through mead acid production in mice. Allergy 2019; 74:1522-1532. [PMID: 30843234 DOI: 10.1111/all.13762] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 12/19/2018] [Accepted: 01/13/2019] [Indexed: 01/03/2023]
Abstract
Coconut oil is used as a dietary oil worldwide, and its healthy effects are recognized by the fact that coconut oil is easy to digest, helps in weight management, increases healthy cholesterol, and provides instant energy. Although topical application of coconut oil is known to reduce skin infection and inflammation, whether dietary coconut oil has any role in decreasing skin inflammation is unknown. In this study, we showed the impact of dietary coconut oil in allergic skin inflammation by using a mouse model of contact hypersensitivity (CHS). Mice maintained on coconut oil showed amelioration of skin inflammation and increased levels of cis-5, 8, 11-eicosatrienoic acid (mead acid) in serum. Intraperitoneal injection of mead acid inhibited CHS and reduced the number of neutrophils infiltrating to the skin. Detailed mechanistic studies unveiled that mead acid inhibited the directional migration of neutrophils by inhibiting the filamentous actin polymerization and leukotriene B4 production required for secondary recruitment of neutrophils. Our findings provide valuable insights into the preventive roles of coconut oil and mead acid against skin inflammation, thereby offering attractive therapeutic possibilities.
Collapse
Affiliation(s)
- Prabha Tiwari
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN) Ibaraki‐city, Osaka Japan
| | - Takahiro Nagatake
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN) Ibaraki‐city, Osaka Japan
| | - So‐ichiro Hirata
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN) Ibaraki‐city, Osaka Japan
- Department of Microbiology and Immunology Kobe University Graduate School of Medicine Kobe‐city, Hyogo Japan
| | - Kento Sawane
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN) Ibaraki‐city, Osaka Japan
- Graduate School of Pharmaceutical Sciences Osaka University Suita‐city, Osaka Japan
- Innovation Center Nippon Flour Mills Co., Ltd Atsugi-city, Kanagawa Japan
| | - Azusa Saika
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN) Ibaraki‐city, Osaka Japan
- Graduate School of Pharmaceutical Sciences Osaka University Suita‐city, Osaka Japan
| | - Yuki Shibata
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN) Ibaraki‐city, Osaka Japan
- Graduate School of Pharmaceutical Sciences Osaka University Suita‐city, Osaka Japan
| | - Sakiko Morimoto
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN) Ibaraki‐city, Osaka Japan
| | - Tetsuya Honda
- Department of Dermatology Kyoto University Graduate School of Medicine Kyoto-city, Kyoto Japan
| | - Jun Adachi
- Laboratory of Proteome Research NIBIOHN Ibaraki‐city, Osaka Japan
| | - Yuichi Abe
- Laboratory of Proteome Research NIBIOHN Ibaraki‐city, Osaka Japan
| | - Junko Isoyama
- Laboratory of Proteome Research NIBIOHN Ibaraki‐city, Osaka Japan
| | - Takeshi Tomonaga
- Laboratory of Proteome Research NIBIOHN Ibaraki‐city, Osaka Japan
| | - Hiroshi Kiyono
- International Research and Development Center for Mucosal Vaccines The Institute of Medical Science, The University of Tokyo Minato-ku, Tokyo Japan
- Department of Immunology, Graduate School of Medicine Chiba University Chiba‐city, Chiba Japan
- Division of Gastroenterology, Department of Medicine University of California, San Diego (UCSD) San Diego California
- CU‐UCSD Center for Mucosal Immunology, Allergy and Vaccines (cMAV) UCSD San Diego California
| | - Kenji Kabashima
- Department of Dermatology Kyoto University Graduate School of Medicine Kyoto-city, Kyoto Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN) Ibaraki‐city, Osaka Japan
- Department of Microbiology and Immunology Kobe University Graduate School of Medicine Kobe‐city, Hyogo Japan
- Graduate School of Pharmaceutical Sciences Osaka University Suita‐city, Osaka Japan
- International Research and Development Center for Mucosal Vaccines The Institute of Medical Science, The University of Tokyo Minato-ku, Tokyo Japan
- Graduate School of Medicine and Graduate School of Dentistry Osaka University Suita‐city, Osaka Japan
| |
Collapse
|
11
|
Shi J, Li P, Zhou L, Qi S, Wang B, Li D, Duan L, Chen WX, Xia J, Zou L, Yang S. Potential biomarkers for antidiastole of tuberculous and malignant pleural effusion by proteome analysis. Biomark Med 2019; 13:123-133. [PMID: 30791695 DOI: 10.2217/bmm-2018-0200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
AIM To investigate novel potential biomarkers for antidiastole of tuberculous pleural effusion (TPE) from malignant pleural effusion (MPE). MATERIALS & METHODS iTRAQTM-coupled LC-MS/MS were applied to analyze the proteome of TPE and MPE samples. The candidate proteins were verified by enzyme-linked immunosorbent assay. RESULTS A total of 432 differential proteins were identified. Enzyme-linked immunosorbent assay revealed significantly higher levels of fibronectin (FN) and cathepsin G (CTSG) in MPE than in TPE, but lower levels of leukotriene-A4 hydrolase (LTA4H). The receiver operator characteristic values were 0.285 for FN, 0.64 for LTA4H, 0.337 for CTSG and 0.793 for a combination of these candidate markers. CONCLUSION FN, LTA4H and CTSG were identified as potential biomarkers to differentiate TPE from MPE and their combination exhibited higher diagnostic capacity.
Collapse
Affiliation(s)
- Jing Shi
- Department of Laboratory Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Pu Li
- Department of Laboratory Medicine, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Lijin Zhou
- Department of Laboratory Medicine, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Suwen Qi
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements & Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, PR China
| | - Bo Wang
- Department of Laboratory Medicine, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Dandan Li
- Department of Laboratory Medicine, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Liang Duan
- Department of Laboratory Medicine, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Wei Xian Chen
- Department of Laboratory Medicine, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Jirong Xia
- Department of Laboratory Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Lin Zou
- Department of Laboratory Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Shuangshuang Yang
- Department of Laboratory Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| |
Collapse
|
12
|
Kim YH, Kim YS, Kim BH, Lee KS, Park HS, Lim CH. Remote ischemic preconditioning ameliorates indirect acute lung injury by modulating phosphorylation of IκBα in mice. J Int Med Res 2019; 47:936-950. [PMID: 30614352 PMCID: PMC6381478 DOI: 10.1177/0300060518818300] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 11/19/2018] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE Acute lung injury is responsible for mortality in seriously ill patients. Previous studies have shown that systemic inflammation is attenuated by remote ischemic preconditioning (RIPC) via reducing nuclear factor-kappa B (NF-κB). Therefore, we investigated whether lipopolysaccharide (LPS)-induced indirect acute lung injury (ALI) can be protected by RIPC. METHODS RIPC was accomplished by 10 minutes of occlusion using a tourniquet on the right hind limb of mice, followed by 10 minutes of reperfusion. This process was repeated three times. Intraperitoneal LPS (20 mg/kg) was administered to induce indirect ALI. Inflammatory cytokines in bronchoalveolar lavage fluid were analyzed using an enzyme-linked immunosorbent assay. Pulmonary tissue was excised for histological examination, and for examining NF-κB activity and phosphorylation of inhibitor of κBα (IκBα). RESULTS NF-κB activation and LPS-induced histopathological changes in the lungs were significantly alleviated in the RIPC group. RIPC reduced phosphorylation of IκBα in lung tissue of ALI mice. CONCLUSIONS RIPC attenuates endotoxin-induced indirect ALI. This attenuation might occur through modification of NF-κB mediation of cytokines by modulating phosphorylation of IκBα.
Collapse
Affiliation(s)
- Yun-Hee Kim
- Department of Anesthesiology and Pain Medicine, Korea University Ansan Hospital, Ansan, Korea
| | - Young-Sung Kim
- Department of Anesthesiology and Pain Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Byung-Hwa Kim
- Department of Anesthesiology and Pain Medicine, Korea University Ansan Hospital, Ansan, Korea
| | - Kuen-Su Lee
- Department of Anesthesiology and Pain Medicine, Korea University Ansan Hospital, Ansan, Korea
| | - Hyung-Sun Park
- Department of Anesthesiology and Pain Medicine, Korea University Anam Hospital, Seoul, Korea
| | - Choon-Hak Lim
- Department of Anesthesiology and Pain Medicine, Korea University Anam Hospital, Seoul, Korea
| |
Collapse
|
13
|
Chakraborty A, Boer JC, Selomulya C, Plebanski M, Royce SG. Insights into endotoxin-mediated lung inflammation and future treatment strategies. Expert Rev Respir Med 2018; 12:941-955. [PMID: 30221563 DOI: 10.1080/17476348.2018.1523009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Airway inflammatory disorders are prevalent diseases in need of better management and new therapeutics. Immunotherapies offer a solution to the problem of corticosteroid resistance. Areas covered: The current review focuses on lipopolysaccharide (Gram-negative bacterial endotoxin)-mediated inflammation in the lung and the animal models used to study related diseases. Endotoxin-induced lung pathology is usually initiated by antigen presenting cells (APC). We will discuss different subsets of APC including lung dendritic cells and macrophages, and their role in responding to endotoxin and environmental challenges. Expert commentary: The pharmacotherapeutic considerations to combat airway inflammation should cost-effectively improve quality of life with sustainable and safe strategies. Selectively targeting APCs in the lung offer the potential for a promising new strategy for the better management and treatment of inflammatory lung disease.
Collapse
Affiliation(s)
- Amlan Chakraborty
- a Department of Chemical Engineering , Monash University , Clayton , Australia.,b Department of Immunology and Pathology , Central Clinical School, Monash University , Melbourne , Australia
| | - Jennifer C Boer
- b Department of Immunology and Pathology , Central Clinical School, Monash University , Melbourne , Australia
| | - Cordelia Selomulya
- a Department of Chemical Engineering , Monash University , Clayton , Australia
| | - Magdalena Plebanski
- b Department of Immunology and Pathology , Central Clinical School, Monash University , Melbourne , Australia.,c School of Health and Biomedical Sciences and Enabling Capability platforms, Biomedical and Health Innovation , RMIT University , Melbourne , Australia
| | - Simon G Royce
- d Central Clinical School , Monash University , Clayton , Victoria , Australia.,e Department of Pharmacology , Monash University , Clayton , Australia
| |
Collapse
|
14
|
Dang X, Du G, Hu W, Ma L, Wang P, Li Y. Peroxisome proliferator‐activated receptor gamma coactivator‐1α/HSF1 axis effectively alleviates lipopolysaccharide‐induced acute lung injury via suppressing oxidative stress and inflammatory response. J Cell Biochem 2018; 120:544-551. [PMID: 30216506 DOI: 10.1002/jcb.27409] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/11/2018] [Indexed: 11/05/2022]
Affiliation(s)
- Xingbo Dang
- Department of Emergency Surgery Shannxi Provincial People‘s Hospital Xi’an Shannxi China
| | - Gongliang Du
- Department of Emergency Surgery Shannxi Provincial People‘s Hospital Xi’an Shannxi China
| | - Wei Hu
- Department of Emergency Surgery Shannxi Provincial People‘s Hospital Xi’an Shannxi China
| | - Longyang Ma
- Department of Emergency Surgery Shannxi Provincial People‘s Hospital Xi’an Shannxi China
| | - Pei Wang
- Department of Emergency Surgery Shannxi Provincial People‘s Hospital Xi’an Shannxi China
| | - Yi Li
- Department of Emergency Surgery Shannxi Provincial People‘s Hospital Xi’an Shannxi China
| |
Collapse
|
15
|
Wang C, Meng Y, Wang Y, Jiang Z, Xu M, Bo L, Deng X. Ouabain Protects Mice Against Lipopolysaccharide-Induced Acute Lung Injury. Med Sci Monit 2018; 24:4455-4464. [PMID: 29953424 PMCID: PMC6053945 DOI: 10.12659/msm.908627] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Ouabain, an inhibitor of Na+/K+-ATPase, is a type of endogenous hormone synthesized in the adrenal cortex and hypothalamus. Previous studies found that ouabain potently inhibited inflammatory reactions and regulated immunological processes. Our present study aimed to investigate the therapeutic role of ouabain on lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice. Material/Methods Ouabain (0.1 mg/kg) or vehicles were intraperitoneally injected into male C57BL/6J mice once a day for 3 consecutive days. One hour after the last injection of ouabain, LPS (5 mg/kg) was administrated through intranasal instillation to induce ALI. 6 hours and 24 hours later, bronchoalveolar lavage fluid (BALF) and lung tissues were harvested to detect the protective effects of ouabain, including protein concentration, inflammation cell counts, lung wet-to-dry ratio, and lung damage. Results The results showed that ouabain attenuated LPS-induced ALI in mice, which was indicated by alleviated pathological changes, downregulated TNF-α, IL-1β, and IL-6 production, inhibited neutrophils infiltration and macrophages, and ameliorated pulmonary edema and permeability. Further results found the activation of nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways were suppressed by ouabain in LPS-induced ALI. Conclusions These results suggest that ouabain negatively modulates the severity of LPS-induced ALI.
Collapse
Affiliation(s)
- Changli Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| | - Yan Meng
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| | - Yuanyuan Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland).,Department of Anesthesiology, Women and Children's Health Care Hospital of Linyi City, Linyi, Shandong, China (mainland)
| | - Zhengyu Jiang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| | - Mengda Xu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| | - Lulong Bo
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| | - Xiaoming Deng
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| |
Collapse
|
16
|
Lee JW, Seo KH, Ryu HW, Yuk HJ, Park HA, Lim Y, Ahn KS, Oh SR. Anti-inflammatory effect of stem bark of Paulownia tomentosa Steud. in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages and LPS-induced murine model of acute lung injury. JOURNAL OF ETHNOPHARMACOLOGY 2018; 210:23-30. [PMID: 28843892 DOI: 10.1016/j.jep.2017.08.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 08/09/2017] [Accepted: 08/23/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The leaves, bark, and flowers of Paulownia tomentosa Steud. have been widely used as a traditional medicine in East Asia to treat inflammatory and infectious diseases. AIM OF THE STUDY We investigated the protective effect of the methanol stem bark extract of P. tomentosa using an animal model of lipopolysaccharide (LPS)-induced acute lung injury (ALI). MATERIALS AND METHODS The UPLC Q-TOF-MS profiles for the methanol extract of P. tomentosa stem bark showed that verbascoside and isoverbascoside were the predominant compounds. Raw 264.7 cells were used for inhibitory effects of cytokine production in vitro. C57BL/6N mice were administered intranasally with LPS (10μg/per mouse) to induce ALI. H&E staining was used to evaluate histological changes in the lung. RESULTS Treatment with P. tomentosa stem bark extract (PTBE) suppressed the production of IL-6 and TNF-α in LPS-stimulated RAW 264.7 macrophages, and the recruitment of neutrophils and macrophages in the BALF of mice with LPS-induced ALI. PTBE also decreased the levels of reactive oxygen species (ROS) and pro-inflammatory cytokines in the BALF. PTBE reduced the levels of nitric oxide (NO) in the serum and of inducible nitric oxide synthase (iNOS) in the lung of ALI mice. PTBE also attenuated the infiltration of inflammatory cells and the expression of monocyte chemoattractant protein-1 (MCP-1) in the lung. In addition, PTBE suppressed the activation of NF-κB and the reduced expression of superoxide dismutase 3 (SOD3) in the lung. CONCLUSION The results suggest that PTBE has a protective effect on LPS-induced ALI.
Collapse
Affiliation(s)
- Jae-Won Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Chungju-si, Chungbuk 363-883, Republic of Korea.
| | - Kyeong-Hwa Seo
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Chungju-si, Chungbuk 363-883, Republic of Korea.
| | - Hyung Won Ryu
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Chungju-si, Chungbuk 363-883, Republic of Korea.
| | - Heung Joo Yuk
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Chungju-si, Chungbuk 363-883, Republic of Korea.
| | - Hyun Ah Park
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Chungju-si, Chungbuk 363-883, Republic of Korea.
| | - YouRim Lim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Chungju-si, Chungbuk 363-883, Republic of Korea.
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Chungju-si, Chungbuk 363-883, Republic of Korea.
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Chungju-si, Chungbuk 363-883, Republic of Korea.
| |
Collapse
|
17
|
Artemisia argyi attenuates airway inflammation in lipopolysaccharide induced acute lung injury model. Lab Anim Res 2017; 33:209-215. [PMID: 29046695 PMCID: PMC5645598 DOI: 10.5625/lar.2017.33.3.209] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/30/2017] [Accepted: 09/01/2017] [Indexed: 01/03/2023] Open
Abstract
Artemisia argyi is used as a health supplement, tea, and food source in Korea. This study aimed to evaluate the effect of Artemisia argyi (AA) and its active compound, dehydromatricarin A (DA), on the attenuation of airway inflammation in a murine model of lipopolysaccharide (LPS)-induced acute lung injury (ALI). The C57BL/6 mice were administered AA (50 mg/kg or 100 mg/kg) and DA (10 mg/kg or 20 mg/kg) by oral gavage from day 0 to 7 days and LPS treated by intranasal instillation 48 hours before the sacrifice. The treatment of AA and DA markedly decreased inflammatory cells in the bronchoalveolar lavage fluid (BALF) compared with that in ALI-induced mice, which was accompanied by a significant reduction in the levels of tumor necrosis factor (TNF)-α and interleukin (IL)-6 in BALF. Furthermore, the administration of AA and DA clearly decreased inducible nitric oxide synthase (iNOS) expression and nuclear factor kappa B (NF-κB) phosphorylation in comparison with that in the ALI-induced mice. The histological examination of the lung tissue revealed that the administration of AA and DA suppressed the inflammatory cell infiltration into the peribronchial and alveolar lesions induced by LPS instillation. Collectively, our results indicated that AA and DA effectively decreased the airway inflammatory response induced by LPS instillation. Therefore, AA and DA may offer a potential therapy for airway inflammatory disease.
Collapse
|
18
|
Yuk HJ, Lee JW, Park HA, Kwon OK, Seo KH, Ahn KS, Oh SR, Ryu HW. Protective effects of coumestrol on lipopolysaccharide-induced acute lung injury via the inhibition of proinflammatory mediators and NF-κB activation. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.04.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
19
|
Zhang F, Wu D, Wang GL, Hou S, Ou-Yang P, Huang J, Xu XY. Synthesis and biological evaluation of novel 1,2,3-benzotriazin-4-one derivatives as leukotriene A 4 hydrolase aminopeptidase inhibitors. CHINESE CHEM LETT 2017. [DOI: 10.1016/j.cclet.2016.12.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
20
|
Drinkwater N, Lee J, Yang W, Malcolm TR, McGowan S. M1 aminopeptidases as drug targets: broad applications or therapeutic niche? FEBS J 2017; 284:1473-1488. [PMID: 28075056 PMCID: PMC7164018 DOI: 10.1111/febs.14009] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/12/2016] [Accepted: 01/09/2017] [Indexed: 12/30/2022]
Abstract
M1 aminopeptidase enzymes are a diverse family of metalloenzymes characterized by conserved structure and reaction specificity. Excluding viruses, M1 aminopeptidases are distributed throughout all phyla, and have been implicated in a wide range of functions including cell maintenance, growth and development, and defense. The structure and catalytic mechanism of M1 aminopeptidases are well understood, and make them ideal candidates for the design of small‐molecule inhibitors. As a result, many research groups have assessed their utility as therapeutic targets for both infectious and chronic diseases of humans, and many inhibitors with a range of target specificities and potential therapeutic applications have been developed. Herein, we have aimed to address these studies, to determine whether the family of M1 aminopeptidases does in fact present a universal target for the treatment of a diverse range of human diseases. Our analysis indicates that early validation of M1 aminopeptidases as therapeutic targets is often overlooked, which prevents the enzymes from being confirmed as drug targets. This validation cannot be neglected, and needs to include a thorough characterization of enzymes’ specific roles within complex physiological pathways. Furthermore, any chemical probes used in target validation must be carefully designed to ensure that specificity over the closely related enzymes has been achieved. While many drug discovery programs that target M1 aminopeptidases remain in their infancy, certain inhibitors have shown promise for the treatment of a range of conditions including malaria, hypertension, and cancer.
Collapse
Affiliation(s)
- Nyssa Drinkwater
- Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne, Vic., Australia
| | - Jisook Lee
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic., Australia
| | - Wei Yang
- Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne, Vic., Australia
| | - Tess R Malcolm
- Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne, Vic., Australia
| | - Sheena McGowan
- Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne, Vic., Australia
| |
Collapse
|