Andrographolide Sulfonate Attenuates Acute Lung Injury by Reducing Expression of Myeloperoxidase and Neutrophil-Derived Proteases in Mice

Andrographolide sulfonate (Andro-S), a sulfonation derivative of andrographolide, is known to be effective in treating inflammation-related diseases, while the underlying mechanisms and global protein alterations in response to Andro-S remain unknown. This study aimed to investigate the pharmacologi...

Full description

Bibliographic Details
Main Authors: Fei Gao, Xing Liu, Ziying Shen, Xiaohui Jia, Han He, Jing Gao, Jianhong Wu, Chunhong Jiang, Hu Zhou, Yiping Wang
Format: Article
Language:English
Published: Frontiers Media S.A. 2018-08-01
Series:Frontiers in Physiology
Subjects:
Online Access:https://www.frontiersin.org/article/10.3389/fphys.2018.00939/full
_version_ 1811283309810941952
author Fei Gao
Xing Liu
Ziying Shen
Xiaohui Jia
Han He
Jing Gao
Jianhong Wu
Chunhong Jiang
Hu Zhou
Yiping Wang
author_facet Fei Gao
Xing Liu
Ziying Shen
Xiaohui Jia
Han He
Jing Gao
Jianhong Wu
Chunhong Jiang
Hu Zhou
Yiping Wang
author_sort Fei Gao
collection DOAJ
description Andrographolide sulfonate (Andro-S), a sulfonation derivative of andrographolide, is known to be effective in treating inflammation-related diseases, while the underlying mechanisms and global protein alterations in response to Andro-S remain unknown. This study aimed to investigate the pharmacological effects and potential targets of Andro-S in a murine model of acute lung injury (ALI). ALI was induced by aerosolized lipopolysaccharide (LPS) exposure before treatment with Andro-S. Inflammatory state of each treatment group was determined by histological analysis and quantification of inflammatory markers. Differentially expressed proteins in lung tissues were identified by an iTRAQ-based quantitative proteomic approach and further confirmed by immunohistochemistry analysis. Administration of Andro-S alleviated LPS-induced histological changes in the lung and reduced the expression of inflammatory markers in serum, bronchoalveolar fluid and lung tissues. Proteomic analysis identified 31 differentially expressed proteins from a total of 2,234 quantified proteins in the lung. According to bioinformatics analysis, neutrophil elastase (ELANE), cathepsin G (CTSG) and myeloperoxidase (MPO), three neutrophil-derived proteases related to immune system process and defense responses to fungi were chosen as potential targets of Andro-S. Further immunohistochemistry analysis confirmed the inhibitory effects of Andro-S on LPS-induced ELANE, CTSG and MPO up-regulation. These results indicate that Andro-S suppressed the severity of LPS-induced ALI, possibly by attenuating the expression of and neutrophil-derived proteases.
first_indexed 2024-04-13T02:09:07Z
format Article
id doaj.art-632b20a9055140e39e53b3d4c184579a
institution Directory Open Access Journal
issn 1664-042X
language English
last_indexed 2024-04-13T02:09:07Z
publishDate 2018-08-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Physiology
spelling doaj.art-632b20a9055140e39e53b3d4c184579a2022-12-22T03:07:21ZengFrontiers Media S.A.Frontiers in Physiology1664-042X2018-08-01910.3389/fphys.2018.00939346859Andrographolide Sulfonate Attenuates Acute Lung Injury by Reducing Expression of Myeloperoxidase and Neutrophil-Derived Proteases in MiceFei Gao0Xing Liu1Ziying Shen2Xiaohui Jia3Han He4Jing Gao5Jianhong Wu6Chunhong Jiang7Hu Zhou8Yiping Wang9State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, ChinaDepartment of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, ChinaState Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, ChinaState Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, ChinaDepartment of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, ChinaDepartment of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, ChinaDepartment of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, ChinaState Key Laboratory of Innovative Natural Medicine and TCM Injections, Ganzhou, ChinaDepartment of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, ChinaState Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, ChinaAndrographolide sulfonate (Andro-S), a sulfonation derivative of andrographolide, is known to be effective in treating inflammation-related diseases, while the underlying mechanisms and global protein alterations in response to Andro-S remain unknown. This study aimed to investigate the pharmacological effects and potential targets of Andro-S in a murine model of acute lung injury (ALI). ALI was induced by aerosolized lipopolysaccharide (LPS) exposure before treatment with Andro-S. Inflammatory state of each treatment group was determined by histological analysis and quantification of inflammatory markers. Differentially expressed proteins in lung tissues were identified by an iTRAQ-based quantitative proteomic approach and further confirmed by immunohistochemistry analysis. Administration of Andro-S alleviated LPS-induced histological changes in the lung and reduced the expression of inflammatory markers in serum, bronchoalveolar fluid and lung tissues. Proteomic analysis identified 31 differentially expressed proteins from a total of 2,234 quantified proteins in the lung. According to bioinformatics analysis, neutrophil elastase (ELANE), cathepsin G (CTSG) and myeloperoxidase (MPO), three neutrophil-derived proteases related to immune system process and defense responses to fungi were chosen as potential targets of Andro-S. Further immunohistochemistry analysis confirmed the inhibitory effects of Andro-S on LPS-induced ELANE, CTSG and MPO up-regulation. These results indicate that Andro-S suppressed the severity of LPS-induced ALI, possibly by attenuating the expression of and neutrophil-derived proteases.https://www.frontiersin.org/article/10.3389/fphys.2018.00939/fullandrographolide sulfonateacute lung injuryiTRAQquantitative proteomicsimmunohistochemistry
spellingShingle Fei Gao
Xing Liu
Ziying Shen
Xiaohui Jia
Han He
Jing Gao
Jianhong Wu
Chunhong Jiang
Hu Zhou
Yiping Wang
Andrographolide Sulfonate Attenuates Acute Lung Injury by Reducing Expression of Myeloperoxidase and Neutrophil-Derived Proteases in Mice
Frontiers in Physiology
andrographolide sulfonate
acute lung injury
iTRAQ
quantitative proteomics
immunohistochemistry
title Andrographolide Sulfonate Attenuates Acute Lung Injury by Reducing Expression of Myeloperoxidase and Neutrophil-Derived Proteases in Mice
title_full Andrographolide Sulfonate Attenuates Acute Lung Injury by Reducing Expression of Myeloperoxidase and Neutrophil-Derived Proteases in Mice
title_fullStr Andrographolide Sulfonate Attenuates Acute Lung Injury by Reducing Expression of Myeloperoxidase and Neutrophil-Derived Proteases in Mice
title_full_unstemmed Andrographolide Sulfonate Attenuates Acute Lung Injury by Reducing Expression of Myeloperoxidase and Neutrophil-Derived Proteases in Mice
title_short Andrographolide Sulfonate Attenuates Acute Lung Injury by Reducing Expression of Myeloperoxidase and Neutrophil-Derived Proteases in Mice
title_sort andrographolide sulfonate attenuates acute lung injury by reducing expression of myeloperoxidase and neutrophil derived proteases in mice
topic andrographolide sulfonate
acute lung injury
iTRAQ
quantitative proteomics
immunohistochemistry
url https://www.frontiersin.org/article/10.3389/fphys.2018.00939/full
work_keys_str_mv AT feigao andrographolidesulfonateattenuatesacutelunginjurybyreducingexpressionofmyeloperoxidaseandneutrophilderivedproteasesinmice
AT xingliu andrographolidesulfonateattenuatesacutelunginjurybyreducingexpressionofmyeloperoxidaseandneutrophilderivedproteasesinmice
AT ziyingshen andrographolidesulfonateattenuatesacutelunginjurybyreducingexpressionofmyeloperoxidaseandneutrophilderivedproteasesinmice
AT xiaohuijia andrographolidesulfonateattenuatesacutelunginjurybyreducingexpressionofmyeloperoxidaseandneutrophilderivedproteasesinmice
AT hanhe andrographolidesulfonateattenuatesacutelunginjurybyreducingexpressionofmyeloperoxidaseandneutrophilderivedproteasesinmice
AT jinggao andrographolidesulfonateattenuatesacutelunginjurybyreducingexpressionofmyeloperoxidaseandneutrophilderivedproteasesinmice
AT jianhongwu andrographolidesulfonateattenuatesacutelunginjurybyreducingexpressionofmyeloperoxidaseandneutrophilderivedproteasesinmice
AT chunhongjiang andrographolidesulfonateattenuatesacutelunginjurybyreducingexpressionofmyeloperoxidaseandneutrophilderivedproteasesinmice
AT huzhou andrographolidesulfonateattenuatesacutelunginjurybyreducingexpressionofmyeloperoxidaseandneutrophilderivedproteasesinmice
AT yipingwang andrographolidesulfonateattenuatesacutelunginjurybyreducingexpressionofmyeloperoxidaseandneutrophilderivedproteasesinmice