Cargando…

SU5416 attenuated lipopolysaccharide-induced acute lung injury in mice by modulating properties of vascular endothelial cells

Background and aim: A potent and selective vascular endothelial growth factor receptor (VEGFR) inhibitor SU5416, has been developed for the treatment of solid human tumors. The binding of VEGF to VEGFR plays a crucial role in the pathophysiology of respiratory disorders. However, the impact of SU541...

Descripción completa

Detalles Bibliográficos
Autores principales: Huang, Xuqing, Zhu, Junqi, Jiang, Yuyue, Xu, Changqing, Lv, Qun, Yu, Dongwei, Shi, Kai, Ruan, Zhaoyang, Wang, Yan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Dove 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6536715/
https://www.ncbi.nlm.nih.gov/pubmed/31213766
http://dx.doi.org/10.2147/DDDT.S188858
_version_ 1783421822505582592
author Huang, Xuqing
Zhu, Junqi
Jiang, Yuyue
Xu, Changqing
Lv, Qun
Yu, Dongwei
Shi, Kai
Ruan, Zhaoyang
Wang, Yan
author_facet Huang, Xuqing
Zhu, Junqi
Jiang, Yuyue
Xu, Changqing
Lv, Qun
Yu, Dongwei
Shi, Kai
Ruan, Zhaoyang
Wang, Yan
author_sort Huang, Xuqing
collection PubMed
description Background and aim: A potent and selective vascular endothelial growth factor receptor (VEGFR) inhibitor SU5416, has been developed for the treatment of solid human tumors. The binding of VEGF to VEGFR plays a crucial role in the pathophysiology of respiratory disorders. However, the impact of SU5416 on lipopolysaccharide (LPS)-induced acute lung injury (ALI) remains unclear. Thus, this study aimed to illuminate the biofunction of SU5416 in the mouse model of ALI. Methods: Wild-type (WT) and toll-like receptor 4 (TLR4)-deficient (TLR4(−/-)) C57BL/6 mice were used to establish LPS-induced ALI model. The primary pulmonary microvascular endothelial cell (PMVEC) was extracted for detection of endothelial barrier function. Results: LPS significantly increased the number of inflammatory cells and inflammatory cytokines in bronchoalveolar lavage fluid (BALF). In addition, LPS increased alveolar epithelial cells injury, inflammation infiltration and vascular permeability of PMVEC in WT and TLR4(−/-) mice. Western blotting experiment indicated VEGF/VEGFR and TLR4/NF-κB pathways were involved in the progression of LPS-stimulated ALI. Consistent with previous research, dexamethasone treatment appeared to be an effective therapeutic for mice with ALI. Moreover, treatment with SU5416 dramatically attenuated LPS-induced immune responses in mice lung tissues via inhibiting VEGF/VEGFR and TLR4/NF-κB pathways. Finally, SU5416 also decreased vascular permeability of PMVEC in vitro. Conclusion: SU5416 ameliorated alveolar epithelial cells injury and histopathological changes in mice lung via inhibiting VEGF/VEGFR and TLR4/NF-κB signaling pathways. We also confirmed that SU5416 could restrain vascular permeability in PMVEC through improving the integrity of endothelial cell. These findings suggested that SU5416 may serve as a potential agent for the treatment of patients with ALI.
format Online
Article
Text
id pubmed-6536715
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Dove
record_format MEDLINE/PubMed
spelling pubmed-65367152019-06-18 SU5416 attenuated lipopolysaccharide-induced acute lung injury in mice by modulating properties of vascular endothelial cells Huang, Xuqing Zhu, Junqi Jiang, Yuyue Xu, Changqing Lv, Qun Yu, Dongwei Shi, Kai Ruan, Zhaoyang Wang, Yan Drug Des Devel Ther Original Research Background and aim: A potent and selective vascular endothelial growth factor receptor (VEGFR) inhibitor SU5416, has been developed for the treatment of solid human tumors. The binding of VEGF to VEGFR plays a crucial role in the pathophysiology of respiratory disorders. However, the impact of SU5416 on lipopolysaccharide (LPS)-induced acute lung injury (ALI) remains unclear. Thus, this study aimed to illuminate the biofunction of SU5416 in the mouse model of ALI. Methods: Wild-type (WT) and toll-like receptor 4 (TLR4)-deficient (TLR4(−/-)) C57BL/6 mice were used to establish LPS-induced ALI model. The primary pulmonary microvascular endothelial cell (PMVEC) was extracted for detection of endothelial barrier function. Results: LPS significantly increased the number of inflammatory cells and inflammatory cytokines in bronchoalveolar lavage fluid (BALF). In addition, LPS increased alveolar epithelial cells injury, inflammation infiltration and vascular permeability of PMVEC in WT and TLR4(−/-) mice. Western blotting experiment indicated VEGF/VEGFR and TLR4/NF-κB pathways were involved in the progression of LPS-stimulated ALI. Consistent with previous research, dexamethasone treatment appeared to be an effective therapeutic for mice with ALI. Moreover, treatment with SU5416 dramatically attenuated LPS-induced immune responses in mice lung tissues via inhibiting VEGF/VEGFR and TLR4/NF-κB pathways. Finally, SU5416 also decreased vascular permeability of PMVEC in vitro. Conclusion: SU5416 ameliorated alveolar epithelial cells injury and histopathological changes in mice lung via inhibiting VEGF/VEGFR and TLR4/NF-κB signaling pathways. We also confirmed that SU5416 could restrain vascular permeability in PMVEC through improving the integrity of endothelial cell. These findings suggested that SU5416 may serve as a potential agent for the treatment of patients with ALI. Dove 2019-05-23 /pmc/articles/PMC6536715/ /pubmed/31213766 http://dx.doi.org/10.2147/DDDT.S188858 Text en © 2019 Huang et al. http://creativecommons.org/licenses/by-nc/3.0/ This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution – Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/). By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms (https://www.dovepress.com/terms.php).
spellingShingle Original Research
Huang, Xuqing
Zhu, Junqi
Jiang, Yuyue
Xu, Changqing
Lv, Qun
Yu, Dongwei
Shi, Kai
Ruan, Zhaoyang
Wang, Yan
SU5416 attenuated lipopolysaccharide-induced acute lung injury in mice by modulating properties of vascular endothelial cells
title SU5416 attenuated lipopolysaccharide-induced acute lung injury in mice by modulating properties of vascular endothelial cells
title_full SU5416 attenuated lipopolysaccharide-induced acute lung injury in mice by modulating properties of vascular endothelial cells
title_fullStr SU5416 attenuated lipopolysaccharide-induced acute lung injury in mice by modulating properties of vascular endothelial cells
title_full_unstemmed SU5416 attenuated lipopolysaccharide-induced acute lung injury in mice by modulating properties of vascular endothelial cells
title_short SU5416 attenuated lipopolysaccharide-induced acute lung injury in mice by modulating properties of vascular endothelial cells
title_sort su5416 attenuated lipopolysaccharide-induced acute lung injury in mice by modulating properties of vascular endothelial cells
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6536715/
https://www.ncbi.nlm.nih.gov/pubmed/31213766
http://dx.doi.org/10.2147/DDDT.S188858
work_keys_str_mv AT huangxuqing su5416attenuatedlipopolysaccharideinducedacutelunginjuryinmicebymodulatingpropertiesofvascularendothelialcells
AT zhujunqi su5416attenuatedlipopolysaccharideinducedacutelunginjuryinmicebymodulatingpropertiesofvascularendothelialcells
AT jiangyuyue su5416attenuatedlipopolysaccharideinducedacutelunginjuryinmicebymodulatingpropertiesofvascularendothelialcells
AT xuchangqing su5416attenuatedlipopolysaccharideinducedacutelunginjuryinmicebymodulatingpropertiesofvascularendothelialcells
AT lvqun su5416attenuatedlipopolysaccharideinducedacutelunginjuryinmicebymodulatingpropertiesofvascularendothelialcells
AT yudongwei su5416attenuatedlipopolysaccharideinducedacutelunginjuryinmicebymodulatingpropertiesofvascularendothelialcells
AT shikai su5416attenuatedlipopolysaccharideinducedacutelunginjuryinmicebymodulatingpropertiesofvascularendothelialcells
AT ruanzhaoyang su5416attenuatedlipopolysaccharideinducedacutelunginjuryinmicebymodulatingpropertiesofvascularendothelialcells
AT wangyan su5416attenuatedlipopolysaccharideinducedacutelunginjuryinmicebymodulatingpropertiesofvascularendothelialcells