Cargando…

Improvement of gut-vascular barrier by terlipressin reduces bacterial translocation and remote organ injuries in gut-derived sepsis

Gut-vascular barrier (GVB) serves as the last barrier to limit the migration of intestinal toxins into the blood circulation. The efficacy of terlipressin (a vasopressin V1 receptor agonist) in reducing GVB and multiple organ damage in gut-derived sepsis is unknown. In this study, we hypothesized th...

Descripción completa

Detalles Bibliográficos
Autores principales: Chang, Zenan, Zhang, Yinan, Lin, Ming, Wen, Shihong, Lai, Hanjin, Zhan, Yaqing, Zhu, Xiufen, Huang, Zhikun, Zhang, Xuyu, Liu, Zimeng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9585222/
https://www.ncbi.nlm.nih.gov/pubmed/36278213
http://dx.doi.org/10.3389/fphar.2022.1019109
_version_ 1784813443463774208
author Chang, Zenan
Zhang, Yinan
Lin, Ming
Wen, Shihong
Lai, Hanjin
Zhan, Yaqing
Zhu, Xiufen
Huang, Zhikun
Zhang, Xuyu
Liu, Zimeng
author_facet Chang, Zenan
Zhang, Yinan
Lin, Ming
Wen, Shihong
Lai, Hanjin
Zhan, Yaqing
Zhu, Xiufen
Huang, Zhikun
Zhang, Xuyu
Liu, Zimeng
author_sort Chang, Zenan
collection PubMed
description Gut-vascular barrier (GVB) serves as the last barrier to limit the migration of intestinal toxins into the blood circulation. The efficacy of terlipressin (a vasopressin V1 receptor agonist) in reducing GVB and multiple organ damage in gut-derived sepsis is unknown. In this study, we hypothesized that, besides other intestinal barriers, GVB play a key role in gut-derived sepsis and terlipressin improve GVB damage and then reduce bacterial translocation and organ injuries. In vivo, a cecal ligation and puncture mouse model was established. The mice were subjected to examine the damage of GVB determined by intestinal plasmalemma vesicle-associated protein-1(PV-1) and vascular endothelial-cadherin. And the intestinal permeability was assessed by translocation of intestinal bacteria and macromolecules. In vitro, transendothelial electrical resistance (TER) during interleukin (IL)-1β stimulation was measured on endothelial cells with or without small interfering RNA targeting β-catenin (si β-catenin). Terlipressin significantly improved GVB damage and reduced translocation of intestinal macromolecules and bacteria by activating PI3K signaling. Of note, intestinal PV-1 expression was significantly correlated with translocation of macromolecules, and dramatic increase of macromolecules was observed in intestinal tissues whereas fewer macromolecules and bacteria were observed in blood, liver and lung following terlipressin treatment. In vitro, terlipressin restored TER during IL-1β stimulation and si β-catenin transfection blocked the changes delivered by terlipressin. Collectively, terlipressin alleviated GVB damage and subsequent bacterial translocation via blood vessels after sepsis challenge, resulting in reduced distant organ injuries and the responsible mechanisms may involve the activation of PI3K/β-catenin pathway.
format Online
Article
Text
id pubmed-9585222
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-95852222022-10-22 Improvement of gut-vascular barrier by terlipressin reduces bacterial translocation and remote organ injuries in gut-derived sepsis Chang, Zenan Zhang, Yinan Lin, Ming Wen, Shihong Lai, Hanjin Zhan, Yaqing Zhu, Xiufen Huang, Zhikun Zhang, Xuyu Liu, Zimeng Front Pharmacol Pharmacology Gut-vascular barrier (GVB) serves as the last barrier to limit the migration of intestinal toxins into the blood circulation. The efficacy of terlipressin (a vasopressin V1 receptor agonist) in reducing GVB and multiple organ damage in gut-derived sepsis is unknown. In this study, we hypothesized that, besides other intestinal barriers, GVB play a key role in gut-derived sepsis and terlipressin improve GVB damage and then reduce bacterial translocation and organ injuries. In vivo, a cecal ligation and puncture mouse model was established. The mice were subjected to examine the damage of GVB determined by intestinal plasmalemma vesicle-associated protein-1(PV-1) and vascular endothelial-cadherin. And the intestinal permeability was assessed by translocation of intestinal bacteria and macromolecules. In vitro, transendothelial electrical resistance (TER) during interleukin (IL)-1β stimulation was measured on endothelial cells with or without small interfering RNA targeting β-catenin (si β-catenin). Terlipressin significantly improved GVB damage and reduced translocation of intestinal macromolecules and bacteria by activating PI3K signaling. Of note, intestinal PV-1 expression was significantly correlated with translocation of macromolecules, and dramatic increase of macromolecules was observed in intestinal tissues whereas fewer macromolecules and bacteria were observed in blood, liver and lung following terlipressin treatment. In vitro, terlipressin restored TER during IL-1β stimulation and si β-catenin transfection blocked the changes delivered by terlipressin. Collectively, terlipressin alleviated GVB damage and subsequent bacterial translocation via blood vessels after sepsis challenge, resulting in reduced distant organ injuries and the responsible mechanisms may involve the activation of PI3K/β-catenin pathway. Frontiers Media S.A. 2022-10-07 /pmc/articles/PMC9585222/ /pubmed/36278213 http://dx.doi.org/10.3389/fphar.2022.1019109 Text en Copyright © 2022 Chang, Zhang, Lin, Wen, Lai, Zhan, Zhu, Huang, Zhang and Liu. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Pharmacology
Chang, Zenan
Zhang, Yinan
Lin, Ming
Wen, Shihong
Lai, Hanjin
Zhan, Yaqing
Zhu, Xiufen
Huang, Zhikun
Zhang, Xuyu
Liu, Zimeng
Improvement of gut-vascular barrier by terlipressin reduces bacterial translocation and remote organ injuries in gut-derived sepsis
title Improvement of gut-vascular barrier by terlipressin reduces bacterial translocation and remote organ injuries in gut-derived sepsis
title_full Improvement of gut-vascular barrier by terlipressin reduces bacterial translocation and remote organ injuries in gut-derived sepsis
title_fullStr Improvement of gut-vascular barrier by terlipressin reduces bacterial translocation and remote organ injuries in gut-derived sepsis
title_full_unstemmed Improvement of gut-vascular barrier by terlipressin reduces bacterial translocation and remote organ injuries in gut-derived sepsis
title_short Improvement of gut-vascular barrier by terlipressin reduces bacterial translocation and remote organ injuries in gut-derived sepsis
title_sort improvement of gut-vascular barrier by terlipressin reduces bacterial translocation and remote organ injuries in gut-derived sepsis
topic Pharmacology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9585222/
https://www.ncbi.nlm.nih.gov/pubmed/36278213
http://dx.doi.org/10.3389/fphar.2022.1019109
work_keys_str_mv AT changzenan improvementofgutvascularbarrierbyterlipressinreducesbacterialtranslocationandremoteorganinjuriesingutderivedsepsis
AT zhangyinan improvementofgutvascularbarrierbyterlipressinreducesbacterialtranslocationandremoteorganinjuriesingutderivedsepsis
AT linming improvementofgutvascularbarrierbyterlipressinreducesbacterialtranslocationandremoteorganinjuriesingutderivedsepsis
AT wenshihong improvementofgutvascularbarrierbyterlipressinreducesbacterialtranslocationandremoteorganinjuriesingutderivedsepsis
AT laihanjin improvementofgutvascularbarrierbyterlipressinreducesbacterialtranslocationandremoteorganinjuriesingutderivedsepsis
AT zhanyaqing improvementofgutvascularbarrierbyterlipressinreducesbacterialtranslocationandremoteorganinjuriesingutderivedsepsis
AT zhuxiufen improvementofgutvascularbarrierbyterlipressinreducesbacterialtranslocationandremoteorganinjuriesingutderivedsepsis
AT huangzhikun improvementofgutvascularbarrierbyterlipressinreducesbacterialtranslocationandremoteorganinjuriesingutderivedsepsis
AT zhangxuyu improvementofgutvascularbarrierbyterlipressinreducesbacterialtranslocationandremoteorganinjuriesingutderivedsepsis
AT liuzimeng improvementofgutvascularbarrierbyterlipressinreducesbacterialtranslocationandremoteorganinjuriesingutderivedsepsis