多杀性巴氏杆菌感染会降低相邻脑微血管内皮细胞之间的紧密连接和粘附连接,从而诱发血脑屏障破坏。

IF 3.7 1区 农林科学 Q1 VETERINARY SCIENCES
Lin Lin, Haixin Bi, Jie Yang, Yuyao Shang, Qingjie Lv, Dajun Zhang, Xi Huang, Mengfei Zhao, Fei Wang, Lin Hua, Huanchun Chen, Bin Wu, Xiangru Wang, Zhong Peng
{"title":"多杀性巴氏杆菌感染会降低相邻脑微血管内皮细胞之间的紧密连接和粘附连接,从而诱发血脑屏障破坏。","authors":"Lin Lin, Haixin Bi, Jie Yang, Yuyao Shang, Qingjie Lv, Dajun Zhang, Xi Huang, Mengfei Zhao, Fei Wang, Lin Hua, Huanchun Chen, Bin Wu, Xiangru Wang, Zhong Peng","doi":"10.1186/s13567-024-01351-5","DOIUrl":null,"url":null,"abstract":"<p><p>Meningitis induced by Pasteurella multocida has been substantially described in clinical practice in both human and veterinary medicine, but the underlying mechanisms have not been previously reported. In this study, we investigated the influence of P. multocida infection on the permeability of the blood-brain barrier (BBB) using different models. Our in vivo tests in a mouse model and in vitro tests using human brain microvascular endothelial cell (hBMEC) model showed that P. multocida infection increased murine BBB permeability in mice and hBMEC monolayer permeability. Furthermore, we observed that P. multocida infection resulted in decreased expression of tight junctions (ZO1, claudin-5, occludin) and adherens junctions (E-cadherin) between neighboring hBMECs. Subsequent experiments revealed that P. multocida infection promoted the activation of hypoxia inducible factor-1α (HIF-1α)/vascular endothelial growth factor A (VEGFA) signaling and NF-κB signaling, and suppressed the HIF-1α/VEGFA significantly remitted the decrease in ZO1/E-cadherin induced by P. multocida infection (P < 0.001). NF-κB signaling was found to contribute to the production of chemokines such as TNF-1α, IL-β, and IL-6. Additionally, transmission electron microscopy revealed that paracellular migration might be the strategy employed by P. multocida to cross the BBB. This study provides the first evidence of the migration strategy used by P. multocida to traverse the mammalian BBB. The data presented herein will contribute to a better understanding of the pathogenesis of the zoonotic pathogen P. multocida.</p>","PeriodicalId":23658,"journal":{"name":"Veterinary Research","volume":null,"pages":null},"PeriodicalIF":3.7000,"publicationDate":"2024-08-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11363436/pdf/","citationCount":"0","resultStr":"{\"title\":\"Pasteurella multocida infection induces blood-brain barrier disruption by decreasing tight junctions and adherens junctions between neighbored brain microvascular endothelial cells.\",\"authors\":\"Lin Lin, Haixin Bi, Jie Yang, Yuyao Shang, Qingjie Lv, Dajun Zhang, Xi Huang, Mengfei Zhao, Fei Wang, Lin Hua, Huanchun Chen, Bin Wu, Xiangru Wang, Zhong Peng\",\"doi\":\"10.1186/s13567-024-01351-5\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>Meningitis induced by Pasteurella multocida has been substantially described in clinical practice in both human and veterinary medicine, but the underlying mechanisms have not been previously reported. In this study, we investigated the influence of P. multocida infection on the permeability of the blood-brain barrier (BBB) using different models. Our in vivo tests in a mouse model and in vitro tests using human brain microvascular endothelial cell (hBMEC) model showed that P. multocida infection increased murine BBB permeability in mice and hBMEC monolayer permeability. Furthermore, we observed that P. multocida infection resulted in decreased expression of tight junctions (ZO1, claudin-5, occludin) and adherens junctions (E-cadherin) between neighboring hBMECs. Subsequent experiments revealed that P. multocida infection promoted the activation of hypoxia inducible factor-1α (HIF-1α)/vascular endothelial growth factor A (VEGFA) signaling and NF-κB signaling, and suppressed the HIF-1α/VEGFA significantly remitted the decrease in ZO1/E-cadherin induced by P. multocida infection (P < 0.001). NF-κB signaling was found to contribute to the production of chemokines such as TNF-1α, IL-β, and IL-6. Additionally, transmission electron microscopy revealed that paracellular migration might be the strategy employed by P. multocida to cross the BBB. This study provides the first evidence of the migration strategy used by P. multocida to traverse the mammalian BBB. The data presented herein will contribute to a better understanding of the pathogenesis of the zoonotic pathogen P. multocida.</p>\",\"PeriodicalId\":23658,\"journal\":{\"name\":\"Veterinary Research\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":3.7000,\"publicationDate\":\"2024-08-29\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11363436/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Veterinary Research\",\"FirstCategoryId\":\"97\",\"ListUrlMain\":\"https://doi.org/10.1186/s13567-024-01351-5\",\"RegionNum\":1,\"RegionCategory\":\"农林科学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"VETERINARY SCIENCES\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Veterinary Research","FirstCategoryId":"97","ListUrlMain":"https://doi.org/10.1186/s13567-024-01351-5","RegionNum":1,"RegionCategory":"农林科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"VETERINARY SCIENCES","Score":null,"Total":0}
引用次数: 0

摘要

由多杀性巴氏杆菌诱发的脑膜炎在人类和兽医的临床实践中都有大量描述,但其潜在机制以前却未见报道。在这项研究中,我们使用不同的模型研究了多杀性巴氏杆菌感染对血脑屏障(BBB)通透性的影响。我们在小鼠模型中进行的体内试验和使用人脑微血管内皮细胞(hBMEC)模型进行的体外试验表明,多杀霉素感染会增加小鼠血脑屏障的通透性和 hBMEC 单层的通透性。此外,我们还观察到,多杀螨病毒感染导致相邻 hBMEC 之间紧密连接(ZO1、claudin-5、occludin)和粘连连接(E-cadherin)的表达减少。随后的实验发现,多杀螨病毒感染促进了缺氧诱导因子-1α(HIF-1α)/血管内皮生长因子A(VEGFA)信号传导和NF-κB信号传导的激活,而抑制HIF-1α/VEGFA能显著缓解多杀螨病毒感染引起的ZO1/E-cadherin的减少(P<0.05)。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Pasteurella multocida infection induces blood-brain barrier disruption by decreasing tight junctions and adherens junctions between neighbored brain microvascular endothelial cells.

Meningitis induced by Pasteurella multocida has been substantially described in clinical practice in both human and veterinary medicine, but the underlying mechanisms have not been previously reported. In this study, we investigated the influence of P. multocida infection on the permeability of the blood-brain barrier (BBB) using different models. Our in vivo tests in a mouse model and in vitro tests using human brain microvascular endothelial cell (hBMEC) model showed that P. multocida infection increased murine BBB permeability in mice and hBMEC monolayer permeability. Furthermore, we observed that P. multocida infection resulted in decreased expression of tight junctions (ZO1, claudin-5, occludin) and adherens junctions (E-cadherin) between neighboring hBMECs. Subsequent experiments revealed that P. multocida infection promoted the activation of hypoxia inducible factor-1α (HIF-1α)/vascular endothelial growth factor A (VEGFA) signaling and NF-κB signaling, and suppressed the HIF-1α/VEGFA significantly remitted the decrease in ZO1/E-cadherin induced by P. multocida infection (P < 0.001). NF-κB signaling was found to contribute to the production of chemokines such as TNF-1α, IL-β, and IL-6. Additionally, transmission electron microscopy revealed that paracellular migration might be the strategy employed by P. multocida to cross the BBB. This study provides the first evidence of the migration strategy used by P. multocida to traverse the mammalian BBB. The data presented herein will contribute to a better understanding of the pathogenesis of the zoonotic pathogen P. multocida.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Veterinary Research
Veterinary Research 农林科学-兽医学
CiteScore
7.00
自引率
4.50%
发文量
92
审稿时长
3 months
期刊介绍: Veterinary Research is an open access journal that publishes high quality and novel research and review articles focusing on all aspects of infectious diseases and host-pathogen interaction in animals.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信