Helicobacter pylori‐derived outer membrane vesicles contribute to Alzheimer's disease pathogenesis via C3‐C3aR signalling

Author:

Xie Junhua123ORCID,Cools Lien124,Van Imschoot Griet12,Van Wonterghem Elien12,Pauwels Marie J.12,Vlaeminck Ine56ORCID,De Witte Chloë3,EL Andaloussi Samir7,Wierda Keimpe56ORCID,De Groef Lies4ORCID,Haesebrouck Freddy3ORCID,Van Hoecke Lien12ORCID,Vandenbroucke Roosmarijn E.12ORCID

Affiliation:

1. VIB Center for Inflammation Research VIB Ghent Belgium

2. Department of Biomedical Molecular Biology Ghent University Ghent Belgium

3. Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine Ghent University Merelbeke Belgium

4. Cellular Communication and Neurodegeneration Research Group, Department of Biology, Leuven Brain Institute KU Leuven Leuven Belgium

5. VIB Center for Brain & Disease Research Electrophysiology Expertise Unit Leuven Belgium

6. KU Leuven ‐ Department of Neurosciences Leuven Belgium

7. Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden

Abstract

AbstractThe gut microbiota represents a diverse and dynamic population of microorganisms that can influence the health of the host. Increasing evidence supports the role of the gut microbiota as a key player in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease (AD). Unfortunately, the mechanisms behind the interplay between gut pathogens and AD are still elusive. It is known that bacteria‐derived outer membrane vesicles (OMVs) act as natural carriers of virulence factors that are central players in the pathogenesis of the bacteria. Helicobacter pylori (H. pylori) is a common gastric pathogen and H. pylori infection has been associated with an increased risk to develop AD. Here, we are the first to shed light on the role of OMVs derived from H. pylori on the brain in healthy conditions and on disease pathology in the case of AD. Our results reveal that H. pylori OMVs can cross the biological barriers, eventually reaching the brain. Once in the brain, these OMVs are taken up by astrocytes, which induce activation of glial cells and neuronal dysfunction, ultimately leading to exacerbated amyloid‐β pathology and cognitive decline. Mechanistically, we identified a critical role for the complement component 3 (C3)‐C3a receptor (C3aR) signalling in mediating the interaction between astrocytes, microglia and neurons upon the presence of gut H. pylori OMVs. Taken together, our study reveals that H. pylori has a detrimental effect on brain functionality and accelerates AD development via OMVs and C3‐C3aR signalling.

Publisher

Wiley

Subject

Cell Biology,Histology

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3