Rhythmicity of intestinal IgA responses confers oscillatory commensal microbiota mutualism

Author:

Penny Hugo A.12ORCID,Domingues Rita G.12ORCID,Krauss Maria Z.12ORCID,Melo-Gonzalez Felipe12ORCID,Lawson Melissa A. E.12ORCID,Dickson Suzanna123ORCID,Parkinson James12ORCID,Hurry Madeleine12ORCID,Purse Catherine12ORCID,Jegham Emna12ORCID,Godinho-Silva Cristina4ORCID,Rendas Miguel4ORCID,Veiga-Fernandes Henrique4ORCID,Bechtold David A.3ORCID,Grencis Richard K.125ORCID,Toellner Kai-Michael6ORCID,Waisman Ari7ORCID,Swann Jonathan R.8ORCID,Gibbs Julie E.123ORCID,Hepworth Matthew R.12ORCID

Affiliation:

1. Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9PL, UK.

2. School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK.

3. Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK.

4. Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon 1400-038, Portugal.

5. Wellcome Centre for Cell Matrix Research, University of Manchester, Manchester M13 9PL, UK.

6. Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham B15 2TT, UK.

7. Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.

8. School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK.

Abstract

Interactions between the mammalian host and commensal microbiota are enforced through a range of immune responses that confer metabolic benefits and promote tissue health and homeostasis. Immunoglobulin A (IgA) responses directly determine the composition of commensal species that colonize the intestinal tract but require substantial metabolic resources to fuel antibody production by tissue-resident plasma cells. Here, we demonstrate that IgA responses are subject to diurnal regulation over the course of a circadian day. Specifically, the magnitude of IgA secretion, as well as the transcriptome of intestinal IgA+plasma cells, was found to exhibit rhythmicity. Oscillatory IgA responses were found to be entrained by time of feeding and were also found to be in part coordinated by the plasma cell–intrinsic circadian clock via deletion of the master clock geneArntl. Moreover, reciprocal interactions between the host and microbiota dictated oscillatory dynamics among the commensal microbial community and its associated transcriptional and metabolic activity in an IgA-dependent manner. Together, our findings suggest that circadian networks comprising intestinal IgA, diet, and the microbiota converge to align circadian biology in the intestinal tract and to ensure host-microbial mutualism.

Publisher

American Association for the Advancement of Science (AAAS)

Subject

General Medicine,Immunology

Cited by 28 articles. 订阅此论文施引文献 订阅此论文施引文献,注册后可以免费订阅5篇论文的施引文献,订阅后可以查看论文全部施引文献

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