A succinate/SUCNR1-brush cell defense program in the tracheal epithelium

Author:

Perniss Alexander12ORCID,Boonen Brett34ORCID,Tonack Sarah5,Thiel Moritz12,Poharkar Krupali12,Alnouri Mohamad Wessam5,Keshavarz Maryam12ORCID,Papadakis Tamara12,Wiegand Silke12,Pfeil Uwe12ORCID,Richter Katrin6ORCID,Althaus Mike7ORCID,Oberwinkler Johannes8ORCID,Schütz Burkhard9ORCID,Boehm Ulrich10ORCID,Offermanns Stefan25ORCID,Leinders-Zufall Trese3ORCID,Zufall Frank3ORCID,Kummer Wolfgang12ORCID

Affiliation:

1. Institute of Anatomy and Cell Biology, German Center for Lung Research, Justus Liebig University Giessen; Giessen, Germany.

2. Excellence Cluster The Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany.

3. Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany.

4. Laboratory of Ion Channel Research, VIB Center for Brain and Disease, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium.

5. Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.

6. Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, Justus-Liebig-University, Giessen, Germany.

7. Physiology Group, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany.

8. Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, Marburg, Germany.

9. Institute of Anatomy and Cell Biology, Philipps University Marburg, Marburg, Germany.

10. Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University, Homburg, Germany.

Abstract

Host-derived succinate accumulates in the airways during bacterial infection. Here, we show that luminal succinate activates murine tracheal brush (tuft) cells through a signaling cascade involving the succinate receptor 1 (SUCNR1), phospholipase Cβ2, and the cation channel transient receptor potential channel subfamily M member 5 (TRPM5). Stimulated brush cells then trigger a long-range Ca 2+ wave spreading radially over the tracheal epithelium through a sequential signaling process. First, brush cells release acetylcholine, which excites nearby cells via muscarinic acetylcholine receptors. From there, the Ca 2+ wave propagates through gap junction signaling, reaching also distant ciliated and secretory cells. These effector cells translate activation into enhanced ciliary activity and Cl secretion, which are synergistic in boosting mucociliary clearance, the major innate defense mechanism of the airways. Our data establish tracheal brush cells as a central hub in triggering a global epithelial defense program in response to a danger-associated metabolite.

Publisher

American Association for the Advancement of Science (AAAS)

Subject

Multidisciplinary

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