Cargando…

F0F1 ATP synthase regulates extracellular calcium influx in human neutrophils by interacting with Ca(v)2.3 and modulates neutrophil accumulation in the lipopolysaccharide-challenged lung

BACKGROUND: Neutrophils form the first line of innate host defense against invading microorganisms. We previously showed that F0F1 ATP synthase (F-ATPase), which is widely known as mitochondrial respiratory chain complex V, is expressed in the plasma membrane of human neutrophils and is involved in...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhu, Baoyi, Feng, Zhengfu, Guo, Yan, Zhang, Tian, Mai, Ai, Kang, Zhanfang, Weijen, Ting, Wang, Dai, Yin, Dazhong, Zhu, Dongxing, Gao, Jun
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7001235/
https://www.ncbi.nlm.nih.gov/pubmed/32019549
http://dx.doi.org/10.1186/s12964-020-0515-3
_version_ 1783494198513631232
author Zhu, Baoyi
Feng, Zhengfu
Guo, Yan
Zhang, Tian
Mai, Ai
Kang, Zhanfang
Weijen, Ting
Wang, Dai
Yin, Dazhong
Zhu, Dongxing
Gao, Jun
author_facet Zhu, Baoyi
Feng, Zhengfu
Guo, Yan
Zhang, Tian
Mai, Ai
Kang, Zhanfang
Weijen, Ting
Wang, Dai
Yin, Dazhong
Zhu, Dongxing
Gao, Jun
author_sort Zhu, Baoyi
collection PubMed
description BACKGROUND: Neutrophils form the first line of innate host defense against invading microorganisms. We previously showed that F0F1 ATP synthase (F-ATPase), which is widely known as mitochondrial respiratory chain complex V, is expressed in the plasma membrane of human neutrophils and is involved in regulating cell migration. Whether F-ATPase performs cellular functions through other pathways remains unknown. METHODS: Blue native polyacrylamide gel electrophoresis followed by nano-ESI-LC MS/MS identification and bioinformatic analysis were used to identify protein complexes containing F-ATPase. Then, the identified protein complexes containing F-ATPase were verified by immunoblotting, immunofluorescence colocalization, immunoprecipitation, real-time RT-PCR and agarose gel electrophoresis. Immunoblotting, flow cytometry and a LPS-induced mouse lung injury model were used to assess the effects of the F-ATPase-containing protein complex in vitro and in vivo. RESULTS: We found that the voltage-gated calcium channel (VGCC) α2δ-1 subunit is a binding partner of cell surface F-ATPase in human neutrophils. Further investigation found that the physical connection between the two proteins may exist between the F1 part (α and β subunits) of F-ATPase and the α2 part of VGCC α2δ-1. Real-time RT-PCR and PCR analyses showed that Ca(v)2.3 (R-type) is the primary type of VGCC expressed in human neutrophils. Research on the F-ATPase/Ca(v)2.3 functional complex indicated that it can regulate extracellular Ca(2+) influx, thereby modulating ERK1/2 phosphorylation and reactive oxygen species production, which are typical features of neutrophil activation. In addition, the inhibition of F-ATPase can reduce neutrophil accumulation in the lungs of mice that were intratracheally instilled with lipopolysaccharide, suggesting that the inhibition of F-ATPase may prevent neutrophilic inflammation-induced tissue damage. CONCLUSIONS: In this study, we identified a mechanism by which neutrophil activity is modulated, with simultaneous regulation of neutrophil-mediated pulmonary damage. These results show that surface F-ATPase of neutrophils is a potential innate immune therapeutic target. GRAPHICAL ABSTRACT: [Image: see text]
format Online
Article
Text
id pubmed-7001235
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-70012352020-02-10 F0F1 ATP synthase regulates extracellular calcium influx in human neutrophils by interacting with Ca(v)2.3 and modulates neutrophil accumulation in the lipopolysaccharide-challenged lung Zhu, Baoyi Feng, Zhengfu Guo, Yan Zhang, Tian Mai, Ai Kang, Zhanfang Weijen, Ting Wang, Dai Yin, Dazhong Zhu, Dongxing Gao, Jun Cell Commun Signal Research BACKGROUND: Neutrophils form the first line of innate host defense against invading microorganisms. We previously showed that F0F1 ATP synthase (F-ATPase), which is widely known as mitochondrial respiratory chain complex V, is expressed in the plasma membrane of human neutrophils and is involved in regulating cell migration. Whether F-ATPase performs cellular functions through other pathways remains unknown. METHODS: Blue native polyacrylamide gel electrophoresis followed by nano-ESI-LC MS/MS identification and bioinformatic analysis were used to identify protein complexes containing F-ATPase. Then, the identified protein complexes containing F-ATPase were verified by immunoblotting, immunofluorescence colocalization, immunoprecipitation, real-time RT-PCR and agarose gel electrophoresis. Immunoblotting, flow cytometry and a LPS-induced mouse lung injury model were used to assess the effects of the F-ATPase-containing protein complex in vitro and in vivo. RESULTS: We found that the voltage-gated calcium channel (VGCC) α2δ-1 subunit is a binding partner of cell surface F-ATPase in human neutrophils. Further investigation found that the physical connection between the two proteins may exist between the F1 part (α and β subunits) of F-ATPase and the α2 part of VGCC α2δ-1. Real-time RT-PCR and PCR analyses showed that Ca(v)2.3 (R-type) is the primary type of VGCC expressed in human neutrophils. Research on the F-ATPase/Ca(v)2.3 functional complex indicated that it can regulate extracellular Ca(2+) influx, thereby modulating ERK1/2 phosphorylation and reactive oxygen species production, which are typical features of neutrophil activation. In addition, the inhibition of F-ATPase can reduce neutrophil accumulation in the lungs of mice that were intratracheally instilled with lipopolysaccharide, suggesting that the inhibition of F-ATPase may prevent neutrophilic inflammation-induced tissue damage. CONCLUSIONS: In this study, we identified a mechanism by which neutrophil activity is modulated, with simultaneous regulation of neutrophil-mediated pulmonary damage. These results show that surface F-ATPase of neutrophils is a potential innate immune therapeutic target. GRAPHICAL ABSTRACT: [Image: see text] BioMed Central 2020-02-04 /pmc/articles/PMC7001235/ /pubmed/32019549 http://dx.doi.org/10.1186/s12964-020-0515-3 Text en © The Author(s). 2020 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Zhu, Baoyi
Feng, Zhengfu
Guo, Yan
Zhang, Tian
Mai, Ai
Kang, Zhanfang
Weijen, Ting
Wang, Dai
Yin, Dazhong
Zhu, Dongxing
Gao, Jun
F0F1 ATP synthase regulates extracellular calcium influx in human neutrophils by interacting with Ca(v)2.3 and modulates neutrophil accumulation in the lipopolysaccharide-challenged lung
title F0F1 ATP synthase regulates extracellular calcium influx in human neutrophils by interacting with Ca(v)2.3 and modulates neutrophil accumulation in the lipopolysaccharide-challenged lung
title_full F0F1 ATP synthase regulates extracellular calcium influx in human neutrophils by interacting with Ca(v)2.3 and modulates neutrophil accumulation in the lipopolysaccharide-challenged lung
title_fullStr F0F1 ATP synthase regulates extracellular calcium influx in human neutrophils by interacting with Ca(v)2.3 and modulates neutrophil accumulation in the lipopolysaccharide-challenged lung
title_full_unstemmed F0F1 ATP synthase regulates extracellular calcium influx in human neutrophils by interacting with Ca(v)2.3 and modulates neutrophil accumulation in the lipopolysaccharide-challenged lung
title_short F0F1 ATP synthase regulates extracellular calcium influx in human neutrophils by interacting with Ca(v)2.3 and modulates neutrophil accumulation in the lipopolysaccharide-challenged lung
title_sort f0f1 atp synthase regulates extracellular calcium influx in human neutrophils by interacting with ca(v)2.3 and modulates neutrophil accumulation in the lipopolysaccharide-challenged lung
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7001235/
https://www.ncbi.nlm.nih.gov/pubmed/32019549
http://dx.doi.org/10.1186/s12964-020-0515-3
work_keys_str_mv AT zhubaoyi f0f1atpsynthaseregulatesextracellularcalciuminfluxinhumanneutrophilsbyinteractingwithcav23andmodulatesneutrophilaccumulationinthelipopolysaccharidechallengedlung
AT fengzhengfu f0f1atpsynthaseregulatesextracellularcalciuminfluxinhumanneutrophilsbyinteractingwithcav23andmodulatesneutrophilaccumulationinthelipopolysaccharidechallengedlung
AT guoyan f0f1atpsynthaseregulatesextracellularcalciuminfluxinhumanneutrophilsbyinteractingwithcav23andmodulatesneutrophilaccumulationinthelipopolysaccharidechallengedlung
AT zhangtian f0f1atpsynthaseregulatesextracellularcalciuminfluxinhumanneutrophilsbyinteractingwithcav23andmodulatesneutrophilaccumulationinthelipopolysaccharidechallengedlung
AT maiai f0f1atpsynthaseregulatesextracellularcalciuminfluxinhumanneutrophilsbyinteractingwithcav23andmodulatesneutrophilaccumulationinthelipopolysaccharidechallengedlung
AT kangzhanfang f0f1atpsynthaseregulatesextracellularcalciuminfluxinhumanneutrophilsbyinteractingwithcav23andmodulatesneutrophilaccumulationinthelipopolysaccharidechallengedlung
AT weijenting f0f1atpsynthaseregulatesextracellularcalciuminfluxinhumanneutrophilsbyinteractingwithcav23andmodulatesneutrophilaccumulationinthelipopolysaccharidechallengedlung
AT wangdai f0f1atpsynthaseregulatesextracellularcalciuminfluxinhumanneutrophilsbyinteractingwithcav23andmodulatesneutrophilaccumulationinthelipopolysaccharidechallengedlung
AT yindazhong f0f1atpsynthaseregulatesextracellularcalciuminfluxinhumanneutrophilsbyinteractingwithcav23andmodulatesneutrophilaccumulationinthelipopolysaccharidechallengedlung
AT zhudongxing f0f1atpsynthaseregulatesextracellularcalciuminfluxinhumanneutrophilsbyinteractingwithcav23andmodulatesneutrophilaccumulationinthelipopolysaccharidechallengedlung
AT gaojun f0f1atpsynthaseregulatesextracellularcalciuminfluxinhumanneutrophilsbyinteractingwithcav23andmodulatesneutrophilaccumulationinthelipopolysaccharidechallengedlung