F0F1 ATP synthase regulates extracellular calcium influx in human neutrophils by interacting with Cav2.3 and modulates neutrophil accumulation in the lipopolysaccharide-challenged lung

Cell Commun Signal. 2020 Feb 4;18(1):19. doi: 10.1186/s12964-020-0515-3.

Abstract

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 Cav2.3 (R-type) is the primary type of VGCC expressed in human neutrophils. Research on the F-ATPase/Cav2.3 functional complex indicated that it can regulate extracellular Ca2+ 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.

Keywords: F0F1 ATP synthase; Inflammation; Lung; Neutrophil; Voltage-gated calcium channel.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Acute Lung Injury / complications
  • Acute Lung Injury / metabolism
  • Acute Lung Injury / pathology
  • Adult
  • Amino Acid Sequence
  • Animals
  • Calcium / metabolism*
  • Calcium Channels, R-Type / metabolism*
  • Cation Transport Proteins / metabolism*
  • Cell Membrane / metabolism
  • Extracellular Signal-Regulated MAP Kinases / metabolism
  • Extracellular Space / metabolism*
  • Humans
  • Lipopolysaccharides
  • Lung / metabolism*
  • Lung / pathology
  • Mice
  • Models, Biological
  • Neutrophil Activation
  • Neutrophils / metabolism*
  • Peptides / chemistry
  • Peptides / metabolism
  • Phosphorylation
  • Pneumonia / complications
  • Pneumonia / metabolism
  • Pneumonia / pathology
  • Protein Binding
  • Protein Isoforms / metabolism
  • Protein Subunits / metabolism
  • Proton-Translocating ATPases / metabolism*
  • Reactive Oxygen Species / metabolism

Substances

  • CACNA1E protein, human
  • Calcium Channels, R-Type
  • Cation Transport Proteins
  • Lipopolysaccharides
  • Peptides
  • Protein Isoforms
  • Protein Subunits
  • Reactive Oxygen Species
  • Extracellular Signal-Regulated MAP Kinases
  • Proton-Translocating ATPases
  • Calcium