Abstract

The vacuolar H(+)-ATPase (v-ATPase) complex is instrumental in establishing and maintaining acidification of some cellular compartments, thereby ensuring their functionality. Recently it has been proposed that the transmembrane V0 sector of v-ATPase and its a-subunits promote membrane fusion in the endocytic and exocytic pathways independent of their acidification functions. Here, we tested if such a proton-pumping independent role of v-ATPase also applies to phagosome-lysosome fusion. Surprisingly, endo(lyso)somes in mouse embryonic fibroblasts lacking the V0 a3 subunit of the v-ATPase acidified normally, and endosome and lysosome marker proteins were recruited to phagosomes with similar kinetics in the presence or absence of the a3 subunit. Further experiments used macrophages with a knockdown of v-ATPase accessory protein 2 (ATP6AP2) expression, resulting in a strongly reduced level of the V0 sector of the v-ATPase. However, acidification appeared undisturbed, and fusion between latex bead-containing phagosomes and lysosomes, as analyzed by electron microscopy, was even slightly enhanced, as was killing of non-pathogenic bacteria by V0 mutant macrophages. Pharmacologically neutralized lysosome pH did not affect maturation of phagosomes in mouse embryonic cells or macrophages. Finally, locking the two large parts of the v-ATPase complex together by the drug saliphenylhalamide A did not inhibit in vitro and in cellulo fusion of phagosomes with lysosomes. Hence, our data do not suggest a fusion-promoting role of the v-ATPase in the formation of phagolysosomes.

Highlights

  • The vacuolar Hϩ-ATPase complex is thought to contribute to membrane fusion

  • Beads that have not been taken up were stained with anti-IgG antibodies coupled to Alexa Fluor 594 for 1 min, and the samples were processed for immunofluorescence staining of the marker proteins early endosome antigen 1 (EEA1) or LAMP-2, respectively

  • V0 Subunit a3 Deficiency Despite Normal pH of the Lysosome— In a genetic-based approach to analyze the role of the v-ATPase subunit a3 in membrane fusion, we used cells from Tcirg1oc/oc mice, which are homozygous for the osteosclerosis mutation and naturally lack the transmembrane a3-subunit of the v-ATPase [24]

Read more

Summary

Background

The vacuolar Hϩ-ATPase complex is thought to contribute to membrane fusion. Results: v-ATPase complex knock-out experiments in mice revealed that its absence does not affect phagosome-lysosome fusion. In the past few years evidence has accumulated that the transmembrane V0 part of the v-ATPase, without participation of the V1 sector, can play active and critical roles in membrane fusion along the endocytic and exocytic pathways independent of its proton-translocating activities Such evidence stems from experiments on synaptic vesicle exocytosis in Drosophila [13], secretion in Caenorhabditis [14], osteoclast fusion [15], and vesicle fusion in zebrafish microglia cells after ingestion of neuron-derived apoptotic bodies [16]. As tools we used the Tcirg1oc/oc mouse line and conditionally deleted Atp6ap knock-out mice Cells derived from these mice either lacked the v-ATPase subunit a3 or showed an almost complete absence of the v-ATPase V0 sector yet allowed phagosome-lysosome as well as endosome-lysosome fusion to progress. All animal experiments were conducted in agreement with local guidelines for the use of animals and their care

Experimental Procedures
Results
Discussion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call