Abstract

The formation of adhesion complexes is the rate-limiting step for collagen phagocytosis by fibroblasts, but the role of Ca(2+) and the potential interactions of actin-binding proteins in regulating collagen phagocytosis are not well defined. We found that the binding of collagen beads to fibroblasts was temporally and spatially associated with actin assembly at nascent phagosomes, which was absent in gelsolin null cells. Analysis of tryptic digests isolated from gelsolin immunoprecipitates indicated that non-muscle (NM) myosin IIA may bind to gelsolin. Immunostaining and immunoprecipitation showed that gelsolin and NM myosin IIA associated at collagen adhesion sites. Gelsolin and NM myosin IIA were both required for collagen binding and internalization. Collagen binding to cells initiated a prolonged increase of [Ca(2+)](i), which was absent in cells null for gelsolin or NM myosin IIA. Collagen bead-induced increases of [Ca(2+)](i) were associated with phosphorylation of the myosin light chain, which was dependent on gelsolin. NM myosin IIA filament assembly, which was dependent on myosin light chain phosphorylation and increased [Ca(2+)](i), also required gelsolin. Ionomycin-induced increases of [Ca(2+)](i) overcame the block of myosin filament assembly in gelsolin null cells. We conclude that gelsolin and NM myosin IIA interact at collagen adhesion sites to enable NM myosin IIA filament assembly and localized, Ca(2+)-dependent remodeling of actin at the nascent phagosome and that these steps are required for collagen phagocytosis.

Highlights

  • Fibroblasts can remodel the extracellular matrix by phagocytosis, but how the adhesion mechanisms are regulated is not well understood

  • We conclude that gelsolin and NM myosin IIA interact at collagen adhesion sites to enable NM myosin IIA filament assembly and localized, Ca2؉-dependent remodeling of actin at the nascent phagosome and that these steps are required for collagen phagocytosis

  • Effect of Gelsolin and NMIIA on Collagen Bead Binding and Phagocytosis—Gelsolin null cells exhibit much more prominent actin filaments than WT cells [40], which may be important in regulating collagen phagocytosis

Read more

Summary

Introduction

Fibroblasts can remodel the extracellular matrix by phagocytosis, but how the adhesion mechanisms are regulated is not well understood. Results: Non-muscle myosin IIA provides a docking site for the actin-severing protein to localize at collagen adhesion sites and remodel actin filaments there. We found that the binding of collagen beads to fibroblasts was temporally and spatially associated with actin assembly at nascent phagosomes, which was absent in gelsolin null cells. Collagen bead-induced increases of [Ca2؉]i were associated with phosphorylation of the myosin light chain, which was dependent on gelsolin. NM myosin IIA filament assembly, which was dependent on myosin light chain phosphorylation and increased [Ca2؉]i, required gelsolin. Ionomycin-induced increases of [Ca2؉]i overcame the block of myosin filament assembly in gelsolin null cells. We conclude that gelsolin and NM myosin IIA interact at collagen adhesion sites to enable NM myosin IIA filament assembly and localized, Ca2؉-dependent remodeling of actin at the nascent phagosome and that these steps are required for collagen phagocytosis

Methods
Results
Conclusion
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