Abstract

BackgroundNon-muscle myosin II (NM II) regulates a wide range of cellular functions, including neuronal differentiation, which requires precise spatio-temporal activation of Rho GTPases. The molecular mechanism underlying the NM II-mediated activation of Rho GTPases is poorly understood. The present study explored the possibility that NM II regulates neuronal differentiation, particularly morphological changes in growth cones and the distal axon, through guanine nucleotide exchange factors (GEFs) of the Dbl family.Principal FindingsNM II colocalized with GEFs, such as βPIX, kalirin and intersectin, in growth cones. Inactivation of NM II by blebbistatin (BBS) led to the increased formation of short and thick filopodial actin structures at the periphery of growth cones. In line with these observations, FRET analysis revealed enhanced Cdc42 activity in BBS-treated growth cones. BBS treatment also induced aberrant targeting of various GEFs to the distal axon where GEFs were seldom observed under physiological conditions. As a result, numerous protrusions and branches were generated on the shaft of the distal axon. The disruption of the NM II–GEF interactions by overexpression of the DH domains of βPIX or Tiam1, or by βPIX depletion with specific siRNAs inhibited growth cone formation and induced slender axons concomitant with multiple branches in cultured hippocampal neurons. Finally, stimulation with nerve growth factor induced transient dissociation of the NM II–GEF complex, which was closely correlated with the kinetics of Cdc42 and Rac1 activation.ConclusionOur results suggest that NM II maintains proper morphology of neuronal growth cones and the distal axon by regulating actin dynamics through the GEF–Rho GTPase signaling pathway.

Highlights

  • The neuronal growth cone is a specialized motile apparatus, which is located at the tips of extending neurites

  • Our results suggest that Non-muscle myosin II (NM II) maintains proper morphology of neuronal growth cones and the distal axon by regulating actin dynamics through the guanine nucleotide exchange factors (GEFs)–Rho GTPase signaling pathway

  • To examine whether these altered actin structures might be due to BBS-induced activation of Cdc42/Rac1, we performed fluorescence resonance energy transfer (FRET) analysis using Raichu-Cdc42 and Rac1 probes (Fig. 1B, S1A and S1B) and a GST-PBD pulldown assay using the p21-binding domain (PBD) of p21activated kinase 1 (PAK1) (Fig. 1C, 1D and S1C)

Read more

Summary

Introduction

The neuronal growth cone is a specialized motile apparatus, which is located at the tips of extending neurites (axons or dendrites). The assembly of new actin at the leading edge induces local protrusion of lamellipodia and filopodia, and promotes motility or turning responses of growth cones [1]. This assembled F-actin undergoes retrograde actin flow and depolymerization at the pointed end near the actin arc [2,3,4,5]. These processes are important in the modulation of growth cone behavior [6]. The present study explored the possibility that NM II regulates neuronal differentiation, morphological changes in growth cones and the distal axon, through guanine nucleotide exchange factors (GEFs) of the Dbl family

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