4.6 Article

JIP3 Activates Kinesin-1 Motility to Promote Axon Elongation

Journal

JOURNAL OF BIOLOGICAL CHEMISTRY
Volume 290, Issue 25, Pages 15512-15525

Publisher

AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC
DOI: 10.1074/jbc.M115.651885

Keywords

axon; kinesin; molecular motor; neurite outgrowth; regeneration; JIP3; KHC; motility

Funding

  1. National Institutes of Health, NINDS [R01 NS082446]
  2. National Science Foundation [MCB-1121287]
  3. Direct For Biological Sciences
  4. Div Of Molecular and Cellular Bioscience [1121287] Funding Source: National Science Foundation

Ask authors/readers for more resources

Background: Axon growth and regeneration depend on kinesin-1-dependent transport. Results: JIP3 binding to KHC promotes kinesin-1 motility along microtubules and is essential for axon elongation and regeneration. Conclusion: JIP3 regulation of kinesin-1 motility is critical for axon elongation and regeneration. Significance: Regulation of intracellular transport is important for proper neuronal development. Kinesin-1 is a molecular motor responsible for cargo transport along microtubules and plays critical roles in polarized cells, such as neurons. Kinesin-1 can function as a dimer of two kinesin heavy chains (KHC), which harbor the motor domain, or as a tetramer in combination with two accessory light chains (KLC). To ensure proper cargo distribution, kinesin-1 activity is precisely regulated. Both KLC and KHC subunits bind cargoes or regulatory proteins to engage the motor for movement along microtubules. We previously showed that the scaffolding protein JIP3 interacts directly with KHC in addition to its interaction with KLC and positively regulates dimeric KHC motility. Here we determined the stoichiometry of JIP3-KHC complexes and observed approximately four JIP3 molecules binding per KHC dimer. We then determined whether JIP3 activates tetrameric kinesin-1 motility. Using an in vitro motility assay, we show that JIP3 binding to KLC engages kinesin-1 with microtubules and that JIP3 binding to KHC promotes kinesin-1 motility along microtubules. We tested the in vivo relevance of these findings using axon elongation as a model for kinesin-1-dependent cellular function. We demonstrate that JIP3 binding to KHC, but not KLC, is essential for axon elongation in hippocampal neurons as well as axon regeneration in sensory neurons. These findings reveal that JIP3 regulation of kinesin-1 motility is critical for axon elongation and regeneration.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available