Login / Signup

An AAGAB-to-CCDC32 handover mechanism controls the assembly of the AP2 adaptor complex.

Chun WanHarrison PuscherYan OuyangJingyi WuYuan TianSuzhao LiQian YinJingshi Shen
Published in: Proceedings of the National Academy of Sciences of the United States of America (2024)
Vesicular transport relies on multimeric trafficking complexes to capture cargo and drive vesicle budding and fusion. Faithful assembly of the trafficking complexes is essential to their functions but remains largely unexplored. Assembly of AP2 adaptor, a heterotetrameric protein complex regulating clathrin-mediated endocytosis, is assisted by the chaperone AAGAB. Here, we found that AAGAB initiates AP2 assembly by stabilizing its α and σ2 subunits, but the AAGAB:α:σ2 complex cannot recruit additional AP2 subunits. We identified CCDC32 as another chaperone regulating AP2 assembly. CCDC32 recognizes the AAGAB:α:σ2 complex, and its binding leads to the formation of an α:σ2:CCDC32 ternary complex. The α:σ2:CCDC32 complex serves as a template that sequentially recruits the µ2 and β2 subunits of AP2 to complete AP2 assembly, accompanied by CCDC32 release. The AP2-regulating function of CCDC32 is disrupted by a disease-causing mutation. These findings demonstrate that AP2 is assembled by a handover mechanism switching from AAGAB-based initiation complexes to CCDC32-based template complexes. A similar mechanism may govern the assembly of other trafficking complexes exhibiting the same configuration as AP2.
Keyphrases
  • transcription factor
  • small molecule
  • gold nanoparticles
  • mass spectrometry
  • heat shock protein
  • heat shock
  • binding protein
  • heat stress
  • simultaneous determination