Mitotic spindle disassembly in human cells relies on CRIPT having hierarchical redox signals

J Cell Sci. 2022 Sep 15;135(18):jcs259657. doi: 10.1242/jcs.259657. Epub 2022 Sep 23.

Abstract

Swift and complete spindle disassembly in late mitosis is essential for cell survival, yet how it happens is largely unknown in mammalian cells. Here we used real-time live cell microscopy and biochemical assays to show that the primordial dwarfism (PD)-related cysteine-rich protein CRIPT dictates the spindle disassembly in a redox-dependent manner in human cells. This previously reported cytoplasmic protein was found to have a confined nuclear localization with a nucleolar concentration during interphase but was distributed to spindles and underwent redox modifications to form disulfide bonds in CXXC pairs during mitosis. Then, it directly interacted with, and might transfer a redox response to, tubulin subunits via a putative redox exchange among cysteine residues to induce microtubule depolymerization. Expression of CRIPT proteins with mutations of these cysteine residues blocked spindle disassembly, generating two cell types with long-lasting metaphase spindles or spindle remnants. Live-cell recordings of a disease-relevant mutant (CRIPTC3Y) revealed that microtubule depolymerization at spindle ends during anaphase and the entire spindle dissolution during telophase might share a common CRIPT-bearing redox-controlled mechanism.

Keywords: CRIPT; Mitosis; Redox regulation; Spindle; Spindle disassembly.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adaptor Proteins, Signal Transducing / metabolism
  • Anaphase
  • Animals
  • Cysteine / metabolism
  • Disulfides / metabolism
  • Humans
  • Mammals / metabolism
  • Metaphase
  • Microtubules / metabolism
  • Mitosis
  • Oxidation-Reduction
  • Spindle Apparatus* / metabolism
  • Tubulin* / metabolism

Substances

  • Adaptor Proteins, Signal Transducing
  • CRIPT protein, human
  • Disulfides
  • Tubulin
  • Cysteine