As erythroid progenitors differentiate into precursors and finally mature red blood cells, they induce the expression of lineage-specific genes, and their proliferation declines until cell cycle exit. CUL-4A encodes a core subunit of a ubiquitin ligase that targets proteins for ubiquitin-mediated degradation, and CUL-4A haploinsufficient mice display hematopoietic dysregulation with fewer multipotential and erythroid committed progenitors (3-fold and 5-fold, respectively). In this study, stress induced by 5-fluorouracil or phenylhydrazine reveals a delay in the ability of CUL-4A +/− mice to recover erythroid progenitors and precursors and to reestablish normal hematocrits. Conversely, over-expression of CUL-4A in a growth factor-dependent, proerythroblast cell line increases proliferation 34% (tritiated thymidine incorporation) and the proportion of cells in S-phase 5%. When these cells are induced to terminally differentiate, endogenous CUL-4A protein expression declines 3.6-fold. Its enforced expression interferes with erythrocyte maturation (beta-globin induction) and cell cycle exit, and instead promotes proliferation. Furthermore, p27 accumulates during erythroid terminal differentiation, but CUL-4A enforced expression increases p27 protein turnover nearly 7-fold and attenuates its accumulation. CUL-4A and p27 proteins co-immunoprecipitate, indicating that a Cul-4A ubiquitin ligase targets p27 for degradation. These findings suggest that a Cul-4A ubiquitin ligase positively regulates proliferation by targeting p27 for degradation and that CUL-4A down-regulation during erythroid terminal differentiation allows p27 to accumulate and signal cell cycle exit.

Author notes

Corresponding author

Sign in via your Institution