Abstract

FBXL5 (F-box and leucine-rich repeat protein 5) is the F-box protein subunit of, and therefore responsible for substrate recognition by, the SCF(FBXL5) ubiquitin-ligase complex, which targets iron regulatory protein 2 (IRP2) for proteasomal degradation. IRP2 plays a central role in the maintenance of cellular iron homeostasis in mammals through posttranscriptional regulation of proteins that contribute to control of the intracellular iron concentration. The FBXL5-IRP2 axis is integral to control of iron metabolism in vivo, given that mice lacking FBXL5 die during early embryogenesis as a result of unrestrained IRP2 activity and oxidative stress attributable to excessive iron accumulation. Despite its pivotal role in the control of iron homeostasis, however, little is known of the upstream regulation of FBXL5 activity. We now show that FBXL5 undergoes constitutive ubiquitin-dependent degradation at the steady state. With the use of a proteomics approach to the discovery of proteins that regulate the stability of FBXL5, we identified the large HECT-type ubiquitin ligase HERC2 (HECT and RLD domain containing E3 ubiquitin protein ligase 2) as an FBXL5-associated protein. Inhibition of the HERC2-FBXL5 interaction or depletion of endogenous HERC2 by RNA interference resulted in the stabilization of FBXL5 and a consequent increase in its abundance. Such accumulation of FBXL5 in turn led to a decrease in the intracellular content of ferrous iron. Our results thus suggest that HERC2 regulates the basal turnover of FBXL5, and that this ubiquitin-dependent degradation pathway contributes to the control of mammalian iron metabolism.

Highlights

  • FBXL5, the F-box protein subunit of an SCF-type ubiquitin-ligase complex, is a regulator of mammalian iron homeostasis

  • With the use of a proteomics approach to the discovery of proteins that regulate the stability of FBXL5, we identified the large HECT-type ubiquitin ligase HERC2 (HECT and RLD domain containing E3 ubiquitin protein ligase 2) as an FBXL5-associated protein

  • To investigate the requirement for the hemerythrin-like domain in the basal degradation of FBXL5, we measured the stability of the FBXL5(⌬1–161) mutant in HEK293T cells cultured in the presence of cycloheximide and in the absence or presence of MG132

Read more

Summary

Background

FBXL5, the F-box protein subunit of an SCF-type ubiquitin-ligase complex, is a regulator of mammalian iron homeostasis. Our results suggest that HERC2 regulates the basal turnover of FBXL5, and that this ubiquitin-dependent degradation pathway contributes to the control of mammalian iron metabolism. Under iron-limiting conditions, FBXL5 itself is targeted for ubiquitylation and degradation by the proteasome, resulting in the stabilization of apo-IRP1 and IRP2 and their interaction with IREs. Indicative of the pivotal role of FBXL5 in the control of mammalian iron homeostasis, we have recently shown that mice lacking FBXL5 die early in embryogenesis as a result of deleterious effects of the accumulation of free iron [13]. The hemerythrin-like domain of FBXL5 is considered to be pivotal to the iron-dependent stability of the protein It remains unclear how FBXL5 is regulated at the steady state, . Our results indicate that HERC2 regulates the basal turnover of FBXL5 and thereby modulates iron metabolism

EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
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