Abstract

Iron efflux from mammalian cells is supported by the synergistic actions of the ferrous iron efflux transporter, ferroportin (Fpn) and a multicopper ferroxidase, that is, hephaestin (Heph), ceruloplasmin (Cp) or both. The two proteins stabilize Fpn in the plasma membrane and catalyze extracellular Fe3+ release. The membrane stabilization of Fpn is also stimulated by its interaction with a 22-amino acid synthetic peptide based on a short sequence in the extracellular E2 domain of the amyloid precursor protein (APP). However, whether APP family members interact with Fpn in vivo is unclear. Here, using cyan fluorescent protein (CFP)-tagged Fpn in conjunction with yellow fluorescent protein (YFP) fusions of Heph and APP family members APP, APLP1, and APLP2 in HEK293T cells we used fluorescence and surface biotinylation to quantify Fpn membrane occupancy and also measured 59Fe efflux. We demonstrate that Fpn and Heph co-localize, and FRET analysis indicated that the two proteins form an iron-efflux complex. In contrast, none of the full-length, cellular APP proteins exhibited Fpn co-localization or FRET. Moreover, iron supplementation increased surface expression of the iron-efflux complex, and copper depletion knocked down Heph activity and decreased Fpn membrane localization. Whereas cellular APP species had no effects on Fpn and Heph localization, addition of soluble E2 elements derived from APP and APLP2, but not APLP1, increased Fpn membrane occupancy. We conclude that a ferroportin-targeting sequence, (K/R)EWEE, present in APP and APLP2, but not APLP1, helps modulate Fpn-dependent iron efflux in the presence of an active multicopper ferroxidase.

Highlights

  • Iron efflux from mammalian cells is supported by the synergistic actions of the ferrous iron efflux transporter, ferroportin (Fpn) and a multicopper ferroxidase, that is, hephaestin (Heph), ceruloplasmin (Cp) or both

  • Whereas cellular amyloid precursor protein (APP) species had no effects on Fpn and Heph localization, addition of soluble E2 elements derived from APP and APLP2, but not APLP1, increased Fpn membrane occupancy

  • Two proteins are broadly recognized to contribute to the membrane occupancy of Fpn: 1) the multicopper ferroxidase, hephaestin (Heph), a type Ia membrane protein expressed by enterocytes that appears to interact with Fpn and likely retards Fpn’s normal retrograde internalization (4 –7); and 2) hepcidin, a peptide hormone secreted primarily by the liver, that binds to Fpn, triggering Fpn’s ubiquitination and degradation (8 –13)

Read more

Summary

Introduction

Iron efflux from mammalian cells is supported by the synergistic actions of the ferrous iron efflux transporter, ferroportin (Fpn) and a multicopper ferroxidase, that is, hephaestin (Heph), ceruloplasmin (Cp) or both. Two proteins are broadly recognized to contribute to the membrane occupancy of Fpn: 1) the multicopper ferroxidase, hephaestin (Heph), a type Ia membrane protein expressed by enterocytes (among other tissues) that appears to interact with Fpn and likely retards Fpn’s normal retrograde internalization (4 –7); and 2) hepcidin, a peptide hormone secreted primarily by the liver, that binds to Fpn, triggering Fpn’s ubiquitination and degradation (8 –13) This dual regulatory pattern appears to obtain irrespective of the cell type involved, e.g. intestinal enterocytes or the macrophages that recycle red cell iron where circulating ceruloplasmin (sCp) complements the activity of hephaestin [1, 2, 14, 15].

Objectives
Results
Conclusion
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