We previously described a zebrafish mutant, frascati (frs), with hypochromic anemia and erythroid maturation arrest. Using positional cloning, we identified mitoferrin1 (mfrn1, slc25a37) as the gene disrupted in frs mutants (Shaw GC, et al. 2006 Nature 440:96–100). Mfrn1, the principle iron-importer in the mitochondria, is essential for heme and iron-sulphur (Fe/S) cluster syntheses in erythroblasts, and is required for primitive and definitive erythropoiesis. The biochemistry for Mfrn1-mediated iron acquisition into the mitochondria of developing erythroblasts, however, is poorly understood. In order to identify Mfrn1-associated proteins involved in mitochondrial iron homeostasis, we employed the strategy of in vivo epitope-tagged affinity purification and mass spectrometry (MS). A “bait protein,” Biotag-mouse Mfrn1 (BT-Mfrn1), was engineered to affinity-purify the associated proteins. A series of control experiments were first conducted to demonstrate that the BT-Mfrn1 protein properly targeted to the mitochondria and functionally complemented the anemia in frs embryos. A two-dimensional Blue Native gel followed by SDS-PAGE and western blot showed Mfrn1 forms higher-order protein complexes with interacting proteins in the mitochondrial fraction. A stable mouse erythroleukemia (MEL) clone expressing BT-Mfrn1 protein was derived as a source for protein purification. Affinity-purified BT-Mfrn1 protein complexes were analyzed by MS in triplicate runs in comparison to control MEL cells. Abcb10, a GATA-1 induced ATP-binding cassette transporter highly expressed in hematopoietic tissues and involved in heme biosynthesis (

Shirihai OS, et al.
2000
EMBO J.
19
:
2492
–2502
), was found as one of the proteins which physically associated with Mfrn1 during MEL differentiation. The Abcb10:Mfrn1 interaction was confirmed by immunoprecipitation (IP) and western analysis with endogenously expressed proteins in MEL cells and proteins expressed by transient transfection in heterologous cells. Mfrn1 was previously shown to have longer protein half-life in differentiated MEL cells compared to undifferentiated cells (Paradkar PN, et al. submitted). To test our hypothesis that the Abcb10:Mfrn1 interaction may enhance Mfrn1 protein stability, a pulse-chase assay using 35S-methionine labeled cells was performed, followed by IP. Abcb10 was found to stabilize Mfrn1 protein in COS7 cells co-transfected with Abcb10 and Mfrn1 compared to control cells transfected with Mfrn1 alone. Similar results were obtained when total steady-state Mfrn1 protein was evaluated by western analysis comparing lysates from Abcb10:Mfrn1 cotransfected and Mfrn1 transfected cells. Our data suggest that Abcb10 physically interacts with Mfrn1 to enhance its stability and promote Mfrn1-dependent mitochondrial heme biogenesis.

Disclosures: No relevant conflicts of interest to declare.

Author notes

Corresponding author

Sign in via your Institution