Abcb10 physically interacts with mitoferrin-1 (Slc25a37) to enhance its stability and function in the erythroid mitochondria.

Bibliographic Collection: 
MOCA Reference, APE
Publication Type: Journal Article
Authors: Chen, Wen; Paradkar, Prasad N; Li, Liangtao; Pierce, Eric L; Langer, Nathaniel B; Takahashi-Makise, Naoko; Hyde, Brigham B; Shirihai, Orian S; Ward, Diane M; Kaplan, Jerry; Paw, Barry H
Year of Publication: 2009
Journal: Proc Natl Acad Sci U S A
Volume: 106
Issue: 38
Pagination: 16263-8
Date Published: 2009 Sep 22
Publication Language: eng
ISSN: 1091-6490
Keywords: Animals, Binding Sites, Biological Transport, Blotting, Western, Cell Differentiation, Cell Line, Tumor, Cercopithecus aethiops, COS Cells, Immunoprecipitation, Iron, Leukemia, Erythroblastic, Acute, Membrane Transport Proteins, Mice, Microscopy, Confocal, Mitochondria, Multidrug Resistance-Associated Proteins, Protein Binding, Protein Stability, Transfection
Abstract:

Mitoferrin-1 (Mfrn1; Slc25a37), a member of the solute carrier family localized in the mitochondrial inner membrane, functions as an essential iron importer for the synthesis of mitochondrial heme and iron-sulfur clusters in erythroblasts. The biochemistry of Mfrn1-mediated iron transport into the mitochondria, however, is poorly understood. Here, we used the strategy of in vivo epitope-tagging affinity purification and mass spectrometry to investigate Mfrn1-mediated mitochondrial iron homeostasis. Abcb10, a mitochondrial inner membrane ATP-binding cassette transporter highly induced during erythroid maturation in hematopoietic tissues, was found as one key protein that physically interacts with Mfrn1 during mouse erythroleukemia (MEL) cell differentiation. Mfrn1 was shown previously to have a longer protein half-life in differentiated MEL cells compared with undifferentiated cells. In this study, Abcb10 was found to enhance the stabilization of Mfrn1 protein in MEL cells and transfected heterologous COS7 cells. In undifferentiated MEL cells, cotransfected Abcb10 specifically interacts with Mfrn1 to enhance its protein stability and promote Mfrn1-dependent mitochondrial iron importation. The structural stabilization of the Mfrn1-Abcb10 complex demonstrates a previously uncharacterized function for Abcb10 in mitochondria. Furthermore, the binding domain of Mfrn1-Abcb10 interaction maps to the N terminus of Mfrn1. These results suggest the tight regulation of mitochondrial iron acquisition and heme synthesis in erythroblasts is mediated by both transcriptional and posttranslational mechanisms, whereby the high level of Mfrn1 is stabilized by oligomeric protein complexes.

DOI: 10.1073/pnas.0904519106
Alternate Journal: Proc. Natl. Acad. Sci. U.S.A.