دورية أكاديمية

Apo- and holo-transferrin differentially interact with hephaestin and ferroportin in a novel mechanism of cellular iron release regulation

التفاصيل البيبلوغرافية
العنوان: Apo- and holo-transferrin differentially interact with hephaestin and ferroportin in a novel mechanism of cellular iron release regulation
المؤلفون: Baringer, Stephanie L., Palsa, Kondaiah, Spiegelman, Vladimir S., Simpson, Ian A., Connor, James R.
المساهمون: National Institute of Health
المصدر: Journal of Biomedical Science ; volume 30, issue 1 ; ISSN 1423-0127
بيانات النشر: Springer Science and Business Media LLC
سنة النشر: 2023
مصطلحات موضوعية: Pharmacology (medical), Biochemistry (medical), Cell Biology, Clinical Biochemistry, Molecular Biology, General Medicine, Endocrinology, Diabetes and Metabolism
الوصف: Background Apo- (iron free) and holo- (iron bound) transferrin (Tf) participate in precise regulation of brain iron uptake at endothelial cells of the blood–brain barrier. Apo-Tf indicates an iron-deficient environment and stimulates iron release, while holo-Tf indicates an iron sufficient environment and suppresses additional iron release. Free iron is exported through ferroportin, with hephaestin as an aid to the process. Until now, the molecular mechanisms of apo- and holo-Tf influence on iron release was largely unknown. Methods Here we use a variety of cell culture techniques, including co-immunoprecipitation and proximity ligation assay, in iPSC-derived endothelial cells and HEK 293 cells to investigate the mechanism by which apo- and holo-Tf influence cellular iron release. Given the established role of hepcidin in regulating cellular iron release, we further explored the relationship of hepcidin to transferrin in this model. Results We demonstrate that holo-Tf induces the internalization of ferroportin through the established ferroportin degradation pathway. Furthermore, holo-Tf directly interacts with ferroportin, whereas apo-Tf directly interacts with hephaestin. Only pathophysiological levels of hepcidin disrupt the interaction between holo-Tf and ferroportin, but similar hepcidin levels are unable to interfere with the interaction between apo-Tf and hephaestin. The disruption of the holo-Tf and ferroportin interaction by hepcidin is due to hepcidin’s ability to more rapidly internalize ferroportin compared to holo-Tf. Conclusions These novel findings provide a molecular mechanism for apo- and holo-Tf regulation of iron release from endothelial cells. They further demonstrate how hepcidin impacts these protein–protein interactions, and offer a model for how holo-Tf and hepcidin cooperate to suppress iron release. These results expand on our previous reports on mechanisms mediating regulation of brain iron uptake to provide a more thorough understanding of the regulatory mechanisms mediating ...
نوع الوثيقة: article in journal/newspaper
اللغة: English
DOI: 10.1186/s12929-023-00934-2
DOI: 10.1186/s12929-023-00934-2.pdf
DOI: 10.1186/s12929-023-00934-2/fulltext.html
الإتاحة: https://doi.org/10.1186/s12929-023-00934-2Test
حقوق: https://creativecommons.org/licenses/by/4.0Test ; https://creativecommons.org/licenses/by/4.0Test
رقم الانضمام: edsbas.EA306B55
قاعدة البيانات: BASE