Abstract

SLC39A8 encodes ZIP8, a divalent metal ion transporter. Mutations in the SLC39A8 gene are associated with congenital disorder of glycosylation type II and Leigh syndrome. Notably, affected patients with both disorders exhibited severe manganese (Mn) deficiency. The cellular function of human SLC39A8 (hSLC39A8) and the mechanisms by which mutations in this protein lead to human diseases are unclear. Herein, we show that hSLC39A8 mediates 54Mn uptake by the cells, and its expression is regulated by Mn. While expression of wild-type hSLC39A8 increased 54Mn uptake activity, disease-associated mutations abrogated the ability of the transporter to mediate Mn uptake into the cells, thereby providing a causal link to severe Mn deficiency. All mutants failed to localize on the cell surface and were retained within the endoplasmic reticulum. Interestingly, expression of hSLC39A8 mutants of both CDG type II and Leigh syndrome reduced mitochondrial 54Mn levels and activity of Mn-dependent mitochondrial superoxide dismutase MnSOD, and in turn increased oxidative stress. The expression of wild-type hSLC39A8, but not the disease-associated mutants, promoted mitochondrial functions. Moreover, loss of function analyses further corroborate hSLC39A8’s critical role in mediating Mn uptake and mitochondrial function. Our results provide a potential pathogenic mechanism of diseases that are associated with hSLC39A8 mutations.

Highlights

  • Manganese (Mn) is an essential nutrient that acts as a cofactor for a variety of enzymes involved in numerous cellular physiological processes[1]

  • The expression of hSLC39A8-WT potentiates mitochondrial functions, while hSLC39A8 mutants are less capable of doing so and, rather, increase oxidative stress. Both loss of function and gain of function analysis suggest that hSLC39A8 plays a critical role in Mn uptake and mitochondrial function, which likely contribute to the pathogenesis of diseases that are associated with hSLC39A8 mutations

  • These results indicate that hSLC39A8 mediates Mn uptake into the cells with a very high affinity, and cadmium has the strongest inhibitory effect on Mn uptake

Read more

Summary

Introduction

Manganese (Mn) is an essential nutrient that acts as a cofactor for a variety of enzymes involved in numerous cellular physiological processes[1]. Affected patients with both disorders exhibited severe Mn deficiency in the blood[8,9,11] These observations raised the possibility that human SLC39A8 (hSLC39A8) is a Mn transporter and the disease-associated hSLC39A8 mutations dysregulate Mn homeostasis. This hypothesis has not been tested directly. The expression of hSLC39A8-WT potentiates mitochondrial functions, while hSLC39A8 mutants are less capable of doing so and, rather, increase oxidative stress Both loss of function and gain of function analysis suggest that hSLC39A8 plays a critical role in Mn uptake and mitochondrial function, which likely contribute to the pathogenesis of diseases that are associated with hSLC39A8 mutations

Objectives
Methods
Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.