1. Academic Validation
  2. Hepatic metal ion transporter ZIP8 regulates manganese homeostasis and manganese-dependent enzyme activity

Hepatic metal ion transporter ZIP8 regulates manganese homeostasis and manganese-dependent enzyme activity

  • J Clin Invest. 2017 Jun 1;127(6):2407-2417. doi: 10.1172/JCI90896.
Wen Lin 1 David R Vann 2 Paschalis-Thomas Doulias 3 Tao Wang 1 Gavin Landesberg 4 Xueli Li 5 Emanuela Ricciotti 6 Rosario Scalia 4 Miao He 5 7 Nicholas J Hand 8 Daniel J Rader 1 6 8
Affiliations

Affiliations

  • 1 Department of Medicine, Perelman School of Medicine, and.
  • 2 Department of Earth and Environmental Science, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
  • 3 Children's Hospital of Philadelphia Research Institute and Department of Pharmacology, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, USA.
  • 4 Department of Physiology, Temple University, Philadelphia, Pennsylvania, USA.
  • 5 Palmieri Metabolic Disease Laboratory, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.
  • 6 Institute for Translational Medicine and Therapeutics.
  • 7 Department of Pathology and Laboratory Medicine, Perelman School of Medicine, and.
  • 8 Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Abstract

Genetic variants at the solute carrier family 39 member 8 (SLC39A8) gene locus are associated with the regulation of whole-blood manganese (Mn) and multiple physiological traits. SLC39A8 encodes ZIP8, a divalent metal ion transporter best known for zinc transport. Here, we hypothesized that ZIP8 regulates Mn homeostasis and Mn-dependent enzymes to influence metabolism. We generated Slc39a8-inducible global-knockout (ZIP8-iKO) and liver-specific-knockout (ZIP8-LSKO) mice and observed markedly decreased Mn levels in multiple organs and whole blood of both mouse models. By contrast, liver-specific overexpression of human ZIP8 (adeno-associated virus-ZIP8 [AAV-ZIP8]) resulted in increased tissue and whole blood Mn levels. ZIP8 expression was localized to the hepatocyte canalicular membrane, and bile Mn levels were increased in ZIP8-LSKO and decreased in AAV-ZIP8 mice. ZIP8-LSKO mice also displayed decreased liver and kidney activity of the Mn-dependent enzyme Arginase. Both ZIP8-iKO and ZIP8-LSKO mice had defective protein N-glycosylation, and humans homozygous for the minor allele at the lead SLC39A8 variant showed hypogalactosylation, consistent with decreased activity of another Mn-dependent Enzyme, β-1,4-galactosyltransferase. In summary, hepatic ZIP8 reclaims Mn from bile and regulates whole-body Mn homeostasis, thereby modulating the activity of Mn-dependent enzymes. This work provides a mechanistic basis for the association of SLC39A8 with whole-blood Mn, potentially linking SLC39A8 variants with other physiological traits.

Figures