4.7 Article

Role of hepcidin in oxidative stress and cell death of cultured mouse renal collecting duct cells: protection against iron and sensitization to cadmium

Journal

ARCHIVES OF TOXICOLOGY
Volume 95, Issue 8, Pages 2719-2735

Publisher

SPRINGER HEIDELBERG
DOI: 10.1007/s00204-021-03106-z

Keywords

DFO; Metallothionein; Ca(V)3; 1; DMT1

Categories

Funding

  1. Deutsche Forschungsgemeinschaft [DFG TH345]
  2. BMBF [BMBF 01DN16039]
  3. Centre for Biomedical Training and Research (ZBAF) of the University of Witten/Herdecke
  4. Projekt DEAL

Ask authors/readers for more resources

Hepcidin plays a critical role in protecting against Fe2+-induced cell death in the kidney, while exacerbating oxidative stress and cell death caused by Cd2+. The differential effects of hepcidin on Fe2+ and Cd2+ toxicity are mediated by its impact on catalase activity, ROS levels, and downstream cell processes.
The liver hormone hepcidin regulates systemic iron homeostasis. Hepcidin is also expressed by the kidney, but exclusively in distal nephron segments. Several studies suggest hepcidin protects against kidney damage involving Fe2+ overload. The nephrotoxic non-essential metal ion Cd2+ can displace Fe2+ from cellular biomolecules, causing oxidative stress and cell death. The role of hepcidin in Fe2+ and Cd2+ toxicity was assessed in mouse renal cortical [mCCD(cl.1)] and inner medullary [mIMCD(3)] collecting duct cell lines. Cells were exposed to equipotent Cd2+ (0.5-5 mu mol/l) and/or Fe2+ (50-100 mu mol/l) for 4-24 h. Hepcidin (Hamp1) was transiently silenced by RNAi or overexpressed by plasmid transfection. Hepcidin or catalase expression were evaluated by RT-PCR, qPCR, immunoblotting or immunofluorescence microscopy, and cell fate by MTT, apoptosis and necrosis assays. Reactive oxygen species (ROS) were detected using CellROX (TM) Green and catalase activity by fluorometry. Hepcidin upregulation protected against Fe2+-induced mIMCD(3) cell death by increasing catalase activity and reducing ROS, but exacerbated Cd2+-induced catalase dysfunction, increasing ROS and cell death. Opposite effects were observed with Hamp1 siRNA. Similar to Hamp1 silencing, increased intracellular Fe2+ prevented Cd2+ damage, ROS formation and catalase disruption whereas chelation of intracellular Fe2+ with desferrioxamine augmented Cd2+ damage, corresponding to hepcidin upregulation. Comparable effects were observed in mCCD(cl.1) cells, indicating equivalent functions of renal hepcidin in different collecting duct segments. In conclusion, hepcidin likely binds Fe2+, but not Cd2+. Because Fe2+ and Cd2+ compete for functional binding sites in proteins, hepcidin affects their free metal ion pools and differentially impacts downstream processes and cell fate.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available