Prion protein (PrPC) is a normal glycoprotein implicated in the pathogenesis of prion disorders, a group of fatal neurodegenerative conditions of humans and animals. A change in the conformation of PrPC to an aggregated, PrP-scrapie (PrPSc) form is believed to be the principal cause of neurotoxicity in these disorders. Recently, we demonstrated that PrPC plays a functional role in cellular iron uptake and transport, and selective deletion of PrPC in PrP knock-out (PrPKO) mice induces a state of systemic iron deficiency in these animals relative to wild-type (wt) controls. Specifically, PrPKO mice show impaired transport of orally introduced iron from the duodenum to the blood stream, and inefficient uptake of plasma iron by hematopoietic precursor cells and parenchymal cells of major organs. Together with our recent report demonstrating a phenotype of iron deficiency that correlates with PrPSc deposits in prion disease affected brains, these observations suggest that loss of normal function of PrPC due to aggregation to the PrPSc form may be responsible for brain iron dys- homeostasis in diseased brains. In this application, we will focus on the mechanism of iron modulation by PrPC, and hypothesize that PrPC is a novel iron uptake and transport protein, and modulates cellular iron metabolism either directly or by interacting with other iron transport protein(s). The proposed studies will test this hypothesis using two complementary approaches: 1) by evaluating the transport of different sources of iron that utilize distinct pathways of transport in the same tissue, and 2) by investigating the transport of same source of iron across tissues that utilize distinct pathways of uptake and transport. Mouse models that express no PrPC (PrPKO), wt levels (wt), and 10-fold higher than wt levels of PrPC (PrPOV) will be used for this analysis. This approach will allow identification of pathways of iron transport by PrPC, and the point where PrPC intersects with known pathways of iron metabolism.
Three specific aims are proposed to accomplish these goals.
In aim 1, the functional role of PrPC in brain iron metabolism will be evaluated in wt, PrPKO, and PrPOV mice, and the underlying mechanism will be investigated in neuroblastoma cells expressing normal and mutant forms of PrP defective in iron transport. In addition, the correlation between PrPC, PrPSc, and brain iron status will be assessed in scrapie infected wt and PrPOV mice.
In aim 2, the transport of different sources of iron will be checked in hematopoietic and reticuloendothelial cells isolated from wt, PrPKO, and PrPOV mice, and the underlying mechanism will be investigated in K562 erythroleukemia cells transfected with normal and mutant PrP forms.
In aim 3, the uptake and transport of different sources of iron across the intestinal epithelium will be checked in wt, PrPKO, and PrPOV mice, and compared with aim 2 above. Based on in vivo results, polarized Caco-2 cells transfected to express PrPC or mutant PrP forms will be used as models of absorptive enterocytes to understand the mechanism of iron transport by PrPC. Successful completion of these studies will uncover novel pathway(s) of iron modulation by PrPC, and improve our understanding of the mechanism(s) underlying brain iron imbalance and associated neurotoxicity in prion disorders.

Public Health Relevance

The long term goal of this application is to understand the role of prion protein (PrPC) in iron uptake, transport, and utilization. Preliminary data from my laboratory demonstrate that PrPC is involved in iron uptake and transport, and loss of this function contributes to brain iron dyshomeostasis in prion disorders. Successful completion of the proposed studies will improve our understanding of the functional role of PrPC in iron metabolism, and the underlying cause of brain iron imbalance in prion disorders.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK088390-04
Application #
8466314
Study Section
Cellular and Molecular Biology of Neurodegeneration Study Section (CMND)
Program Officer
Bishop, Terry Rogers
Project Start
2010-05-15
Project End
2014-04-30
Budget Start
2013-05-01
Budget End
2014-04-30
Support Year
4
Fiscal Year
2013
Total Cost
$311,229
Indirect Cost
$112,994
Name
Case Western Reserve University
Department
Pathology
Type
Schools of Medicine
DUNS #
077758407
City
Cleveland
State
OH
Country
United States
Zip Code
44106
Tripathi, Ajai K; Karmakar, Shilpita; Asthana, Abhishek et al. (2017) Transport of Non-Transferrin Bound Iron to the Brain: Implications for Alzheimer's Disease. J Alzheimers Dis 58:1109-1119
Tripathi, Ajai K; Singh, Neena (2016) Prion Protein-Hemin Interaction Upregulates Hemoglobin Synthesis: Implications for Cerebral Hemorrhage and Sporadic Creutzfeldt-Jakob Disease. J Alzheimers Dis 51:107-21
Haldar, Swati; Tripathi, Ajai; Qian, Juan et al. (2015) Prion protein promotes kidney iron uptake via its ferrireductase activity. J Biol Chem 290:5512-22
Tripathi, Ajai K; Haldar, Swati; Qian, Juan et al. (2015) Prion protein functions as a ferrireductase partner for ZIP14 and DMT1. Free Radic Biol Med 84:322-330
Singh, Neena; Asthana, Abhishek; Baksi, Shounak et al. (2015) The prion-ZIP connection: From cousins to partners in iron uptake. Prion 9:420-8
Singh, Neena (2014) The role of iron in prion disease and other neurodegenerative diseases. PLoS Pathog 10:e1004335
Singh, Ajay; Haldar, Swati; Horback, Katharine et al. (2013) Prion protein regulates iron transport by functioning as a ferrireductase. J Alzheimers Dis 35:541-52
Haldar, Swati; Beveridge, 'alim J; Wong, Joseph et al. (2013) A low-molecular-weight ferroxidase is increased in the CSF of sCJD cases: CSF ferroxidase and transferrin as diagnostic biomarkers for sCJD. Antioxid Redox Signal 19:1662-75
Singh, Ajay; Qing, Liuting; Kong, Qingzhong et al. (2012) Change in the characteristics of ferritin induces iron imbalance in prion disease affected brains. Neurobiol Dis 45:930-8
Singh, Ajay; Beveridge, Alim J; Singh, Neena (2011) Decreased CSF transferrin in sCJD: a potential pre-mortem diagnostic test for prion disorders. PLoS One 6:e16804

Showing the most recent 10 out of 13 publications