Sulfur assimilation is an evolutionarily conserved pathway that plays an essential role in cellular and metabolic processes including sulfation, amino-acid biosynthesis and organismal development. Disruption of the sulfur assimilation pathway has been shown to result in a number of pathologies in drosophila, mice and humans. Here, we report that loss of a key enzymatic component of the pathway, bisphosphate 3'-nucleotidase BPNT1, in mice, both whole animal and intestine specific, leads to iron-deficiency anemia. The long-term goal of this research is to define the molecular mechanisms by which Bpnt1 loss-of-function, and subsequent accumulation of its substrate 3'-phosphoadenosine 5'-phosphate (PAP), leads to dysfunction of the iron regulatory machinery. Analysis of mutant enterocytes from intestine-specific Bpnt1 mutants demonstrate modulation of PAP influences levels of key iron homeostasis factors involved in dietary iron reduction, import and transport, that in part mimic those reported for the loss of hypoxic-induced transcription factor, HIF-2?. Our general hypothesis is that PAP accumulation leads to alterations in the iron-regulatory pathway, leading to iron deficiency anemia. Specifically, I hypothesize that PAP accumulation inhibits HIF- 2??dependent and independent signaling. To test this hypothesis, I will employ biochemical, molecular, proteomic, and animal model approaches to obtain a mechanistic understanding of the molecular consequences of PAP accumulation. Then, I will apply these mechanistic insights to translational human genetics studies. I propose three aims: 1) Elucidate the molecular mechanisms by which PAP accumulation perturbs iron metabolism, 2) Characterize the role of Bpnt1-HIF-2? mediated iron homeostasis in mice and 3) Determine the role of Bpnt1 in human iron metabolism. The proposed studies are expected to define a new genetic basis for iron-deficiency anemia, delineate a molecular approach for rescuing the pathophysiology and provide an unanticipated link between nucleotide hydrolysis in the sulfur assimilation pathway and iron homeostasis. !

Public Health Relevance

Regulation of iron homeostasis is an essential homeostatic process that can be perturbed in numerous pathologic states. Here, we link a key component of the sulfur assimilation pathway, bisphosphate 3'- nucleotidase (BPNT1), to broad modulation of iron metabolism in part due to inhibition of hypoxia-inducible factor 2-?. Reduction of BPNT1's substrate, phosphoadenosine phosphate (PAP), through introduction of a hypomorphic mutation in 3'-phosphoadenosine 5-phosphosulfate synthase 2 gene (Papss2, the enzyme responsible for PAP production) rescues the iron deficiency phenotype, and thus implicates the sulfur assimilation pathway as a potential opportunity for new therapeutic development. !

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Individual Predoctoral NRSA for M.D./Ph.D. Fellowships (ADAMHA) (F30)
Project #
1F30HL143826-01
Application #
9608622
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Chang, Henry
Project Start
2018-09-01
Project End
2023-08-31
Budget Start
2018-09-01
Budget End
2019-08-30
Support Year
1
Fiscal Year
2018
Total Cost
Indirect Cost
Name
Vanderbilt University Medical Center
Department
Type
Schools of Medicine
DUNS #
965717143
City
Nashville
State
TN
Country
United States
Zip Code
37240
Hudson, Benjamin H; Hale, Andrew T; Irving, Ryan P et al. (2018) Modulation of intestinal sulfur assimilation metabolism regulates iron homeostasis. Proc Natl Acad Sci U S A 115:3000-3005