Deficiency of fumarylacetoacetate hydrolase (FAH) causes tyrosinemia type I (hepatorenal tyrosinemia, HT 1) in humans and is characterized by severe liver dysfunction, renal tubular damage and liver cancer. In the murine model of HT 1 hepatocytes stably expressing Fah have a strong selective growth advantage. Fah mutant mice are therefore uniquely suited for the study of the biology of integrating gene therapy vectors in the liver. We have succeeded in the synthesis of 2 potent small inhibitors of Fah, which work in mice in vivo. Because of the powerful selection found in genetic Fah deficiency, pharmacological inhibition of Fah may be a useful strategy for the in vivo selection of genetically modified hepatocytes. FAH deficient humans and mice have a high propensity to the development of hepatocarcinoma. Our recent studies demonstrate that oxidative stress induced by fumarylacetoacetate (FAA) creates generalized apoptosis resistance in Fah mutant hepatocytes. Apoptosis resistance may therefore be an important early factor in the evolution of HCC in HT 1. The overall theme of this application is the utilization of the tyrosine catabolic pathway for in vivo metabolic selection of genetically modified epithelial cells in the liver. The feasibility and biology of various strategies for hepatic gene therapy will be explored.
In aim 1, we will test in vivo gene targeting in the liver using AAV vectors.
In aim 2, we will use small molecule inhibitors of Fah to select for genetically modified hepatocytes in vivo.
In aim 3, the pathophysiology of hepatic cancer in HT 1 and strategies for its chemoprevention will be studied. ? ?

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
2R01DK048252-12
Application #
6876809
Study Section
Special Emphasis Panel (ZRG1-GNM (02))
Program Officer
Mckeon, Catherine T
Project Start
1993-12-01
Project End
2008-11-30
Budget Start
2005-02-15
Budget End
2005-11-30
Support Year
12
Fiscal Year
2005
Total Cost
$342,200
Indirect Cost
Name
Oregon Health and Science University
Department
Genetics
Type
Schools of Medicine
DUNS #
096997515
City
Portland
State
OR
Country
United States
Zip Code
97239
Paulk, Nicole K; Pekrun, Katja; Zhu, Erhua et al. (2018) Bioengineered AAV Capsids with Combined High Human Liver Transduction In Vivo and Unique Humoral Seroreactivity. Mol Ther 26:289-303
Nygaard, Sean; Barzel, Adi; Haft, Annelise et al. (2016) A universal system to select gene-modified hepatocytes in vivo. Sci Transl Med 8:342ra79
Hickey, Raymond D; Mao, Shennen A; Glorioso, Jaime et al. (2014) Fumarylacetoacetate hydrolase deficient pigs are a novel large animal model of metabolic liver disease. Stem Cell Res 13:144-53
Yin, Hao; Xue, Wen; Chen, Sidi et al. (2014) Genome editing with Cas9 in adult mice corrects a disease mutation and phenotype. Nat Biotechnol 32:551-3
Harding, Cary O; Winn, Shelley R; Gibson, K Michael et al. (2014) Pharmacologic inhibition of L-tyrosine degradation ameliorates cerebral dopamine deficiency in murine phenylketonuria (PKU). J Inherit Metab Dis 37:735-43
Lisowski, Leszek; Dane, Allison P; Chu, Kirk et al. (2014) Selection and evaluation of clinically relevant AAV variants in a xenograft liver model. Nature 506:382-6
Dorrell, Craig; Tarlow, Branden; Wang, Yuhan et al. (2014) The organoid-initiating cells in mouse pancreas and liver are phenotypically and functionally similar. Stem Cell Res 13:275-83
Grompe, Markus; Strom, Stephen (2013) Mice with human livers. Gastroenterology 145:1209-14
Gramignoli, Roberto; Tahan, Veysel; Dorko, Kenneth et al. (2013) New potential cell source for hepatocyte transplantation: discarded livers from metabolic disease liver transplants. Stem Cell Res 11:563-73
Taylor, A M; Preston, A J; Paulk, N K et al. (2012) Ochronosis in a murine model of alkaptonuria is synonymous to that in the human condition. Osteoarthritis Cartilage 20:880-6

Showing the most recent 10 out of 38 publications