Cytochrome P450 1b1 (Cyp1b1) participates in embryogenesis and is expressed in endothelia, fibroblasts and adipocytes. Genes involved in liver fatty acid metabolism are remarkably affected by deletion of Cyp1b1 in mice, despite only minimal expression in hepatocytes. Two gene clusters, distinguished by responses to dietary fat, are both suppressed by Cyp1b1 deletion and regulated by PPAR? Preliminary data that indicates that Cyp1b1 participates in the generation of endogenous PPAR? ligands will be explored further. Cyp1b1 deletion also suppresses diet-induced obesity and non-alcoholic hepatic steatosis. The increased liver fatty acid metabolism may be associated with decreased oxidative stress, as indicated by improved insulin sensitivity. Cyp1b1 may, therefore, be an unexpected contributor to Type 2 diabetes. Many dietary flavanoids are potent inhibitors of Cyp1b1. Select Cyp1b1 inhibitors, including natural compounds, may have therapeutic value for diabetes. Human Cyp1b1 suppression also enhances liver adenomas, consistent with a role in developmental processes. The proposed research compares the liver gene expression, metabolism responses and adiposity changes in WT and Cyp1b1-ko mice administered low fat/high carbohydrate or high fat/low carbohydrate diets. The goal is to determine how hepatocytes can be indirectly affected by Cyp1b1 metabolism in other cell types, particularly the endothelia of liver sinusoids and adipose tissue. The expression of Cyp1b1 in non-parenchymal liver cells and in fetal liver will be characterized. In order to define the site and timing of Cypb1 intervention, we have introduced a Floxed Cyp1b1 allele into mice. This will ultimately provide the means for controlled Cyp1b1 deletions in mice. We will use this defining set of liver and adipose responses to determine the effectiveness of Cyp1b1 deletions specifically targeted to endothelia by Tie2-Cre. We have shown that Cyp1b1 deletion affects the functions of endothelial cells and suppresses angiogenesis in vivo. Cyp1b1 deletion may cause these adult changes through active participation of Cyp1b1 in the fetal liver development that imprints adult liver regulation. Cre-targeting of Floxed Cyp1b1 will be activated through a tamoxifen-dependent promoter at selected times during development, in order to test whether this early expression contributes to the general deletion effects. Metabolomic studies using 2D-NMR and mass spectrometry analyses on, respectively, liver and serum extracts will focus on establishing that Cyp1b1 deletion increases mitochondrial fatty acid oxidation and glycogen synthesis, while lowering oxidative stress. The roles of PPAR? and PPAR? loss in these liver gene changes of Cyp1b1-ko mice will be tested by comparisons to effects of deleting these PPAR genes in mouse liver. Serum markers applicable to clinical studies will be used in probing these liver changes.

Public Health Relevance

Loss of Cytochrome P450 1B1 (CYP1B1) in humans is linked to Glaucoma and Liver tumors. CYP1B1 variants are common in humans. We find that CYP1B1 losses can counter obesity and improve insulin sensitivity. These studies address how CYP1B1 deletion remarkably increases liver fat oxidation and glycogen synthesis through suppression of PPAR?activity. The pathway offers therapeutic possibilities, including through dietary compounds that bind to CYP1B1.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK090249-02
Application #
8429375
Study Section
Xenobiotic and Nutrient Disposition and Action Study Section (XNDA)
Program Officer
Serrano, Jose
Project Start
2012-04-01
Project End
2017-03-31
Budget Start
2013-04-01
Budget End
2014-03-31
Support Year
2
Fiscal Year
2013
Total Cost
$312,489
Indirect Cost
$102,601
Name
University of Wisconsin Madison
Department
Anatomy/Cell Biology
Type
Schools of Medicine
DUNS #
161202122
City
Madison
State
WI
Country
United States
Zip Code
53715
Jefcoate, Colin R; Lee, Jinwoo (2018) Cholesterol signaling in single cells: lessons from STAR and sm-FISH. J Mol Endocrinol 60:R213-R235
Lee, Jinwoo; Yamazaki, Takeshi; Dong, Hui et al. (2017) A single cell level measurement of StAR expression and activity in adrenal cells. Mol Cell Endocrinol 441:22-30
Maguire, Meghan; Bushkofsky, Justin R; Larsen, Michele Campaigne et al. (2017) Diet-dependent retinoid effects on liver gene expression include stellate and inflammation markers and parallel effects of the nuclear repressor Shp. J Nutr Biochem 47:63-74
Maguire, Meghan; Larsen, Michele Campaigne; Foong, Yee Hoon et al. (2017) Cyp1b1 deletion and retinol deficiency coordinately suppress mouse liver lipogenic genes and hepcidin expression during post-natal development. Mol Cell Endocrinol 454:50-68
Lee, Jinwoo; Tong, Tiegang; Duan, Haichuan et al. (2016) Regulation of StAR by the N-terminal Domain and Coinduction of SIK1 and TIS11b/Znf36l1 in Single Cells. Front Endocrinol (Lausanne) 7:107
Lee, Jinwoo; Foong, Yee Hoon; Musaitif, Ibrahim et al. (2016) Analysis of specific RNA in cultured cells through quantitative integration of q-PCR and N-SIM single cell FISH images: Application to hormonal stimulation of StAR transcription. Mol Cell Endocrinol 429:93-105
Rondelli, Catherine M; Larsen, Michele Campaigne; N'jai, Alhaji et al. (2016) PAHs Target Hematopoietic Linages in Bone Marrow through Cyp1b1 Primarily in Mesenchymal Stromal Cells but Not AhR: A Reconstituted In Vitro Model. Stem Cells Int 2016:1753491
Dinu, Daniela; Chu, Chun; Veith, Alex et al. (2016) Mechanistic role of cytochrome P450 (CYP)1B1 in oxygen-mediated toxicity in pulmonary cells: A novel target for prevention of hyperoxic lung injury. Biochem Biophys Res Commun 476:346-351
Bushkofsky, Justin R; Maguire, Meghan; Larsen, Michele Campaigne et al. (2016) Cyp1b1 affects external control of mouse hepatocytes, fatty acid homeostasis and signaling involving HNF4? and PPAR?. Arch Biochem Biophys 597:30-47
Larsen, Michele Campaigne; Bushkofsky, Justin R; Gorman, Tyler et al. (2015) Cytochrome P450 1B1: An unexpected modulator of liver fatty acid homeostasis. Arch Biochem Biophys 571:21-39

Showing the most recent 10 out of 13 publications