Cholestatic liver disease has many causes, including autoimmune destruction of bile duct epithelial cells, genetic disorders, and physical obstruction of the bile duct. Currently, the only approved treatment for many types of cholestatic liver disease is usodeoxycholic acid, which is only efficacious in a small fraction of patients. Although, several studies indicate an important role for inflammation in the development of hepatocellular injury and fibrosis during cholestasis, glucocorticoids and other potent anti-inflammatory drugs that target classical inflammatory cytokines are only modestly effective at ameliorating liver disease in patients and are often associated with severe side-effects. The failure of current anti-inflammatory treatment regimens to treat cholestatic liver disease underscores the importance of elucidating the mechanism(s) involved in the initiation of hepatic inflammation in this type of liver disease. Our preliminary studies demonstrate that pathological concentrations of the bile acid, taurocholic acid (TCA), stimulate hepatocytes to produce the cytokines, IL-23 and macrophage inflammatory protein-2 (MIP-2). Activation of Akt by TCA is required for upregulation of IL-23 and MIP-2, whereas activation of p38 by TCA inhibits upregulation of these cytokines. IL-23 contributes to hepatic inflammation by stimulating Th17 cells to produce IL-17A. This cytokine inhibits TCA-mediated activation of p38 in hepatocytes leading to enhanced production of MIP-2 and IL-23. Our studies demonstrate further that activation of lysophosphatidic acid receptor 1 (LPAR1) is required for upregulation of IL-23 and MIP-2 in TCA-treated hepatocytes, and that LPAR1 is activated independent of its ligand, LPA. This indicates that LPAR1 is activated by a novel mechanism that may involve direct activation of LPAR1 by TCA. Based upon these studies, the central hypothesis of this application is that during cholestasis, elevated concentrations of bile acids activate LPAR1 which increases expression of IL-23 and MIP-2, a process which is enhanced by IL-17A-mediated inhibition of p38. The studies in this proposal aim to test the central hypothesis by: (1) determining the mechanism by bile acids increase expression of IL-23 and MIP-2 in hepatocytes, (2) determining the mechanism by which IL-17A inhibits activation of p38 by TCA, and (3) determine the mechanism by which p38 negatively regulates IL-23 and MIP-2 induction by TCA. Collectively, these studies will increase our current knowledge of regulation of hepatic inflammation during cholestasis, which will allow for the identification of new drug targets to tret cholestatic liver disease.

Public Health Relevance

Currently, there are limited therapies for patients with cholestatic liver disease, and therefore, many patients die from this disease. The inflammatory system plays a key role in the development of liver disease during cholestasis. The studies in this proposal will determine the mechanism by which inflammation is initiated in the liver during cholestasis, in order to identify ways to dampen the inflammatory response in patients with this disease.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK073566-09
Application #
9305051
Study Section
Hepatobiliary Pathophysiology Study Section (HBPP)
Program Officer
Burgess-Beusse, Bonnie L
Project Start
2007-05-01
Project End
2019-07-31
Budget Start
2017-08-01
Budget End
2019-07-31
Support Year
9
Fiscal Year
2017
Total Cost
Indirect Cost
Name
Michigan State University
Department
Pharmacology
Type
Schools of Medicine
DUNS #
193247145
City
East Lansing
State
MI
Country
United States
Zip Code
48824
Copple, Bryan L; Li, Tiangang (2016) Pharmacology of bile acid receptors: Evolution of bile acids from simple detergents to complex signaling molecules. Pharmacol Res 104:9-21
Roth, Katherine J; Copple, Bryan L (2015) Role of Hypoxia-Inducible Factors in the Development of Liver Fibrosis. Cell Mol Gastroenterol Hepatol 1:589-597
Mochizuki, Akie; Pace, Aaron; Rockwell, Cheryl E et al. (2014) Hepatic stellate cells orchestrate clearance of necrotic cells in a hypoxia-inducible factor-1?-dependent manner by modulating macrophage phenotype in mice. J Immunol 192:3847-3857
Yang, Min; Ramachandran, Anup; Yan, Hui-Min et al. (2014) Osteopontin is an initial mediator of inflammation and liver injury during obstructive cholestasis after bile duct ligation in mice. Toxicol Lett 224:186-95
Ni, Hong-Min; Woolbright, Benjamin L; Williams, Jessica et al. (2014) Nrf2 promotes the development of fibrosis and tumorigenesis in mice with defective hepatic autophagy. J Hepatol 61:617-25
Ni, Hong-Min; Bhakta, Amar; Wang, Shaogui et al. (2014) Role of hypoxia inducing factor-1? in alcohol-induced autophagy, steatosis and liver injury in mice. PLoS One 9:e115849
O'Brien, Kate M; Allen, Katryn M; Rockwell, Cheryl E et al. (2013) IL-17A synergistically enhances bile acid-induced inflammation during obstructive cholestasis. Am J Pathol 183:1498-1507
Kassel, Karen M; Sullivan, Bradley P; Cui, Wei et al. (2012) Therapeutic administration of the direct thrombin inhibitor argatroban reduces hepatic inflammation in mice with established fatty liver disease. Am J Pathol 181:1287-95
Copple, Bryan L; Kaska, Sophia; Wentling, Callie (2012) Hypoxia-inducible factor activation in myeloid cells contributes to the development of liver fibrosis in cholestatic mice. J Pharmacol Exp Ther 341:307-16
Zhang, Youcai; Hong, Ji-Young; Rockwell, Cheryl E et al. (2012) Effect of bile duct ligation on bile acid composition in mouse serum and liver. Liver Int 32:58-69

Showing the most recent 10 out of 19 publications