Retinoic Acid Receptor β Loss in Hepatocytes Increases Steatosis and Elevates the Integrated Stress Response in Alcohol-Associated Liver Disease.

TitleRetinoic Acid Receptor β Loss in Hepatocytes Increases Steatosis and Elevates the Integrated Stress Response in Alcohol-Associated Liver Disease.
Publication TypeJournal Article
Year of Publication2023
AuthorsMelis M, Trasino SE, Tang X-H, Rappa A, Zhang T, Qin L, Gudas LJ
JournalInt J Mol Sci
Volume24
Issue15
Date Published2023 Jul 27
ISSN1422-0067
KeywordsAnimals, Ethanol, Fatty Liver, Hepatocytes, Humans, Liver, Liver Diseases, Alcoholic, Mice, Mice, Knockout, Receptors, Retinoic Acid, Vitamin A
Abstract

In alcohol-associated liver disease (ALD), hepatic reductions in vitamin A and perturbations in vitamin A metabolism are common. However, the roles that the vitamin A receptors, termed retinoic acid receptors (RARs), may have in preventing the pathophysiology of ALD remains unclear. Our prior data indicate that a RARβ agonist limits the pathology of alcohol-related liver disease. Thus, we generated liver-specific AlbCre-RARβ knockout (BKO) mice and compared them to wild type (WT) mice in an early ALD model. Both strains showed similar blood ethanol concentrations and ETOH-metabolizing enzymes. However, the livers of pair-fed-BKO and ETOH-BKO mice developed higher levels of steatosis and triglycerides than pair-fed-WT and ETOH-WT mice. The increased hepatic steatosis observed in the pair-fed-BKO and ETOH-BKO mice was associated with higher lipid synthesis/trafficking transcripts and lower beta-oxidation transcripts. ETOH-BKO mice also exhibited a higher integrated stress response (ISR) signature, including higher transcript and protein levels of ATF4 and its target, 4-EBP1. In human hepatocytes (HepG2) that lack RARβ (RARβ-KO), ETOH treatments resulted in greater reactive oxygen species compared to their parental cells. Notably, even without ETOH, ATF4 and 4-EBP1 protein levels were higher in the RARβ-KO cells than in their parental cells. These 4-EBP1 increases were greatly attenuated in cultured ATF4-deficient and RARβ/ATF4-deficient HepG2, suggesting that RARβ is a crucial negative regulator of 4-EBP1 through ATF4 in cultured hepatocytes. Here, we identify RARβ as a negative regulator of lipid metabolism and cellular stress in ALD.

DOI10.3390/ijms241512035
Alternate JournalInt J Mol Sci
PubMed ID37569418
PubMed Central IDPMC10418449
Grant ListR21 AA027637 / AA / NIAAA NIH HHS / United States
SC2 GM127206 / GM / NIGMS NIH HHS / United States
5SC2GM127206-0 / GM / NIGMS NIH HHS / United States