Skip to main content

Nitro-fatty acids protect against steatosis and fibrosis during development of nonalcoholic fatty liver disease in mice.

Citation
Rom, O., et al. “Nitro-Fatty Acids Protect Against Steatosis And Fibrosis During Development Of Nonalcoholic Fatty Liver Disease In Mice.”. Ebiomedicine, pp. 62-72.
Center University of Michigan
Author Oren Rom, Guan Xu, Yanhong Guo, Yunhao Zhu, Huilun Wang, Jifeng Zhang, Yanbo Fan, Wenying Liang, Haocheng Lu, Yuhao Liu, Michael Aviram, Zhipeng Liu, Seongho Kim, Wanqing Liu, Xueding Wang, Eugene Chen, Luis Villacorta
Keywords Nitro-fatty acids, Non-alcoholic Steatohepatitis, non-alcoholic fatty liver disease, Non-invasive liver imaging, liver fibrosis
Abstract

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD) and resulting nonalcoholic steatohepatitis (NASH) are reaching global epidemic proportions. Lack of non-invasive diagnostic tools and effective therapies constitute two of the major hurdles for a bona fide treatment and a reversal of NASH progression and/or regression of the disease. Nitro-oleic acid (OA-NO) has been proven effective in multiple experimental models of inflammation and fibrosis. Thus, the potential benefit of in vivo administration of OA-NO to treat advanced NAFLD was tested herein in a model of long-term NASH diet-induced liver damage.

METHODS: Non-invasive imaging (e.g. photoacustic-ultrasound (PA-US)) was pursued to establish advanced experimental model of NASH in mice in which both steatosis and fibrosis were diagnosed prior experimental therapy with OA-NO. Experimental controls included equimolar amounts of the non-nitrated oleic acid (OA). CLAMS and NMR-based analysis was used for energy metabolism.

FINDINGS: CLAMS and NMR-based analysis demonstrates that OA-NO improves body composition and energy metabolism and inhibits hepatic triglyceride (TG) accumulation. Photoacoustic-ultrasound imaging revealed a robust inhibition of liver steatosis and fibrosis by OA-NO. RNA-sequencing analysis uncovered inflammation and fibrosis as major pathways suppressed by OA-NO administration, as well as regulation of lipogenesis and lipolysis pathways, with a robust inhibition of SREBP1 proteolytic activation and subsequent lipogenesis gene expression by OA-NO. These results were further supported by histological analysis and quantification of lipid accumulation, lobular inflammation (F4/80 staining) and fibrosis (collagen deposition, αSMA staining) as well as established parameters of liver damage (ALT). In vitro studies indicate that OA-NO inhibits TG biosynthesis and accumulation in hepatocytes and inhibits fibrogenesis in human stellate cells.

INTERPRETATION: OA-NO improve steatohepatitis and fibrosis and may constitute an effective therapeutic approach against advanced NAFLD that warrants further clinical evaluation.

Year of Publication
2019
Journal
EBioMedicine
Volume
41
Number of Pages
62-72
Date Published
03/2019
ISSN Number
2352-3964
DOI
10.1016/j.ebiom.2019.02.019
Alternate Journal
EBioMedicine
PMID
30772307
PMCID
PMC6444056
Download citation