Ablation of IFNγ in Myeloid Cells Suppresses Liver Inflammation and Fibrogenesis in Mice with Hepatic Small Heterodimer Partner (SHP) Deletion

Elsevier

Available online 6 April 2024, 101932

Molecular MetabolismAuthor links open overlay panel, , , , , , , , , Highlights•

Hepatic SHP is required to limit lipotoxicity under nutrient excess conditions.

Hepatic SHP is required to suppress mRNA levels of genes which products may promote

liver inflammation.

Blocking IFNg in myeloid cells suppresses inflammation and fibrosis in NASH liver.

Sex differences in NASH transition with a fructose diet.

AbstractBackground

Metabolic dysfunction-associated steatotic liver disease (MASLD) is a common complication of obesity and in severe cases progresses to metabolic dysfunction-associated steatohepatitis (MASH). Small heterodimer partner (SHP) is an orphan member of the nuclear receptor superfamily and regulates metabolism and inflammation in the liver via a variety of pathways. In this study, we investigate the molecular foundation of MASH progression in mice with hepatic SHP deletion and explore possible therapeutic means to reduce MASH.

Methods

Hepatic SHP knockout mice (SHPΔhep) and their wild-type littermates (SHPfl/fl) of both sexes were fed a fructose diet for 14 weeks and subjected to an oral glucose tolerance test. Then, plasma lipids were determined, and liver lipid metabolism and inflammation pathways were analyzed with immunoblotting, RNAseq, and qPCR assays. To explore possible therapeutic intersections of SHP and inflammatory pathways, SHPΔhep mice were reconstituted with bone marrow lacking interferon γ (IFNγ-/-) to suppress inflammation.

Results

Hepatic deletion of SHP in mice fed a fructose diet decreased liver fat and increased proteins for fatty acid oxidation and liver lipid uptake including UCP1, CPT1α, ACDAM, and SRBI. Despite lower liver fat, hepatic SHP deletion increased liver inflammatory F4/80+ cells and mRNA levels of inflammatory cytokines (IL-12, IL-6, Ccl2, and IFNγ) in both sexes and elevated endoplasmic reticulum stress markers of Cox2 and CHOP in female mice. Liver bulk RNAseq data showed upregulation of genes whose protein products regulate lipid transport, fatty acid oxidation, and inflammation in SHPΔhep mice. The increased inflammation and fibrosis in SHPΔhep mice were corrected with transplantation of bone marrow-derived IFNγ-/- myeloid cells.

Conclusion

Hepatic deletion of SHP improves fatty liver but worsens hepatic inflammation possibly by driving excess fatty acid oxidation, which is corrected by deletion of IFNγ specifically in myeloid cells. This suggests that hepatic SHP limits fatty acid oxidation during fructose diet feeding, but in doing so prevents pro-MASH pathways. The IFNγ-mediated inflammation in myeloid cells appears to be a potential therapeutic target to suppress MASH.

© 2024 Published by Elsevier GmbH.

留言 (0)

沒有登入
gif