Absence of Either Ripk3 or Mlkl Reduces Incidence of Hepatocellular Carcinoma Independent of Liver Fibrosis

Nonalcoholic fatty liver disease (NAFLD) is one of the etiologies that contribute to hepatocellular carcinoma (HCC), and chronic inflammation is one of the proposed mediators of HCC. Because necroptosis is a cell death pathway that induces inflammation, we tested whether necroptosis-induced inflamma...

Full description

Saved in:
Bibliographic Details
Published inMolecular cancer research Vol. 21; no. 9; pp. 933 - 946
Main Authors Mohammed, Sabira, Thadathil, Nidheesh, Ohene-Marfo, Phoebe, Tran, Albert L, Van Der Veldt, Michael, Georgescu, Constantin, Oh, Sangphil, Nicklas, Evan H, Wang, Dawei, Haritha, Nair Hariprasad, Luo, Wenyi, Janknecht, Ralf, Miller, Benjamin F, Wren, Jonathan D, Freeman, Willard M, Deepa, Sathyaseelan S
Format Journal Article
LanguageEnglish
Published United States American Association for Cancer Research 01.09.2023
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Nonalcoholic fatty liver disease (NAFLD) is one of the etiologies that contribute to hepatocellular carcinoma (HCC), and chronic inflammation is one of the proposed mediators of HCC. Because necroptosis is a cell death pathway that induces inflammation, we tested whether necroptosis-induced inflammation contributes to the progression of NAFLD to HCC in a mouse model of diet-induced HCC. Male and female wild-type (WT) mice and mouse models where necroptosis is blocked (Ripk3-/- or Mlkl-/- mice) were fed either a control diet, choline-deficient low-fat diet or choline-deficient high-fat diet. Blocking necroptosis reduced markers of inflammation [proinflammatory cytokines (TNFα, IL6, and IL1β), F4/80+ve macrophages, CCR2+ve infiltrating monocytes], inflammation-associated oncogenic pathways (JNK, PD-L1/PD-1, β-catenin), and HCC in male mice. We demonstrate that hepatic necroptosis promotes recruitment and activation of liver macrophages leading to chronic inflammation, which in turn trigger oncogenic pathways leading to the progression of NAFLD to HCC in male mice. Whereas in female mice, blocking necroptosis reduced HCC independent of inflammation. Our data show a sex-specific difference in the development of inflammation, fibrosis, and HCC in WT mice. However, blocking necroptosis reduced HCC in both males and females without altering liver fibrosis. Thus, our study suggests that necroptosis is a valid therapeutic target for NAFLD-mediated HCC. Necroptosis is a major contributor to hepatic inflammation that drives the progression of NAFLD to HCC and therefore represents a valid target for NAFLD-mediated HCC.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
Mol Cancer Res 2023;21:933–46
ISSN:1541-7786
1557-3125
DOI:10.1158/1541-7786.MCR-22-0820