File Download

There are no files associated with this item.

  Links for fulltext
     (May Require Subscription)
Supplementary

Article: METTL3 drives NAFLD-related hepatocellular carcinoma and is a therapeutic target for boosting immunotherapy

TitleMETTL3 drives NAFLD-related hepatocellular carcinoma and is a therapeutic target for boosting immunotherapy
Authors
Issue Date15-Aug-2023
PublisherElsevier
Citation
Cell Reports Medicine, 2023, v. 4, n. 8 How to Cite?
Abstract

Non-alcoholic fatty liver disease (NAFLD) is an emerging risk factor of hepatocellular carcinoma (HCC). However, the mechanism and target therapy of NAFLD-HCC are still unclear. Here, we identify that the N6-methyladenosine (m6A) methyltransferase METTL3 promotes NAFLD-HCC. Hepatocyte-specific Mettl3 knockin exacerbated NAFLD-HCC formation, while Mettl3 knockout exerted the opposite effect in mice. Single-cell RNA sequencing revealed that METTL3 suppressed antitumor immune response by reducing granzyme B (GZMB+) and interferon gamma-positive (IFN-γ+) CD8+ T cell infiltration, thereby facilitating immune escape. Mechanistically, METTL3 mediates sterol regulatory element-binding protein (SREBP) cleavage-activating protein (SCAP) mRNA m6A to promote its translation, leading to the activation of cholesterol biosynthesis. This enhanced secretion of cholesterol and cholesteryl esters that impair CD8+ T cell function in the tumor microenvironment. Targeting METTL3 by single-guide RNA, nanoparticle small interfering RNA (siRNA), or pharmacological inhibitor (STM2457) in combination with anti-programmed cell death protein 1 (PD-1) synergized to reinvigorate cytotoxic CD8+ T cells and mediate tumor regression. Together, METTL3 is a therapeutic target in NAFLD-HCC, especially in conjunction with immune checkpoint blockade (ICB) therapy.


Persistent Identifierhttp://hdl.handle.net/10722/331891

 

DC FieldValueLanguage
dc.contributor.authorPan, Y-
dc.contributor.authorChen, H-
dc.contributor.authorZhang, X-
dc.contributor.authorLiu, W-
dc.contributor.authorDing, Y-
dc.contributor.authorHuang, D-
dc.contributor.authorZhai, J-
dc.contributor.authorWei, W-
dc.contributor.authorWen, J-
dc.contributor.authorChen, D-
dc.contributor.authorZhou, Y-
dc.contributor.authorLiang, C-
dc.contributor.authorWong, N-
dc.contributor.authorMan, K-
dc.contributor.authorCheung, AH-
dc.contributor.authorWong, CC-
dc.contributor.authorYu, J-
dc.date.accessioned2023-09-28T04:59:24Z-
dc.date.available2023-09-28T04:59:24Z-
dc.date.issued2023-08-15-
dc.identifier.citationCell Reports Medicine, 2023, v. 4, n. 8-
dc.identifier.urihttp://hdl.handle.net/10722/331891-
dc.description.abstract<p>Non-alcoholic fatty liver disease (NAFLD) is an emerging risk factor of hepatocellular carcinoma (HCC). However, the mechanism and target therapy of NAFLD-HCC are still unclear. Here, we identify that the N<sup>6</sup>-methyladenosine (m<sup>6</sup>A) methyltransferase METTL3 promotes NAFLD-HCC. Hepatocyte-specific <em>Mettl3</em> knockin exacerbated NAFLD-HCC formation, while <em>Mettl3</em> knockout exerted the opposite effect in mice. Single-cell RNA sequencing revealed that METTL3 suppressed antitumor immune response by reducing granzyme B (GZMB<sup>+</sup>) and interferon gamma-positive (IFN-γ<sup>+</sup>) CD8<sup>+</sup> T cell infiltration, thereby facilitating immune escape. Mechanistically, METTL3 mediates sterol regulatory element-binding protein (SREBP) cleavage-activating protein (SCAP) mRNA m<sup>6</sup>A to promote its translation, leading to the activation of cholesterol biosynthesis. This enhanced secretion of cholesterol and cholesteryl esters that impair CD8<sup>+</sup> T cell function in the tumor microenvironment. Targeting METTL3 by single-guide RNA, nanoparticle small interfering RNA (siRNA), or pharmacological inhibitor (STM2457) in combination with anti-programmed cell death protein 1 (PD-1) synergized to reinvigorate cytotoxic CD8<sup>+</sup> T cells and mediate tumor regression. Together, METTL3 is a therapeutic target in NAFLD-HCC, especially in conjunction with immune checkpoint blockade (ICB) therapy.</p>-
dc.languageeng-
dc.publisherElsevier-
dc.relation.ispartofCell Reports Medicine-
dc.rightsThis work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.-
dc.titleMETTL3 drives NAFLD-related hepatocellular carcinoma and is a therapeutic target for boosting immunotherapy-
dc.typeArticle-
dc.identifier.doi10.1016/j.xcrm.2023.101144-
dc.identifier.scopuseid_2-s2.0-85167971166-
dc.identifier.volume4-
dc.identifier.issue8-
dc.identifier.eissn2666-3791-
dc.identifier.issnl2666-3791-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats