File Download

There are no files associated with this item.

Supplementary

Conference Paper: The oncogenic m6A demethylase FTO promotes tumorigenesis and immune escape by upregulating GPNMB in hepatocellular carcinoma

TitleThe oncogenic m6A demethylase FTO promotes tumorigenesis and immune escape by upregulating GPNMB in hepatocellular carcinoma
Authors
Issue Date6-Jun-2024
Abstract

Background and Aims: Hepatocellular carcinoma (HCC) is the 3rd leading cause of cancer-related 
death worldwide. Immune checkpoint inhibitor (ICI) targeting PD-L1 combined with VEGF is the first-
line therapy for advanced HCC. However, the efficiency of Immune checkpoint inhibitor (ICI) is not 
satisfactory in HCC patients. FTO (Fat mass and obesity-associated protein) is the first identified N6-
methyladenosine (m6A) demethylase and has been reported to play an oncogenic or tumor-
suppressive role in liver cancer. A better understanding of the functional role and mechanistic basis of 
FTO in HCC is needed. In this study, we aimed to functionally characterize the roles of m6A 
demethylase FTO in HCC. 

Method: To investigate the tumorigenic functions of FTO, colony formation, proliferation, migration 
and invasion assays were conducted in vitro. Co-culture system of HCC cells and CD8 T cells or THP-
1 cells was used to examine the activation of effector memory T cells and polarization and recruitment 
of macrophages upon stable knockdown of FTO (FTO-KD). Multiple xenograft and spontaneous HCC 
forming mouse models were used to evaluate the effect of FTO on self-renewal, tumor growth, and 
response to ICI treatment. RNA-sequencing and m6A-sequincing were done to identify the gene 
targets related to m6A modification by FTO. 

Results: FTO expression was up-regulated in patients’ HCC tumors, and high FTO expression (2-fold 
cutoff) was significantly associated with poorer survival rates. Depletion of FTO resulted in reduced 
pro-oncogenic functions in vitro, and suppressed tumor formation, progression, metastasis in vivo. 
Stable FTO-KD significantly increased tumor-infiltrating immune cells and cytotoxic CD8+ T cells in 
mouse tumor tissues and was accompanied by a significant reduction of PD1+ T cell exhaustion. We 
also observed a trend of increase in anti-tumoral M1 macrophages in mouse tumors. Consistently, in 
vitro co-culture assays of human HCC cells with T cells isolated from PBMC of healthy human 
subjects showed that FTO KD increased the proportion of effector memory T cells and enhanced the 
proliferation of CD8+ T cells. FTO silencing increased the anti-tumoral polarization and migration of 
M1 macrophages. We identified GPNMB as a novel downstream target of FTO, which reduced the 
m6A abundance of GPNMB, hence stabilizing it from degradation by YTHDF2. GPNMB was able to 
inhibit CD8+ T cell activation. Furthermore, targeting FTO by its potential inhibitor CS2 promoted the 
therapeutic effect of anti-PD1 in a well-established ‘cold-tumor’ mouse model (Trp53KO/C-MycOE), 
resulting in significantly smaller tumors and higher tumor-infiltrating CD8 T cells in the liver tumor 
tissues. 

Conclusion:  We demonstrated that FTO exerts oncogenic function and promotes immune escape in 
HCC by regulating the m6A demethylation of GPNMB mRNA. Targeting FTO may provide a new 
therapeutic approach for treating HCC.


Persistent Identifierhttp://hdl.handle.net/10722/343994

 

DC FieldValueLanguage
dc.contributor.authorZhang, Vanilla Xin-
dc.contributor.authorChen, Ao-
dc.contributor.authorZhang, Qingyang-
dc.contributor.authorSze, Karen Man-Fong-
dc.contributor.authorTian, Lu-
dc.contributor.authorHuang, Hongyang-
dc.contributor.authorLee, Eva-
dc.contributor.authorLu, Jingyi-
dc.contributor.authorLyu, Xueying-
dc.contributor.authorLee, Joyce Man Fong-
dc.contributor.authorWong, Jack Chun-Ming-
dc.contributor.authorHo, Daniel Wai-Hung-
dc.contributor.authorNg, Irene Oi-Lin-
dc.date.accessioned2024-06-25T03:29:28Z-
dc.date.available2024-06-25T03:29:28Z-
dc.date.issued2024-06-06-
dc.identifier.urihttp://hdl.handle.net/10722/343994-
dc.description.abstract<p>Background and Aims: Hepatocellular carcinoma (HCC) is the 3rd leading cause of cancer-related <br>death worldwide. Immune checkpoint inhibitor (ICI) targeting PD-L1 combined with VEGF is the first-<br>line therapy for advanced HCC. However, the efficiency of Immune checkpoint inhibitor (ICI) is not <br>satisfactory in HCC patients. FTO (Fat mass and obesity-associated protein) is the first identified N6-<br>methyladenosine (m6A) demethylase and has been reported to play an oncogenic or tumor-<br>suppressive role in liver cancer. A better understanding of the functional role and mechanistic basis of <br>FTO in HCC is needed. In this study, we aimed to functionally characterize the roles of m6A <br>demethylase FTO in HCC. </p><p>Method: To investigate the tumorigenic functions of FTO, colony formation, proliferation, migration <br>and invasion assays were conducted in vitro. Co-culture system of HCC cells and CD8 T cells or THP-<br>1 cells was used to examine the activation of effector memory T cells and polarization and recruitment <br>of macrophages upon stable knockdown of FTO (FTO-KD). Multiple xenograft and spontaneous HCC <br>forming mouse models were used to evaluate the effect of FTO on self-renewal, tumor growth, and <br>response to ICI treatment. RNA-sequencing and m6A-sequincing were done to identify the gene <br>targets related to m6A modification by FTO. </p><p>Results: FTO expression was up-regulated in patients’ HCC tumors, and high FTO expression (2-fold <br>cutoff) was significantly associated with poorer survival rates. Depletion of FTO resulted in reduced <br>pro-oncogenic functions in vitro, and suppressed tumor formation, progression, metastasis in vivo. <br>Stable FTO-KD significantly increased tumor-infiltrating immune cells and cytotoxic CD8+ T cells in <br>mouse tumor tissues and was accompanied by a significant reduction of PD1+ T cell exhaustion. We <br>also observed a trend of increase in anti-tumoral M1 macrophages in mouse tumors. Consistently, in <br>vitro co-culture assays of human HCC cells with T cells isolated from PBMC of healthy human <br>subjects showed that FTO KD increased the proportion of effector memory T cells and enhanced the <br>proliferation of CD8+ T cells. FTO silencing increased the anti-tumoral polarization and migration of <br>M1 macrophages. We identified GPNMB as a novel downstream target of FTO, which reduced the <br>m6A abundance of GPNMB, hence stabilizing it from degradation by YTHDF2. GPNMB was able to <br>inhibit CD8+ T cell activation. Furthermore, targeting FTO by its potential inhibitor CS2 promoted the <br>therapeutic effect of anti-PD1 in a well-established ‘cold-tumor’ mouse model (Trp53KO/C-MycOE), <br>resulting in significantly smaller tumors and higher tumor-infiltrating CD8 T cells in the liver tumor <br>tissues. </p><p>Conclusion:  We demonstrated that FTO exerts oncogenic function and promotes immune escape in <br>HCC by regulating the m6A demethylation of GPNMB mRNA. Targeting FTO may provide a new <br>therapeutic approach for treating HCC.</p>-
dc.languageeng-
dc.relation.ispartofEuropean Association for the Study of the Liver Congress (EASL) Congress 2024 (05/06/2024-08/06/2024, , , Milan)-
dc.titleThe oncogenic m6A demethylase FTO promotes tumorigenesis and immune escape by upregulating GPNMB in hepatocellular carcinoma-
dc.typeConference_Paper-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats