4.7 Article

Targeting HNRNPM Inhibits Cancer Stemness and Enhances Antitumor Immunity in Wnt-activated Hepatocellular Carcinoma

Journal

Publisher

ELSEVIER INC
DOI: 10.1016/j.jcmgh.2022.02.006

Keywords

Cancer Stem Cell; Hepatocellular Carcinoma; Im-mune Escape; Immunotherapy; RNA Splicing

Funding

  1. National Natural Science Foundation of China [82072670, 81871916]
  2. Leading Project of the Science and Technology Commission of Shanghai Municipality [21Y21900100]
  3. Project of Shanghai Municipal Health Commission [202140269]

Ask authors/readers for more resources

This study reveals the role of HNRNPM in regulating the positive association between cancer stemness and immune evasion in hepatocellular carcinoma. By controlling the alternative splicing of MBD2, HNRNPM affects the activation of the WNT/beta-catenin pathway and further regulates the expression of OCT4 and SOX2. Inhibition of HNRNPM promotes the activation of CD8+ T cells and enhances the effectiveness of anti-PD-1 immunotherapy. These findings provide new insights for the development of tailored immunotherapeutic approaches for hepatocellular carcinoma.
BACKGROUND & AIMS: Cancer stemness and immune evasion are closely associated and play critical roles in tumor development and resistance to immunotherapy. However, little is known about the underlying molecular mechanisms that coordinate this association. METHODS: The expressions of heterogeneous nuclear ribonucleoprotein M (HNRNPM) in 240 hepatocellular carcinoma (HCC) samples, public databases, and liver development databases were analyzed. Chromatin immunoprecipitation assays were performed to explore the associations between stem-cell transcription factors and HNRNPM. HNRNPM-regulated alternative splicing (AS) and its binding motif were identified by RNA-seq and RIP-seq. HNRNPM-specific antisense oligonucleotides were developed to explore potential therapeutic targets in HCC. CD8+ T cells that were co-cultured with tumor cells were sorted by flow cytometry assays. RESULTS: We identified an elevated oncofetal splicing factor in HCC, HNRNPM, that unifies and regulates the positive association between cancer stemness and immune evasion. HNRNPM knockdown abolished HCC tumorigenesis and diminished cancer stem cell properties in vitro and in vivo. Mechanistically, HNRNPM regulated the AS of MBD2 by binding its flanking introns, whose isoforms played opposing roles. Although MBD2a and MBD2c competitively bound to CpG islands in the FZD3 promoter, MBD2a preferentially increased FZD3 expression and then activated the WNT/beta-catenin pathway. Interestingly, FZD3 and b-catenin further provided additional regulation by targeting OCT4 and SOX2. We found that HNRNPM inhibition significantly promoted CD8+ T cell activation and that HNRNPM-antisense oligonucleotides effectively inhibited WNT/beta-catenin to enhance anti-programmed cell death protein-1 immunotherapy by promoting CD8+ T cell infiltration. CONCLUSIONS: HNRNPM has a tumor-intrinsic function in generating an immunosuppressive HCC environment through an AS-dependent mechanism and demonstrates proof of the concept of targeting HNRNPM in tailoring HCC immunotherapeutic approaches.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available