4.7 Article

METTL3/IGF2BP3 axis inhibits tumor immune surveillance by upregulating N6-methyladenosine modification of PD-L1 mRNA in breast cancer

Journal

MOLECULAR CANCER
Volume 21, Issue 1, Pages -

Publisher

BMC
DOI: 10.1186/s12943-021-01447-y

Keywords

Breast cancer; PD-L1; m(6)A; METTL3; Immune surveillance

Funding

  1. National Key RD Program [2018YFC1313400]
  2. Science and Technology Innovation Enhancement Project of Army Medical University [2019CXJSB017, 2019XYY21]
  3. Chonqing Science and Technology Bureau [2021MSXM338]

Ask authors/readers for more resources

The study found that METTL3 can post-transcriptionally upregulate PD-L1 expression in an m(6)A-IGF2BP3-dependent manner to promote stabilization of PD-L1 mRNA. This may have important implications for new and efficient therapeutic strategies in tumor immunotherapy.
Background Continual expression of PD-L1 in tumor cells is critical for tumor immune escape and host T cell exhaustion, however, knowledge on its clinical benefits through inhibition is limited in breast cancer. N-6-methyladenosine (m(6)A) plays a crucial role in multiple biological activities. Our study aimed to investigate the regulatory role of the m(6)A modification in PD-L1 expression and immune surveillance in breast cancer. Methods MeRIP-seq and epitranscriptomic microarray identified that PD-L1 is the downstream target of METTL3. MeRIP-qPCR, absolute quantification of m(6)A modification assay, and RIP-qPCR were used to examine the molecular mechanism underlying METTL3/m(6)A/IGF2BP3 signaling axis in PD-L1 expression. B-NDG and BALB/c mice were used to construct xenograft tumor models to verify the phenotypes upon METTL3 and IGF2BP3 silencing. In addition, breast cancer tissue microarray was used to analyze the correlation between PD-L1 and METTL3 or IGF2BP3 expression. Results We identified that PD-L1 was a downstream target of METTL3-mediated m(6)A modification in breast cancer cells. METTL3 knockdown significantly abolished m(6)A modification and reduced stabilization of PD-L1 mRNA. Additionally, METTL3-mediated PD-L1 mRNA activation was m(6)A-IGF2BP3-dependent. Moreover, inhibition of METTL3 or IGF2BP3 enhanced anti-tumor immunity through PD-L1-mediated T cell activation, exhaustion, and infiltration both in vitro and in vivo. PD-L1 expression was also positively correlated with METTL3 and IGF2BP3 expression in breast cancer tissues. Conclusion Our study suggested that METTL3 could post-transcriptionally upregulate PD-L1 expression in an m(6)A-IGF2BP3-dependent manner to further promote stabilization of PD-L1 mRNA, which may have important implications for new and efficient therapeutic strategies in the tumor immunotherapy.

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