4.7 Article

SRSF10 stabilizes CDC25A by triggering exon 6 skipping to promote hepatocarcinogenesis

出版社

BMC
DOI: 10.1186/s13046-022-02558-0

关键词

SRSF10; CDC25A; Alternative splicing; Exon skipping; Hepatocellular carcinoma

类别

向作者/读者索取更多资源

The study found that SRSF10 is enriched in HCC tissues and promotes HCC proliferation, cell cycle, and invasion. SRSF10 promotes exon 6 exclusion of CDC25A pre-mRNA. CDC25A(oE6) is identified as a crucial cell cycle mediator that is stabilized and retained in the nucleus due to the deletion of two ubiquitination sites in exon 6.
Background: Alternative splicing (AS) events are extensively involved in the progression of diverse tumors, but how serine/arginine-rich splicing Factor 10 (SRSF10) behaves in hepatocellular carcinoma (HCC) has not been sufficiently studied. We aimed to determine SRSF10 associated AS mechanisms and their effects on HCC progression. Methods: The expression of SRSF10 in HCC tissues was examined, and the in vitro and in vivo functions of SRSF10 were investigated. The downstream AS targets were screened using RNA sequencing. The interaction between SRSF10 protein and exclusion of cell division cycle 25 A (CDC25A) mRNA was identified using RNA immunoprecipitation and crosslinking immunoprecipitation q-PCR. The effects of SRSF10 on CDC25A posttranslational modification, subcellular distribution, and protein stability were verified through coimmunoprecipitation, immunofluorescence, and western blotting. Results: SRSF10 was enriched in HCC tissues and facilitated HCC proliferation, cell cycle, and invasion. RNA sequencing showed that SRSF10 promotes exon 6 exclusion of CDC25A pre-mRNA splicing. As a crucial cell cycle mediator, the exon-skipped isoform CDC25A(oE6) was identified to be stabilized and retained in the nucleus due to the deletion of two ubiquitination (Lys150, Lys169) sites in exon 6. The stabilized isoform CDC25A(oE6) derived from AS had stronger cell cycle effects on HCC tumorigenesis, and playing a more significant role than the commonly expressed longer variant CDC25A(L). Interestingly, SRSF10 activated the carcinogenesis role of CDC25A through Ser178 dephosphorylation to cause nuclear retention. Moreover, CDC25A(oE6) was verified to be indispensable for SRSF10 to promote HCC development in vitro and in vivo. Conclusions: We reveal a regulatory pattern whereby SRSF10 contributes to a large proportion of stabilized CDC25A(delta E6) production, which is indispensable for SRSF10 to promote HCC development. Our findings uncover AS mechanisms such as CDC25A that might serve as potential therapeutic targets to treat HCC.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据