Abstract

Poorly differentiated tumors usually exhibit phenotypes similar to that of their developmental precursor cells. Tumor cells that acquire the lineage progenitor cells feature usually exploit developmental signaling to potentiate cancer progression. However, the underlying molecular events remain elusive. In this study, based on analysis of an in vitro hepatocyte differentiation model, the maternal factor PGC7 (also known as DPPA3, STELLA) was found closely associated with liver development and tumor differentiation in hepatocellular carcinoma (HCC). Expression of PGC7 decreased during hepatocyte maturation and increased progressively from well-differentiated HCCs to poorly differentiated HCCs. Whole-genome methylation sequencing found that PGC7 could induce promoter demethylation of genes related to development. Pathway-based network analysis indicated that downstream targets of PGC7 might form networks associated with developmental transcription factor activation. Overexpression of PGC7 conferred progenitor-like features of HCC cells both in vitro and in vivo. Mechanism studies revealed that PGC7 could impede nuclear translocation of UHRF1, and thus facilitate promoter demethylation of GLI1 and MYCN, both of which are important regulators of HCC self-renewal and differentiation. Depletion or inhibition of GLI1 effectively downregulated MYCN, abolished the effect of PGC7, and sensitized HCC cells to sorafenib treatment. In addition, we found a significant correlation of PGC7 with GLI1/MYCN and lineage differentiation markers in clinical HCC patients. PGC7 expression might drive HCC toward a “dedifferentiated” progenitor lineage through facilitating promoter demethylation of key developmental transcription factors; further inhibition of PGC7/GLI1/MYCN might reverse poorly differentiated HCCs and provide novel therapeutic strategies.

Highlights

  • It has been well-studied that cancer cells usually exhibited phenotypic similarity with their lineage precursor cells during development [1]

  • We proposed a novel mechanism in which ectopic expression of PGC7 could promote hepatocellular carcinoma (HCC) oncogenic dedifferentiation and maintain an epigenetic status suitable for liver progenitor cells, which further contributed to metastasis and poor prognosis of HCC

  • Bioinformatics-aided pathway and gene ontology analysis revealed that this cluster of genes was closely associated with cellular development, cell differentiation, and signaling regulating pluripotency of stem cells (Supplementary Fig. 1b), among which PGC7 arouse our interest as it ranks at the top of the list

Read more

Summary

Introduction

It has been well-studied that cancer cells usually exhibited phenotypic similarity with their lineage precursor cells during development [1]. The differentiation process comprises four stages—embryonic stem cell, endoderm, liver progenitor cell, and premature hepatocyte stages. Based on the selection criteria, the maternal factor PGC7 arouse our interest since it ranks at the top of the list, and has a unique expression pattern strongly associated with tumor differentiation and poor clinical outcomes. PGC7/DPPA3 belongs to the developmental pluripotency-associated protein (DPPA) family and was found frequently expressed in pluripotent cells [7]. PGC7 protects DNA methylation pattern in early embryogenesis [8] by binding to either dimethylated histone H3K9 (H3K9me2) [9] or TET2/TET3 [10]. Expression of PGC7 was found indispensable for the generation of induced pluripotent stem cells (iPSCs) [13]. All of the evidence indicated that PGC7 might be critical in lineage-related tumorigenesis through remodeling DNA methylation patterns

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call