1. Academic Validation
  2. PCGF6 controls neuroectoderm specification of human pluripotent stem cells by activating SOX2 expression

PCGF6 controls neuroectoderm specification of human pluripotent stem cells by activating SOX2 expression

  • Nat Commun. 2022 Aug 6;13(1):4601. doi: 10.1038/s41467-022-32295-z.
Xianchun Lan 1 Song Ding 1 Tianzhe Zhang 1 Ying Yi 1 Conghui Li 2 Wenwen Jin 1 Jian Chen 3 Kaiwei Liang 2 Hengbin Wang 4 Wei Jiang 5 6 7
Affiliations

Affiliations

  • 1 Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, RNA Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China.
  • 2 Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China.
  • 3 Chinese Institute for Brain Research (Beijing), Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, 102206, Beijing, China.
  • 4 Department of Internal Medicine, Division of Hematology, Oncology, and Palliative Care, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA.
  • 5 Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, RNA Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China. [email protected].
  • 6 Human Genetics Resource Preservation Center of Wuhan University, Wuhan, China. [email protected].
  • 7 Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China. [email protected].
Abstract

Polycomb group (PcG) proteins are known to repress developmental genes during embryonic development and tissue homeostasis. Here, we report that PCGF6 controls neuroectoderm specification of human pluripotent stem cells (PSCs) by activating SOX2 gene. Human PSCs with PCGF6 depletion display impaired neuroectoderm differentiation coupled with increased mesendoderm outcomes. Transcriptome analysis reveals that de-repression of the Wnt/β-catenin signaling pathway is responsible for the differentiation of PSC toward the mesendodermal lineage. Interestingly, PCGF6 and MYC directly interact and co-occupy a distal regulatory element of SOX2 to activate SOX2 expression, which likely accounts for the regulation in neuroectoderm differentiation. Supporting this notion, genomic deletion of the SOX2-regulatory element phenocopies the impaired neuroectoderm differentiation, while overexpressing SOX2 rescues the neuroectoderm phenotype caused by PCGF6-depletion. Together, our study reveals that PCGF6 can function as lineage switcher between mesendoderm and neuroectoderm in human PSCs by both suppression and activation mechanisms.

Figures
Products