Extensive NEUROG3 occupancy in the human pancreatic endocrine gene regulatory network

Publikation: Bidrag til tidsskriftTidsskriftartikelForskningfagfællebedømt

Dokumenter

  • Valérie Schreiber
  • Reuben Mercier
  • Sara Jiménez
  • Tao Ye
  • Emmanuel García-Sánchez
  • Annabelle Klein
  • Aline Meunier
  • Sabitri Ghimire
  • Catherine Birck
  • Bernard Jost
  • Kristian Honnens de Lichtenberg
  • Christian Honoré
  • Serup, Palle
  • Gérard Gradwohl

Objective: Mice lacking the bHLH transcription factor (TF) Neurog3 do not form pancreatic islet cells, including insulin-secreting beta cells, the absence of which leads to diabetes. In humans, homozygous mutations of NEUROG3 manifest with neonatal or childhood diabetes. Despite this critical role in islet cell development, the precise function of and downstream genetic programs regulated directly by NEUROG3 remain elusive. Therefore, we mapped genome-wide NEUROG3 occupancy in human induced pluripotent stem cell (hiPSC)–derived endocrine progenitors and determined NEUROG3 dependency of associated genes to uncover direct targets. Methods: We generated a novel hiPSC line (NEUROG3-HA-P2A-Venus) where NEUROG3 is HA-tagged and fused to a self-cleaving fluorescent VENUS reporter. We used the CUT&RUN technique to map NEUROG3 occupancy and epigenetic marks in pancreatic endocrine progenitors (PEP) that were differentiated from this hiPSC line. We integrated NEUROG3 occupancy data with chromatin status and gene expression in PEPs as well as their NEUROG3-dependence. In addition, we investigated whether NEUROG3 binds type 2 diabetes mellitus (T2DM)–associated variants at the PEP stage. Results: CUT&RUN revealed a total of 863 NEUROG3 binding sites assigned to 1263 unique genes. NEUROG3 occupancy was found at promoters as well as at distant cis-regulatory elements that frequently overlapped within PEP active enhancers. De novo motif analyses defined a NEUROG3 consensus binding motif and suggested potential co-regulation of NEUROG3 target genes by FOXA or RFX transcription factors. We found that 22% of the genes downregulated in NEUROG3−/− PEPs, and 10% of genes enriched in NEUROG3-Venus positive endocrine cells were bound by NEUROG3 and thus likely to be directly regulated. NEUROG3 binds to 138 transcription factor genes, some with important roles in islet cell development or function, such as NEUROD1, PAX4, NKX2-2, SOX4, MLXIPL, LMX1B, RFX3, and NEUROG3 itself, and many others with unknown islet function. Unexpectedly, we uncovered that NEUROG3 targets genes critical for insulin secretion in beta cells (e.g., GCK, ABCC8/KCNJ11, CACNA1A, CHGA, SCG2, SLC30A8, and PCSK1). Thus, analysis of NEUROG3 occupancy suggests that the transient expression of NEUROG3 not only promotes islet destiny in uncommitted pancreatic progenitors, but could also initiate endocrine programs essential for beta cell function. Lastly, we identified eight T2DM risk SNPs within NEUROG3-bound regions. Conclusion: Mapping NEUROG3 genome occupancy in PEPs uncovered unexpectedly broad, direct control of the endocrine genes, raising novel hypotheses on how this master regulator controls islet and beta cell differentiation.

OriginalsprogEngelsk
Artikelnummer101313
TidsskriftMolecular Metabolism
Vol/bind53
Antal sider15
ISSN2212-8778
DOI
StatusUdgivet - 2021

Bibliografisk note

Funding Information:
The authors thank the members of the Gradwohl team and the Genomeast platform (particularly Christelle Thibault-Carpentier and David Rodriguez), Flow cytometry, and Cell culture facilities for the sequencing of the CUT&RUN samples, cell sorting, and hiPSC maintenance respectively. The authors are grateful to I. Cebola for providing ChIP-seq data and R. Scharfmann for helpful discussions. The Gradwohl lab is funded by the Novo Nordisk Foundation (Challenge Grant NNF14OC0013655). Sequencing was performed by the GenomEast platform, a member of the ?France G?nomique? consortium (ANR-10-INBS-0009). This work used the Integrated Structural Biology platform of the Strasbourg Instruct-ERIC center IGBMC-CBI supported by FRISBI (ANR-10-INBS-0005-001). IGBMC is supported by the grant ANR-10-LABX-0030-INRT, a French State fund managed by the Agence Nationale de la Recherche under the frame program Investissements d'Avenir ANR-10-IDEX-0002-02.

Funding Information:
The authors thank the members of the Gradwohl team and the Genomeast platform (particularly Christelle Thibault-Carpentier and David Rodriguez), Flow cytometry, and Cell culture facilities for the sequencing of the CUT&RUN samples, cell sorting, and hiPSC maintenance respectively. The authors are grateful to I. Cebola for providing ChIP-seq data and R. Scharfmann for helpful discussions. The Gradwohl lab is funded by the Novo Nordisk Foundation (Challenge Grant NNF14OC0013655 ). Sequencing was performed by the GenomEast platform, a member of the ‘France Génomique’ consortium (ANR-10-INBS-0009). This work used the Integrated Structural Biology platform of the Strasbourg Instruct-ERIC center IGBMC-CBI supported by FRISBI ( ANR-10-INBS-0005-001 ). IGBMC is supported by the grant ANR-10-LABX-0030-INRT, a French State fund managed by the Agence Nationale de la Recherche under the frame program Investissements d'Avenir ANR-10-IDEX-0002-02 .

Publisher Copyright:
© 2021 The Authors

Antal downloads er baseret på statistik fra Google Scholar og www.ku.dk


Ingen data tilgængelig

ID: 280174137