Skip to main content

Advertisement

Log in

Essential role of Notch/Hes1 signaling in postnatal pancreatic exocrine development

  • Original Article―Liver, Pancreas, and Biliary Tract
  • Published:
Journal of Gastroenterology Aims and scope Submit manuscript

Abstract

Background

Notch/Hes1 signaling has been shown to play a role in determining the fate of pancreatic progenitor cells. However, its function in postnatal pancreatic maturation is not fully elucidated.

Methods

We generated conditional Hes1 knockout and/or Notch intracellular domain (NICD) overexpression mice in Ptf1a- or Pdx1-positive pancreatic progenitor cells and analyzed pancreatic tissues.

Results

Both Ptf1acre/+; Hes1f/f and Ptf1acre/+; Rosa26NICD mice showed normal pancreatic development at P0. However, exocrine tissue of the pancreatic tail in Ptf1acre/+; Hes1f/f mice atrophied and was replaced by fat tissue by 4 weeks of age, with increased apoptotic cells and fewer centroacinar cells. This impaired exocrine development was completely rescued by NICD overexpression in Ptf1acre/+; Hes1f/f; Rosa26NICD mice, suggesting compensation by a Notch signaling pathway other than Hes1. Conversely, Pdx1-Cre; Hes1f/f mice showed impaired postnatal exocrine development in both the pancreatic head and tail, revealing that the timing and distribution of embryonic Hes1 expression affects postnatal exocrine tissue development.

Conclusions

Notch signaling has an essential role in pancreatic progenitor cells for the postnatal maturation of exocrine tissue, partly through the formation of centroacinar cells.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Kawaguchi Y, Cooper B, Gannon M, et al. The role of the transcriptional regulator Ptf1a in converting intestinal to pancreatic progenitors. Nat Genet. 2002;32:128–34.

    Article  CAS  Google Scholar 

  2. Jørgensen MC, Ahnfelt-Rønne J, Hald J, et al. An illustrated review of early pancreas development in the mouse. Endocr Rev. 2007;28:685–705.

    Article  Google Scholar 

  3. Kageyama R, Ohtsuka T, Kobayashi T. The Hes gene family: repressors and oscillators that orchestrate embryogenesis. Development. 2007;134:1243–51.

    Article  CAS  Google Scholar 

  4. Jensen J, Pedersen EE, Galante P, et al. Control of endodermal endocrine development by Hes-1. Nat Genet. 2000;24:36–44.

    Article  CAS  Google Scholar 

  5. Fukuda A, Kawaguchi Y, Furuyama K, et al. Ectopic pancreas formation in Hes1-knockout mice reveals plasticity of endodermal progenitors of the gut, bile duct, and pancreas. J Clin Invest. 2006;116:1484–93.

    Article  CAS  Google Scholar 

  6. Murtaugh LC, Stanger BZ, Kwan KM, et al. Notch signaling controls multiple steps of pancreatic differentiation. Proc Natl Acad Sci USA. 2003;100:14920–5.

    Article  CAS  Google Scholar 

  7. Hidalgo-Sastre A, Brodylo RL, Lubeseder-Martellato C, et al. Hes1 controls exocrine cell plasticity and restricts development of pancreatic ductal adenocarcinoma in a mouse model. Am J Pathol. 2016;186:2934–44.

    Article  CAS  Google Scholar 

  8. Rovira M, Scott SG, Liss AS, et al. Isolation and characterization of centroacinar/terminal ductal progenitor cells in adult mouse pancreas. Proc Natl Acad Sci USA. 2010;107:75–80.

    Article  CAS  Google Scholar 

  9. Furuyama K, Kawaguchi Y, Akiyama H, et al. Continuous cell supply from a Sox9-expressing progenitor zone in adult liver, exocrine pancreas and intestine. Nat Genet. 2011;43:34–41.

    Article  CAS  Google Scholar 

  10. Miyamoto Y, Maitra A, Ghosh B, et al. Notch mediates TGF alpha-induced changes in epithelial differentiation during pancreatic tumorigenesis. Cancer Cell. 2003;3:565–76.

    Article  CAS  Google Scholar 

  11. Nakano Y, Negishi N, Gocho S, et al. Disappearance of centroacinar cells in the Notch ligand-deficient pancreas. Genes Cells. 2015;20:500–11.

    Article  CAS  Google Scholar 

  12. Gu G, Dubauskaite J, Melton DA. Direct evidence for the pancreatic lineage: NGN3+ cells are islet progenitors and are distinct from duct progenitors. Development. 2002;129:2447–57.

    Article  CAS  Google Scholar 

  13. Imayoshi I, Shimogori T, Ohtsuka T, et al. Hes genes and neurogenin regulate non-neural versus neural fate specification in the dorsal telencephalic midline. Development. 2008;135:2531–41.

    Article  CAS  Google Scholar 

  14. Nishikawa Y, Kodama Y, Shiokawa M, et al. Hes1 plays an essential role in Kras-driven pancreatic tumorigenesis. Oncogene. 2019;38:4283–96.

    Article  CAS  Google Scholar 

  15. Gukovskaya AS, Gukovsky I. Autophagy and pancreatitis. Am J Physiol Gastrointest Liver Physiol. 2012;303:993–1003.

    Article  Google Scholar 

  16. Arda HE, Benitez CM, Kim SK. Gene regulatory networks governing pancreas development. Dev Cell. 2013;25:5–13.

    Article  CAS  Google Scholar 

  17. Mameishvili E, Serafimidis I, Iwaszkiewicz S, et al. Aldh1b1 expression defines progenitor cells in the adult pancreas and is required for Kras-induced pancreatic cancer. Proc Natl Acad Sci USA. 2019;116:20679–88.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Y Kawaguchi (Kyoto University, Kyoto, Japan) for supplying Ptf1acre/+ mice. We thank D Melton (Harvard University, MA, USA) for supplying Rosa26NICD mice. We thank Yuta Kawamata and Taichi Ito for the excellent technical support.

Funding

This work was supported by the Japan Society for the Promotion of Science (JSPS) KAKENHI Grant Numbers JP16K09395.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yuzo Kodama.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

535_2021_1779_MOESM1_ESM.pdf

Supplementary Figure S1 Mouse strains. a The protein-coding region of Ptf1a was precisely replaced with that of Cre, encoding the recombinase Cre. b Hes1 is floxed by loxp and conditionally knocked out in the Cre recombinase promoter site. c Rosa-26, Lox-STOP-Lox, and LacZ construct. LacZ expresses in the Cre recombinase promoter site. d Rosa-26, Lox-STOP-Lox, and NICD construct. NICD expresses in the Cre recombinase promoter site. e Cre recombinase is under the transcriptional control of the mouse Pdx1 promoter. Supplementary Figure S2. Hes1 immunohistochemistry Hes1 expression in pancreatic tissue was analyzed by immunohistochemistry in Hes1 cKO (Ptf1acre/+;Hes1f/f ;Rosa26LacZ) and control (Ctrl, Ptf1acre/+;Hes1f/f ;Rosa26LacZ ) mice at P0. Hes1 expression was deleted in Hes1 cKO mice (b), while it was positive in the ducts and centroacinar cells of control mice (a). Supplementary Figure S3 Changes in body weight and blood glucose level Changes in body weight (a) and blood glucose level (b) after birth in Hes1 cKO (Ptf1acre/+;Hes1f/f ;Rosa26LacZ) and control (Ctrl, Ptf1a+/+;Hes1f/f;Rosa26LacZ ) mice. The median and the standard error of mean are shown. Supplementary Figure S4 Hes1 knockout in Ptf1a-positive progenitor cells. X-gal staining of Hes1 conditional knockout mice (Ptf1acre/+;Hes1f/f;Rosa26LacZ) and control mice (Ptf1acre/+;Hes1f/+;Rosa26LacZ) at 4 weeks of age. b and d are a magnification of the broken line in a and c. Arrow head: islet, arrow: duct. Scale bars: 50 μm (b, d). Supplementary Figure S5 a Western blot analysis for LC3-I and its lapidated form LC3-II. We found expression of LC3-I but not its lipidated form LC3-II in both pancreatic head and tail of Hes1 cKO mice and control mice at P4. Supplementary Figure S6 a Immunofluorescence images of Hes1 and Aldh1a1 in Hes1 cKO (Ptf1acre/+;Hes1f/f) and control (Ctrl, Ptf1a+/+;Hes1f/f) mice at P3. Scale bars: 50 μm. b Microarray analysis of pancreatic head and tail tissues of Hes1 cKO and control mice at P7. A heatmap of genes related to pancreatic development is shown. Supplementary Figure S7 a FACS isolation of Aldh-positive cells with Aldefluor reagent. FACS gating was established by negative control. The number of Aldh-positive cells were decreased in Hes1 cKO (Ptf1acre/+;Hes1f/f) mice compared with that in control mice at P7 (b). The number of organoid formation from pancreatic cells (c, d). *P < 0.05. Supplementary Figure S8 a H&E staining and immunohistochemical staining for Hes5 and Hey1 of control mice, Ptf1acre/+;Hes1f/f mice (cKO), Ptf1acre/+;Rosa26NICD mice, and Ptf1acre/+;Hes1f/f;Rosa26NICD mice at P0. Marked increase of Hes5 expression was observed in Ptf1acre/+;Hes1f/f;Rosa26NICD mice. Scale bars: 50 μm. Supplementary Table 1. Primary antibodies used in this study. (PDF 4987 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kuriyama, K., Kodama, Y., Shiokawa, M. et al. Essential role of Notch/Hes1 signaling in postnatal pancreatic exocrine development. J Gastroenterol 56, 673–687 (2021). https://doi.org/10.1007/s00535-021-01779-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00535-021-01779-y

Keywords

Navigation