Obestatin enhances in vitro generation of pancreatic islets through regulation of developmental pathways

PLoS One. 2013 May 31;8(5):e64374. doi: 10.1371/journal.pone.0064374. Print 2013.

Abstract

Availability of large amounts of in vitro generated β-cells may support replacement therapy in diabetes. However, methods to obtain β-cells from stem/progenitor cells are limited by inefficient endocrine differentiation. We have recently shown that the ghrelin gene product obestatin displays beneficial effects on pancreatic β-cell survival and function. Obestatin prevents β-cell apoptosis, preserves β-cell mass and stimulates insulin secretion in vitro and in vivo, in both normal and diabetic conditions. In the present study, we investigated whether obestatin may promote in vitro β-cell generation from mouse pancreatic islet-derived precursor cells. Treatment of cultured islets of Langerhans with obestatin (i) enriched cells expressing the mesenchymal/neuronal marker nestin, which is associated with pancreatic precursors; (ii) increased cell survival and reduced apoptosis during precursor selection; (iii) promoted the generation of islet-like cell clusters (ICCs) with increased insulin gene expression and C-peptide secretion. Furthermore, obestatin modulated the expression of fibroblast growth factor receptors (FGFRs), Notch receptors and neurogenin 3 (Ngn3) during islet-derived precursor cell selection and endocrine differentiation. These results indicate that obestatin improves the generation of functional β-cells/ICCs in vitro, suggesting implications for cell-based replacement therapy in diabetes. Moreover, obestatin may play a role in regulating pathways involved in pancreas development and regeneration.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Basic Helix-Loop-Helix Transcription Factors / genetics
  • Basic Helix-Loop-Helix Transcription Factors / metabolism
  • Biomarkers / metabolism
  • C-Peptide / biosynthesis
  • C-Peptide / metabolism
  • Cell Differentiation / drug effects
  • Cell Survival / drug effects
  • Cells, Cultured
  • Gene Expression Regulation, Developmental / drug effects*
  • Insulin / biosynthesis
  • Insulin / metabolism
  • Insulin Secretion
  • Islets of Langerhans / cytology
  • Islets of Langerhans / drug effects*
  • Islets of Langerhans / metabolism
  • Mice
  • Nerve Tissue Proteins / genetics
  • Nerve Tissue Proteins / metabolism
  • Nestin / genetics
  • Nestin / metabolism
  • Peptide Hormones / metabolism
  • Peptide Hormones / pharmacology*
  • Receptors, Fibroblast Growth Factor / genetics
  • Receptors, Fibroblast Growth Factor / metabolism
  • Receptors, Notch / genetics
  • Receptors, Notch / metabolism
  • Signal Transduction

Substances

  • Basic Helix-Loop-Helix Transcription Factors
  • Biomarkers
  • C-Peptide
  • Insulin
  • Nerve Tissue Proteins
  • Nes protein, mouse
  • Nestin
  • Neurog3 protein, mouse
  • Peptide Hormones
  • Receptors, Fibroblast Growth Factor
  • Receptors, Notch
  • obestatin, mouse

Grants and funding

This work was supported by the following grants: Regione Piemonte 2008, Brain Drain University of Turin 2008, PRIN 2008 (to R.G.); Compagnia di San Paolo 2008 (to E.G.) and 2011 (to R.G.); Studio delle Malattie Endocrino Metaboliche (SMEM) Foundation (Turin, Italy); NIT (Neuroscience Institute of Turin), and GISNe (Italian Group of Neuroendocrine Sciences). There are no conflicts of interest. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.