C-Abl inhibitor imatinib enhances insulin production by β cells: c-Abl negatively regulates insulin production via interfering with the expression of NKx2.2 and GLUT-2

PLoS One. 2014 May 16;9(5):e97694. doi: 10.1371/journal.pone.0097694. eCollection 2014.

Abstract

Chronic myelogenous leukemia patients treated with tyrosine kinase inhibitor, Imatinib, were shown to have increased serum levels of C-peptide. Imatinib specifically inhibits the tyrosine kinase, c-Abl. However, the mechanism of how Imatinib treatment can lead to increased insulin level is unclear. Specifically, there is little investigation into whether Imatinib directly affects β cells to promote insulin production. In this study, we showed that Imatinib significantly induced insulin expression in both glucose-stimulated and resting β cells. In line with this finding, c-Abl knockdown by siRNA and overexpression of c-Abl markedly enhanced and inhibited insulin expression in β cells, respectively. Unexpectedly, high concentrations of glucose significantly induced c-Abl expression, suggesting c-Abl may play a role in balancing insulin production during glucose stimulation. Further studies demonstrated that c-Abl inhibition did not affect the major insulin gene transcription factor, pancreatic and duodenal homeobox-1 (PDX-1) expression. Of interest, inhibition of c-Abl enhanced NKx2.2 and overexpression of c-Abl in β cells markedly down-regulated NKx2.2, which is a positive regulator for insulin gene expression. Additionally, we found that c-Abl inhibition significantly enhanced the expression of glucose transporter GLUT2 on β cells. Our study demonstrates a previously unrecognized mechanism that controls insulin expression through c-Abl-regulated NKx2.2 and GLUT2. Therapeutic targeting β cell c-Abl could be employed in the treatment of diabetes or β cell tumor, insulinoma.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Benzamides / pharmacology*
  • Cell Line
  • Gene Expression Regulation
  • Glucose / metabolism
  • Glucose Transporter Type 2 / genetics
  • Glucose Transporter Type 2 / metabolism*
  • Homeobox Protein Nkx-2.2
  • Homeodomain Proteins / genetics
  • Homeodomain Proteins / metabolism*
  • Imatinib Mesylate
  • Insulin / biosynthesis*
  • Insulin / genetics
  • Insulin-Secreting Cells / drug effects*
  • Leukemia, Myeloid / metabolism
  • Mice
  • Peptide Biosynthesis / drug effects
  • Piperazines / pharmacology*
  • Protein Kinase Inhibitors / pharmacology*
  • Proto-Oncogene Proteins c-abl / antagonists & inhibitors*
  • Proto-Oncogene Proteins c-abl / genetics
  • Proto-Oncogene Proteins c-abl / physiology
  • Pyrimidines / pharmacology*
  • RNA, Small Interfering / metabolism
  • Trans-Activators / metabolism
  • Transcription Factors / genetics
  • Transcription Factors / metabolism*
  • Zebrafish Proteins

Substances

  • Benzamides
  • Glucose Transporter Type 2
  • Homeobox Protein Nkx-2.2
  • Homeodomain Proteins
  • Insulin
  • Nkx2-2 protein, mouse
  • Piperazines
  • Protein Kinase Inhibitors
  • Pyrimidines
  • RNA, Small Interfering
  • Slc2a2 protein, mouse
  • Trans-Activators
  • Transcription Factors
  • Zebrafish Proteins
  • nkx2.2b protein, zebrafish
  • pancreatic and duodenal homeobox 1 protein
  • Imatinib Mesylate
  • Proto-Oncogene Proteins c-abl
  • Glucose

Grants and funding

This work was partially supported by JDRF academic R&D award (17-2008-1036 to CQX) and National Natural Science Foundation of China (81172854 and 81241015 to CQX). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.