Abstract

ObjectiveCongenital hyperinsulinism (CHI) is a rare disease characterized by persistent hypoglycemia as a result of inappropriate insulin secretion, which can lead to irreversible neurological defects in infants. Poor efficacy and strong adverse effects of the current medications impede successful treatment. The aim of the study was to investigate new approaches to silence β-cells and thus attenuate insulin secretion.Research Design and MethodsIn the scope of our research, we tested substances more selective and more potent than the gold standard diazoxide that also interact with neuroendocrine ATP-sensitive K+ (KATP) channels. Additionally, KATP channel-independent targets as Ca2+-activated K+ channels of intermediate conductance (KCa3.1) and L-type Ca2+ channels were investigated. Experiments were performed using human islet cell clusters isolated from tissue of CHI patients (histologically classified as pathological) and islet cell clusters obtained from C57BL/6N (WT) or SUR1 knockout (SUR1-/-) mice. The cytosolic Ca2+ concentration ([Ca2+]c) was used as a parameter for the pathway regulated by electrical activity and was determined by fura-2 fluorescence. The mitochondrial membrane potential (ΔΨ) was determined by rhodamine 123 fluorescence and single channel currents were measured by the patch-clamp technique.ResultsThe selective KATP channel opener NN414 (5 µM) diminished [Ca2+]c in isolated human CHI islet cell clusters and WT mouse islet cell clusters stimulated with 10 mM glucose. In islet cell clusters lacking functional KATP channels (SUR1-/-) the drug was without effect. VU0071063 (30 µM), another KATP channel opener considered to be selective, lowered [Ca2+]c in human CHI islet cell clusters. The compound was also effective in islet cell clusters from SUR1-/- mice, showing that [Ca2+]c is influenced by additional effects besides KATP channels. Contrasting to NN414, the drug depolarized ΔΨ in murine islet cell clusters pointing to severe interference with mitochondrial metabolism. An opener of KCa3.1 channels, DCEBIO (100 µM), significantly decreased [Ca2+]c in SUR1-/- and human CHI islet cell clusters. To target L-type Ca2+ channels we tested two already approved drugs, dextromethorphan (DXM) and simvastatin. DXM (100 µM) efficiently diminished [Ca2+]c in stimulated human CHI islet cell clusters as well as in stimulated SUR1-/- islet cell clusters. Similar effects on [Ca2+]c were observed in experiments with simvastatin (7.2 µM).ConclusionsNN414 seems to provide a good alternative to the currently used KATP channel opener diazoxide. Targeting KCa3.1 channels by channel openers or L-type Ca2+ channels by DXM or simvastatin might be valuable approaches for treatment of CHI caused by mutations of KATP channels not sensitive to KATP channel openers.

Highlights

  • Congenital hyperinsulinism (CHI) is a rare heterogeneous genetic disorder, but the most frequent cause of severe, persistent hypoglycemia in neonates, infants and children

  • Glucosestimulated insulin secretion in human pancreatic b-cells is completely suppressed by pharmacologic blockage of L-type Ca2+ channels [61]

  • As control we tested the effect of the Ltype Ca2+ channel blocker nifedipine on [Ca2+]c in human islet cell clusters isolated from tissue of a patient with diffuse CHI (Table 1: 7)

Read more

Summary

Introduction

Congenital hyperinsulinism (CHI) is a rare heterogeneous genetic disorder, but the most frequent cause of severe, persistent hypoglycemia in neonates, infants and children. The main reasons for developing CHI are defects in important genes regulating pancreatic b-cell function. ABCC8 and KCNJ11 genes encode the KATP channel subunits SUR1 and Kir6.2, respectively, and mutations in these genes represent the most prevalent cause of CHI. Defects in these genes are responsible for the failure of b-cells to respond to normal regulatory mechanisms, leading to inappropriate and excessive insulin release despite low blood glucose concentrations resulting in frequent episodes of hypoglycemia [1, 2]. There are some excellent reviews giving detailed information about molecular mechanisms underlying the pathophysiology of CHI [1,2,3,4,5]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call