Abstract

ATP-sensitive potassium (KATP) channels play a key role in mediating glucose-stimulated insulin secretion by coupling metabolic signals to β-cell membrane potential. Loss of KATP channel function due to mutations in ABCC8 or KCNJ11, genes encoding the sulfonylurea receptor 1 (SUR1) or the inwardly rectifying potassium channel Kir6.2, respectively, results in congenital hyperinsulinism. Many SUR1 mutations prevent trafficking of channel proteins from the endoplasmic reticulum to the cell surface. Channel inhibitors, including sulfonylureas and carbamazepine, have been shown to correct channel trafficking defects. In the present study, we identified 13 novel SUR1 mutations that cause channel trafficking defects, the majority of which are amenable to pharmacological rescue by glibenclamide and carbamazepine. By contrast, none of the mutant channels were rescued by KATP channel openers. Cross-linking experiments showed that KATP channel inhibitors promoted interactions between the N terminus of Kir6.2 and SUR1, whereas channel openers did not, suggesting the inhibitors enhance intersubunit interactions to overcome channel biogenesis and trafficking defects. Functional studies of rescued mutant channels indicate that most mutants rescued to the cell surface exhibited WT-like sensitivity to ATP, MgADP, and diazoxide. In intact cells, recovery of channel function upon trafficking rescue by reversible sulfonylureas or carbamazepine was facilitated by the KATP channel opener diazoxide. Our study expands the list of KATP channel trafficking mutations whose function can be recovered by pharmacological ligands and provides further insight into the structural mechanism by which channel inhibitors correct channel biogenesis and trafficking defects.

Highlights

  • Protein function relies on the proper folding, assembly, and trafficking to specific cellular compartments

  • We have previously found that sulfonylureas (SUs); KATP channel inhibitors, such as glibenclamide (GBC); and tolbutamide could act as pharmacological chaperones to correct KATP channel trafficking defects [13,14,15,16]

  • Our previous studies have shown that several compounds that bind to the channel and inhibit channel activity are able to correct trafficking defects of KATP channels [13,14,15, 17, 19]

Read more

Summary

Results

Identification and Pharmacological Rescue of Novel HI-causing KATP Trafficking Mutants—In a previous study [17], we identified a new KATP channel pharmacological chaperone. The KATP Channel Opener Diazoxide Does Not Enhance SUR1-Kir6.2 Subunit Interactions as Assessed by p-Azidophenylalanine-mediated Photocross-linking—Using a genetically encoded photocross-linkable amino acid, p-azidophenylalanine, engineered into the distal N terminus of Kir6.2, we have recently shown that both GBC and CBZ promote cross-linking of the Kir6.2 N terminus to SUR1 [23] Based on this observation and findings that deleting the N terminus of Kir6.2 severely compromised the biogenesis efficiency of WT channels and prevented rescue of trafficking-impaired mutants by GBC or CBZ [23], we proposed that GBC and CBZ rescue TMD0-SUR1 trafficking mutants by promoting physical interactions between the N terminus of Kir6.2 and SUR1 to overcome channel protein folding and assembly defects caused by SUR1 TMD0 mutations. A and B, whereas overnight treatment with GBC and CBZ increased the intensity of the cross-linked Kir6.2-SUR1 band compared with control cells treated overnight with 0.1% DMSO as reported previously [23], overnight treatment with 200 ␮M diazoxide did not. Further confirm the efficacy of CBZ in rescuing this class of trafficking mutations to the cell surface

Gating Properties of Mutant Channels Rescued to the Cell
FIGURE tamide
Discussion
Experimental Procedures
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