1
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Possible New Strategies for the Treatment of Congenital Hyperinsulinism

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Objective

          Congenital 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 Methods

          In 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 + (K ATP) channels. Additionally, K ATP channel-independent targets as Ca 2+-activated K + channels of intermediate conductance (K Ca3.1) and L-type Ca 2+ 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 Ca 2+ concentration ([Ca 2+] 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.

          Results

          The selective K ATP channel opener NN414 (5 µM) diminished [Ca 2+] 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 K ATP channels (SUR1 -/-) the drug was without effect. VU0071063 (30 µM), another K ATP channel opener considered to be selective, lowered [Ca 2+] c in human CHI islet cell clusters. The compound was also effective in islet cell clusters from SUR1 -/- mice, showing that [Ca 2+] c is influenced by additional effects besides K ATP channels. Contrasting to NN414, the drug depolarized ΔΨ in murine islet cell clusters pointing to severe interference with mitochondrial metabolism. An opener of K Ca3.1 channels, DCEBIO (100 µM), significantly decreased [Ca 2+] c in SUR1 -/- and human CHI islet cell clusters. To target L-type Ca 2+ channels we tested two already approved drugs, dextromethorphan (DXM) and simvastatin. DXM (100 µM) efficiently diminished [Ca 2+] c in stimulated human CHI islet cell clusters as well as in stimulated SUR1 -/- islet cell clusters. Similar effects on [Ca 2+] c were observed in experiments with simvastatin (7.2 µM).

          Conclusions

          NN414 seems to provide a good alternative to the currently used K ATP channel opener diazoxide. Targeting K Ca3.1 channels by channel openers or L-type Ca 2+ channels by DXM or simvastatin might be valuable approaches for treatment of CHI caused by mutations of K ATP channels not sensitive to K ATP channel openers.

          Related collections

          Most cited references122

          • Record: found
          • Abstract: found
          • Article: not found

          TOR signaling in growth and metabolism.

          The target of rapamycin (TOR) is a conserved Ser/Thr kinase that regulates cell growth and metabolism in response to environmental cues. Here, highlighting contributions from studies in model organisms, we review mammalian TOR complexes and the signaling branches they mediate. TOR is part of two distinct multiprotein complexes, TOR complex 1 (TORC1), which is sensitive to rapamycin, and TORC2, which is not. The physiological consequences of mammalian TORC1 dysregulation suggest that inhibitors of mammalian TOR may be useful in the treatment of cancer, cardiovascular disease, autoimmunity, and metabolic disorders.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Statin-Associated Side Effects.

            Hydroxy-methyl-glutaryl-coenzyme A (HMG-CoA) reductase inhibitors or statins are well tolerated, but associated with various statin-associated symptoms (SAS), including statin-associated muscle symptoms (SAMS), diabetes mellitus (DM), and central nervous system complaints. These are "statin-associated symptoms" because they are rare in clinical trials, making their causative relationship to statins unclear. SAS are, nevertheless, important because they prompt dose reduction or discontinuation of these life-saving mediations. SAMS is the most frequent SAS, and mild myalgia may affect 5% to 10% of statin users. Clinically important muscle symptoms, including rhabdomyolysis and statin-induced necrotizing autoimmune myopathy (SINAM), are rare. Antibodies against HMG-CoA reductase apparently provoke SINAM. Good evidence links statins to DM, but evidence linking statins to other SAS is largely anecdotal. Management of SAS requires making the possible diagnosis, altering or discontinuing the statin treatment, and using alternative lipid-lowering therapy.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Voltage-gated ion channels in human pancreatic beta-cells: electrophysiological characterization and role in insulin secretion.

              To characterize the voltage-gated ion channels in human beta-cells from nondiabetic donors and their role in glucose-stimulated insulin release. Insulin release was measured from intact islets. Whole-cell patch-clamp experiments and measurements of cell capacitance were performed on isolated beta-cells. The ion channel complement was determined by quantitative PCR. Human beta-cells express two types of voltage-gated K(+) currents that flow through delayed rectifying (K(V)2.1/2.2) and large-conductance Ca(2+)-activated K(+) (BK) channels. Blockade of BK channels (using iberiotoxin) increased action potential amplitude and enhanced insulin secretion by 70%, whereas inhibition of K(V)2.1/2.2 (with stromatoxin) was without stimulatory effect on electrical activity and secretion. Voltage-gated tetrodotoxin (TTX)-sensitive Na(+) currents (Na(V)1.6/1.7) contribute to the upstroke of action potentials. Inhibition of Na(+) currents with TTX reduced glucose-stimulated (6-20 mmol/l) insulin secretion by 55-70%. Human beta-cells are equipped with L- (Ca(V)1.3), P/Q- (Ca(V)2.1), and T- (Ca(V)3.2), but not N- or R-type Ca(2+) channels. Blockade of L-type channels abolished glucose-stimulated insulin release, while inhibition of T- and P/Q-type Ca(2+) channels reduced glucose-induced (6 mmol/l) secretion by 60-70%. Membrane potential recordings suggest that L- and T-type Ca(2+) channels participate in action potential generation. Blockade of P/Q-type Ca(2+) channels suppressed exocytosis (measured as an increase in cell capacitance) by >80%, whereas inhibition of L-type Ca(2+) channels only had a minor effect. Voltage-gated T-type and L-type Ca(2+) channels as well as Na(+) channels participate in glucose-stimulated electrical activity and insulin secretion. Ca(2+)-activated BK channels are required for rapid membrane repolarization. Exocytosis of insulin-containing granules is principally triggered by Ca(2+) influx through P/Q-type Ca(2+) channels.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Endocrinol (Lausanne)
                Front Endocrinol (Lausanne)
                Front. Endocrinol.
                Frontiers in Endocrinology
                Frontiers Media S.A.
                1664-2392
                27 October 2020
                2020
                : 11
                : 545638
                Affiliations
                [1] 1Department of Pharmacology, Institute of Pharmacy, University of Tübingen , Tübingen, Germany
                [2] 2Department of Pharmacology, Institute of Pharmaceutical and Medicinal Chemistry, University of Münster , Münster, Germany
                [3] 3Department of Pediatric Surgery, University Medicine Greifswald , Greifswald, Germany
                [4] 4Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald , Greifswald, Germany
                [5] 5Institute of Human Genetics, University Hospital Magdeburg , Magdeburg, Germany
                Author notes

                Edited by: Jeff M. P. Holly, University of Bristol, United Kingdom

                Reviewed by: Charles Alfred Stanley, Children’s Hospital of Philadelphia, United States; Hana Zemkova, Institute of Physiology (ASCR), Czechia

                *Correspondence: Peter Krippeit-Drews, peter.krippeit-drews@ 123456uni-tuebingen.de

                This article was submitted to Cellular Endocrinology, a section of the journal Frontiers in Endocrinology

                Article
                10.3389/fendo.2020.545638
                7653201
                33193079
                1ccf1c55-c563-4945-ba53-a50821007edd
                Copyright © 2020 Sikimic, Hoffmeister, Gresch, Kaiser, Barthlen, Wolke, Wieland, Lendeckel, Krippeit-Drews, Düfer and Drews

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 25 March 2020
                : 02 October 2020
                Page count
                Figures: 7, Tables: 1, Equations: 0, References: 122, Pages: 16, Words: 8096
                Funding
                Funded by: Deutsche Forschungsgemeinschaft 10.13039/501100001659
                Categories
                Endocrinology
                Original Research

                Endocrinology & Diabetes
                congenital hyperinsulinism,katp channels,diazoxide,nn414,l-type ca2+ channels,kca3.1 channels

                Comments

                Comment on this article