8
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The LRRC8 volume‐regulated anion channel inhibitor, DCPIB, inhibits mitochondrial respiration independently of the channel

      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

          There has been a resurgence of interest in the volume‐regulated anion channel (VRAC) since the recent cloning of the LRRC8A‐E gene family that encodes VRAC. The channel is a heteromer comprised of LRRC8A and at least one other family member; disruption of LRRC8A expression abolishes VRAC activity. The best‐in‐class VRAC inhibitor, DCPIB, suffers from off‐target activity toward several different channels and transporters. Considering that some anion channel inhibitors also suppress mitochondrial respiration, we systematically explored whether DCPIB inhibits respiration in wild type (WT) and LRRC8A‐knockout HAP‐1 and HEK‐293 cells. Knockout of LRRC8A had no apparent effects on cell morphology, proliferation rate, mitochondrial content, or expression of several mitochondrial genes in HAP‐1 cells. Addition of 10 µM DCPIB, a concentration typically used to inhibit VRAC, suppressed basal and ATP‐linked respiration in part through uncoupling the inner mitochondrial membrane (IMM) proton gradient and membrane potential. Additionally, DCPIB inhibits the activity of complex I, II, and III of the electron transport chain (ETC). Surprisingly, the effects of DCPIB on mitochondrial function are also observed in HAP‐1 and HEK‐293 cells which lack LRRC8A expression. Finally, we demonstrate that DCPIB activates ATP‐inhibitable potassium channels comprised of heterologously expressed Kir6.2 and SUR1 subunits. These data indicate that DCPIB suppresses mitochondrial respiration and ATP production by dissipating the mitochondrial membrane potential and inhibiting complexes I‐III of the ETC. They further justify the need for the development of sharper pharmacological tools for evaluating the integrative physiology and therapeutic potential of VRAC in human diseases.

          Abstract

          There has been a resurgence of interest in the volume‐regulated anion channel (VRAC) since the recent cloning of the LRRC8A‐E gene family that encodes VRAC. We demonstrate that the best‐in‐class VRAC inhibitor, DCPIB, inhibits mitochondrial respiration and ATP production by dissipating the mitochondrial membrane potential and inhibiting complexes I, II, and III of the electron transport chain. More specific pharmacological tools should be developed for exploring the integrative physiology and therapeutic potential of VRAC in human diseases.

          Related collections

          Most cited references44

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

          KATP channels as molecular sensors of cellular metabolism.

          In responding to cytoplasmic nucleotide levels, ATP-sensitive potassium (K(ATP)) channel activity provides a unique link between cellular energetics and electrical excitability. Over the past ten years, a steady drumbeat of crystallographic and electrophysiological studies has led to detailed structural and kinetic models that define the molecular basis of channel activity. In parallel, the uncovering of disease-causing mutations of K(ATP) has led to an explanation of the molecular basis of disease and, in turn, to a better understanding of the structural basis of channel function.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            SWELL1, a plasma membrane protein, is an essential component of volume-regulated anion channel.

            Maintenance of a constant cell volume in response to extracellular or intracellular osmotic changes is critical for cellular homeostasis. Activation of a ubiquitous volume-regulated anion channel (VRAC) plays a key role in this process; however, its molecular identity in vertebrates remains unknown. Here, we used a cell-based fluorescence assay and performed a genome-wide RNAi screen to find components of VRAC. We identified SWELL1 (LRRC8A), a member of a four-transmembrane protein family with unknown function, as essential for hypotonicity-induced iodide influx. SWELL1 is localized to the plasma membrane, and its knockdown dramatically reduces endogenous VRAC currents and regulatory cell volume decrease in various cell types. Furthermore, point mutations in SWELL1 cause a significant change in VRAC anion selectivity, demonstrating that SWELL1 is an essential VRAC component. These findings enable further molecular characterization of the VRAC channel complex and genetic studies for understanding the function of VRAC in normal physiology and disease. Copyright © 2014 Elsevier Inc. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Reconstitution of IKATP: an inward rectifier subunit plus the sulfonylurea receptor.

              A member of the inwardly rectifying potassium channel family was cloned here. The channel, called BIR (Kir6.2), was expressed in large amounts in rat pancreatic islets and glucose-responsive insulin-secreting cell lines. Coexpression with the sulfonylurea receptor SUR reconstituted an inwardly rectifying potassium conductance of 76 picosiemens that was sensitive to adenosine triphosphate (ATP) (IKATP) and was inhibited by sulfonylureas and activated by diazoxide. The data indicate that these pancreatic beta cell potassium channels are a complex composed of at least two subunits--BIR, a member of the inward rectifier potassium channel family, and SUR, a member of the ATP-binding cassette superfamily. Gene mapping data show that these two potassium channel subunit genes are clustered on human chromosome 11 at position 11p15.1.
                Bookmark

                Author and article information

                Contributors
                aqeela.afzal@vumc.org
                jerod.s.denton@vumc.org
                Journal
                Physiol Rep
                Physiol Rep
                10.1002/(ISSN)2051-817X
                PHY2
                physreports
                Physiological Reports
                John Wiley and Sons Inc. (Hoboken )
                2051-817X
                09 December 2019
                December 2019
                : 7
                : 23 ( doiID: 10.14814/phy2.v7.23 )
                : e14303
                Affiliations
                [ 1 ] Department of Neurological Surgery Vanderbilt University Nashville Tennessee
                [ 2 ] Department of Medicine Vanderbilt University Nashville Tennessee
                [ 3 ] Department of Pharmacology Vanderbilt University Nashville Tennessee
                [ 4 ] Department of Anesthesiology Vanderbilt University Medical Center Nashville Tennessee
                [ 5 ] Agilent Technologies Santa Clara California
                [ 6 ] Vanderbilt Vaccine Center Vanderbilt University Medical Center Nashville Tennessee
                Author notes
                [*] [* ] Correspondence

                Aqeela Afzal, T4224, Medical Center North, 1161 21st Avenue South, Vanderbilt University Medical Center, Nashville, TN 37232.

                Email: aqeela.afzal@ 123456vumc.org

                and

                Jerod S. Denton, T4208 Medical Center North, 1161 21st Avenue South, Vanderbilt University Medical Center, Nashville, TN 37232.

                Email: jerod.s.denton@ 123456vumc.org

                Author information
                https://orcid.org/0000-0003-0032-8586
                Article
                PHY214303
                10.14814/phy2.14303
                6900491
                31814333
                88535362-664f-4a5b-8d2f-9e9ffac7f6da
                © 2019 The Authors. Physiological Reports published by Wiley Periodicals, Inc. on behalf of The Physiological Society and the American Physiological Society.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                Page count
                Figures: 8, Tables: 1, Pages: 15, Words: 17000
                Funding
                Funded by: National Institute of Diabetes and Digestive and Kidney Diseases , open-funder-registry 10.13039/100000062;
                Award ID: 1F31DK120225‐01
                Award ID: R01 DK51610
                Categories
                Cellular Physiology
                Membrane Physiology
                Original Research
                Original Research
                Custom metadata
                2.0
                December 2019
                Converter:WILEY_ML3GV2_TO_JATSPMC version:5.7.2 mode:remove_FC converted:09.12.2019

                dcpib,lrrc8,mitochondria,respiration
                dcpib, lrrc8, mitochondria, respiration

                Comments

                Comment on this article