22
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Drug Repurposing: The Anthelmintics Niclosamide and Nitazoxanide Are Potent TMEM16A Antagonists That Fully Bronchodilate Airways

      research-article
      1 , 2 , 1 , 3 , 1 , 1 , 1 , 4 , 4 , 5 , 6 , 7 , 8 , 6 , 7 , 8 , 1 , 9 , 1 , 1 , 1 , 1 , 5 , 1 , 5 , 5 , 5 , 5 , 1 , 1 , 3 , 2 , 3 , 10 , 1 , * , 2 , * , 1 , *
      Frontiers in Pharmacology
      Frontiers Media S.A.
      TMEM16A antagonist, niclosamide, nitazoxanide, bronchodilator, desensitization, airway smooth muscle (ASM), calcium-activated chloride channel, drug repositioning

      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 is an unmet need in severe asthma where approximately 40% of patients exhibit poor β-agonist responsiveness, suffer daily symptoms and show frequent exacerbations. Antagonists of the Ca 2+-activated Cl channel, TMEM16A, offers a new mechanism to bronchodilate airways and block the multiple contractiles operating in severe disease. To identify TMEM16A antagonists we screened a library of ∼580,000 compounds. The anthelmintics niclosamide, nitazoxanide, and related compounds were identified as potent TMEM16A antagonists that blocked airway smooth muscle depolarization and contraction. To evaluate whether TMEM16A antagonists resist use- and inflammatory-desensitization pathways limiting β-agonist action, we tested their efficacy under harsh conditions using maximally contracted airways or airways pretreated with a cytokine cocktail. Stunningly, TMEM16A antagonists fully bronchodilated airways, while the β-agonist isoproterenol showed only partial effects. Thus, antagonists of TMEM16A and repositioning of niclosamide and nitazoxanide represent an important additional treatment for patients with severe asthma and COPD that is poorly controlled with existing therapies. It is of note that drug repurposing has also attracted wide interest in niclosamide and nitazoxanide as a new treatment for cancer and infectious disease. For the first time we identify TMEM16A as a molecular target for these drugs and thus provide fresh insights into their mechanism for the treatment of these disorders in addition to respiratory disease.

          Related collections

          Most cited references94

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

          TMEM16A confers receptor-activated calcium-dependent chloride conductance.

          Calcium (Ca(2+))-activated chloride channels are fundamental mediators in numerous physiological processes including transepithelial secretion, cardiac and neuronal excitation, sensory transduction, smooth muscle contraction and fertilization. Despite their physiological importance, their molecular identity has remained largely unknown. Here we show that transmembrane protein 16A (TMEM16A, which we also call anoctamin 1 (ANO1)) is a bona fide Ca(2+)-activated chloride channel that is activated by intracellular Ca(2+) and Ca(2+)-mobilizing stimuli. With eight putative transmembrane domains and no apparent similarity to previously characterized channels, ANO1 defines a new family of ionic channels. The biophysical properties as well as the pharmacological profile of ANO1 are in full agreement with native Ca(2+)-activated chloride currents. ANO1 is expressed in various secretory epithelia, the retina and sensory neurons. Furthermore, knockdown of mouse Ano1 markedly reduced native Ca(2+)-activated chloride currents as well as saliva production in mice. We conclude that ANO1 is a candidate Ca(2+)-activated chloride channel that mediates receptor-activated chloride currents in diverse physiological processes.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            TMEM16A, a membrane protein associated with calcium-dependent chloride channel activity.

            Calcium-dependent chloride channels are required for normal electrolyte and fluid secretion, olfactory perception, and neuronal and smooth muscle excitability. The molecular identity of these membrane proteins is still unclear. Treatment of bronchial epithelial cells with interleukin-4 (IL-4) causes increased calcium-dependent chloride channel activity, presumably by regulating expression of the corresponding genes. We performed a global gene expression analysis to identify membrane proteins that are regulated by IL-4. Transfection of epithelial cells with specific small interfering RNA against each of these proteins shows that TMEM16A, a member of a family of putative plasma membrane proteins with unknown function, is associated with calcium-dependent chloride current, as measured with halide-sensitive fluorescent proteins, short-circuit current, and patch-clamp techniques. Our results indicate that TMEM16A is an intrinsic constituent of the calcium-dependent chloride channel. Identification of a previously unknown family of membrane proteins associated with chloride channel function will improve our understanding of chloride transport physiopathology and allow for the development of pharmacological tools useful for basic research and drug development.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Expression cloning of TMEM16A as a calcium-activated chloride channel subunit.

              Calcium-activated chloride channels (CaCCs) are major regulators of sensory transduction, epithelial secretion, and smooth muscle contraction. Other crucial roles of CaCCs include action potential generation in Characean algae and prevention of polyspermia in frog egg membrane. None of the known molecular candidates share properties characteristic of most CaCCs in native cells. Using Axolotl oocytes as an expression system, we have identified TMEM16A as the Xenopus oocyte CaCC. The TMEM16 family of "transmembrane proteins with unknown function" is conserved among eukaryotes, with family members linked to tracheomalacia (mouse TMEM16A), gnathodiaphyseal dysplasia (human TMEM16E), aberrant X segregation (a Drosophila TMEM16 family member), and increased sodium tolerance (yeast TMEM16). Moreover, mouse TMEM16A and TMEM16B yield CaCCs in Axolotl oocytes and mammalian HEK293 cells and recapitulate the broad CaCC expression. The identification of this new family of ion channels may help the development of CaCC modulators for treating diseases including hypertension and cystic fibrosis.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Pharmacol
                Front Pharmacol
                Front. Pharmacol.
                Frontiers in Pharmacology
                Frontiers Media S.A.
                1663-9812
                14 February 2019
                2019
                : 10
                : 51
                Affiliations
                [1] 1Department of Inflammation Research, Amgen Inc. , Thousand Oaks, CA, United States
                [2] 2Department of Therapeutic Discovery, Amgen Inc. , Regensburg, Germany
                [3] 3Department of Therapeutic Discovery, Amgen Inc. , Thousand Oaks, CA, United States
                [4] 4Department of Medicinal Chemistry, Amgen Inc. , Thousand Oaks, CA, United States
                [5] 5Department of Inflammation Research, Amgen Inc. , Seattle, WA, United States
                [6] 6Department of Comparative Biology and Safety Sciences, Amgen Inc. , Seattle, WA, United States
                [7] 7Department of Comparative Biology and Safety Sciences, Amgen Inc. , Thousand Oaks, CA, United States
                [8] 8Department of Comparative Biology and Safety Sciences, Amgen Inc. , South San Francisco, CA, United States
                [9] 9Genome Analysis Unit, Amgen Inc. , South San Francisco, CA, United States
                [10] 10Institut für Physiologie, Universität Regensburg , Regensburg, Germany
                Author notes

                Edited by: Heike Wulff, University of California, Davis, United States

                Reviewed by: George Chandy, University of California, Irvine, United States; Peter Bradding, University of Leicester, United Kingdom

                Present address: Katja Labitzke, Clariant Produkte (Deutschland) GmbH, Planegg, Germany, Longbin Liu, CHDI Management, CHDI Foundation, Los Angeles, CA, United States, Anh Leith, Department of Genome Sciences, University of Washington, Seattle, WA, United States, Esther Trueblood, Seattle Genetics, Bothell, WA, United States, Kelly Hensley, Seattle Genetics, Bothell, WA, United States, Teresa L. Born, Sartorius Stedim BioOutsource, Cambridge, MA, United States, Alison Budelsky, Immunology Research, Lilly Research Laboratories, San Diego, CA, United States, Dirk Smith, Insight BioConsulting, Bainbridge Island, WA, United States, Kerstin Weikl, Assay.Works GmbH, Regensburg, Germany, Andreas Hochheimer, ISAR Bioscience GmbH, Planegg, Germany, John K. Sullivan, PolestarBio LLC, Newbury Park, CA, United States

                This article was submitted to Respiratory Pharmacology, a section of the journal Frontiers in Pharmacology

                Article
                10.3389/fphar.2019.00051
                6382696
                30837866
                2836610d-68db-4cf8-9895-c4261dee3f06
                Copyright © 2019 Miner, Labitzke, Liu, Wang, Henckels, Gaida, Elliott, Chen, Liu, Leith, Trueblood, Hensley, Xia, Homann, Bennett, Fiorino, Whoriskey, Yu, Escobar, Wong, Born, Budelsky, Comeau, Smith, Phillips, Johnston, McGivern, Weikl, Powers, Kunzelmann, Mohn, Hochheimer and Sullivan.

                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
                : 23 November 2018
                : 18 January 2019
                Page count
                Figures: 11, Tables: 1, Equations: 0, References: 112, Pages: 34, Words: 0
                Funding
                Funded by: Amgen 10.13039/100002429
                Categories
                Pharmacology
                Original Research

                Pharmacology & Pharmaceutical medicine
                tmem16a antagonist,niclosamide,nitazoxanide,bronchodilator,desensitization,airway smooth muscle (asm),calcium-activated chloride channel,drug repositioning

                Comments

                Comment on this article