3
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Impact of arrhythmogenic calmodulin variants on small conductance Ca 2+‐activated K + (SK3) channels

      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

          Calmodulin (CaM) is a ubiquitous Ca 2+‐sensing protein regulating many important cellular processes. Several CaM‐associated variants have been identified in a small group of patients with cardiac arrhythmias. The mechanism remains largely unknown, even though a number of ion channels, including the ryanodine receptors and the L‐type calcium channels have been shown to be functionally affected by the presence of mutant CaM. CaM is constitutively bound to the SK channel, which underlies the calcium‐gated I SK contributing to cardiac repolarization. The CaM binding to SK channels is essential for gating, correct assembly, and membrane expression. To elucidate the effect of nine different arrhythmogenic CaM variants on SK3 channel function, HEK293 cells stably expressing SK3 were transiently co‐transfected with CaM WT or variant and whole‐cell patch‐clamp recordings were performed with a calculated free Ca 2+ concentration of 400 nmol/L. MDCK cells were transiently transfected with SK3 and/or CaM WT or variant to address SK3 and CaM localization by immunocytochemistry. The LQTS‐associated variants CaM D96V, CaM D130G, and CaM F142L reduced I SK,Ca compared with CaM WT ( P < 0.01, P < 0.001, and P < 0.05, respectively ). The CPVT associated variant CaM N54I also reduced the I SK,Ca ( P < 0.05), which was linked to an accumulation of SK3/CaM N54I channel complexes in intracellular compartments ( P < 0.05). The CPVT associated variants, CaM A103V and CaM D132E only revealed a tendency toward reduced current, while the variants CaM F90L and CaM N98S, causing LQTS syndrome, did not have any impact on I SK,Ca. In conclusion, we found that the arrhythmogenic CaM variants CaM N54I, CaM D96V, CaM D130G, and CaM F142L significantly down‐regulate the SK3 channel current, but with distinct mechanism.

          Related collections

          Most cited references42

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

          Structure of the gating domain of a Ca2+-activated K+ channel complexed with Ca2+/calmodulin.

          Small-conductance Ca2+-activated K+ channels (SK channels) are independent of voltage and gated solely by intracellular Ca2+. These membrane channels are heteromeric complexes that comprise pore-forming alpha-subunits and the Ca2+-binding protein calmodulin (CaM). CaM binds to the SK channel through the CaM-binding domain (CaMBD), which is located in an intracellular region of the alpha-subunit immediately carboxy-terminal to the pore. Channel opening is triggered when Ca2+ binds the EF hands in the N-lobe of CaM. Here we report the 1.60 A crystal structure of the SK channel CaMBD/Ca2+/CaM complex. The CaMBD forms an elongated dimer with a CaM molecule bound at each end; each CaM wraps around three alpha-helices, two from one CaMBD subunit and one from the other. As only the CaM N-lobe has bound Ca2+, the structure provides a view of both calcium-dependent and -independent CaM/protein interactions. Together with biochemical data, the structure suggests a possible gating mechanism for the SK channel.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The many faces of calmodulin in cell proliferation, programmed cell death, autophagy, and cancer.

            Calmodulin (CaM) is a ubiquitous Ca(2+) receptor protein mediating a large number of signaling processes in all eukaryotic cells. CaM plays a central role in regulating a myriad of cellular functions via interaction with multiple target proteins. This review focuses on the action of CaM and CaM-dependent signaling systems in the control of vertebrate cell proliferation, programmed cell death and autophagy. The significance of CaM and interconnected CaM-regulated systems for the physiology of cancer cells including tumor stem cells, and processes required for tumor progression such as growth, tumor-associated angiogenesis and metastasis are highlighted. Furthermore, the potential targeting of CaM-dependent signaling processes for therapeutic use is discussed.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Small-conductance, calcium-activated potassium channels from mammalian brain.

              Members of a previously unidentified family of potassium channel subunits were cloned from rat and human brain. The messenger RNAs encoding these subunits were widely expressed in brain with distinct yet overlapping patterns, as well as in several peripheral tissues. Expression of the messenger RNAs in Xenopus oocytes resulted in calcium-activated, voltage-independent potassium channels. The channels that formed from the various subunits displayed differential sensitivity to apamin and tubocurare. The distribution, function, and pharmacology of these channels are consistent with the SK class of small-conductance, calcium-activated potassium channels, which contribute to the afterhyperpolarization in central neurons and other cell types.
                Bookmark

                Author and article information

                Contributors
                arnela@sund.ku.dk
                Journal
                Physiol Rep
                Physiol Rep
                10.1002/(ISSN)2051-817X
                PHY2
                physreports
                Physiological Reports
                John Wiley and Sons Inc. (Hoboken )
                2051-817X
                06 October 2019
                October 2019
                : 7
                : 19 ( doiID: 10.14814/phy2.v7.19 )
                : e14210
                Affiliations
                [ 1 ] Laboratory of Cardiac Physiology, Faculty of Health and Medical Sciences, Department of Biomedical Sciences University of Copenhagen Copenhagen Denmark
                [ 2 ] Cell Biology and Physiology, Department of Biology University of Copenhagen Copenhagen Denmark
                [ 3 ] H. Lundbech A/S Valby Denmark
                Author notes
                [*] [* ] Correspondence

                Arnela Saljic, Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK‐2200 Copenhagen, Denmark.

                Tel: +45 42360411

                E‐mail: arnela@ 123456sund.ku.dk

                Author information
                https://orcid.org/0000-0001-8910-0902
                Article
                PHY214210
                10.14814/phy2.14210
                6778599
                31587513
                db537af2-fcca-4f25-bce9-9ac00e2a9ea4
                © 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
                : 26 July 2019
                : 31 July 2019
                Page count
                Figures: 9, Tables: 0, Pages: 13, Words: 14515
                Funding
                Funded by: European Union’s Horizon 2020 research and innovation programme under the Marie Sklodowska‐Curie grant
                Award ID: No. 675351
                Funded by: Novo Nordisk Foundation Synergy program
                Categories
                Cardiovascular Conditions, Disorders and Treatments
                Cellular and Molecular Physiology
                Original Research
                Original Research
                Custom metadata
                2.0
                phy214210
                October 2019
                Converter:WILEY_ML3GV2_TO_NLMPMC version:5.7.0 mode:remove_FC converted:06.10.2019

                calmodulin,cardiac arrhythmias,channelopathies,cpvt,lqts,small conductance ca+‐activated k+ channels

                Comments

                Comment on this article