4
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Myosin-II mediated traction forces evoke localized Piezo1-dependent Ca 2+ flickers

      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

          Piezo channels transduce mechanical stimuli into electrical and chemical signals to powerfully influence development, tissue homeostasis, and regeneration. Studies on Piezo1 have largely focused on transduction of “outside-in” mechanical forces, and its response to internal, cell-generated forces remains poorly understood. Here, using measurements of endogenous Piezo1 activity and traction forces in native cellular conditions, we show that cellular traction forces generate spatially-restricted Piezo1-mediated Ca 2+ flickers in the absence of externally-applied mechanical forces. Although Piezo1 channels diffuse readily in the plasma membrane and are widely distributed across the cell, their flicker activity is enriched near force-producing adhesions. The mechanical force that activates Piezo1 arises from Myosin II phosphorylation by Myosin Light Chain Kinase. We propose that Piezo1 Ca 2+ flickers allow spatial segregation of mechanotransduction events, and that mobility allows Piezo1 channels to explore a large number of mechanical microdomains and thus respond to a greater diversity of mechanical cues.

          all

          Ellefsen et al. monitor Piezo-dependent Calcium signals in live cells by TIRF and super-resolution microscopy and find that Ca 2+ flickers localize to areas of high traction force. They show that Myosin II activity and MLCK are needed for the generation of Piezo Ca 2+ signals and that Piezo1 channels are mobile in the plasma membrane.

          Related collections

          Most cited references50

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

          Piezos are pore-forming subunits of mechanically activated channels

          Mechanotransduction plays a crucial role in physiology. Biological processes including sensing touch and sound waves require yet unidentified cation channels that detect pressure. Mouse piezo1 (mpiezo1) and mpiezo2 induce mechanically activated cationic currents in cells; however, it is unknown if piezos are pore-forming ion channels or modulate ion channels. We show that Drosophila piezo (dpiezo) also induces mechanically activated currents in cells, but through channels with remarkably distinct pore properties including sensitivity to the pore blocker ruthenium red and single channel conductances. mpiezo1 assembles as a ~1.2 million-Dalton tetramer, with no evidence of other proteins in this complex. Finally, purified mpiezo1 reconstituted into asymmetric lipid bilayers and liposomes forms ruthenium red-sensitive ion channels. These data demonstrate that piezos are an evolutionarily conserved ion channel family involved in mechanotransduction.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Piezo1, a mechanically activated ion channel, is required for vascular development in mice.

            Mechanosensation is perhaps the last sensory modality not understood at the molecular level. Ion channels that sense mechanical force are postulated to play critical roles in a variety of biological processes including sensing touch/pain (somatosensation), sound (hearing), and shear stress (cardiovascular physiology); however, the identity of these ion channels has remained elusive. We previously identified Piezo1 and Piezo2 as mechanically activated cation channels that are expressed in many mechanosensitive cell types. Here, we show that Piezo1 is expressed in endothelial cells of developing blood vessels in mice. Piezo1-deficient embryos die at midgestation with defects in vascular remodeling, a process critically influenced by blood flow. We demonstrate that Piezo1 is activated by shear stress, the major type of mechanical force experienced by endothelial cells in response to blood flow. Furthermore, loss of Piezo1 in endothelial cells leads to deficits in stress fiber and cellular orientation in response to shear stress, linking Piezo1 mechanotransduction to regulation of cell morphology. These findings highlight an essential role of mammalian Piezo1 in vascular development during embryonic development.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Mechanical stretch triggers rapid epithelial cell division through Piezo1

              Despite acting as a barrier for the organs they encase, epithelial cells turnover at some of the fastest rates in the body. Yet, epithelial cell division must be tightly linked to cell death to preserve barrier function and prevent tumour formation. How do the number of dying cells match those dividing to maintain constant numbers? We previously found that when epithelial cells become too crowded, they activate the stretch-activated channel Piezo1 to trigger extrusion of cells that later die 1 . Conversely, what controls epithelial cell division to balance cell death at steady state? Here, we find that cell division occurs in regions of low cell density, where epithelial cells are stretched. By experimentally stretching epithelia, we find that mechanical stretch itself rapidly stimulates cell division through activation of the same Piezo1 channel. To do so, stretch triggers cells paused in early G2 to activate calcium-dependent ERK1/2 phosphorylation that activates cyclin B transcription necessary to drive cells into mitosis. Although both epithelial cell division and cell extrusion require Piezo1 at steady state, the type of mechanical force controls the outcome: stretch induces cell division whereas crowding induces extrusion. How Piezo1-dependent calcium transients activate two opposing processes may depend on where and how Piezo1 is activated since it accumulates in different subcellular sites with increasing cell density. In sparse epithelial regions where cells divide, Piezo1 localizes to the plasma membrane and cytoplasm whereas in dense regions where cells extrude, it forms large cytoplasmic aggregates. Because Piezo1 senses both mechanical crowding and stretch, it may act as a homeostatic sensor to control epithelial cell numbers, triggering extrusion/apoptosis in crowded regions and cell division in sparse regions.
                Bookmark

                Author and article information

                Contributors
                medhap@uci.edu
                Journal
                Commun Biol
                Commun Biol
                Communications Biology
                Nature Publishing Group UK (London )
                2399-3642
                7 August 2019
                7 August 2019
                2019
                : 2
                : 298
                Affiliations
                [1 ]ISNI 0000 0001 0668 7243, GRID grid.266093.8, Department of Neurobiology & Behavior, UC Irvine, ; Irvine, CA 92697 USA
                [2 ]ISNI 0000 0001 0668 7243, GRID grid.266093.8, Department of Physiology & Biophysics, UC Irvine, ; Irvine, CA 92697 USA
                [3 ]ISNI 0000 0001 0668 7243, GRID grid.266093.8, Sue and Bill Gross Stem Cell Research Center, UC Irvine, ; Irvine, CA 92697 USA
                [4 ]ISNI 0000 0001 0668 7243, GRID grid.266093.8, Center for Complex Biological Systems, UC Irvine, ; Irvine, CA 92697 USA
                [5 ]ISNI 0000000419368956, GRID grid.168010.e, Department of Chemical Engineering, , Stanford University, ; Stanford, CA 94305 USA
                [6 ]ISNI 0000 0001 0668 7243, GRID grid.266093.8, Department of Biomedical Engineering, UC Irvine, ; Irvine, CA 92697 USA
                [7 ]ISNI 0000 0001 0668 7243, GRID grid.266093.8, Department of Neurology, UC Irvine, ; Irvine, CA 92697 USA
                [8 ]ISNI 0000000419368956, GRID grid.168010.e, Stanford Cardiovascular Institute, Stanford School of Medicine, ; Stanford, CA 94305 USA
                Author information
                http://orcid.org/0000-0002-4019-3797
                http://orcid.org/0000-0001-8136-6394
                http://orcid.org/0000-0003-2537-0879
                http://orcid.org/0000-0002-9315-0352
                http://orcid.org/0000-0002-6518-3085
                Article
                514
                10.1038/s42003-019-0514-3
                6685976
                31396578
                a041fbae-d848-4ee7-b1af-617ea14f8f18
                © The Author(s) 2019

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 14 December 2018
                : 18 June 2019
                Funding
                Funded by: FundRef https://doi.org/10.13039/100006955, U.S. Department of Health & Human Services | NIH | Office of Extramural Research, National Institutes of Health (OER);
                Award ID: DP2 AT010376
                Award ID: R01 NS109810
                Award ID: R21 NS085628
                Award ID: R37 GM048071
                Award ID: F31 GM119330
                Award ID: R01 GM112998
                Award ID: T32 NS082174
                Award Recipient :
                Funded by: FundRef https://doi.org/10.13039/100000011, Howard Hughes Medical Institute (HHMI);
                Award ID: Faculty Scholars Award
                Award Recipient :
                Categories
                Article
                Custom metadata
                © The Author(s) 2018

                ion channel signalling,ion transport
                ion channel signalling, ion transport

                Comments

                Comment on this article