201
|
Liu Y, Wang K. Exploiting the Diversity of Ion Channels: Modulation of Ion Channels for Therapeutic Indications. Handb Exp Pharmacol 2019; 260:187-205. [PMID: 31820177 DOI: 10.1007/164_2019_333] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Ion channels are macromolecular proteins that form water-filled pores in cell membranes and they are critical for a variety of physiological and pharmacological functions. Dysfunctional ion channels can cause diseases known as channelopathies. Ion channels are encoded by approximately 400 genes, representing the second largest class of proven drug targets for therapeutic areas including neuropsychiatric disorders, cardiovascular and metabolic diseases, immunological diseases, nephrological diseases, gastrointestinal diseases, pulmonary/respiratory diseases, and many cancers. With more ion channel structures are being solved and functional robust assays are being developed, there are tremendous opportunities for identifying specific modulators targeting ion channels for new therapy.
Collapse
Affiliation(s)
- Yani Liu
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - KeWei Wang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China.
| |
Collapse
|
202
|
Brunner JD, Schenck S. Preparation of Proteoliposomes with Purified TMEM16 Protein for Accurate Measures of Lipid Scramblase Activity. Methods Mol Biol 2019; 1949:181-199. [PMID: 30790257 DOI: 10.1007/978-1-4939-9136-5_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The distribution of different lipid species between the two leaflets is tightly regulated and underlies the concerted action of distinct catalytic entities. While flippases and floppases establish membrane asymmetry, scramblases randomize the lipid distribution and play pivotal roles during blood clotting, apoptosis, and in processes such as N-linked glycosylation of proteins. The recent discovery of TMEM16 family members acting as scramblases has led to an increasing demand for developing protocols tailored for TMEM16 proteins to enable functional investigations of their scrambling activity. Here we describe a protocol for the expression, purification, and functional reconstitution of TMEM16 proteins into preformed liposomes and measurement of their scrambling activity using fluorescence-labeled lipid derivatives. The reconstitution involves extrusion of liposomes through a membrane, destabilization of liposomes using Triton X-100, and stepwise detergent removal by adsorption on styryl-beads. The scrambling assay is based on the selective bleaching of nitrobenzoxadiazol fluorescent lipids on the outer leaflet of liposomes by the membrane-impermeant reducing agent sodium dithionite. The assay allows conclusions on the substrate specificity and on the kinetics of the transported lipids as shown with the example of a Ca2+-activated TMEM16 scramblase from the fungus Nectria haematococca (nhTMEM16).
Collapse
Affiliation(s)
- Janine Denise Brunner
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland.
| | - Stephan Schenck
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland
| |
Collapse
|
203
|
Zwicker VE, Oliveira BL, Yeo JH, Fraser ST, Bernardes GJL, New EJ, Jolliffe KA. A Fluorogenic Probe for Cell Surface Phosphatidylserine Using an Intramolecular Indicator Displacement Sensing Mechanism. Angew Chem Int Ed Engl 2018; 58:3087-3091. [DOI: 10.1002/anie.201812489] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Indexed: 11/11/2022]
Affiliation(s)
| | - Bruno L. Oliveira
- University of Cambridge Department of Chemistry Lensfield Road CB2 1EW Cambridge UK
- Universidade de Lisboa Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina Avenida Professor Egas Moniz 1649-028 Lisboa Portugal
| | - Jia Hao Yeo
- University of Sydney School of Chemistry Sydney NSW 2006 Australia
- University of Sydney School of Medical Sciences Camperdown NSW 2050 Australia
| | - Stuart T. Fraser
- University of Sydney School of Medical Sciences Camperdown NSW 2050 Australia
| | - Gonçalo J. L. Bernardes
- University of Cambridge Department of Chemistry Lensfield Road CB2 1EW Cambridge UK
- Universidade de Lisboa Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina Avenida Professor Egas Moniz 1649-028 Lisboa Portugal
| | - Elizabeth J. New
- University of Sydney School of Chemistry Sydney NSW 2006 Australia
| | | |
Collapse
|
204
|
Zwicker VE, Oliveira BL, Yeo JH, Fraser ST, Bernardes GJL, New EJ, Jolliffe KA. A Fluorogenic Probe for Cell Surface Phosphatidylserine Using an Intramolecular Indicator Displacement Sensing Mechanism. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201812489] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
| | - Bruno L. Oliveira
- University of Cambridge Department of Chemistry Lensfield Road CB2 1EW Cambridge UK
- Universidade de Lisboa Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina Avenida Professor Egas Moniz 1649-028 Lisboa Portugal
| | - Jia Hao Yeo
- University of Sydney School of Chemistry Sydney NSW 2006 Australia
- University of Sydney School of Medical Sciences Camperdown NSW 2050 Australia
| | - Stuart T. Fraser
- University of Sydney School of Medical Sciences Camperdown NSW 2050 Australia
| | - Gonçalo J. L. Bernardes
- University of Cambridge Department of Chemistry Lensfield Road CB2 1EW Cambridge UK
- Universidade de Lisboa Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina Avenida Professor Egas Moniz 1649-028 Lisboa Portugal
| | - Elizabeth J. New
- University of Sydney School of Chemistry Sydney NSW 2006 Australia
| | | |
Collapse
|
205
|
Teulon J, Planelles G, Sepúlveda FV, Andrini O, Lourdel S, Paulais M. Renal Chloride Channels in Relation to Sodium Chloride Transport. Compr Physiol 2018; 9:301-342. [DOI: 10.1002/cphy.c180024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
206
|
Roland BP, Naito T, Best JT, Arnaiz-Yépez C, Takatsu H, Yu RJ, Shin HW, Graham TR. Yeast and human P4-ATPases transport glycosphingolipids using conserved structural motifs. J Biol Chem 2018; 294:1794-1806. [PMID: 30530492 DOI: 10.1074/jbc.ra118.005876] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 11/29/2018] [Indexed: 12/21/2022] Open
Abstract
Lipid transport is an essential process with manifest importance to human health and disease. Phospholipid flippases (P4-ATPases) transport lipids across the membrane bilayer and are involved in signal transduction, cell division, and vesicular transport. Mutations in flippase genes cause or contribute to a host of diseases, such as cholestasis, neurological deficits, immunological dysfunction, and metabolic disorders. Genome-wide association studies have shown that ATP10A and ATP10D variants are associated with an increased risk of diabetes, obesity, myocardial infarction, and atherosclerosis. Moreover, ATP10D SNPs are associated with elevated levels of glucosylceramide (GlcCer) in plasma from diverse European populations. Although sphingolipids strongly contribute to metabolic disease, little is known about how GlcCer is transported across cell membranes. Here, we identify a conserved clade of P4-ATPases from Saccharomyces cerevisiae (Dnf1, Dnf2), Schizosaccharomyces pombe (Dnf2), and Homo sapiens (ATP10A, ATP10D) that transport GlcCer bearing an sn2 acyl-linked fluorescent tag. Further, we establish structural determinants necessary for recognition of this sphingolipid substrate. Using enzyme chimeras and site-directed mutagenesis, we observed that residues in transmembrane (TM) segments 1, 4, and 6 contribute to GlcCer selection, with a conserved glutamine in the center of TM4 playing an essential role. Our molecular observations help refine models for substrate translocation by P4-ATPases, clarify the relationship between these flippases and human disease, and have fundamental implications for membrane organization and sphingolipid homeostasis.
Collapse
Affiliation(s)
- Bartholomew P Roland
- From the Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37235 and
| | - Tomoki Naito
- the Graduate School of Pharmaceutical Science, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Jordan T Best
- From the Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37235 and
| | - Cayetana Arnaiz-Yépez
- From the Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37235 and
| | - Hiroyuki Takatsu
- the Graduate School of Pharmaceutical Science, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Roger J Yu
- From the Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37235 and
| | - Hye-Won Shin
- the Graduate School of Pharmaceutical Science, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Todd R Graham
- From the Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37235 and
| |
Collapse
|
207
|
Ji Q, Guo S, Wang X, Pang C, Zhan Y, Chen Y, An H. Recent advances in TMEM16A: Structure, function, and disease. J Cell Physiol 2018; 234:7856-7873. [PMID: 30515811 DOI: 10.1002/jcp.27865] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 11/13/2018] [Indexed: 12/13/2022]
Abstract
TMEM16A (also known as anoctamin 1, ANO1) is the molecular basis of the calcium-activated chloride channels, with ten transmembrane segments. Recently, atomic structures of the transmembrane domains of mouse TMEM16A (mTMEM16A) were determined by single-particle electron cryomicroscopy. This gives us a solid ground to discuss the electrophysiological properties and functions of TMEM16A. TMEM16A is reported to be dually regulated by Ca2+ and voltage. In addition, the dysfunction of TMEM16A has been found to be involved in many diseases including cystic fibrosis, various cancers, hypertension, and gastrointestinal motility disorders. TMEM16A is overexpressed in many cancers, including gastrointestinal stromal tumors, gastric cancer, head and neck squamous cell carcinoma (HNSCC), colon cancer, pancreatic ductal adenocarcinoma, and esophageal cancer. Furthermore, overexpression of TMEM16A is related to the occurrence, proliferation, and migration of tumor cells. To date, several studies have shown that many natural compounds and synthetic compounds have regulatory effects on TMEM16A. These small molecule compounds might be novel drugs for the treatment of diseases caused by TMEM16A dysfunction in the future. In addition, recent studies have shown that TMEM16A plays different roles in different diseases through different signal transduction pathways. This review discusses the topology, electrophysiological properties, modulators and functions of TMEM16A in mediates nociception, gastrointestinal dysfunction, hypertension, and cancer and focuses on multiple regulatory mechanisms regarding TMEM16A.
Collapse
Affiliation(s)
- Qiushuang Ji
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Shuai Guo
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Xuzhao Wang
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Chunli Pang
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Yong Zhan
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Yafei Chen
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Hailong An
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| |
Collapse
|
208
|
Liu X, Wang J, Sun L. Structure of the hyperosmolality-gated calcium-permeable channel OSCA1.2. Nat Commun 2018; 9:5060. [PMID: 30498218 PMCID: PMC6265326 DOI: 10.1038/s41467-018-07564-5] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/09/2018] [Indexed: 01/08/2023] Open
Abstract
In plants, hyperosmolality stimuli triggers opening of the osmosensitive channels, leading to a rapid downstream signaling cascade initiated by cytosolic calcium concentration elevation. Members of the OSCA family in Arabidopsis thaliana, identified as the hyperosmolality-gated calcium-permeable channels, have been suggested to play a key role during the initial phase of hyperosmotic stress response. Here, we report the atomic structure of Arabidopsis OSCA1.2 determined by single-particle cryo-electron microscopy. It contains 11 transmembrane helices and forms a homodimer. It is in an inactivated state, and the pore-lining residues are clearly identified. Its cytosolic domain contains a RNA recognition motif and two unique long helices. The linker between these two helices forms an anchor in the lipid bilayer and may be essential to osmosensing. The structure of AtOSCA1.2 serves as a platform for the study of the mechanism underlying osmotic stress responses and mechanosensing. In plants, hyperosmolality stimuli triggers opening of the osmosensitive channels, leading to a rapid downstream signaling cascade. Here, the authors solve the cryo-EM structure of an osmosensitive channel from Arabidopsis OSCA1.2 in its inactivated state.
Collapse
Affiliation(s)
- Xin Liu
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, China
| | - Jiawei Wang
- State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Centre for Structural Biology, School of Life Sciences, Tsinghua University, 100084, Beijing, China.
| | - Linfeng Sun
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, 230027, Hefei, China. .,CAS Centre for Excellence in Molecular Cell Science, University of Science and Technology of China, Chinese Academy of Sciences, 230027, Hefei, China.
| |
Collapse
|
209
|
Wang L, Iwasaki Y, Andra KK, Pandey K, Menon AK, Bütikofer P. Scrambling of natural and fluorescently tagged phosphatidylinositol by reconstituted G protein-coupled receptor and TMEM16 scramblases. J Biol Chem 2018; 293:18318-18327. [PMID: 30287690 PMCID: PMC6254352 DOI: 10.1074/jbc.ra118.004213] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 10/03/2018] [Indexed: 12/12/2022] Open
Abstract
Members of the G protein-coupled receptor and TMEM16 (transmembrane protein 16) protein families are phospholipid scramblases that facilitate rapid, bidirectional movement of phospholipids across a membrane bilayer in an ATP-independent manner. On reconstitution into large unilamellar vesicles, these proteins scramble more than 10,000 lipids/protein/s as measured with co-reconstituted fluorescent nitrobenzoxadiazole (NBD)-labeled phospholipids. Although NBD-labeled phospholipids are ubiquitously used as reporters of scramblase activity, it remains unclear whether the NBD modification influences the quantitative outcomes of the scramblase assay. We now report a refined biochemical approach for measuring the activity of scramblase proteins with radiolabeled natural phosphatidylinositol ([3H]PI) and exploiting the hydrolytic activity of bacterial PI-specific phospholipase C (PI-PLC) to detect the transbilayer movement of PI. PI-PLC rapidly hydrolyzed 50% of [3H]PI in large symmetric, unilamellar liposomes, corresponding to the lipid pool in the outer leaflet. On reconstitution of a crude preparation of yeast endoplasmic reticulum scramblase, purified bovine opsin, or purified Nectria haematococca TMEM16, the extent of [3H]PI hydrolysis increased, indicating that [3H]PI from the inner leaflet had been scrambled to the outer leaflet. Using transphosphatidylation, we synthesized acyl-NBD-PI and used it to compare our PI-PLC-based assay with conventional fluorescence-based methods. Our results revealed quantitative differences between the two assays that we attribute to the specific features of the assays themselves rather than to the nature of the phospholipid. In summary, we have developed an assay that measures scrambling of a chemically unmodified phospholipid by a reconstituted scramblase.
Collapse
Affiliation(s)
- Lei Wang
- From the Institute of Biochemistry and Molecular Medicine and; Graduate School for Cellular and Biochemical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Yugo Iwasaki
- the Laboratory of Molecular Biotechnology, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Nagoya 464-8601, Japan, and
| | - Kiran K Andra
- the Department of Biochemistry, Weill Cornell Medical College, New York, New York 10065
| | - Kalpana Pandey
- the Department of Biochemistry, Weill Cornell Medical College, New York, New York 10065
| | - Anant K Menon
- the Department of Biochemistry, Weill Cornell Medical College, New York, New York 10065.
| | - Peter Bütikofer
- From the Institute of Biochemistry and Molecular Medicine and.
| |
Collapse
|
210
|
Patel SJ, Van Lehn RC. Characterizing the Molecular Mechanisms for Flipping Charged Peptide Flanking Loops across a Lipid Bilayer. J Phys Chem B 2018; 122:10337-10348. [PMID: 30376710 DOI: 10.1021/acs.jpcb.8b06613] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The cell membrane largely prevents the passive diffusion of charged molecules due to the large free energy barrier associated with translocating charged groups across the hydrophobic lipid bilayer core. Despite this barrier, some peptides can interconvert between transmembrane and surface-adsorbed states by "flipping" charged flanking loops across the bilayer on a surprisingly rapid second-minute time scale. The transmembrane helices of some multispanning membrane proteins undergo similar reorientation processes, suggesting that loop-flipping may be a mechanism for regulating membrane protein topology; however, the molecular mechanisms underlying this behavior remain unknown. In this work, we study the loop-flipping behavior exhibited by a peptide with a hydrophobic transmembrane helix, charged flanking loops, and a central, membrane-exposed aspartate residue of varying protonation state. We utilize all-atom temperature accelerated molecular dynamics simulations to predict the likelihood of loop-flipping without predefining specific loop-flipping pathways. We demonstrate that this approach can identify multiple possible flipping pathways, with the prevalence of each pathway depending on the protonation state of the central residue. In particular, we find that a charged central residue facilitates loop-flipping by stabilizing membrane water defects, enabling the "self-catalysis" of charge translocation. These findings provide detailed molecular-level insights into charged loop-flipping pathways that may generalize to other charge translocation processes, such as lipid flip-flop or the large-scale conformational rearrangements of multispanning membrane proteins.
Collapse
Affiliation(s)
- Samarthaben J Patel
- Department of Chemical and Biological Engineering , University of Wisconsin-Madison , Madison , Wisconsin 53706 , United States
| | - Reid C Van Lehn
- Department of Chemical and Biological Engineering , University of Wisconsin-Madison , Madison , Wisconsin 53706 , United States
| |
Collapse
|
211
|
Jojoa-Cruz S, Saotome K, Murthy SE, Tsui CCA, Sansom MS, Patapoutian A, Ward AB. Cryo-EM structure of the mechanically activated ion channel OSCA1.2. eLife 2018; 7:41845. [PMID: 30382939 PMCID: PMC6235563 DOI: 10.7554/elife.41845] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 10/11/2018] [Indexed: 12/15/2022] Open
Abstract
Mechanically activated ion channels underlie touch, hearing, shear-stress sensing, and response to turgor pressure. OSCA/TMEM63s are a newly-identified family of eukaryotic mechanically activated ion channels opened by membrane tension. The structural underpinnings of OSCA/TMEM63 function are not explored. Here, we elucidate high resolution cryo-electron microscopy structures of OSCA1.2, revealing a dimeric architecture containing eleven transmembrane helices per subunit and surprising topological similarities to TMEM16 proteins. We locate the ion permeation pathway within each subunit by demonstrating that a conserved acidic residue is a determinant of channel conductance. Molecular dynamics simulations reveal membrane interactions, suggesting the role of lipids in OSCA1.2 gating. These results lay a foundation to decipher how the structural organization of OSCA/TMEM63 is suited for their roles as MA ion channels.
Collapse
Affiliation(s)
- Sebastian Jojoa-Cruz
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, California, United States
| | - Kei Saotome
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, California, United States.,Department of Neuroscience, Dorris Neuroscience Center, Howard Hughes Medical Institute, The Scripps Research Institute, California, United States
| | - Swetha E Murthy
- Department of Neuroscience, Dorris Neuroscience Center, Howard Hughes Medical Institute, The Scripps Research Institute, California, United States
| | - Che Chun Alex Tsui
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, California, United States.,Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Mark Sp Sansom
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Ardem Patapoutian
- Department of Neuroscience, Dorris Neuroscience Center, Howard Hughes Medical Institute, The Scripps Research Institute, California, United States
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, California, United States
| |
Collapse
|
212
|
Lam AK, Dutzler R. Calcium-dependent electrostatic control of anion access to the pore of the calcium-activated chloride channel TMEM16A. eLife 2018; 7:39122. [PMID: 30311910 PMCID: PMC6195346 DOI: 10.7554/elife.39122] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/11/2018] [Indexed: 12/14/2022] Open
Abstract
TMEM16A is a ligand-gated anion channel that is activated by intracellular Ca2+. This channel comprises two independent pores and closely apposed Ca2+ binding sites that are contained within each subunit of a homodimeric protein. Previously we characterized the influence of positively charged pore-lining residues on anion conduction (Paulino et al., 2017a). Here, we demonstrate the electrostatic control of permeation by the bound calcium ions in mouse TMEM16A using electrophysiology and Poisson-Boltzmann calculations. The currents of constitutively active mutants lose their outward rectification as a function of Ca2+ concentration due to the alleviation of energy barriers for anion conduction. This phenomenon originates from Coulombic interactions between the bound Ca2+ and permeating anions and thus demonstrates that an electrostatic gate imposed by the vacant binding site present in the sterically open pore, is released by Ca2+ binding to enable an otherwise sub-conductive pore to conduct with full capacity.
Collapse
Affiliation(s)
- Andy Km Lam
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Raimund Dutzler
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| |
Collapse
|
213
|
Effects of Passive Phospholipid Flip-Flop and Asymmetric External Fields on Bilayer Phase Equilibria. Biophys J 2018; 115:1956-1965. [PMID: 30393103 DOI: 10.1016/j.bpj.2018.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/21/2018] [Accepted: 10/01/2018] [Indexed: 11/20/2022] Open
Abstract
Compositional asymmetry between the leaflets of bilayer membranes modifies their phase behavior and is thought to influence other important features such as mechanical properties and protein activity. We address here how phase behavior is affected by passive phospholipid flip-flop, such that the compositional asymmetry is not fixed. We predict transitions from "pre-flip-flop" behavior to a restricted set of phase equilibria that can persist in the presence of passive flip-flop. Surprisingly, such states are not necessarily symmetric. We further account for external symmetry breaking, such as a preferential substrate interaction, and show how this can stabilize strongly asymmetric equilibrium states. Our theory explains several experimental observations of flip-flop-mediated changes in phase behavior and shows how domain formation and compositional asymmetry can be controlled in concert, by manipulating passive flip-flop rates and applying external fields.
Collapse
|
214
|
Choi J, Jang Y, Kim H, Wee J, Cho S, Son WS, Kim SM, Yang YD. Functional roles of glutamic acid E143 and E705 residues in the N-terminus and transmembrane domain 7 of Anoctamin 1 in calcium and noxious heat sensing. BMB Rep 2018; 51:236-241. [PMID: 29335069 PMCID: PMC5988578 DOI: 10.5483/bmbrep.2018.51.5.199] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Indexed: 01/12/2023] Open
Abstract
Anoctamin 1 (ANO1) is an anion channel that is activated by changes in cytosolic Ca2+ concentration and noxious heat. Although the critical roles of ANO1 have been elucidated in various cell types, the control of its gating mechanisms by Ca2+ and heat remain more elusive. To investigate critical amino acid residues for modulation of Ca2+ and heat sensing, we constructed a randomized mutant library for ANO1. Among 695 random mutants, reduced Ca2+ sensitivity was observed in two mutants (mutant 84 and 87). Consequently, the E143A mutant showed reduced sensitivity to Ca2+ but not to high temperatures, whereas the E705V mutant exhibited reduced sensitivity to both Ca2+ and noxious heat. These results suggest that the glutamic acids (E) at 143 and 705 residues in ANO1 are critical for modulation of Ca2+ and/or heat responses. Furthermore, these findings help to provide a better understanding of the Ca2+-mediated activation and heat-sensing mechanism of ANO1.
Collapse
Affiliation(s)
- Jonghyun Choi
- College of Pharmacy, CHA University, Sungnam 13488, Korea
| | - Yongwoo Jang
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
| | - Haedong Kim
- College of Pharmacy, CHA University, Sungnam 13488, Korea
| | - Jungwon Wee
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Korea
| | - Sinyoung Cho
- College of Pharmacy, CHA University, Sungnam 13488, Korea
| | - Woo Sung Son
- College of Pharmacy, CHA University, Sungnam 13488, Korea
| | - Sung Min Kim
- Department of Physical Education, College of Performing Arts and Sport, Hanyang University, Seoul 04763; Department of Active Aging Industry, Graduate School, Hanyang University, Seoul 04763, Korea
| | - Young Duk Yang
- College of Pharmacy, CHA University, Sungnam 13488, Korea
| |
Collapse
|
215
|
Whitlock JM, Yu K, Cui YY, Hartzell HC. Anoctamin 5/TMEM16E facilitates muscle precursor cell fusion. J Gen Physiol 2018; 150:1498-1509. [PMID: 30257928 PMCID: PMC6219693 DOI: 10.1085/jgp.201812097] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/12/2018] [Accepted: 09/10/2018] [Indexed: 12/19/2022] Open
Abstract
Limb-girdle muscular dystrophy type 2L arises from mutations in the anoctamin ANO5, whose role in muscle physiology is unknown. Whitlock et al. show that loss of ANO5 perturbs phosphatidylserine exposure and cell–cell fusion in muscle precursor cells, which is an essential step in muscle repair. Limb-girdle muscular dystrophy type 2L (LGMD2L) is a myopathy arising from mutations in ANO5; however, information about the contribution of ANO5 to muscle physiology is lacking. To explain the role of ANO5 in LGMD2L, we previously hypothesized that ANO5-mediated phospholipid scrambling facilitates cell–cell fusion of mononucleated muscle progenitor cells (MPCs), which is required for muscle repair. Here, we show that heterologous overexpression of ANO5 confers Ca2+-dependent phospholipid scrambling to HEK-293 cells and that scrambling is associated with the simultaneous development of a nonselective ionic current. MPCs isolated from adult Ano5−/− mice exhibit defective cell fusion in culture and produce muscle fibers with significantly fewer nuclei compared with controls. This defective fusion is associated with a decrease of Ca2+-dependent phosphatidylserine exposure on the surface of Ano5−/− MPCs and a decrease in the amplitude of Ca2+-dependent outwardly rectifying ionic currents. Viral introduction of ANO5 in Ano5−/− MPCs restores MPC fusion competence, ANO5-dependent phospholipid scrambling, and Ca2+-dependent outwardly rectifying ionic currents. ANO5-rescued MPCs produce myotubes having numbers of nuclei similar to wild-type controls. These data suggest that ANO5-mediated phospholipid scrambling or ionic currents play an important role in muscle repair.
Collapse
Affiliation(s)
- Jarred M Whitlock
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - Kuai Yu
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - Yuan Yuan Cui
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - H Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
216
|
Dai WJ, Qiu J, Sun J, Ma CL, Huang N, Jiang Y, Zeng J, Ren BC, Li WC, Li YH. Downregulation of microRNA-9 reduces inflammatory response and fibroblast proliferation in mice with idiopathic pulmonary fibrosis through the ANO1-mediated TGF-β-Smad3 pathway. J Cell Physiol 2018; 234:2552-2565. [PMID: 30144053 DOI: 10.1002/jcp.26961] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 06/13/2018] [Indexed: 12/23/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease with increasing occurrence, high death rates and unfavorable treatment regimens. In the current study, we identified the expression of microRNA-9 (miR-9) and anoctamin-1 (ANO1) in IPF mouse models induced by bleomycin, and their effects on inflammation and fibroblast proliferation through the transforming growth factor-β (TGF-β)-Smad3 pathway. To verify the targeting relationship between miR-9 and ANO1, we used bioinformatics prediction and conducted a dual-luciferase reporter gene assay. The underlying regulatory mechanisms of miR-9 and the target gene ANO1 were investigated mainly with the treatment of miR-9 mimic, miR-9 inhibitor, or siRNA against ANO1 in fibroblasts isolated from IPF mice. Enzyme-linked immunosorbent assay was performed to investigate the effect of miR-9 or ANO1 on inflammatory factors. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and flow cytometry were used to detect fibroblast proliferation and apoptosis. Reverse transcription quantitative polymerase chain reaction and western blot analysis were applied to measure the expression of the TGF-β-Smad3 pathway-related genes. The determination of luciferase activity suggested that miR-9 targets ANO1. Upregulation of miR-9 or silencing of ANO1 intensified inflammation in IPF, promoted proliferation and inhibited apoptotic ability of lung fibroblasts. MiR-9 negatively modulated ANO1, and thus activated the TGF-β-Smad3 pathway. These findings suggest that miR-9 can indirectly activate the TGF-β-Smad3 pathway by inhibiting the expression of ANO1, thereby aggravating inflammation, promotes proliferation and suppressing apoptosis of lung fibroblasts in mice models of IPF.
Collapse
Affiliation(s)
- Wen-Jing Dai
- Department of Respiratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jing Qiu
- Department of Respiratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jian Sun
- Department of Respiratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Chun-Lan Ma
- Department of Respiratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Na Huang
- Department of Respiratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Yi Jiang
- Department of Respiratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jun Zeng
- Department of Respiratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Bo-Chen Ren
- Department of Respiratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Wan-Cheng Li
- Department of Respiratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Yun-Hui Li
- Department of Respiratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
217
|
Gating mechanism of the extracellular entry to the lipid pathway in a TMEM16 scramblase. Nat Commun 2018; 9:3251. [PMID: 30108217 PMCID: PMC6092359 DOI: 10.1038/s41467-018-05724-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 07/04/2018] [Indexed: 12/22/2022] Open
Abstract
Members of the TMEM16/ANO family of membrane proteins are Ca2+-activated phospholipid scramblases and/or Cl− channels. A membrane-exposed hydrophilic groove in these proteins serves as a shared translocation pathway for ions and lipids. However, the mechanism by which lipids gain access to and permeate through the groove remains poorly understood. Here, we combine quantitative scrambling assays and molecular dynamic simulations to identify the key steps regulating lipid movement through the groove. Lipid scrambling is limited by two constrictions defined by evolutionarily conserved charged and polar residues, one extracellular and the other near the membrane mid-point. The region between these constrictions is inaccessible to lipids and water molecules, suggesting that the groove is in a non-conductive conformation. A sequence of lipid-triggered reorganizations of interactions between these residues and the permeating lipids propagates from the extracellular entryway to the central constriction, allowing the groove to open and coordinate the headgroups of transiting lipids. Some TMEM16 family members are Ca2+-dependent phospholipid scramblases, which also mediate non-selective ion transport; however, the mechanism how lipids permeate through the TMEM16 remains poorly understood. Here, the authors combine biochemical assays and simulations to identify the key steps regulating lipid movement through the membrane-exposed groove.
Collapse
|
218
|
Ballesteros A, Fenollar-Ferrer C, Swartz KJ. Structural relationship between the putative hair cell mechanotransduction channel TMC1 and TMEM16 proteins. eLife 2018; 7:38433. [PMID: 30063209 PMCID: PMC6067890 DOI: 10.7554/elife.38433] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/09/2018] [Indexed: 02/03/2023] Open
Abstract
The hair cell mechanotransduction (MET) channel complex is essential for hearing, yet it's molecular identity and structure remain elusive. The transmembrane channel-like 1 (TMC1) protein localizes to the site of the MET channel, interacts with the tip-link responsible for mechanical gating, and genetic alterations in TMC1 alter MET channel properties and cause deafness, supporting the hypothesis that TMC1 forms the MET channel. We generated a model of TMC1 based on X-ray and cryo-EM structures of TMEM16 proteins, revealing the presence of a large cavity near the protein-lipid interface that also harbors the Beethoven mutation, suggesting that it could function as a permeation pathway. We also find that hair cells are permeable to 3 kDa dextrans, and that dextran permeation requires TMC1/2 proteins and functional MET channels, supporting the presence of a large permeation pathway and the hypothesis that TMC1 is a pore forming subunit of the MET channel complex.
Collapse
Affiliation(s)
- Angela Ballesteros
- Molecular Physiology and Biophysics Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, United States
| | - Cristina Fenollar-Ferrer
- Laboratory of Molecular & Cellular Neurobiology, National Institute of Mental Health, National Institutes of Health, Bethesda, United States.,Laboratory of Molecular Genetics, National Institute of Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, United States.,Molecular Biology and Genetics Section, National Institute of Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, United States
| | - Kenton Jon Swartz
- Molecular Physiology and Biophysics Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, United States
| |
Collapse
|
219
|
Affiliation(s)
- Angela Ballesteros
- Molecular Physiology and Biophysics Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Kenton J Swartz
- Molecular Physiology and Biophysics Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
220
|
Malvezzi M, Andra KK, Pandey K, Lee BC, Falzone ME, Brown A, Iqbal R, Menon AK, Accardi A. Out-of-the-groove transport of lipids by TMEM16 and GPCR scramblases. Proc Natl Acad Sci U S A 2018; 115:E7033-E7042. [PMID: 29925604 PMCID: PMC6065010 DOI: 10.1073/pnas.1806721115] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Phospholipid scramblases externalize phosphatidylserine to facilitate numerous physiological processes. Several members of the structurally unrelated TMEM16 and G protein-coupled receptor (GPCR) protein families mediate phospholipid scrambling. The structure of a TMEM16 scramblase shows a membrane-exposed hydrophilic cavity, suggesting that scrambling occurs via the ‟credit-card" mechanism where lipid headgroups permeate through the cavity while their tails remain associated with the membrane core. Here we show that afTMEM16 and opsin, representatives of the TMEM16 and GCPR scramblase families, transport phospholipids with polyethylene glycol headgroups whose globular dimensions are much larger than the width of the cavity. This suggests that transport of these large headgroups occurs outside rather than within the cavity. These large lipids are scrambled at rates comparable to those of normal phospholipids and their presence in the reconstituted vesicles promotes scrambling of normal phospholipids. This suggests that both large and small phospholipids can move outside the cavity. We propose that the conformational rearrangements underlying TMEM16- and GPCR-mediated credit-card scrambling locally deform the membrane to allow transbilayer lipid translocation outside the cavity and that both mechanisms underlie transport of normal phospholipids.
Collapse
Affiliation(s)
- Mattia Malvezzi
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY 10065
| | - Kiran K Andra
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - Kalpana Pandey
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - Byoung-Cheol Lee
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY 10065
| | - Maria E Falzone
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - Ashley Brown
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - Rabia Iqbal
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - Anant K Menon
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - Alessio Accardi
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY 10065;
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10065
| |
Collapse
|
221
|
Yoo JI, O’Malley MA. Tuning Vector Stability and Integration Frequency Elevates Functional GPCR Production and Homogeneity in Saccharomyces cerevisiae. ACS Synth Biol 2018; 7:1763-1772. [PMID: 29871481 DOI: 10.1021/acssynbio.8b00036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Membrane proteins play a valuable role in biotechnology, yet the difficulty of producing high yields of functional membrane protein limits their use in synthetic biology. The practical application of G protein-coupled receptors in whole cell biosensors, for example, is restricted to those that are functionally produced at the cell surface in the chosen host, limiting the range of detectable molecules. Here, we present a facile approach to significantly improve the yield and homogeneity of functional membrane proteins in Saccharomyces cerevisiae by altering only the choice of expression vector. Expression of a model GPCR, the human adenosine A2a receptor, from commonly used centromeric and episomal vectors leads to low yields and cellular heterogeneity due to plasmid loss in 20-90% of the cell population. In contrast, homogeneous production of GPCR is attained using a multisite integrating vector or a novel, modified high copy vector that does not require genomic integration or addition of any selection agents. Finally, we introduce a FACS-based screen, which enables rapid isolation of cells with 4- to 15-fold increases in gene dosage and up to a 9-fold increase in functional protein yield without loss of homogeneity compared to a strain isolated through conventional, low-throughput methods. These results can be extended to improve the cellular homogeneity and yield of other membrane proteins, expanding the repertoire of useful receptors for synthetic biology applications.
Collapse
Affiliation(s)
- Justin I. Yoo
- Department of Chemical Engineering, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Michelle A. O’Malley
- Department of Chemical Engineering, University of California Santa Barbara, Santa Barbara, California 93106, United States
| |
Collapse
|
222
|
Wozniak KL, Phelps WA, Tembo M, Lee MT, Carlson AE. The TMEM16A channel mediates the fast polyspermy block in Xenopus laevis. J Gen Physiol 2018; 150:1249-1259. [PMID: 30012842 PMCID: PMC6122928 DOI: 10.1085/jgp.201812071] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/12/2018] [Indexed: 12/03/2022] Open
Abstract
In their preceding paper, Wozniak et al. show that fertilization increases intracellular Ca2+ in Xenopus laevis eggs by activating an IP3 signaling cascade. Here, they reveal that Ca2+ subsequently opens the Cl− channel TMEM16A to allow Cl− efflux, cell depolarization, and fast block to polyspermy. In externally fertilizing animals, such as sea urchins and frogs, prolonged depolarization of the egg immediately after fertilization inhibits the entry of additional sperm—a phenomenon known as the fast block to polyspermy. In the African clawed frog Xenopus laevis, this depolarization is driven by Ca2+-activated Cl− efflux. Although the prominent Ca2+-activated Cl− currents generated in immature X. laevis oocytes are mediated by X. laevis transmembrane protein 16a (xTMEM16A) channels, little is known about the channels that contribute to the fast block in mature eggs. Moreover, the gamete undergoes a gross transformation as it develops from an immature oocyte into a fertilization-competent egg. Here, we report the results of our approach to identify the Ca2+-activated Cl− channel that triggers the fast block. By querying published proteomic and RNA-sequencing data, we identify two Ca2+-activated Cl− channels expressed in fertilization-competent X. laevis eggs: xTMEM16A and X. laevis bestrophin 2A (xBEST2A). By exogenously expressing xTMEM16A and xBEST2A in axolotl cells lacking endogenous Ca2+-activated currents, we characterize the effect of inhibitors on currents mediated by these channels. None of the inhibitors tested block xBEST2A currents specifically. However, 2-(4-chloro-2-methylphenoxy)-N-[(2-methoxyphenyl)methylideneamino]-acetamide (Ani9) and N-((4-methoxy)-2-naphthyl)-5-nitroanthranilic acid (MONNA) each reduce xTMEM16A currents by more than 70% while only nominally inhibiting those generated by xBEST2A. Using whole-cell recordings during fertilization, we find that Ani9 and MONNA effectively diminish fertilization-evoked depolarizations. Additionally, these inhibitors lead to increased polyspermy in X. laevis embryos. These results indicate that fertilization activates TMEM16A channels in X. laevis eggs and induces the earliest known event triggered by fertilization: the fast block to polyspermy.
Collapse
Affiliation(s)
| | - Wesley A Phelps
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA
| | - Maiwase Tembo
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA
| | - Miler T Lee
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA
| | - Anne E Carlson
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
223
|
Falzone ME, Malvezzi M, Lee BC, Accardi A. Known structures and unknown mechanisms of TMEM16 scramblases and channels. J Gen Physiol 2018; 150:933-947. [PMID: 29915161 PMCID: PMC6028493 DOI: 10.1085/jgp.201711957] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/29/2018] [Indexed: 12/25/2022] Open
Abstract
Falzone et al. interpret the mechanisms underlying the activity of TMEM16 family members from recent structural and functional work. The TMEM16 family of membrane proteins is composed of both Ca2+-gated Cl− channels and Ca2+-dependent phospholipid scramblases. The functional diversity of TMEM16s underlies their involvement in numerous signal transduction pathways that connect changes in cytosolic Ca2+ levels to cellular signaling networks. Indeed, defects in the function of several TMEM16s cause a variety of genetic disorders, highlighting their fundamental pathophysiological importance. Here, we review how our mechanistic understanding of TMEM16 function has been shaped by recent functional and structural work. Remarkably, the recent determination of near-atomic-resolution structures of TMEM16 proteins of both functional persuasions has revealed how relatively minimal rearrangements in the substrate translocation pathway are sufficient to precipitate the dramatic functional differences that characterize the family. These structures, when interpreted in the light of extensive functional analysis, point to an unusual mechanism for Ca2+-dependent activation of TMEM16 proteins in which substrate permeation is regulated by a combination of conformational rearrangements and electrostatics. These breakthroughs pave the way to elucidate the mechanistic bases of ion and lipid transport by the TMEM16 proteins and unravel the molecular links between these transport activities and their function in human pathophysiology.
Collapse
Affiliation(s)
- Maria E Falzone
- Department of Biochemistry, Weill Cornell Medical School, New York, NY
| | - Mattia Malvezzi
- Department of Anesthesiology, Weill Cornell Medical School, New York, NY
| | - Byoung-Cheol Lee
- Department of Anesthesiology, Weill Cornell Medical School, New York, NY
| | - Alessio Accardi
- Department of Biochemistry, Weill Cornell Medical School, New York, NY .,Department of Anesthesiology, Weill Cornell Medical School, New York, NY.,Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medical School, New York, NY
| |
Collapse
|
224
|
Andra KK, Dorsey S, Royer CA, Menon AK. Structural mapping of fluorescently-tagged, functional nhTMEM16 scramblase in a lipid bilayer. J Biol Chem 2018; 293:12248-12258. [PMID: 29903908 DOI: 10.1074/jbc.ra118.003648] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/12/2018] [Indexed: 01/25/2023] Open
Abstract
Most members of the TransMEMbrane protein 16 (TMEM16) family are Ca2+-regulated scramblases that facilitate the bidirectional movement of phospholipids across membranes necessary for diverse physiological processes. The nhTMEM16 scramblase (from the fungus Nectria hematococca) is a homodimer with a large cytoplasmic region and a hydrophilic, membrane-exposed groove in each monomer. The groove provides the transbilayer conduit for lipids, but the mechanism by which Ca2+ regulates it is not clear. Because fusion of large protein tags at either the N or C terminus abolishes nhTMEM16 activity, we hypothesized that its cytoplasmic portion containing both termini may regulate lipid translocation via a Ca2+-dependent conformational change. To test this hypothesis, here we used fluorescence methods to map key distances within the nhTMEM16 homodimer and between its termini and the membrane. To this end, we developed functional nhTMEM16 variants bearing an acyl carrier protein (ACP) tag at one or both of the termini. These constructs were fluorescently labeled by ACP synthase-mediated insertion of CoA-conjugated fluorophores and reconstituted into vesicles containing fluorescent lipids to obtain the distance of closest approach between the labeled tag and the membrane via FRET. Fluorescence lifetime measurements with phasor analysis were used to determine the distance between the N and C termini of partnering monomers in the nhTMEM16 homodimer. We now report that the measured distances do not vary significantly between Ca2+-replete and EGTA-treated samples, indicating that whereas the cytoplasmic portion of the protein is important for function, it does not appear to regulate scramblase activity via a detectable conformational change.
Collapse
Affiliation(s)
- Kiran K Andra
- Department of Biochemistry, Weill Cornell Medical College, New York, New York 10065
| | - Savanna Dorsey
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York 12180
| | - Catherine A Royer
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York 12180
| | - Anant K Menon
- Department of Biochemistry, Weill Cornell Medical College, New York, New York 10065.
| |
Collapse
|
225
|
Abstract
We report a 49-year-old man who presented with a history of asymmetric weakness. His neurological examination and electromyogram testing suggested the presence of a myopathy. A muscle biopsy confirmed the presence of a myopathy with several lobulated, whorled and ring fibers, and it showed no evidence of inflammation. Genetic testing of more than 50 genes known to cause myopathy was performed and demonstrates the presence of the common founder mutation in ANO5 gene c.191dupA, which he inherited from his unaffected father. In addition, he inherited a novel mutation, c.1063C>T (p.L355F) in exon 11 of ANO5 gene from his unaffected mother. The founder mutation is a known pathogenic variant and, based on our protein modeling analysis, the novel c.1063C>T (p.L355F) variant is likely pathogenic. This indicates that he is a compound heterozygote, providing strong support for the diagnosis of limb-girdle muscular dystrophy 2L.
Collapse
|
226
|
Sui T, Xu L, Lau YS, Liu D, Liu T, Gao Y, Lai L, Han R, Li Z. Development of muscular dystrophy in a CRISPR-engineered mutant rabbit model with frame-disrupting ANO5 mutations. Cell Death Dis 2018; 9:609. [PMID: 29789544 PMCID: PMC5964072 DOI: 10.1038/s41419-018-0674-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/23/2018] [Accepted: 05/07/2018] [Indexed: 12/27/2022]
Abstract
Limb girdle muscular dystrophy type 2L (LGMD2L) and Miyoshi myopathy type 3 (MMD3) are autosomal recessive muscular dystrophy caused by mutations in the gene encoding anoctamin-5 (ANO5), which belongs to the anoctamin protein family. Two independent lines of mice with complete disruption of ANO5 transcripts did not exhibit overt muscular dystrophy phenotypes; instead, one of these mice was observed to present with some abnormality in sperm motility. In contrast, a third line of ANO5-knockout (KO) mice with residual expression of truncated ANO5 expression was reported to display defective membrane repair and very mild muscle pathology. Many of the ANO5-related patients carry point mutations or small insertions/deletions (indels) in the ANO5 gene. To more closely mimic the human ANO5 mutations, we engineered mutant ANO5 rabbits via co-injection of Cas9 mRNA and sgRNA into the zygotes. CRISPR-mediated small indels in the exon 12 and/or 13 in the mutant rabbits lead to the development of typical signs of muscular dystrophy with increased serum creatine kinase (CK), muscle necrosis, regeneration, fatty replacement and fibrosis. This novel ANO5 mutant rabbit model would be useful in studying the disease pathogenesis and therapeutic treatments for ANO5-deficient muscular dystrophy.
Collapse
Affiliation(s)
- Tingting Sui
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Institute of Zoonosis, Jilin University, 130062, Changchun, China
| | - Li Xu
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, United States
| | - Yeh Siang Lau
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, United States
| | - Di Liu
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Institute of Zoonosis, Jilin University, 130062, Changchun, China
| | - Tingjun Liu
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Institute of Zoonosis, Jilin University, 130062, Changchun, China
| | - Yandi Gao
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, United States
| | - Liangxue Lai
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Institute of Zoonosis, Jilin University, 130062, Changchun, China.
| | - Renzhi Han
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, United States.
| | - Zhanjun Li
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Institute of Zoonosis, Jilin University, 130062, Changchun, China.
| |
Collapse
|
227
|
Rottgen TS, Nickerson AJ, Rajendran VM. Calcium-Activated Cl - Channel: Insights on the Molecular Identity in Epithelial Tissues. Int J Mol Sci 2018; 19:E1432. [PMID: 29748496 PMCID: PMC5983713 DOI: 10.3390/ijms19051432] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 12/25/2022] Open
Abstract
Calcium-activated chloride secretion in epithelial tissues has been described for many years. However, the molecular identity of the channel responsible for the Ca2+-activated Cl− secretion in epithelial tissues has remained a mystery. More recently, TMEM16A has been identified as a new putative Ca2+-activated Cl− channel (CaCC). The primary goal of this article will be to review the characterization of TMEM16A, as it relates to the physical structure of the channel, as well as important residues that confer voltage and Ca2+-sensitivity of the channel. This review will also discuss the role of TMEM16A in epithelial physiology and potential associated-pathophysiology. This will include discussion of developed knockout models that have provided much needed insight on the functional localization of TMEM16A in several epithelial tissues. Finally, this review will examine the implications of the identification of TMEM16A as it pertains to potential novel therapies in several pathologies.
Collapse
Affiliation(s)
- Trey S Rottgen
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA.
- Department of Biochemistry and Molecular Pharmacology, West Virginia University School of Medicine, Morgantown, WV 26506, USA.
| | - Andrew J Nickerson
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA.
- Department of Biochemistry and Molecular Pharmacology, West Virginia University School of Medicine, Morgantown, WV 26506, USA.
| | - Vazhaikkurichi M Rajendran
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA.
- Department of Biochemistry and Molecular Pharmacology, West Virginia University School of Medicine, Morgantown, WV 26506, USA.
| |
Collapse
|
228
|
Structural basis for dual-mode inhibition of the ABC transporter MsbA. Nature 2018; 557:196-201. [PMID: 29720648 DOI: 10.1038/s41586-018-0083-5] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 03/20/2018] [Indexed: 11/08/2022]
Abstract
The movement of core-lipopolysaccharide across the inner membrane of Gram-negative bacteria is catalysed by an essential ATP-binding cassette transporter, MsbA. Recent structures of MsbA and related transporters have provided insights into the molecular basis of active lipid transport; however, structural information about their pharmacological modulation remains limited. Here we report the 2.9 Å resolution structure of MsbA in complex with G907, a selective small-molecule antagonist with bactericidal activity, revealing an unprecedented mechanism of ABC transporter inhibition. G907 traps MsbA in an inward-facing, lipopolysaccharide-bound conformation by wedging into an architecturally conserved transmembrane pocket. A second allosteric mechanism of antagonism occurs through structural and functional uncoupling of the nucleotide-binding domains. This study establishes a framework for the selective modulation of ABC transporters and provides rational avenues for the design of new antibiotics and other therapeutics targeting this protein family.
Collapse
|
229
|
Di Zanni E, Gradogna A, Scholz-Starke J, Boccaccio A. Gain of function of TMEM16E/ANO5 scrambling activity caused by a mutation associated with gnathodiaphyseal dysplasia. Cell Mol Life Sci 2018; 75:1657-1670. [PMID: 29124309 PMCID: PMC5897490 DOI: 10.1007/s00018-017-2704-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/10/2017] [Accepted: 10/30/2017] [Indexed: 12/31/2022]
Abstract
Mutations in the human TMEM16E (ANO5) gene are associated both with the bone disease gnathodiaphyseal dysplasia (GDD; OMIM: 166260) and muscle dystrophies (OMIM: 611307, 613319). However, the physiological function of TMEM16E has remained unclear. We show here that human TMEM16E, when overexpressed in mammalian cell lines, displayed partial plasma membrane localization and gave rise to phospholipid scrambling (PLS) as well as non-selective ionic currents with slow time-dependent activation at highly depolarized membrane potentials. While the activity of wild-type TMEM16E depended on elevated cytosolic Ca2+ levels, a mutant form carrying the GDD-causing T513I substitution showed PLS and large time-dependent ion currents even at low cytosolic Ca2+ concentrations. Contrarily, mutation of the homologous position in the Ca2+-activated Cl- channel TMEM16B paralog hardly affected its function. In summary, these data provide the first direct demonstration of Ca2+-dependent PLS activity for TMEM16E and suggest a gain-of-function phenotype related to a GDD mutation.
Collapse
Affiliation(s)
- Eleonora Di Zanni
- Institute of Biophysics, Consiglio Nazionale delle Ricerche, Via de Marini 6, 16149, Genova, Italy
| | - Antonella Gradogna
- Institute of Biophysics, Consiglio Nazionale delle Ricerche, Via de Marini 6, 16149, Genova, Italy
| | - Joachim Scholz-Starke
- Institute of Biophysics, Consiglio Nazionale delle Ricerche, Via de Marini 6, 16149, Genova, Italy.
| | - Anna Boccaccio
- Institute of Biophysics, Consiglio Nazionale delle Ricerche, Via de Marini 6, 16149, Genova, Italy.
| |
Collapse
|
230
|
Nakao H, Hayashi C, Ikeda K, Saito H, Nagao H, Nakano M. Effects of Hydrophilic Residues and Hydrophobic Length on Flip-Flop Promotion by Transmembrane Peptides. J Phys Chem B 2018; 122:4318-4324. [PMID: 29589918 DOI: 10.1021/acs.jpcb.8b00298] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Peptide-induced phospholipid flip-flop (scrambling) was evaluated using transmembrane model peptides in which the central residue was substituted with various amino acid residues (sequence: Ac-GKK(LA) nXW(LA) nLKKA-CONH2). Peptides with a strongly hydrophilic residue (X = Q, N, or H) had higher scramblase activity than that of other peptides, and the activity was also dependent on the length of the peptides. Peptides with a hydrophobic stretch of 17 residues showed high flip-promotion propensity, whereas those of 21 and 25 residues did not, suggesting that membrane thinning under negative mismatch conditions promotes the flipping. Interestingly, a hydrophobic stretch of 19 residues intensively promoted phospholipid scrambling and membrane leakage. The distinctive characteristics of the peptide were ascribed by long-term molecular dynamics simulation to the arrangement of central glutamine and terminal four lysine residues on the same side of the helix. The combination of simulated and experimental data enables understanding of the mechanisms by which transmembrane helices, and ultimately unidentified scramblases in biomembranes, cause lipid scrambling.
Collapse
Affiliation(s)
- Hiroyuki Nakao
- Graduate School of Medicine and Pharmaceutical Sciences , University of Toyama , 2630 Sugitani , Toyama 930-0194 , Japan
| | - Chihiro Hayashi
- Graduate School of Medicine and Pharmaceutical Sciences , University of Toyama , 2630 Sugitani , Toyama 930-0194 , Japan
| | - Keisuke Ikeda
- Graduate School of Medicine and Pharmaceutical Sciences , University of Toyama , 2630 Sugitani , Toyama 930-0194 , Japan
| | - Hiroaki Saito
- Institute of Science and Engineering , Kanazawa University , Kakuma , Kanazawa , Ishikawa 920-1192 , Japan.,Laboratory for Computational Molecular Design , RIKEN Quantitative Biology Center , 6-2-4 Furuedai , Suita , Osaka 565-0874 , Japan
| | - Hidemi Nagao
- Institute of Science and Engineering , Kanazawa University , Kakuma , Kanazawa , Ishikawa 920-1192 , Japan
| | - Minoru Nakano
- Graduate School of Medicine and Pharmaceutical Sciences , University of Toyama , 2630 Sugitani , Toyama 930-0194 , Japan
| |
Collapse
|
231
|
Xu J, Xu L, Lau YS, Gao Y, Moore SA, Han R. A novel ANO5 splicing variant in a LGMD2L patient leads to production of a truncated aggregation-prone Ano5 peptide. J Pathol Clin Res 2018; 4:135-145. [PMID: 29665321 PMCID: PMC5903698 DOI: 10.1002/cjp2.92] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/12/2017] [Accepted: 01/04/2018] [Indexed: 12/12/2022]
Abstract
Mutations in ANO5 cause several human diseases including gnathodiaphyseal dysplasia 1 (GDD1), limb-girdle muscular dystrophy 2L (LGMD2L), and Miyoshi myopathy 3 (MMD3). Previous work showed that complete genetic disruption of Ano5 in mice did not recapitulate human muscular dystrophy, while residual expression of mutant Ano5 in a gene trapped mouse developed muscular dystrophy with defective membrane repair. This suggests that truncated Ano5 expression may be pathogenic. Here, we screened a panel of commercial anti-Ano5 antibodies using a recombinant adenovirus expressing human Ano5 with FLAG and YFP at the N- and C-terminus, respectively. The monoclonal antibody (mAb) N421A/85 was found to specifically detect human Ano5 by immunoblotting and immunofluorescence staining. The antigen epitope was mapped to a region of 28 residues within the N-terminus. Immunofluorescence staining of muscle cryosections from healthy control subjects showed that Ano5 is localized at the sarcoplasmic reticulum. The muscle biopsy from a LGMD2L patient homozygous for the c.191dupA mutation showed no Ano5 signal, confirming the specificity of the N421A/85 antibody. Surprisingly, strong Ano5 signal was detected in a patient with compound heterozygous mutations (c.191dupA and a novel splice donor site variant c.363 + 4A > G at the exon 6-intron 6 junction). Interestingly, insertion of the mutant intron 6, but not the wild-type intron 6, into human ANO5 cDNA resulted in a major transcript that carried the first 158-bp of intron 6. Transfection of the construct encoding the first 121 amino acids into C2C12 cells resulted in protein aggregate formation, suggesting that aggregate-forming Ano5 peptide may contribute to the pathogenesis of muscular dystrophy.
Collapse
Affiliation(s)
- Jing Xu
- Division of Cardiovascular Medicine, Department of Cardiac Surgery, Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOHUSA
| | - Li Xu
- Division of Cardiovascular Medicine, Department of Cardiac Surgery, Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOHUSA
| | - Yeh S Lau
- Division of Cardiovascular Medicine, Department of Cardiac Surgery, Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOHUSA
| | - Yandi Gao
- Division of Cardiovascular Medicine, Department of Cardiac Surgery, Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOHUSA
| | - Steven A Moore
- Department of Pathology, Carver College of MedicineUniversity of IowaIowa CityIAUSA
| | - Renzhi Han
- Division of Cardiovascular Medicine, Department of Cardiac Surgery, Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOHUSA
| |
Collapse
|
232
|
Medrano-Soto A, Moreno-Hagelsieb G, McLaughlin D, Ye ZS, Hendargo KJ, Saier MH. Bioinformatic characterization of the Anoctamin Superfamily of Ca2+-activated ion channels and lipid scramblases. PLoS One 2018; 13:e0192851. [PMID: 29579047 PMCID: PMC5868767 DOI: 10.1371/journal.pone.0192851] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/31/2018] [Indexed: 01/01/2023] Open
Abstract
Our laboratory has developed bioinformatic strategies for identifying distant phylogenetic relationships and characterizing families and superfamilies of transport proteins. Results using these tools suggest that the Anoctamin Superfamily of cation and anion channels, as well as lipid scramblases, includes three functionally characterized families: the Anoctamin (ANO), Transmembrane Channel (TMC) and Ca2+-permeable Stress-gated Cation Channel (CSC) families; as well as four families of functionally uncharacterized proteins, which we refer to as the Anoctamin-like (ANO-L), Transmembrane Channel-like (TMC-L), and CSC-like (CSC-L1 and CSC-L2) families. We have constructed protein clusters and trees showing the relative relationships among the seven families. Topological analyses suggest that the members of these families have essentially the same topologies. Comparative examination of these homologous families provides insight into possible mechanisms of action, indicates the currently recognized organismal distributions of these proteins, and suggests drug design potential for the disease-related channel proteins.
Collapse
Affiliation(s)
- Arturo Medrano-Soto
- Department of Molecular Biology, University of California at San Diego, La Jolla, California, United States of America
| | | | - Daniel McLaughlin
- Department of Molecular Biology, University of California at San Diego, La Jolla, California, United States of America
| | - Zachary S. Ye
- Department of Molecular Biology, University of California at San Diego, La Jolla, California, United States of America
| | - Kevin J. Hendargo
- Department of Molecular Biology, University of California at San Diego, La Jolla, California, United States of America
| | - Milton H. Saier
- Department of Molecular Biology, University of California at San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
233
|
Abstract
Transmembrane protein 16F (TMEM16F) is a Ca2+-dependent phospholipid scramblase that translocates phospholipids bidirectionally between the leaflets of the plasma membrane. Phospholipid scrambling of TMEM16F causes exposure of phosphatidylserine in activated platelets to induce blood clotting and in differentiated osteoblasts to promote bone mineralization. Despite the importance of TMEM16F-mediated phospholipid scrambling in various biological reactions, the fundamental features of the scrambling reaction remain elusive due to technical difficulties in the preparation of a platform for assaying scramblase activity in vitro. Here, we established a method to express and purify mouse TMEM16F as a dimeric molecule by constructing a stable cell line and developed a microarray containing membrane bilayers with asymmetrically distributed phospholipids as a platform for single-molecule scramblase assays. The purified TMEM16F was integrated into the microarray, and monitoring of phospholipid translocation showed that a single TMEM16F molecule transported phospholipids nonspecifically between the membrane bilayers in a Ca2+-dependent manner. Thermodynamic analysis of the reaction indicated that TMEM16F transported 4.5 × 104 lipids per second at 25 °C, with an activation free energy of 47 kJ/mol. These biophysical features were similar to those observed with channels, which transport substrates by facilitating diffusion, and supported the stepping-stone model for the TMEM16F phospholipid scramblase.
Collapse
|
234
|
Peters CJ, Gilchrist JM, Tien J, Bethel NP, Qi L, Chen T, Wang L, Jan YN, Grabe M, Jan LY. The Sixth Transmembrane Segment Is a Major Gating Component of the TMEM16A Calcium-Activated Chloride Channel. Neuron 2018; 97:1063-1077.e4. [PMID: 29478917 DOI: 10.1016/j.neuron.2018.01.048] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 12/03/2017] [Accepted: 01/26/2018] [Indexed: 11/29/2022]
Abstract
Calcium-activated chloride channels (CaCCs) formed by TMEM16A or TMEM16B are broadly expressed in the nervous system, smooth muscles, exocrine glands, and other tissues. With two calcium-binding sites and a pore within each monomer, the dimeric CaCC exhibits voltage-dependent calcium sensitivity. Channel activity also depends on the identity of permeant anions. To understand how CaCC regulates neuronal signaling and how CaCC is, in turn, modulated by neuronal activity, we examined the molecular basis of CaCC gating. Here, we report that voltage modulation of TMEM16A-CaCC involves voltage-dependent occupancy of calcium- and anion-binding site(s) within the membrane electric field as well as a voltage-dependent conformational change intrinsic to the channel protein. These gating modalities all critically depend on the sixth transmembrane segment.
Collapse
Affiliation(s)
- Christian J Peters
- Departments of Physiology, Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - John M Gilchrist
- Departments of Physiology, Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jason Tien
- Departments of Physiology, Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Neville P Bethel
- Department of Pharmaceutical Chemistry and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lijun Qi
- Departments of Physiology, Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Tingxu Chen
- Departments of Physiology, Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lynn Wang
- Departments of Physiology, Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yuh Nung Jan
- Departments of Physiology, Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Michael Grabe
- Department of Pharmaceutical Chemistry and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lily Y Jan
- Departments of Physiology, Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
235
|
Pelz T, Drose DR, Fleck D, Henkel B, Ackels T, Spehr M, Neuhaus EM. An ancestral TMEM16 homolog from Dictyostelium discoideum forms a scramblase. PLoS One 2018; 13:e0191219. [PMID: 29444117 PMCID: PMC5812556 DOI: 10.1371/journal.pone.0191219] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 12/29/2017] [Indexed: 01/11/2023] Open
Abstract
TMEM16 proteins are a recently identified protein family comprising Ca2+-activated Cl- channels that generate outwardly rectifying ionic currents in response to intracellular Ca2+ elevations. Some TMEM16 family members, such as TMEM16F/ANO6 are also essential for Ca2+-dependent phospholipid scrambling. TMEM16-like genes are present in the genomes of most eukaryotic species, the function(s) of TMEM16 family members from evolutionary ancient eukaryotes is not completely clear. Here, we provide insight into the evolution of these TMEM16 proteins by similarity searches for ancestral sequences. All eukaryotic genomes contain TMEM16 homologs, but only vertebrates have the full repertoire of ten distinct subtypes. TMEM16 homologs studied so far belong to the opisthokont branch of the phylogenetic tree, which includes the animal and fungal kingdoms. An organism outside this group is Dictyostelium discoideum, a representative of the amoebozoa group that diverged from the metazoa before fungi. We here functionally investigated the TMEM16 family member from Dictyostelium discoideum. When recombinantly expressed in HEK293 cells, DdTMEM16 induces phospholipid scrambling. However, in several electrophysiological experiments we did not find evidence for a Ca2+-activated Cl- channel function of DdTMEM16.
Collapse
Affiliation(s)
- Thomas Pelz
- Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Daniela R. Drose
- Department of Chemosensation, Institute for Biology II, RWTH-Aachen University, Aachen, Germany
| | - David Fleck
- Department of Chemosensation, Institute for Biology II, RWTH-Aachen University, Aachen, Germany
| | - Bastian Henkel
- Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Tobias Ackels
- Department of Chemosensation, Institute for Biology II, RWTH-Aachen University, Aachen, Germany
| | - Marc Spehr
- Department of Chemosensation, Institute for Biology II, RWTH-Aachen University, Aachen, Germany
| | - Eva M. Neuhaus
- Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
236
|
Abstract
The human body generates 10-100 billion cells every day, and the same number of cells die to maintain homeostasis in our body. Cells infected by bacteria or viruses also die. The cell death that occurs under physiological conditions mainly proceeds by apoptosis, which is a noninflammatory, or silent, process, while pathogen infection induces necroptosis or pyroptosis, which activates the immune system and causes inflammation. Dead cells generated by apoptosis are quickly engulfed by macrophages for degradation. Caspases are a large family of cysteine proteases that act in cascades. A cascade that leads to caspase 3 activation mediates apoptosis and is responsible for killing cells, recruiting macrophages, and presenting an "eat me" signal(s). When apoptotic cells are not efficiently engulfed by macrophages, they undergo secondary necrosis and release intracellular materials that represent a damage-associated molecular pattern, which may lead to a systemic lupus-like autoimmune disease.
Collapse
Affiliation(s)
- Shigekazu Nagata
- Laboratory of Biochemistry and Immunology, World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan;
| |
Collapse
|
237
|
Phosphatidylinositol-(4, 5)-bisphosphate regulates calcium gating of small-conductance cation channel TMEM16F. Proc Natl Acad Sci U S A 2018; 115:E1667-E1674. [PMID: 29382763 DOI: 10.1073/pnas.1718728115] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
TMEM16F, which is activated by elevation of intracellular calcium to trigger phospholipid scrambling and the collapse of lipid bilayer asymmetry to mediate important cellular functions such as blood coagulation, also generates a small-conductance calcium-activated cation current. How TMEM16F activation may be regulated is an open question. By recording TMEM16F Ca2+-activated current, we found that the TMEM16F Ca2+-response is desensitized by a brief exposure to high intracellular Ca2+, which is associated with depletion of phosphatidylinositol-(4, 5)-bisphosphate (PIP2) from the inner leaflet of the membrane. Application of artificial or natural PIP2 restores TMEM16F channel activity. PIP2 modulation of TMEM16F requires the presence of several positively charged amino acids in its cytoplasmic N-terminal domain. TMEM16F interaction with PIP2 works synergistically with membrane depolarization to facilitate Ca2+-gating of TMEM16F. Our study reveals the dependence of TMEM16F activity on phosphoinositides and provides one mechanism for TMEM16F activation to be strictly regulated in the cell membrane.
Collapse
|
238
|
Affiliation(s)
- H Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322
| | - Jarred M Whitlock
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
239
|
Schreiber R, Ousingsawat J, Wanitchakool P, Sirianant L, Benedetto R, Reiss K, Kunzelmann K. Regulation of TMEM16A/ANO1 and TMEM16F/ANO6 ion currents and phospholipid scrambling by Ca 2+ and plasma membrane lipid. J Physiol 2018; 596:217-229. [PMID: 29134661 PMCID: PMC5767690 DOI: 10.1113/jp275175] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 11/08/2017] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS TMEM16 proteins can operate as Ca2+ -activated Cl- channels or scramble membrane phospholipids, which are both highly relevant mechanisms during disease. Overexpression of TMEM16A and TMEM16F were found to be partially active at 37°C and at resting intracellular Ca2+ concentrations. We show that TMEM16 Cl- currents and phospholipid scrambling can be activated by modification of plasma membrane phospholipids, through reactive oxygen species and phospholipase A2. While phospholipids and Cl- ions are likely to use the same pore within TMEM16F, TMEM16A only conducts Cl- ions. Lipid regulation of TMEM16 proteins is highly relevant during inflammation and regulated cell death such as apoptosis and ferroptosis. ABSTRACT TMEM16/anoctamin (ANO) proteins form Ca2+ -activated ion channels or phospholipid scramblases. We found that both TMEM16A/ANO1 and TMEM16F/ANO6 produced Cl- currents when activated by intracellular Ca2+ , but only TMEM16F was able to expose phosphatidylserine to the outer leaflet of the plasma membrane. Mutations within TMEM16F or TMEM16A/F chimeras similarly changed Cl- currents and phospholipid scrambling, suggesting the same intramolecular pathway for Cl- and phospholipids. When overexpressed, TMEM16A and TMEM16F produced spontaneous Cl- currents at 37°C even at resting intracellular Ca2+ levels, which was abolished by inhibition of phospholipase A2 (PLA2 ). Connversely, activation of PLA2 or application of active PLA2 , as well as lipid peroxidation induced by reactive oxygen species (ROS) using staurosporine or tert-butyl hydroperoxide, enhanced ion currents by TMEM16A/F and in addition activated phospholipid scrambling by TMEM16F. Thus, TMEM16 proteins are activated by an increase in intracellular Ca2+ , or independent of intracellular Ca2+ , by modifications occurring in plasma and intracellular membrane phospholipids. These results may help to explain why regions distant to the TMEM16 pore and the Ca2+ binding sites control Cl- currents and phospholipid scrambling. Regulation of TMEM16 proteins through modification of membrane phospholipids occurs during regulated cell death such as apoptosis and ferroptosis. It contributes to inflammatory and nerve injury-induced hypersensitivity and generation of pain and therefore provides a regulatory mechanism that is particularly relevant during disease.
Collapse
Affiliation(s)
- Rainer Schreiber
- Institut für PhysiologieUniversität RegensburgUniversitätsstraße 31D‐93053RegensburgGermany
| | - Jiraporn Ousingsawat
- Institut für PhysiologieUniversität RegensburgUniversitätsstraße 31D‐93053RegensburgGermany
| | | | - Lalida Sirianant
- Institut für PhysiologieUniversität RegensburgUniversitätsstraße 31D‐93053RegensburgGermany
| | - Roberta Benedetto
- Institut für PhysiologieUniversität RegensburgUniversitätsstraße 31D‐93053RegensburgGermany
| | - Karina Reiss
- Department of DermatologyUniversity of KielSchittenhelmstrasse 7Kiel24105Germany
| | - Karl Kunzelmann
- Institut für PhysiologieUniversität RegensburgUniversitätsstraße 31D‐93053RegensburgGermany
| |
Collapse
|
240
|
Anoctamin 9/TMEM16J is a cation channel activated by cAMP/PKA signal. Cell Calcium 2017; 71:75-85. [PMID: 29604966 DOI: 10.1016/j.ceca.2017.12.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/16/2017] [Accepted: 12/26/2017] [Indexed: 12/21/2022]
Abstract
Anoctamins (ANOs) are multifunctional membrane proteins that consist of 10 homologs. ANO1 (TMEM16A) and ANO2 (TMEM16B) are anion channels activated by intracellular calcium that meditate numerous physiological functions. ANO6 is a scramblase that redistributes phospholipids across the cell membrane. The other homologs are not well characterized. We found ANO9/TMEM16J is a cation channel activated by a cAMP-dependent protein kinase A (PKA). Intracellular cAMP-activated robust currents in whole cells expressing ANO9, which were inhibited by a PKA blocker. A cholera toxin that persistently stimulated adenylate cyclase activated ANO9 as did the application of PKA. The cAMP-induced ANO9 currents were permeable to cations. The cAMP-dependent ANO9 currents were augmented by intracellular Ca2+. Ano9 transcripts were predominant in the intestines. Human intestinal SW480 cells expressed high levels of Ano9 transcripts and showed PKA inhibitor-reversible cAMP-dependent currents. We conclude that ANO9 is a cation channel activated by a cAMP/PKA pathway and could play a role in intestine function.
Collapse
|
241
|
Acheson K. TMEM16F: function from (iso)form. J Physiol 2017; 594:2785-6. [PMID: 27246546 DOI: 10.1113/jp271980] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 01/01/2016] [Indexed: 11/08/2022] Open
Affiliation(s)
- Kathryn Acheson
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| |
Collapse
|
242
|
Mechanisms of Lipid Scrambling by the G Protein-Coupled Receptor Opsin. Structure 2017; 26:356-367.e3. [PMID: 29290486 DOI: 10.1016/j.str.2017.11.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/29/2017] [Accepted: 11/27/2017] [Indexed: 01/05/2023]
Abstract
Several class-A G protein-coupled receptor (GPCR) proteins act as constitutive phospholipid scramblases catalyzing the transbilayer translocation of >10,000 phospholipids per second when reconstituted into synthetic vesicles. To address the molecular mechanism by which these proteins facilitate rapid lipid scrambling, we carried out large-scale ensemble atomistic molecular dynamics simulations of the opsin GPCR. We report that, in the process of scrambling, lipid head groups traverse a dynamically revealed hydrophilic pathway in the region between transmembrane helices 6 and 7 of the protein while their hydrophobic tails remain in the bilayer environment. We present quantitative kinetic models of the translocation process based on Markov State Model analysis. As key residues on the lipid translocation pathway are conserved within the class-A GPCR family, our results illuminate unique aspects of GPCR structure and dynamics while providing a rigorous basis for the design of variants of these proteins with defined scramblase activity.
Collapse
|
243
|
Phosphatidylinositol 4,5-bisphosphate, cholesterol, and fatty acids modulate the calcium-activated chloride channel TMEM16A (ANO1). Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1863:299-312. [PMID: 29277655 DOI: 10.1016/j.bbalip.2017.12.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 12/10/2017] [Accepted: 12/18/2017] [Indexed: 11/20/2022]
Abstract
The TMEM16A-mediated Ca2+-activated Cl- current drives several important physiological functions. Membrane lipids regulate ion channels and transporters but their influence on members of the TMEM16 family is poorly understood. Here we have studied the regulation of TMEM16A by phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2), cholesterol, and fatty acids using patch clamp, biochemistry and fluorescence microscopy. We found that depletion of membrane PI(4,5)P2 causes a decline in TMEM16A current that is independent of cytoskeleton, but is partially prevented by removing intracellular Ca2+. On the other hand, supplying PI(4,5)P2 to inside-out patches attenuated channel rundown and/or partially rescued activity after channel rundown. Also, depletion (with methyl-β-cyclodextrin M-βCD) or restoration (with M-βCD+cholesterol) of membrane cholesterol slows down the current decay observed after reduction of PI(4,5)P2. Neither depletion nor restoration of cholesterol change PI(4,5)P2 content. However, M-βCD alone transiently increases TMEM16A activity and dampens rundown whereas M-βCD+cholesterol increases channel rundown. Thus, PI(4,5)P2 is required for TMEM16A function while cholesterol directly and indirectly via a PI(4,5)P2-independent mechanism regulate channel function. Stearic, arachidonic, oleic, docosahexaenoic, and eicosapentaenoic fatty acids as well as methyl stearate inhibit TMEM16A in a dose- and voltage-dependent manner. Phosphatidylserine, a phospholipid whose hydrocarbon tails contain stearic and oleic acids also inhibits TMEM16A. Finally, we show that TMEM16A remains in the plasma membrane after treatment with M-βCD, M-βCD+cholesterol, oleic, or docosahexaenoic acids. Thus, we propose that lipids and fatty acids regulate TMEM16A channels through a membrane-delimited protein-lipid interaction.
Collapse
|
244
|
Cryo-EM structures of the TMEM16A calcium-activated chloride channel. Nature 2017; 552:426-429. [PMID: 29236684 PMCID: PMC5750132 DOI: 10.1038/nature25024] [Citation(s) in RCA: 240] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 11/10/2017] [Indexed: 12/19/2022]
|
245
|
Activation mechanism of the calcium-activated chloride channel TMEM16A revealed by cryo-EM. Nature 2017; 552:421-425. [PMID: 29236691 DOI: 10.1038/nature24652] [Citation(s) in RCA: 209] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 10/18/2017] [Indexed: 12/21/2022]
Abstract
The calcium-activated chloride channel TMEM16A is a ligand-gated anion channel that opens in response to an increase in intracellular Ca2+ concentration. The protein is broadly expressed and contributes to diverse physiological processes, including transepithelial chloride transport and the control of electrical signalling in smooth muscles and certain neurons. As a member of the TMEM16 (or anoctamin) family of membrane proteins, TMEM16A is closely related to paralogues that function as scramblases, which facilitate the bidirectional movement of lipids across membranes. The unusual functional diversity of the TMEM16 family and the relationship between two seemingly incompatible transport mechanisms has been the focus of recent investigations. Previous breakthroughs were obtained from the X-ray structure of the lipid scramblase of the fungus Nectria haematococca (nhTMEM16), and from the cryo-electron microscopy structure of mouse TMEM16A at 6.6 Å (ref. 14). Although the latter structure disclosed the architectural differences that distinguish ion channels from lipid scramblases, its low resolution did not permit a detailed molecular description of the protein or provide any insight into its activation by Ca2+. Here we describe the structures of mouse TMEM16A at high resolution in the presence and absence of Ca2+. These structures reveal the differences between ligand-bound and ligand-free states of a calcium-activated chloride channel, and when combined with functional experiments suggest a mechanism for gating. During activation, the binding of Ca2+ to a site located within the transmembrane domain, in the vicinity of the pore, alters the electrostatic properties of the ion conduction path and triggers a conformational rearrangement of an α-helix that comes into physical contact with the bound ligand, and thereby directly couples ligand binding and pore opening. Our study describes a process that is unique among channel proteins, but one that is presumably general for both functional branches of the TMEM16 family.
Collapse
|
246
|
Pandey K, Ploier B, Goren MA, Levitz J, Khelashvili G, Menon AK. An engineered opsin monomer scrambles phospholipids. Sci Rep 2017; 7:16741. [PMID: 29196630 PMCID: PMC5711885 DOI: 10.1038/s41598-017-16842-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/16/2017] [Indexed: 11/09/2022] Open
Abstract
The G protein-coupled receptor opsin is a phospholipid scramblase that facilitates rapid transbilayer phospholipid exchange in liposomes. The mechanism by which opsin scrambles lipids is unknown. It has been proposed that lipid translocation may occur at protein-protein interfaces of opsin dimers. To test this possibility, we rationally engineered QUAD opsin by tryptophan substitution of four lipid-facing residues in transmembrane helix 4 (TM4) that is known to be important for dimerization. Atomistic molecular dynamics simulations of wild type and QUAD opsins combined with continuum modeling revealed that the tryptophan substitutions lower the energetically unfavorable residual hydrophobic mismatch between TM4 and the membrane, reducing the drive of QUAD opsin to dimerize. We purified thermostable wild type and QUAD opsins, with or without a SNAP tag for fluorescence labeling. Single molecule fluorescence measurements of purified SNAP-tagged constructs revealed that both proteins are monomers. Fluorescence-based activity assays indicated that QUAD opsin is a fully functional scramblase. However, unlike wild type opsin which dimerizes en route to insertion into phospholipid vesicles, QUAD opsin reconstitutes as a monomer. We conclude that an engineered opsin monomer can scramble phospholipids, and that the lipid-exposed face of TM4 is unlikely to contribute to transbilayer phospholipid exchange.
Collapse
Affiliation(s)
- Kalpana Pandey
- Department of Biochemistry, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Birgit Ploier
- Department of Biochemistry, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Michael A Goren
- Department of Biochemistry, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Joshua Levitz
- Department of Biochemistry, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - George Khelashvili
- Department of Physiology and Biophysics, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.,Institute for Computational Biomedicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Anant K Menon
- Department of Biochemistry, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
247
|
Lérias JR, Pinto MC, Botelho HM, Awatade NT, Quaresma MC, Silva IAL, Wanitchakool P, Schreiber R, Pepperkok R, Kunzelmann K, Amaral MD. A novel microscopy-based assay identifies extended synaptotagmin-1 (ESYT1) as a positive regulator of anoctamin 1 traffic. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1865:421-431. [PMID: 29154949 DOI: 10.1016/j.bbamcr.2017.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/02/2017] [Accepted: 11/14/2017] [Indexed: 01/14/2023]
Abstract
An attractive possibility to treat Cystic Fibrosis (CF), a severe condition caused by dysfunctional CFTR, an epithelial anion channel, is through the activation of alternative (non-CFTR) anion channels. Anoctamin 1 (ANO1) was demonstrated to be a Ca2+-activated chloride channel (CaCC) and thus of high potential to replace CFTR. Despite that ANO1 is expressed in human lung CF tissue, it is present at the cell surface at very low levels. In addition, little is known about regulation of ANO1 traffic, namely which factors promote its plasma membrane (PM) localization. Here, we generated a novel cellular model, expressing an inducible 3HA-ANO1-eGFP construct, and validated its usage as a microscopy tool to monitor for ANO1 traffic. We demonstrate the robustness and specificity of this cell-based assay, by the identification of siRNAs acting both as ANO1 traffic enhancer and inhibitor, targeting respectively COPB1 and ESYT1 (extended synaptotagmin-1), the latter involved in coupling of the endoplasmic reticulum to the PM at specific microdomains. We further show that knockdown of ESYT1 (and family members ESYT2 and ESYT3) significantly decreased ANO1 current density. This ANO1 cell-based assay constitutes an important tool to be further used in high-throughput screens and drug discovery of high relevance for CF and cancer.
Collapse
Affiliation(s)
- Joana R Lérias
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal; Department of Physiology, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Madalena C Pinto
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal; Department of Physiology, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany; Cell Biology and Biophysics Unit and Advanced Light Microscopy Facility, European Molecular Biology Laboratory (EMBL), Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Hugo M Botelho
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Nikhil T Awatade
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Margarida C Quaresma
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Iris A L Silva
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Podchanart Wanitchakool
- Department of Physiology, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Rainer Schreiber
- Department of Physiology, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Rainer Pepperkok
- Cell Biology and Biophysics Unit and Advanced Light Microscopy Facility, European Molecular Biology Laboratory (EMBL), Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Karl Kunzelmann
- Department of Physiology, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Margarida D Amaral
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal; Cell Biology and Biophysics Unit and Advanced Light Microscopy Facility, European Molecular Biology Laboratory (EMBL), Meyerhofstraße 1, 69117 Heidelberg, Germany.
| |
Collapse
|
248
|
Zheng L, Lin Y, Lu S, Zhang J, Bogdanov M. Biogenesis, transport and remodeling of lysophospholipids in Gram-negative bacteria. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1404-1413. [PMID: 27956138 PMCID: PMC6162059 DOI: 10.1016/j.bbalip.2016.11.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 11/28/2016] [Accepted: 11/30/2016] [Indexed: 11/18/2022]
Abstract
Lysophospholipids (LPLs) are metabolic intermediates in bacterial phospholipid turnover. Distinct from their diacyl counterparts, these inverted cone-shaped molecules share physical characteristics of detergents, enabling modification of local membrane properties such as curvature. The functions of LPLs as cellular growth factors or potent lipid mediators have been extensively demonstrated in eukaryotic cells but are still undefined in bacteria. In the envelope of Gram-negative bacteria, LPLs are derived from multiple endogenous and exogenous sources. Although several flippases that move non-glycerophospholipids across the bacterial inner membrane were characterized, lysophospholipid transporter LplT appears to be the first example of a bacterial protein capable of facilitating rapid retrograde translocation of lyso forms of glycerophospholipids across the cytoplasmic membrane in Gram-negative bacteria. LplT transports lyso forms of the three bacterial membrane phospholipids with comparable efficiency, but excludes other lysolipid species. Once a LPL is flipped by LplT to the cytoplasmic side of the inner membrane, its diacyl form is effectively regenerated by the action of a peripheral enzyme, acyl-ACP synthetase/LPL acyltransferase (Aas). LplT-Aas also mediates a novel cardiolipin remodeling by converting its two lyso derivatives, diacyl or deacylated cardiolipin, to a triacyl form. This coupled remodeling system provides a unique bacterial membrane phospholipid repair mechanism. Strict selectivity of LplT for lyso lipids allows this system to fulfill efficient lipid repair in an environment containing mostly diacyl phospholipids. A rocker-switch model engaged by a pair of symmetric ion-locks may facilitate alternating substrate access to drive LPL flipping into bacterial cells. This article is part of a Special Issue entitled: Bacterial Lipids edited by Russell E. Bishop.
Collapse
Affiliation(s)
- Lei Zheng
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston McGovern Medical School, 6431 Fannin Street, Houston, TX 77030, USA.
| | - Yibin Lin
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston McGovern Medical School, 6431 Fannin Street, Houston, TX 77030, USA
| | - Shuo Lu
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston McGovern Medical School, 6431 Fannin Street, Houston, TX 77030, USA
| | - Jiazhe Zhang
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston McGovern Medical School, 6431 Fannin Street, Houston, TX 77030, USA
| | - Mikhail Bogdanov
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston McGovern Medical School, 6431 Fannin Street, Houston, TX 77030, USA
| |
Collapse
|
249
|
Li H, Salomon JJ, Sheppard DN, Mall MA, Galietta LJ. Bypassing CFTR dysfunction in cystic fibrosis with alternative pathways for anion transport. Curr Opin Pharmacol 2017; 34:91-97. [PMID: 29065356 DOI: 10.1016/j.coph.2017.10.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/20/2017] [Accepted: 10/04/2017] [Indexed: 12/20/2022]
Abstract
One therapeutic strategy for cystic fibrosis (CF) seeks to restore anion transport to affected epithelia by targeting other apical membrane Cl- channels to bypass dysfunction of the cystic fibrosis transmembrane conductance regulator (CFTR) Cl- channel. The properties and regulation of the Ca2+-activated Cl- channel TMEM16A argue that long-acting small molecules which target directly TMEM16A are required to overcome CFTR loss. Through genetic studies of lung diseases, SLC26A9, a member of the solute carrier 26 family of anion transporters, has emerged as a promising target to bypass CFTR dysfunction. An alternative strategy to circumvent CFTR dysfunction is to deliver to CF epithelia artificial anion transporters that shuttle Cl- across the apical membrane. Recently, powerful, non-toxic, biologically-active artificial anion transporters have emerged.
Collapse
Affiliation(s)
- Hongyu Li
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Johanna J Salomon
- Division of Pediatric Pulmonology and Allergy and Cystic Fibrosis Center, Department of Pediatrics, University Hospital Heidelberg, Heidelberg, Germany; Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - David N Sheppard
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Marcus A Mall
- Division of Pediatric Pulmonology and Allergy and Cystic Fibrosis Center, Department of Pediatrics, University Hospital Heidelberg, Heidelberg, Germany; Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Luis Jv Galietta
- Telethon Institute for Genetics and Medicine (Tigem), Pozzuoli, Italy.
| |
Collapse
|
250
|
Muallem S, Chung WY, Jha A, Ahuja M. Lipids at membrane contact sites: cell signaling and ion transport. EMBO Rep 2017; 18:1893-1904. [PMID: 29030479 DOI: 10.15252/embr.201744331] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/10/2017] [Accepted: 09/21/2017] [Indexed: 12/14/2022] Open
Abstract
Communication between organelles is essential to coordinate cellular functions and the cell's response to physiological and pathological stimuli. Organellar communication occurs at membrane contact sites (MCSs), where the endoplasmic reticulum (ER) membrane is tethered to cellular organelle membranes by specific tether proteins and where lipid transfer proteins and cell signaling proteins are located. MCSs have many cellular functions and are the sites of lipid and ion transfer between organelles and generation of second messengers. This review discusses several aspects of MCSs in the context of lipid transfer, formation of lipid domains, generation of Ca2+ and cAMP second messengers, and regulation of ion transporters by lipids.
Collapse
Affiliation(s)
- Shmuel Muallem
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, Bethesda, MD, USA
| | - Woo Young Chung
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, Bethesda, MD, USA
| | - Archana Jha
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, Bethesda, MD, USA
| | - Malini Ahuja
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, Bethesda, MD, USA
| |
Collapse
|