Expression and Analysis of Recombinant Ion Channels Edited by Jeffrey J. Clare and Derek J.Trezise
Related Titles Dau...
281 downloads
565 Views
4MB Size
Report
This content was uploaded by our users and we assume good faith they have the permission to share this book. If you own the copyright to this book and it is wrongfully on our website, we offer a simple DMCA procedure to remove your content from our site. Start by pressing the button below!
Report copyright / DMCA form
Expression and Analysis of Recombinant Ion Channels Edited by Jeffrey J. Clare and Derek J.Trezise
Related Titles Daunert, S., Deo, S. K. (eds.)
Sibley, D. R.
Photoproteins in Bioanalysis
Receptor Biochemistry and Methodology Series
approx. 300 pages Hardcover ISBN 3-527-31016-9
Dingermann, T., Steinhilber, D., Folkers, G. (eds.)
6 Volume Set approx. 2500 pages 2002 Hardcover ISBN 0-471-32258-X
Molecular Biology in Medicinal Chemistry
Novartis
435 pages with 113 figures and 32 tables 2004 Hardcover ISBN 3-527-30431-2
Novartis Foundation Symposium 245 – Ion Channels – From Atomic Resolution Physiology to Functional Genomics
Babine, R. E., Abdel-Meguid, S. S. (eds.)
Protein Crystallography in Drug Discovery 278 pages with 131 figures and 11 tables 2004 Hardcover ISBN 3-527-30678-1
284 pages 2002 Hardcover ISBN 0-470-84375-6
Expression and Analysis of Recombinant Ion Channels From Structural Studies to Pharmacological Screening
Edited by Jeffrey J. Clare and Derek J.Trezise
The Editors Dr. Jeffrey J. Clare GlaxoSmithKline Department of Gene Expression and Protein Biochemistry Gunnels Wood Road Stevenage, SG1 2NY Great Britain Dr. Derek J. Trezise GlaxoSmithKline Department of Assay Development Gunnels Wood Road Stevenage SG1 2NY Great Britain
& All books published by Wiley-VCH are carefully
produced. Nevertheless, authors, editors, and publisher do not warrant the information contained in these books, including this book, to be free of errors. Readers are advised to keep in mind that statements, data, illustrations, procedural details or other items may inadvertently be inaccurate. Library of Congress Card No.: applied for British Library Cataloguing-in-Publication Data A catalogue record for this book is available from the British Library. Bibliographic information published by Die Deutsche Bibliothek Die Deutsche Bibliothek lists this publication in the Deutsche Nationalbibliografie; detailed bibliographic data is available in the Internet at . 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim All rights reserved (including those of translation into other languages). No part of this book may be reproduced in any form – by photoprinting, microfilm, or any other means – nor transmitted or translated into a machine language without written permission from the publishers. Registered names, trademarks, etc. used in this book, even when not specifically marked as such, are not to be considered unprotected by law. Typesetting ProSatz Unger,Weinheim Printing Strauss GmbH, Mörlenbach Binding J. Schäffer GmbH i.G., Grünstadt Printed in the Federal Republic of Germany Printed on acid-free paper ISBN-13: 978-3-527-31209-2 ISBN-10: 3-527-31209-9
V
Contents Preface XI List of Contributors Color Plates XVII 1 1.1 1.2 1.3 1.4 1.4.1 1.4.1.1 1.4.1.2 1.4.1.3 1.4.1.4 1.4.2 1.4.3 1.4.3.1 1.4.3.2 1.5 1.5.1 1.5.2 1.5.3 1.6
2
2.1 2.2
XIII
Expression of Ion Channels in Xenopus Oocytes 1 Alan L. Goldin Introduction 1 Advantages and Disadvantages of Xenopus Oocytes 2 Procedures for Using Oocytes 3 Types of Analyses 5 Electrophysiological Analysis 5 Two-electrode Whole Cell Voltage-clamp 5 Cut-open Oocyte Voltage-clamp 7 Macropatch Clamp 9 Single Channel Analysis 11 Biochemical Analysis 12 Compound Screening 13 Serial Recording Using the Roboocyte 14 Parallel Recording Using the OpusXpress 16 Examples of Use 17 Characterization of cDNA Clones for a Channel 17 Structure–Function Correlations 18 Studies of Human Disease Mutations 19 Conclusions 21 Acknowledgments 21 References 21 Molecular Biology Techniques for Structure – Function Studies of Ion Channels 27 Louisa Stevens, Andrew J. Powell, and Dennis Wray Introduction 27 Methods for cDNA Subcloning 28
Expression and Analysis of Recombinant Ion Channels. Edited by Jeffrey J. Clare and Derek J. Trezise Copyright # 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim ISBN: 3-527-31209-9
VI
Contents
2.2.1 2.2.2 2.2.3 2.3 2.3.1 2.3.2 2.3.3 2.4 2.4.1 2.4.2 2.5 2.6 2.7
3
3.1 3.2 3.3 3.4 3.4.1 3.4.2 3.5 3.6
4 4.1 4.2 4.3 4.4 4.5 4.6 4.6.1 4.6.2
Conventional Sub-cloning Using Restriction Enzymes and DNA Ligase 28 PCR-based cDNA Sub-cloning 31 Sub-cloning cDNA through Site-specific Recombination 33 Generation of Chimeric Channel cDNAs 36 Use of Restriction Enzymes to Generate Chimeric Channel cDNAs 36 PCR-mediated Overlap Extension for Chimera Generation 39 PCR-mediated Integration or Replacement of cDNA Fragments 43 Site-directed Mutagenesis 43 Examples of the Use of Site-directed Mutagenesis 45 Modification of the QuikChange Method for the Replacement of cDNA Fragments 50 Epitope-tagged Channels and Fusion Partners 50 Channel Subunit Concatamers 52 Concluding Remarks 53 References 54 Unnatural Amino Acids as Probes of Ion Channel Structure – Function and Pharmacology 59 Paul B. Bennett, Niki Zacharias, John B. Nicholas, Sue Dee Sahba, Ashutosh Kulkarni, and Mark Nowak Introduction 59 Unnatural Amino Acid Mutagenesis Methodology 60 Unnatural Amino Acid Mutagenesis for Ion Channel Studies 64 Structure–Function Example Studies 65 Nicotinic Acetylcholine Receptor 65 Drug Interactions with the hERG Voltage-gated Potassium Ion Channel 67 Other Uses of Unnatural Amino Acids as Probes of Protein Structure and Function 72 Conclusions 73 Acknowledgements 74 References 74 Functional Expression of Ion Channels in Mammalian Systems Jeff J. Clare Introduction 79 cDNA Cloning and Manipulation 80 Choice of Host Cell Background 81 Post-translational Processing of Heterologous Expressed Ion Channels 85 Cytotoxicity 90 Transient Expression Systems 91 “Standard” Transient Expression 91 Viral Expression Systems 92
79
Contents
4.7 4.7.1 4.7.2 4.7.3 4.8
Stable Expression of Ion Channels 96 Bicistronic Expression Systems 96 Stable Expression of Multiple Subunits 100 Inducible Expression 101 Summary 103 Acknowledgements 103 References 104
5
Analysis of Electrophysiological Data 111 Michael Pusch 5.1 Overview 111 5.2 Introduction 111 5.3 Expression Systems and Related Recording Techniques 113 5.3.1 Expression in Xenopus Oocytes 113 5.3.2 Expression in Mammalian Cells 115 5.3.3 Leak and Capacitance Subtraction 116 5.4 Macroscopic Recordings 117 5.4.1 Analysis of Pore Properties – Permeation 118 5.4.2 Analysis of Fast Voltage-dependent Block – the Woodhull Model 121 5.4.3 Information on Gating Properties from Macroscopic Measurements 122 5.4.3.1 Equilibrium Properties – Voltage-gated Channels 124 5.4.3.2 Equilibrium Properties – Ligand Gated Channels 126 5.4.3.3 Macroscopic Kinetics 129 5.4.4 Channel Block 132 5.4.5 Nonstationary Noise Analysis 133 5.4.6 Gating Current Measurements in Voltage Gated Channels 135 5.5 Single Channel Analysis 136 5.5.1 Amplitude Histogram Analysis 136 5.5.2 Kinetic Single Channel Analysis 138 5.6 Summary 142 Acknowledgements 142 References 142 6 6.1 6.2 6.3 6.3.1 6.3.2 6.3.3 6.3.4 6.4 6.5 6.6
Automated Planar Array Electrophysiology for Ion Channel Research Derek J Trezise Introduction 145 Overview of Planar Array Recording 145 Experimental Methods and Design 147 Cell Preparation 148 Cell Sealing and Recording 149 Drug Application 152 Experimental Design and Data Analysis 155 Overall Success Rates and Throughput 158 Population Patch Clamp 159 Summary and Perspective 162
145
VII
VIII
Contents
Acknowledgments 162 References 162 7 7.1 7.2 7.2.1 7.2.2 7.2.2.1 7.2.2.2 7.2.2.3 7.2.2.4 7.3 7.3.1 7.3.2 7.3.3 7.3.4
8
8.1 8.2 8.2.1 8.2.2 8.2.3 8.3 8.3.1 8.3.2 8.4 8.4.1 8.4.2 8.4.3 8.4.4 8.4.5 8.5 8.6
Ion Flux and Ligand Binding Assays for Analysisof Ion Channels 165 Georg C. Terstappen Introduction 165 Ion Flux Assays 166 Radioactive Ion Flux Assays 167 Nonradioactive Ion Flux Assays based on Atomic Absorption Spectrometry 168 Nonradioactive Rubidium Efflux Assay 168 Nonradioactive Lithium Influx Assay 174 Nonradioactive Chloride Influx Assay 174 Conclusions 174 Ligand Binding Assays 175 Heterogeneous Binding Assays Employing Radioligands 177 Homogeneous Binding Assays Employing Radioligands 178 Homogeneous Binding Assays Employing Fluorescent-Labeled Ligands and Fluorescence Polarization 180 Conclusions 181 Acknowledgements 182 References 182 Ion Channel Assays Based on Ion and Voltage-sensitive Fluorescent Probes 187 Jesús E. González, Jennings Worley, and Fredrick Van Goor Introduction 187 Membrane Potential Probes 188 Redistribution Probes 188 FRET Probes 190 Advantages and Limitations of Membrane Potential Probes 192 Ion-sensitive Fluorescent Probes 194 Calcium Dyes 194 Indicators of Other Ions 195 Fluorescence Assays for Ion Channels 196 Calcium Channels 196 Non-voltage-gated Calcium Permeable Channels 197 Sodium Channels 200 Potassium Channels 201 Chloride Channels 203 Assays for Monitoring Channel Trafficking 205 Summary 207 References 208
Contents
9 9.1 9.2 9.2.1 9.2.2 9.2.3 9.2.4 9.3 9.4 9.5 9.6 9.7 9.8 9.9 9.10 9.11
10
10.1 10.2 10.3 10.3.1 10.3.2 10.3.3 10.3.4 10.3.5 10.3.6 10.3.7 10.3.8 10.4 10.4.1 10.4.2 10.4.3 10.4.4 10.4.5 10.5
Approaches for Ion Channel Structural Studies 213 Randal B. Bass and Robert H. Spencer Introduction 213 Expression of Membrane Proteins for Structural Studies 216 Mammalian Expression 216 Insect Expression 217 Yeast Expression 217 Bacterial Expression 218 The Detergent Factor 219 Purification 223 Crystallization 227 Use of Antibody Fragments 229 Generation of First Diffraction Datasets 230 Selenomethionine Phasing of Membrane Proteins 232 MAD Phasing and Edge Scanning 233 Negative B- factor Application (Structure Factor Sharpening) 234 Conclusions 235 References 235 Molecular Modeling and Simulations of Ion Channels: Applications to Potassium Channels 241 Daniele Bemporad, Alessandro Grottesi, Shozeb Haider, Zara A. Sands, and Mark S.P. Sansom Introduction 241 Computational Methods 242 Kir Channels 246 Structures 246 Molecular Modeling 247 Simulations 248 Filter Flexibility 248 M2 Helices and Hinge Motion 250 Intracellular Domain Dynamics 251 Interactions with Ligands 251 Towards an Integrated Gating Model 253 Kv Channels 254 Structures 254 S6 Helices, Hinges and Gating 256 The Barrier at the Gate 257 The Nature of the Voltage Sensor 258 A Possible Gating Model 260 Summary and Future Directions 261 Acknowledgements 262 References 262 Subject Index
269
IX
XI
Preface Given the exciting advances that have been made within the last few years there can seldom have been a more opportune time to collate a book on ion channel research methods. Molecular genetics has provided us with the sequence of the human ion channel genome and an understanding of ‘channelopathies’ – the link between ion channel gene mutations and pathology. Advances in gene expression and functional analysis methods have leveraged a greater understanding of ion channel structure/function as well as accelerating the quest for new ion channel drugs. Computational approaches are also providing further insight into the molecular dynamics of ion channels at the atomic level. Excitingly, in June 2005 through the pioneering work of Mackinnon and co-workers, large scale protein purification and X-ray crystallography have revealed for the first time the full structure of a eukaryotic ion channel, Shaker KV1.2, resolved to 2.9 Å. The opportunity for discovery in the ion channel field has never been greater. Presently, we know of approximately 350 ion channel encoding genes in humans. Of these, 143 comprise the voltage-gated ion channel superfamily making this the third largest gene family after the G-protein-coupled receptors and protein kinases (Yu & Catterall, 2004, Sci. STKE 253, 1-17). These channels are specialised for electrical signalling and ionic homeostasis and include the well characterised voltage-gated Na+, Ca2+ and K+ channel subfamilies as well as transient receptor potential (TRP) and cyclic-nucleotide-gated channels. Other ion channels are ligand-gated and well adapted for fast synaptic transmission, notably nicotinic acetylcholine receptors, GABA- and glutamate activated channels and ATP-gated P2X receptors. The remainder are currently categorised as ‘miscellaneous’. These include channels as structurally and functionally diverse as aquaporins, ABC-transporter-like proteins such as the Cystic-fibrosis transmembrane regulator (CFTR) and volume- and Ca2+-activated chloride channels. Heteromeric assembly of different ion channel subunits, and their regulation by alternative splicing and by cellular effectors such as kinases, nuclear receptors, integrins and GPCRs further amplifies this diversity in function. For the research scientist, the challenges ahead lie in rationalising which channel variants and complexes are physiologically and pathologically relevant and how these proteins function, down to the molecular and atomic level. For the drug discoverer the ability to use this knowledge-base and experimental techniques to rapidly find safe and efficacious new ion channel modulators is paramount. Expression and Analysis of Recombinant Ion Channels. Edited by Jeffrey J. Clare and Derek J. Trezise Copyright # 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim ISBN: 3-527-31209-9
XII
Preface
This book is directed at both the new and the experienced ion channel researcher wishing to learn more about the considerations and methods for studying recombinant ion channels. Our aim is that it should be of interest to academic and industrial workers alike. Chapters 1 to 3 cover the use of the Xenopus oocyte expression system for structure-function studies, from basic approaches for manipulating ion channel cDNAs to more specialised but powerful techniques such as unnatural amino acid substitution. This is followed by reviews of strategies and methodologies available for expressing channels in mammalian cells and for their analysis by patch-clamp electrophysiology. In Chapters 6 to 8, the latest methodologies for ion channel drug discovery are reviewed, including high throughput screening using fluorescence and luminescence as well as automated planar array electrophysiology. The remaining 2 chapters focus on approaches for determining ion channel crystal structures and on computational approaches to understanding channel mechanisms at atomic resolution. Our goal is to have covered the spectrum of methods that are relevant to recombinant ion channel expression and analysis. Rather than provide detailed protocols, for which readers are directed to appropriate references in each chapter, the aim is to provide an overview of the techniques involved, reviewing underlying principles and providing working guidelines as well as an understanding of the key theoretical and practical considerations associated with each topic. In each case, this practical advice is illustrated by real life examples, taken either from the author’s own experience or from key examples in the literature. In summary, we hope to have compiled a compendium of practical ion channel information that will prove a valuable resource to the reader. We gratefully acknowledge the efforts of each of the expert authors who have provided contributions, without which this book would not be possible. We would also like to extend our sincere thanks to Jane Sanders from GSK for excellent administrative assistance and Steffen Pauly, Frank Weinreich and co-workers at Wiley for help and guidance through the publication process. Stevenage, October 2005
Jeffrey J. Clare Derek J. Trezise
XIII
List of Contributors Randal B. Bass Amgen Inc. Department of Analytical Sciences 1201 Amgen Court West Seattle WA 98119 USA
Alan L. Goldin Department of Microbiology & Molecular Genetics University of California Irvine CA 92697–4025 USA
Daniele Bemporad Department of Biochemistry University of Oxford South Parks Road Oxford OX1 3QU UK
Jesu´s E. Gonza´lez Vertex Pharmaceuticals, Inc. Discovery Biology 11010 Torreyana Road San Diego CA 92121 USA
Paul B. Bennett Neurion Pharmaceuticals, Inc. 180 N. Vinedo Avenue Pasadena CA 91107 USA
Alessandro Grottesi Department of Biochemistry University of Oxford South Parks Road Oxford OX1 3QU UK
Jeff J. Clare Department of Gene Expression and Protein Biochemistry GlaxoSmithKline Stevenage Herts SG1 2NY UK
Shozeb Haider Department of Biochemistry University of Oxford South Parks Road Oxford OX1 3QU UK
Expression and Analysis of Recombinant Ion Channels. Edited by Jeffrey J. Clare and Derek J. Trezise Copyright # 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim ISBN: 3-527-31209-9
XIV
List of Contributors
Ashutosh Kulkarni Neurion Pharmaceuticals, Inc. 180 N. Vinedo Avenue Pasadena CA 91107 USA John B. Nicholas Neurion Pharmaceuticals, Inc. 180 N. Vinedo Avenue Pasadena CA 91107 USA Mark Nowak Neurion Pharmaceuticals, Inc. 180 N. Vinedo Avenue Pasadena CA 91107 USA Andrew J. Powell Department of Gene Expression and Protein Biochemistry GlaxoSmithKline Stevenage Herts SG1 2NY UK
Zara A. Sands Department of Biochemistry University of Oxford South Parks Road Oxford OX1 3QU UK Mark S.P. Sansom Department of Biochemistry University of Oxford South Parks Road Oxford OX1 3QU UK Robert H. Spencer Department of Molecular Neurology Merck Research Laboratories P.O. Box 4 West Point PA 19486 USA Louisa Stevens Faculty of Biomedical Sciences University of Leeds Leeds LS2 9JT UK
Michael Pusch Istituto di Biofisica Consiglio Nazionale delle Ricerche Via de Marini 6 16149 Genova Italy
Georg C. Terstappen Sienabiotech S.p.A. Discovery Research Via Fiorentina 1 53100 Siena Italy
Sue Dee Sahba Neurion Pharmaceuticals, Inc. 180 N. Vinedo Avenue Pasadena CA 91107 USA
Derek J. Trezise Department of Assay Development GlaxoSmithKline Medicines Research Centre Gunnels Wood Road Stevenage SG1 2NY UK
List of Contributors
Fredrick VanGoor Vertex Pharmaceuticals, Inc. Discovery Biology 11010 Torreyana Road San Diego CA 92121 USA Jennings Worley III Vertex Pharmaceuticals, Inc. Discovery Operations 11010 Torreyana Road San Diego CA 92121 USA
Dennis Wray Faculty of Biomedical Sciences University of Leeds Leeds LS2 9JT UK Niki Zacharias Neurion Pharmaceuticals, Inc. 180 N. Vinedo Avenue Pasadena CA 91107 USA
XV
XVII
Color Plates
Fig. 2.1 Modification of multiple cloning site (MCS) in a vector by insertion of a synthetic oligonucleotide linker. The figure shows, on the left, part of the MCS of a vector, which is cut with BamHI and HindIII, removing a SnaBI site. The linear vector is then dephosphorylated to prevent religation. On the right, two synthetic oligonucleotides are phosphorylated with polynucleotide kinase (PNK) and annealed to form a double-stranded linker.
The annealed linker contains matching BamHI and HindIII sticky ends, as well as the sequences for the new restriction sites to be introduced (XbaI, EcoRI and NotI). After ligation of the linker into the linear vector (bottom of figure), the resulting vector contains XbaI, EcoRI and NotI restriction sites instead of SnaBI. (This figure also appears on page 30.)
Expression and Analysis of Recombinant Ion Channels. Edited by Jeffrey J. Clare and Derek J. Trezise Copyright # 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim ISBN: 3-527-31209-9
Fig. 3.1 The fundamental protocol for incorporating unnatural amino acids through nonsense suppression, showing an “in vivo” translation system with electrophysiology readout. A special nonsense codon is introduced into the cDNA of interest at the position of interest. A tRNA that recognizes
only the nonsense codon is created with the unnatural amino acid appended. The gene of interest (cDNA or in vitro transcribed mRNA) and the tRNA are introduced into a cell where the protein is expressed and detected using electrophysiology. (This figure also appears on page 61.)
XVIII
Color Plates
Color Plates
Fig. 3.3 Correlation between the change in EC50 and calculated binding energy in nicotinic acetylcholine receptor binding pocket. Tryptophan 149 was systematically replaced with fluorinated (F) tryptophan (Trp, 1–4 F) unnatural amino acids. Each added F further deactivates the Trp p electron cloud, resulting
in a decreased binding energy. Binding energy was calculated in the gas phase and the absolute values will be scaled down by the presence of water and other factors but the trend is expected to remain the same. (This figure also appears on page 66.)
XIX
XX
Color Plates
Fig. 3.4 Homology models of hERG closed (left) and open ion conducting (right) states. Only the S5-P-S6 segments are shown. Many drugs enter the open channel and bind to residues along the S6 segment. (This figure also appears on page 68.)
Color Plates
Figure 3.5 hERG MAP astemizole: A hERG MAP elucidates the nature and relative importance of specific drug–channel interactions. The right hand bar graph shows the change in astemizole binding energy (kcal mol–1) at each position of the channel when that position is altered. These changes in binding energy may be interpreted in terms of atomic level interactions such as hydrogen-bonding, cation–p, hydrophobic, and ion pairing. Each hERG mutant is designed to identify a specific
noncovalent binding interaction with the channel. Each compound displays a unique hERG MAP signature. In this example, changes at serine 624 suggest H-bond interactions with the compound. Progressive changes in binding as fluorine (F) is added to phenylalanine at position 652 indicate cation– p or p–p aromatic interactions. The chemical structure of astemizole is shown. (This figure also appears on page 69.)
XXI
XXII
Color Plates
Fig. 4.4 Effect of reduced cell culture temperature on ion channel expression. (A) Confocal images of stable HERG-expressing CHO cells grown at different temperatures, following immunocytochemical staining with a HERG-specific antibody. The level of HERG immunoreactivity is significantly increased in cells grown at 27 8C and 30 8C compared to 37 8C. No immunoreactivity is seen in untransfected CHO cells (CHO-wt). The increase in total HERG protein at 30 8C seen here is also mirrored by an increase in surface-localised functional protein as measured by IonWorks electrophysiology
(see Fig. 6.2). Note that, in the HERG-expressing cells, at 37 8C and 30 8C immunoreactivity is uniform and granular throughout the cytoplasm whereas at 27 8C there is an accumulation in giant lysosome-like bodies. (B) Quantitative analysis of HERG immunoreactivity by flow cytometry confirms the increases seen at lower growth temperatures, as indicated by the rightward shift in peak fluorescence (Xaxis) observed within the populations of cells grown at 27 8C and 30 8C compared to 37 8C. (This figure also appears on page 89.)
Color Plates
Fig. 4.5 The BacMam expression system. (A) Map of the pFastBacMam1 shuttle vector which used to generate recombinant BacMam viruses. The gene of interest is inserted downstream of the CMV promoter where it is flanked by Tn7 inverted repeats that direct site-specific transposition into the viral gen-
ome when transfected into the appropriate E.coli host. (B) Workflow for the generation of recombinant BacMam virus stock and transduction into mammalian cells for expression. Reproduced with permission from Ref. [77]. (This figure also appears on page 94.)
XXIII
XXIV
Color Plates
Fig. 8.4 hTRPV1 response in 3456 microtiter plate using fluorescent Ca2+ indicator Fluo-3/ Fura-red readout. (A) HEK cells stably expressing hTRPV1 were plated in a 3456-nanoplate. Cells were loaded with Fluo-3AM/Fura-red and capsaicin (200 nM) was added to evoke a calcium transient measured using a fluorescent plate reader. Each 6X6 square, one example is
enlarged, represents an 11 point concentration response analysis to a single compound in triplicate as well as capsaicin, positive (capsaicin plus antagonist) and negative (DMSO) controls with N = 1. (B) Concentration–response curve of capsazepine block of capscaicin-stimulated hTRPV1 is shown. (This figure also appears on page 198.)
Color Plates
Fig. 8.7 CFTR membrane potential assay demonstrates efficacy limitations compared to flux assays: Response to CFTR activation between fluorescent-based and electrophysiological assay formats. To monitor the response to increasing amounts of CFTR activation cells expressing wild-type CFTR were mixed with parental cells in the indicated proportions (% wild-type CFTR). The response to CFTR activation using a maximal concentration of forskolin was monitored in both the fluorescence assay (black circles) and in an electrophysiological assay (red circles) under voltage-clamp control. The response in both assays was normalized to the response using
100 % wild-type CFTR-expressing cells. In the fluorescence-based assay, the half maximal response was observed at ~ 3% wild-type CFTR and was nonlinear as the concentration of wild-type CFTR expressing cells was increased. In contrast, the half-maximal response in the using chamber assay was reached at ~ 60 % wild-type CFTR and was linear. These results highlight the nonlinearity of the fluorescence-based assays, which can limit the SAR evaluation of agonist efficacybecause the sensitive response saturates with low amounts of CFTR. (This figure also appears on page 204.)
XXV
XXVI
Color Plates
Color Plates
3 Fig. 8.8 CFTR membrane potential assay demonstrates the dependence of channel density on agonist sensitivity: Effects of CFTR density on agonist activity in fluorescent membrane potential assays. (A) Theoretical Michaelis– Menten type increase in open probability of CFTR by forskolin (Kd = 5 mM). (B) Simulation of the membrane potential response to CFTR activation by forskolin. The concentration dependent effects on membrane potential were calculated using the Goldman–Hodgkin–Katz equation incorporating the open probability at each agonist concentration and CFTR densities of 0.01 to 100 times the background permeability, which was assumed to be due to an endo-
genous K+ conductance. Only at very low CFTR densities did the EC50 approximate the Kd for forskolin. (C) To monitor the effects on CFTR density on agonist stimulation in a fluorescence-based membrane potential assay, CFTRexpressing cells were mixed with parental cells at the indicated proportions and expressed as % wild-type (wt) CFTR. As observed in the simulations, only at very low wt-CFTR proportions did the EC50 for forskolin approximate its Kd. These results illustrate that the potency of ion channel agonists in a fluorescence-based assay is highly sensitive to the channel density. (This figure also appears on page 206.)
XXVII
XXVIII
Color Plates
Fig. 9.4 Examples of X-ray diffraction images. (A) Initial diffraction pattern from a single crystal of the MscL ion channel protein from M. tuberculosis (Tb-MscL) prior to the optimization of crystallization and cryoprotection conditions. (B) Following optimization of the crystallization and cryoprotection conditions for a crystal of Tb-MscL that diffracted to a limiting resolution of 7 Å. (C) Diffraction pattern of a Tb-MscL crystal after soaking with the heavy atom compound, Na3Au(S2O3)2.
Note the significant improvement in the diffraction limit, as compared to (B), extending to 3.5 Å. This image also illustrates the significant anisotropy and intensity decay of the reflections often observed with membrane proteins. (D) Ribbon diagram of the resulting 3.5 Å structure of the M. tuberculosis MscL, a mechanosensitive channel, reproduced with permission from Ref. [15]. (This figure also appears with the color plates.) (This figure also appears on page 231.)
Color Plates
Fig. 10.7 Interactions of PIP2 with Kir6.2. (A) A molecular surface representation of a model of the Kir6.2 channel calculated using GRASP [115], color on electrostatic potential (from –6.6 to 5.4 kT, red to blue). The region of positive electrostatic potential (blue) near the intracellular membrane/water interface (indicated by the circle) corresponds to a pos-
sible binding site for PIP2. (B) Snapshot from a simulation of three PIP2 molecules within a POPC bilayer. The PIP2 molecules are shown in space-filling format whilst the phosphorus atoms of the POPC headgroups are shown as green spheres. (This figure also appears on page 253.)
Fig. 10.13 The KvAP voltage sensor. (A) The voltage sensor embedded in a detergent (DMA) micelle shown at the end (t = 40 ns) of an MD simulation. (B) The structure of the S2–S3 region of the VS at the end of a 40 ns MD simulation at 368 K. The residues are colored according to the magnitude of root mean square fluctuations experienced by the
Cas during the course of each simulation (on a scale from blue = 0.0 Å to red = 4.8 Å). The loss of helicity in S3 a is evident. (C) KvAP S4 helix hinge-swivelling about residue I130, as revealed by eigenvector 1 of an MD simulation at 368 K. The colors indicate the range of motions represented by this eigenvector. (This figure also appears on page 260.)
XXIX
1
1 Expression of Ion Channels in Xenopus Oocytes Alan L. Goldin
1.1 Introduction
Xenopus oocytes have been widely used for studying ion channels in a controlled in vivo environment since the system was initially developed for this purpose by Miledi and coworkers [1, 2]. There have been at least five major types of studies using oocytes to examine ion channel expression. The earliest use was to examine the properties of specific ion channels in a living cell free from other responses. The oocytes were injected with RNA isolated from whole brains, and the responses were analyzed using the two-microelectrode whole cell voltageclamp [2, 3], the patch clamp [4], or a variety of biochemical techniques [5, 6]. Once the responses were isolated, Xenopus oocytes were then used in a second type of study as an assay system to isolate cDNA clones encoding the proteins involved. For example, cDNA clones encoding the 5-HT1C receptor were isolated using electrophysiological assays, both by hybrid depletion [7] and by directly transcribing RNA from a cDNA library and injecting the transcripts into oocytes [8]. These types of studies are much less commonly used now because of the large number of available heterologous expression systems and cDNA clones encoding ion channels. The third major type of study for which the Xenopus oocyte expression system has been, and continues to be, particularly useful is the correlation of molecular structure with electrophysiological function of a specific channel. The two basic approaches have been to construct defined mutations whose effects are determined by expression in oocytes, and to construct chimeric molecules between two closely related channels or receptors followed by expression in oocytes and electrophysiological analysis. These types of approach are still commonly used but with more sophisticated structural alterations and functional analyses. The fourth general approach utilizing expression in oocytes is to determine the functional effects of mutations that cause human diseases. These types of studies are also performed using expression in other heterologous systems such as mammalian cells, and the advantages and disadvantages of each approach will be discussed in Section 1.5.3. Expression and Analysis of Recombinant Ion Channels. Edited by Jeffrey J. Clare and Derek J. Trezise Copyright # 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim ISBN: 3-527-31209-9
2
1 Expression of Ion Channels in Xenopus Oocytes
The final approach that takes advantage of expression in Xenopus oocytes is to screen potential drugs to determine their relative efficacies against specific types of ion channels. These studies have been made feasible by the development of automated voltage-clamp devices. Two such devices are the Roboocyte from Multichannel Systems and the OpusXpress from Axon Instruments, which is now part of Molecular Devices. The features of these instruments are described in Section 1.4.3.
1.2 Advantages and Disadvantages of Xenopus Oocytes
While the Xenopus oocyte system is a valuable tool for the study of ion channel function, there are a number of important factors to consider in deciding whether oocytes are the most appropriate system to use. One of the primary advantages of oocytes is that these cells do not express a large number of ion channels and receptors, so that the exogenous protein can be studied without contamination from endogenous channels. This advantage is not true in all cases, however, as oocytes do express some channels and receptors [9]. These responses are not usually a problem for two reasons. First, only some oocytes express the channels, so that it is frequently possible to obtain oocytes that do not have endogenous current. Second, current from the injected RNA is usually much larger than that through the endogenous channels, so that it is generally possible to record the expressed current without significant contamination from native oocyte currents. In some cases, the presence of an endogenous response can be used to advantage as a second messenger system that is coupled to the initial response that is being studied. Another advantage is that some channels can only be expressed in oocytes and not in mammalian cells. It is not possible to predict which channels fall into this category, and success using mammalian cells often depends very strongly on choosing the appropriate cell type (see Chapter 4). Even when the channels can be expressed in other systems, oocytes may still be advantageous for examination of the roles of different subunits. Since expression in oocytes involves injection of RNA, it is possible to adjust the ratio of RNA encoding each subunit and thus examine channels with a relatively well controlled composition. In contrast, it is more difficult to control the ratios of different subunits expressed by transfection in mammalian cells. There are also some advantages to the use of oocytes with respect to electrophysiological recording. The oocyte system is particularly well suited for the study of many different mutations because injection and two-electrode voltage-clamping can be carried out rapidly and in a semi-automated fashion. In addition, studies involving modulation by second messenger systems such as phosphorylation are particularly well suited to oocytes because it is possible to express multiple proteins in the same cell, and modulators can be injected while recording with the two-electrode voltage-clamp. Finally, some analysis techniques are unique to oo-
1.3 Procedures for Using Oocytes
cytes. For example, the cut-open oocyte voltage-clamp was developed specifically for high resolution electrophysiological recording from oocytes and it is particularly well-suited for analyzing fast ionic and gating currents [10, 11]. There are a number of disadvantages to using oocytes for expression of ion channels. First, as pointed out above, oocytes do express some endogenous channels and receptors. A second major disadvantage is that it has not been possible to express every channel in oocytes. On the other hand, most channels that have not been expressed in oocytes have not been expressed in other heterologous systems either, so that this problem is not unique to the use of oocytes. Another disadvantage of oocytes is that many pharmacological agents are less potent on channels in oocytes compared to the channels in mammalian cells or native tissues. This difference in potency most likely reflects decreased accessibility of the drug because of the large number of invaginations in the oocyte membrane, the vitelline membrane surrounding the oocyte surface, or the follicle cells around the oocyte. However, although the absolute concentration of drug that is required for block is often higher than that required in native tissues, the relative efficacies of drugs against different channels are generally representative of those in native tissues. Other potential disadvantages with the use of oocytes include the need for procedures and equipment beyond that usually found in a standard research laboratory, occasional wide variations in quality due to seasonal and other factors, and the fact that they are best maintained at ambient temperature which may lead to altered synthesis and processing of mammalian channels compared to physiological conditions. The most serious disadvantage of using expression in oocytes as an assay system is that the cells are not the native cells in which the channels are normally expressed. This can be reflected in two major ways. First, the functional properties that are observed may not be identical to those characterized in native tissues, although it is often difficult to make a direct comparison because the native cell usually contains multiple different types of channels. In addition, the functional properties may depend on the subunit composition, in which case the oocyte system can be used to determine which subunits are required for properties similar to those observed in vivo. The second consequence of oocytes not being the native tissue is that cellular trafficking is different, particularly in comparison to neurons. Because of this difference, some channels may not be expressed on the cell surface in oocytes and the effects of mutations that affect trafficking cannot be studied at all.
1.3 Procedures for Using Oocytes
The procedures for maintenance of Xenopus laevis, preparation of oocytes and injection with mRNA have been previously described [12, 13]. In addition, details concerning the maintenance of Xenopus laevis and the use of oocytes can be obtained from the Xenopus Express website (http://www.xenopus.com/links.htm).
3
4
1 Expression of Ion Channels in Xenopus Oocytes
A frog colony does not require elaborate equipment, although there are two important considerations. First, amphibians are sensitive to both chlorine and chloramine, so the water must be purified to remove both compounds. Second, although frogs can tolerate a wide range of temperature fluctuations, inconsistent temperature, and particularly elevated temperatures above 20 8C, greatly diminish oocyte viability. Surgery to remove oocytes is a relatively simple procedure that can be carried out on a bench top in a clean room. After preparation, the follicle cells are usually removed by treatment with collagenase, although oocytes can be injected and voltage-clamped with intact follicle cells around them. Some electrophysiological responses in oocytes either depend on the presence of follicle cells or occur in the follicle cells, which would be a reason to maintain the cells. However, all procedures are technically more difficult because the follicle cells are harder to pierce and it is time-consuming to separate out individual oocytes, so it is usually best to use defolliculated oocytes. It is possible to obtain oocytes that have been surgically removed and prepared for injection by commercial vendors. The advantages of this approach are that there is no need to maintain a frog colony, an animal use protocol is not required because no vertebrate animals will be used, and there is a considerable saving in time. The disadvantages are that the oocytes are significantly more expensive and they are only available in specific geographical areas, although the number of vendors may increase, depending on demand. One commercial source for oocytes is EcoCyte Bioscience in Germany (http://www.ecocyte.de). For most studies, oocytes are injected with RNA that has been transcribed in vitro from a cDNA clone using either a T7 or T3 promoter. Transcription is easily performed using commercially available kits, although there are some important considerations that affect expression levels. First, the RNA needs to be capped for optimal efficiency, which is part of the procedure in most kits. Second, inclusion of Xenopus b-globin 5' and 3' untranslated mRNA regions and a poly(A) tail at the 3' end usually enhances stability and translatability. Third, the length of the untranslated 5' region can have a dramatic effect on the level of current, with shorter regions generally resulting in greater efficiency of expression. It is also possible to inject DNA into the nucleus of oocytes. In this case, the cDNA should be cloned downstream of a eukaryotic promoter such as the commonly used Cytomegalovirus (CMV) promoter. The advantage of injecting DNA is that there is no need to perform in vitro transcription reactions, which saves both time and money. The disadvantage is that the procedure is more difficult and requires a more sophisticated injection apparatus [12]. Cytoplasmic injection is a relatively rapid and easy procedure. The basic requirements are a dissecting microscope, a micromanipulator, and an injector that can be a simple manual or motor driven dispenser (Fig. 1.1) [12]. Alternatively, oocytes can be injected using an automated device such as the Roboocyte from Multichannel Systems, as described in Section 1.4.3.1. This instrument was developed for automated two-electrode voltage-clamping of oocytes, but it can also function for both cytoplasmic and nuclear injection of oocytes.
1.4 Types of Analyses Fig. 1.1 Apparatus for cytoplasmic injection of Xenopus oocytes. The oocytes are distributed on polypropylene mesh in a 35–mm tissue culture dish on the stage of a dissecting microscope. Injection needles are made using a pipette puller to draw out the glass bores that are normally used with the microdispenser. After pulling, the needles are broken off at a tip diameter of 20–40 mm, as measured with a reticle under a dissecting microscope. The injection needle is position over individual oocytes using a micromanipulator and the oocytes are injected with up to 100 nl of RNA solution. Using these procedures, it is possible to inject 20 oocytes with one sample in a few minutes.
1.4 Types of Analyses 1.4.1 Electrophysiological Analysis
The most sensitive approach for analyzing ion channel function in Xenopus oocytes is the use of electrophysiology. Essentially all of the standard electrophysiological techniques can be performed on oocytes, including whole cell recording and patch-clamp recording of both macroscopic and single channel currents. In addition, techniques such as cut-open oocyte voltage-clamp recording have been developed specifically for analyzing currents in Xenopus oocytes [10]. The goal of this chapter is to present the general considerations involved in using the various approaches, as detailed procedures have been previously described [14].
1.4.1.1 Two-electrode Whole Cell Voltage-clamp Whole cell voltage-clamping of oocytes involves using two electrodes inserted into the oocyte, rather than using one electrode to make a patch on the surface followed by rupturing the membrane, as is done in mammalian cells. One electrode is used to measure the internal potential of the oocyte and the other electrode is used to inject current (Fig. 1.2). The large size of the oocyte (about 1 mm in diameter and 0.5–1 ml in volume for stage V oocytes) makes this feasible, and is both the major advantage and disadvantage of the system. One advantage is that the procedure is easy to learn and fast to perform. The electrodes are simple to pre-
5
6
1 Expression of Ion Channels in Xenopus Oocytes
Fig. 1.2 Diagram of the two-electrode voltageclamp. The oocyte is placed in a chamber under a dissecting microscope and two electrodes are gently inserted through the membrane using micromanipulators. One electrode is used to measure the internal potential of the oocyte and the other electrode is used to
inject current for clamping the oocyte at different potentials. The currents can be recorded either through the current electrode or separately through a virtual ground circuit in the bath. The bath solution can be easily and rapidly changed by continuous perfusion from gravity flow reservoirs.
pare and it is generally possible to obtain records from all oocytes if they are healthy, so that there is very little time lost in preparation. In addition, perfusion of the external medium can be easily changed multiple times. These features make the two-electrode voltage-clamp ideal for screening purposes. A second advantage is that the recordings can be stable over long periods of time, which makes it particularly useful for analyzing properties that require long protocols, such as slow inactivation. A third advantage is that it is possible to insert multiple electrodes and injection needles into the same oocyte. Therefore, modulators of channel function can be injected inside the cell while recording, so that a rapid and direct response to an intracellular signal can be observed. The final advantage of the two-electrode voltage-clamp is that it records currents through channels present in the whole cell, so that it is very sensitive. For example, currents as small as 50 nA can be detected, which corresponds to only 56104 molecules if the single channel current is 1 pA. The two-electrode voltage-clamp can be used to record currents over a wide range of amplitudes from about 10 nA to over 100 mA, depending on the amount of RNA that is injected. However, it is important to adjust the amount of RNA to obtain currents that can be reliably clamped because it is difficult to accurately clamp the membrane potential of the oocyte if the currents are larger than 5 mA.
1.4 Types of Analyses
The major disadvantage of recording from the entire oocyte is that the large size and extensive membrane invaginations result in an extremely large membrane capacitance, approximately 150–200 nF. The large capacitance causes a slow clamp settling time when the membrane potential is changed. The capacity transient can be minimized by using electrodes with low impedances of 500 kO or less. This can be accomplished by filling the electrode tips with low-melting temperature agarose, making it possible to have a large tip opening without significant leakage of KCl into the oocyte [15]. However, even with the best electrodes it is difficult to obtain data during the first 1–2 ms of a depolarization, which is the time during which rapidly activating voltage-gated channels such as sodium channels are activated (Fig. 1.3). The large capacity transient is not a problem when recording slow responses or ligand-gated currents that do not require changes in voltage. A second major disadvantage is that there is no control of the internal cellular environment, so that it is difficult to perform quantitative studies, for example examining selective permeability.
1.4.1.2 Cut-open Oocyte Voltage-clamp The cut-open oocyte voltage-clamp was developed to circumvent many of the disadvantages involved in using the two-electrode whole cell voltage-clamp [10, 11]. In this procedure, the oocyte is inserted in a chamber that separates the surface into three regions (Fig. 1.4). The top portion of the oocyte membrane is the region that is clamped and is the section from which currents are actually recorded. The middle portion is a guard that is clamped to the same potential as the top to null leakage currents though the seals. The bottom portion is the region of the oocyte that is “cut-open”, either by permeabilization with saponin or by insertion of a cannula, thus making it possible to perfuse the internal environment and to inject current intracellularly through a low resistance pathway. The internal environment is clamped to ground, as measured by a low resistance electrode inserted through the top of the oocyte, which ensures that the region of the oocyte near the top portion is accurately held at ground. The bath surrounding the top portion of the oocyte is clamped to the command potential, which can be rapidly changed with minimal series resistance. Currents are recorded through low resistance electrodes in the top chamber. There are a number of advantages to the cut-open oocyte voltage-clamp compared to the two-electrode voltage-clamp. First, the capacity transient is minimized so that the clamp can settle in 50 ms, which is fast enough to study activation of even the fastest ion channels (Fig. 1.3). Second, current noise is quite low, approximately 1 nA RMS at 5 kHz bandwidth. Third, it is possible to control the solutions in both external and internal environments. The internal solution can be accurately adjusted with the initial perfusate, but is difficult to change because of the slow perfusion rate, even when using a cannula. In contrast, the external solution can be changed quickly and completely. Finally, the recordings can be stable for hours. These advantages make the clamp particularly well suited for studies involving fast ionic and gating currents.
7
8
1 Expression of Ion Channels in Xenopus Oocytes Fig. 1.3 Representative traces of sodium currents using the different recording techniques. The two-electrode voltage-clamp records from channels throughout the entire oocyte membrane, which results in a large capacitive transient (the gap in the current records) so that data cannot be obtained for 1–2 ms after a depolarization. The cut-open oocyte voltage-clamp records from channels in approximately a third of the oocyte membrane. Therefore, the magnitudes of the currents are comparable, but the time resolution is significantly faster because of the design of the clamp. The macropatch voltage-clamp records from channels in a small patch of oocyte membrane, resulting in fast time resolution but smaller current amplitudes (note the current scale is in pA rather than nA). Small patches can be used to record single channel activity with excellent time resolution. The amplitude of single sodium channels is approximately 1 pA at the potential used for these recordings. All of the data were obtained from oocytes injected with RNA encoding the rat Nav1.2 sodium channel a subunit alone. The macroscopic current traces are shown for depolarizations from a holding potential of –100 mV to a range of potentials between – 30 and +30 mV in 10 mV increments. The single channel current trace is shown for a depolarization from –100 to –30 mV. The arrows indicate the start of the depolarization.
The major disadvantages of the cut-open oocyte voltage-clamp procedure are that it requires specialized equipment and that it is more difficult to use than the twoelectrode voltage-clamp. However, all of the equipment for this procedure, including the voltage-clamp, recording chamber and temperature controller, are commercially available from Dagan Corporation (http://www.dagan.com/ca-1b.htm).
1.4 Types of Analyses
Fig. 1.4 Diagram of the cut-open oocyte voltage-clamp. The oocyte is inserted in a chamber that separates the surface into three regions. The top chamber is clamped to the command potential and is the section from which currents are recorded. The middle chamber is a guard that is clamped to the same potential as the top to null leakage currents though the seals. The bottom
chamber is used to inject current intracellularly through a low resistance pathway. The internal environment is clamped to ground as measured by a low resistance electrode inserted through the top of the oocyte. A cannula inserted into the oocyte through the bottom chamber makes it possible to perfuse the internal environment.
1.4.1.3 Macropatch Clamp An alternative method of circumventing the problems caused by the large size of the oocyte is to record from only a fraction of the membrane in an isolated patch [16]. To record macroscopic currents, an electrode with a relatively large opening of about 10 mm in diameter is used to establish a macropatch on the surface membrane (Fig. 1.5). Recording can be performed in either the cell-attached mode or excised inside-out mode. The cell-attached mode is technically easier and maintains the normal cytoplasmic environment, but the intracellular potential needs to be determined. This potential can be measured by inserting electrodes into the oocyte, which is possible because of its large size. Either a single electrode can be used to measure the potential, or two electrodes can be used to clamp the oocyte at a fixed holding potential. The use of two electrodes has the advantage that channels throughout the remainder of the oocyte are held at the desired potential, minimizing slow inactivation of voltage-gated channels so that the same oocyte can be used for multiple patches. The excised patch technique allows complete control of the potential on both sides of the membrane, but it can be more diffi-
9
10
1 Expression of Ion Channels in Xenopus Oocytes Fig. 1.5 Macropatch versus small patch recording. To record macroscopic currents using the patch clamp, an electrode with a large tip diameter of approximately 10 mm is used to make a seal with the oocyte membrane. Currents can be recorded while the electrode is still attached to the intact oocyte or the electrode can be gently pulled back to record currents through an excised portion of the membrane. More sensitive recordings can be obtained using the same approaches but with a smaller diameter electrode tip (approximately 1 mm).
cult to perform because the seal between the membrane and the large diameter electrode is less stable than with a small electrode. An advantage of using an excised patch is that the internal face of the membrane can be perfused with different solutions. There are a number of advantages in using the macropatch technique to record from ion channels in oocytes. First, the capacity transient is minimized because only a small region of the membrane is depolarized, which makes it possible to record fast ionic and gating currents (Fig. 1.3). Second, there is complete control of the solutions on both sides of the membrane if the patch is excised, although only the internal side can be altered with perfusion because the outside is fixed by the electrode composition. These characteristics of the macropatch technique are similar to those of the cut-open oocyte voltage-clamp. An advantage compared to the cut-open oocyte technique is that the equipment is not as specialized, so that a patch clamp amplifier from any supplier can be used. There are some disadvantages in using the macropatch technique compared to the cut-open oocyte voltage-clamp. First, macropatches are usually not as stable as oocytes in the cut-open oocyte voltage-clamp. The decreased stability is a function of the patch and the fact that it is necessary to remove the vitelline membrane from the oocyte to make a seal with the electrode. Oocytes without the vitelline membrane are less stable than intact oocytes and they will lyse if exposed to air. A second disadvantage of macropatch recording is that it usually requires a high level of expression in the oocyte. In this regard, it is possible to use smaller electrodes by increasing the level of expression, which in turn decreases the technical difficulty. Because the macropatch technique involves either cell-attached or excised inside-out patches, there is no access to the external surface of the portion of the membrane being studied. Therefore, macropatch recording is
1.4 Types of Analyses
not well suited for studying the interactions of toxins that directly bind to a channel from the external surface. On the other hand, it is an excellent system for studying modulation through second messenger systems, particularly in the cellattached mode.
1.4.1.4 Single Channel Analysis Xenopus oocytes can also be used for conventional patch clamp recording, including single channel analysis. This approach differs from macropatch recording only in the size of the electrode, with a tip diameter of about 1 mm compared to about 10 mm for macropatches (Fig. 1.5). To make the seal, the vitelline membrane must first be removed from the oocyte, which is accomplished by placing the oocyte in a hypertonic solution (200 mM NaCl). The oocyte shrinks, leaving the vitelline membrane exposed so that it can be manually removed with forceps. Patches can then be obtained in the cell-attached or excised configuration, as in mammalian cells. One disadvantage of the cell-attached mode is that the intracellular potential must be determined. However, an advantage of using oocytes for this purpose is that additional electrodes can be inserted into the large oocyte, making it possible to accurately measure or clamp the intracellular potential. On the other hand, the additional electrodes and voltage-clamp increase the noise, which can be a significant problem for single channel recording. A second disadvantage of cell-attached recording is that oocytes express a high level of endogenous stretchactivated channels [17], and currents through these channels may interfere with the signal of interest. The patches can be excised in either the inside-out or outside-out configuration, just as with mammalian cells (Fig. 1.6). A difference compared to mammalian cells is that it is more difficult to rupture the oocyte membrane compared to a mammalian cell membrane. Positive pressure is generally the most effective technique, particularly with electrodes that have small electrode tip openings. In addition, the oocyte cannot be clamped with a single electrode after the membrane has been ruptured, unlike the situation with mammalian cells. Once the patch is excised, recording is comparable to recording from mammalian cell patches. Single-channel analysis in oocytes is generally equivalent to single channel analysis using mammalian cells, and it therefore has the same advantages and disadvantages. First, there is excellent time resolution because only a small portion of the membrane is depolarized. Therefore, it is possible to obtain detailed information about the opening and closing of individual channel molecules, which provides the most quantitative information for developing gating models (Fig. 1.3). The major disadvantage is that it is technically difficult to get high quality data, especially with small conductance channels. In addition, single channel recording is very time consuming for both acquisition and analysis. There are also some advantages and disadvantages to using oocytes for this purpose compared to mammalian cells. One advantage is that the level of expression can be adjusted by injecting different quantities of RNA. Therefore, it is possible to maximize the probability of obtaining patches that contain single channels. A
11
12
1 Expression of Ion Channels in Xenopus Oocytes
Fig. 1.6 Inside-out versus outside-out patch recording. Patches of oocyte membrane can be excised in either the inside-out or outside-out configuration. In both cases, the electrode is first placed against the oocyte membrane to obtain a tight seal. For an inside-out patch, the electrode is then gently pulled back,
leaving the cytoplasmic face of the membrane exposed to the bath solution. For an outsideout patch, the oocyte membrane is first ruptured, after which the electrode is gently pulled back. The membrane reforms a vesicle attached to the electrode with the external face exposed to the bath solution.
related advantage is that the oocytes can be screened first using a two-electrode voltage-clamp to determine the levels of expression so that only cells with an appropriate level of current are used to obtain patches. On the other hand, a disadvantage of oocytes is that the vitelline membrane must be removed before patching, which adds an extra step and decreases the viability of the oocyte. 1.4.2 Biochemical Analysis
Ion channels expressed in oocytes can be studied using standard biochemical techniques such as immunoprecipitation or binding assays [18, 19]. The major disadvantage of these techniques in oocytes is that they are significantly less sensitive than the electrophysiological approaches. Using the whole cell voltage-clamp, it is possible to detect as few as 105 channel molecules in a single oocyte (less than 10–18 mole). On the other hand, biochemical techniques are generally reliable down to the level of 10–12 mole, although this depends strongly on the specific activity of the reagents being used. Because of this limitation, it is necessary to express the channels at a much higher level for biochemical analysis than is required for electrophysiological recording.
1.4 Types of Analyses
Another consideration in using biochemical techniques to analyze ion channel expression in oocytes is that solubilization results in isolation of both cytoplasmic and membrane proteins. Functional ion channels are located in the membrane, but there is generally a large intracellular pool of molecules [20]. An advantage of electrophysiological analysis is that it examines only the functional channels in the membrane, whereas immunoprecipitation of total oocyte proteins examines both membrane-bound and cytoplasmic proteins. This problem can be mitigated by using membrane preparations for solubilization [21], but it is difficult to remove all nonmembrane proteins and those bound to internal membranes. Ligand binding assays do not suffer from this limitation and thus they are well suited for the study of ion channels expressed in oocytes. Ligand binding assays using oocytes are very similar to those carried out using mammalian cells (see Chapter 7). The advantage of using oocytes is that it is possible to easily and quickly express many different channel mutations or variations. In addition, the assay can usually be carried out on a single oocyte so that the composition of channels and subunits is relatively homogenous. The use of a single cell is also the major disadvantage of oocytes, in that it is necessary to obtain a sufficiently high level of expression so that the ligand can be detected. Therefore, the assay requires a high affinity ligand that can be labeled to high specific activity. 1.4.3 Compound Screening
An application of Xenopus oocyte expression that has become more common in the past few years is screening the effectiveness of new pharmaceuticals and those in development. In this regard, the use of oocyte expression cannot really be considered high throughput screening but rather, moderate throughput screening. Even though the limitation on throughput is unlikely to be eliminated anytime soon, there are some advantages to using oocyte expression for this purpose. A major advantage of screening in oocytes is that the assay is electrophysiological, which is the most detailed and relevant response to the compound or drug. In this regard, screening ion channels in oocytes is comparable to screening with one of the automated patch clamp systems that have been developed. Because automated patch clamps are designed for screening cell lines that are usually constructed to be stably expressing the genes of interest, oocytes are better suited for analyzing multiple channel variations such as mutations or different compositions of subunits. For this reason, the oocyte system is particularly appropriate for target identification and optimization. There are also some situations for which the oocyte system has a clear advantage. The first is if the channels do not express well (or at all) in mammalian cells. The second is if the mammalian cells express native currents that interfere with detection of the expressed response. A major disadvantage of using oocytes for screening drugs is that the cell is not the physiological target and the responses may differ from those that occur in vivo. However, this is a criticism of most screening systems because the physiological target cells often cannot be used because they express many different chan-
13
14
1 Expression of Ion Channels in Xenopus Oocytes
nels and receptors. One difference between oocytes and mammalian cells is that higher drug concentrations are often required for effective block of ion channels in oocytes compared to mammalian cells. This difference probably reflects limited drug access to the oocyte membrane because of the large number of invaginations. On the other hand, although the actual EC50 is usually higher in oocytes, the rank order of different drugs on the same channel or the relative efficacy of the same drug on different channels is likely to reflect the situation with native tissue. Two different automated systems have been developed for electrophysiological screening of ion channels using Xenopus oocytes. The Roboocyte system uses a serial approach in which oocytes are tested sequentially, and the OpusXpress system uses a parallel approach in which eight oocytes are tested simultaneously.
1.4.3.1 Serial Recording Using the Roboocyte The Roboocyte was developed by Multichannel Systems for the automated screening of oocytes in a serial fashion. Information about the Roboocyte can be obtained from the company web site at the following address: http://www.multichannelsystems.com/products/roboocyte/robointro.htm. The instrument consists of a single head that moves vertically for both injection and recording, with the oocytes located in the chambers of 96 well dishes (Fig. 1.7). The head can be configured with an injection needle or with a recording assembly that contains both voltage and current electrodes and a perfusion needle. The dish sits in a carrier that moves in the X and Y directions to position each oocyte sequentially under the head. The entire instrument is computer-controlled with separate procedures for injection and electrophysiological recording. Perfusion can be controlled using a gravity based system that is part of the apparatus and contains either 8 or 16 valves, or the device can be connected to a Gilson liquid handler that can dispense compounds from multiwell plates. The Roboocyte is ideally suited to a situation in which the same sample will be injected into every oocyte, with alterations in the recording conditions or drug application. A major advantage is that injection can be automated after a single setup configuration. On the other hand, it is not possible to inject different samples without going through the complete set-up procedure again. Oocytes can be obtained already prepared in multiwell plates from EcoCyte Bioscience (http:// www.ecocyte.de), although the availability is limited to specific geographic regions. A unique advantage of the Roboocyte is that it is relatively simple to perform nuclear injection. For this purpose, oocytes are allowed to settle in the 96 well plates, during which time the lighter nucleus rises towards the top surface of the oocyte. Because the oocytes are injected vertically, the instrument can be configured so that the needle pierces the nucleus by increasing the depth of injection. Nuclear injection avoids the time and expense of in vitro transcription. The coding region is cloned following a eukaryotic promoter rather than a T7 or T3 promoter, and the RNA is transcribed in the oocyte nucleus. A disadvantage of nuclear injec-
1.4 Types of Analyses
Fig. 1.7 The Roboocyte automated voltageclamp from Multichannel Systems. A, The instrument consists of a single head that moves vertically for both injection and recording, with the oocytes located in the chambers of a 96 well dish that moves on a cushion of pressur-
ized air above a magnetic steel plate. Perfusion can be controlled with a gravity-based system containing 16 valves. B, Close-up view of an injection needle. C, Close-up view of the recording head, which contains both voltage and current electrodes and a perfusion needle.
tion is that it is difficult to control the amount of RNA that is synthesized and hence the size of the current that is expressed. In addition, it is impossible to inject fixed ratios of different subunits. The Roboocyte performs automated two-electrode electrophysiological recording with semi-automated on-line and off-line analysis. The recording protocol is the same for each oocyte, and the oocytes are tested for viability before recording so that data are not obtained from dead oocytes. The sampling frequency is up to 2–kHz, which is a lower time resolution than that of the OpusXpress. The perfusion system that is included as part of the Roboocyte is limited to a maximum of 16 samples flowing by gravity. The manifold includes the outlet from all reservoirs so there is no lag time for perfusion, but there is a risk of cross-contamination at the tip. The instrument is designed to interface with a more sophisticated liquid handling system from Gilson, in which case samples can be stored in a variety of wells or tubes and the flow rate is controlled by a peristaltic pump. A disadvantage of this system is that there is a significant lag time for drug delivery to the recording chamber, so that the flow rate must be calibrated to determine when the compound reaches the oocyte.
15
16
1 Expression of Ion Channels in Xenopus Oocytes
1.4.3.2 Parallel Recording Using the OpusXpress The OpusXpress was developed by Axon Instruments, which is now part of Molecular Devices. This instrument is designed for automated analysis of oocytes in a parallel configuration. Information about the OpusXpress can be obtained from the company web site at the following address: http://www.axon.com/cs_Opus Xpress.html The instrument consists of eight individual recording chambers that are configured with perfusion and ground assemblies (Fig. 1.8). Separate voltage and current electrodes are positioned in each chamber for a total of 16 electrodes. Manipulation of the electrodes is controlled by 8 separate controllers and recording is carried out using 8 separate voltage-clamp modules. As with the Roboocyte, the entire instrument is computer-controlled. Perfusion is applied from one of two large reservoirs to all of the chambers simultaneously. Individual compounds are applied from 96 well dishes by an automated liquid handling system that uses 8 parallel pipette tips. The OpusXpress is designed purely as a recording instrument with no provision for automated injection, so that injection of the oocytes must be carried out separately. The OpusXpress is particularly well suited for examining oocytes injected with different types of RNA. The parallel design has the potential to increase throughput, as recordings are obtained from 8 oocytes simultaneously. The initial set-up time is longer than for the Roboocyte because 16 electrodes must be prepared and positioned in the holders. However, the electrodes can be reused for a number of days, so that the set-up for additional recordings only involves replacing the
Fig. 1.8 The OpusXpress automated voltageclamp from Molecular Devices. A, The instrument consists of eight individual two-electrode voltage-clamps and a liquid handling system. B, Close-up view of the automated manipulators that control the 8 pairs of electrodes, with
the voltage electrodes on the left and the current electrodes on the right. C, Close-up view of the 8 voltage-clamp recording chambers, each of which is equipped with perfusion and virtual ground assemblies.
1.5 Examples of Use
oocytes, which takes significantly less time. A major advantage of this approach is that compounds can be applied simultaneously to oocytes expressing different channels. Recordings are obtained using the same series of protocols for all 8 oocytes, with automated operation and real-time analysis. In addition, oocytes are tested for viability so that compounds are not delivered to chambers containing dead oocytes. An advantage of the OpusXpress is that the sampling frequency is up to 30 kHz, which is significantly faster than for the Roboocyte. The instrument can be programmed for a variety of recording protocols and solutions exchanges. The compounds are loaded into the chambers of 96 well dishes and relatively small volumes are required. Another advantage of the OpusXpress is that each compound is delivered via an individual pipette tip, so that there is no delay and no cross-contamination.
1.5 Examples of Use 1.5.1 Characterization of cDNA Clones for a Channel
Originally, the oocyte expression system was especially useful for the isolation of cDNA clones encoding ion channels for which no sequence information was available. With the acquisition of complete genomic sequence information from many species, new ion channels are now usually identified as candidate genes based on their similarity with known family members. However, once the sequence has been determined, it is still necessary to demonstrate that the gene encodes a functional channel. This step is critical because the sequence may not correctly predict the properties of the encoded protein. An example of this situation is the characterization of the BSC1 channel from the German cockroach Blattella germanica [22]. BSC1 was originally identified as the orthologue of the DSC1 channel from Drosophila melanogaster [23], which was in turn identified by its sequence similarity to voltage-gated sodium channels [24]. However, neither DSC1 nor BSC1 had been functionally expressed, so that the assignment of these genes as voltagegated sodium channels was based purely on sequence similarity. Zhou et al. [22] succeeded in expressing BSC1 in oocytes and demonstrated that it encodes a functional voltage-gated cation channel whose properties differ significantly in a number of ways from those of voltage-gated sodium channels. First, the channels are more selective for barium than for sodium. Second, the kinetics of activation and inactivation are significantly slower than the kinetics of sodium channel gating. Third, the channels deactivate very slowly with a substantial tail current. Finally, sodium currents through the channel can be blocked by low concentrations of calcium, resulting in an anomalous mole fraction effect. All of these properties are more similar to voltage-gated calcium channels than to voltagegated sodium channels. BSC1 appears to be the prototype of a novel family of in-
17
18
1 Expression of Ion Channels in Xenopus Oocytes
vertebrate voltage-dependent, cation channels with a close structural and evolutionary relationship to voltage-gated sodium and calcium channels. The Xenopus oocyte expression system was both helpful and problematical for the characterization of BSC1. A critical advantage was that the BSC1 channel had only been expressed in oocytes, so that this system was essential to study the channel. Second, currents through BSC1 were too small to measure using isolated patch recording, which made it difficult to compare the permeability of different ions. The cut-open oocyte voltage-clamp made it possible to examine permeability because the ionic composition on both sides of the membrane could be altered while still recording macroscopic currents through most of the cell membrane. The major disadvantage of using oocytes was that they express a robust calciumactivated chloride current that was turned on by calcium entry through BSC1. The chloride current made it significantly more difficult to record calcium current through the slowly gating BSC1 channel, which made it necessary to record barium rather than calcium current. 1.5.2 Structure–Function Correlations
One of the most powerful uses for oocytes in the study of ion channels has been to correlate molecular structure with biochemical and electrophysiological functions. Studies of this type initially involved mutagenesis followed by relatively straightforward analysis using either the two-electrode voltage-clamp or patch clamp. These approaches identified many of the regions involved in activation [25, 26], inactivation [27–30], toxin-binding [31–34] and permeation [35, 36] of voltagegated sodium and potassium channels. More sophisticated approaches have since been developed that take advantage of the features of oocyte expression to investigate the molecular mechanisms involved. One technique that has been particularly powerful has been the combination of fluorescent microscopy with electrophysiological recording to determine the movement of specific regions of the channel. This approach utilizes the substituted cysteine scanning accessibility method originally developed by Javitch et al. [37], which involves replacing amino acids individually with cysteine and then using cysteine-modifying reagents to determine the accessibility of those residues (see Chapter 2). For the fluorescence measurements, the cysteine residue is labeled with a fluorophore that can be used to detect movement of the specific region of the molecule. While cysteine scanning mutagenesis has been performed using a variety of expression systems, the combination of fluorescence microscopy with electrophysiological recording is a unique capability of the oocyte expression system. These approaches have been developed and used extensively by Isacoff and coworkers and Bezanilla and coworkers. The combination of fluorescence and electrophysiology measurements has revealed a great deal about the movement of the voltage sensor in the potassium channel. Cha et al. [38] used lanthanide-based resonance energy transfer to measure the voltage-dependent distance changes near the S4 subunit of the Shaker
1.5 Examples of Use
potassium channel, demonstrating that gating is accompanied by a rotation and possible tilt rather than a large transmembrane movement. Mannuzzu et al. [39] used voltage-clamp fluorometry of oocytes to measure gating rearrangements in the Shaker potassium channel. Their results demonstrated that there are two charge-carrying steps, the first of which takes place independently in each subunit whereas the second involves cooperative interactions between S4 segments. This approach has also been useful for testing structural models of voltage-gated ion channels. For example, Gandhi et al. [40] used accessibility probing and disulfide scanning experiments to demonstrate that the S4 voltage sensor in the bacterial KvAP potassium channel lies in close apposition to the pore domain in the resting and activated state, in contrast to the predictions of the crystal structure for that channel [41, 42]. Similar approaches have revealed details about the interactions of the four S4 voltage sensors in the sodium channel. Chanda et al. [43] used the cut-open oocyte voltage-clamp to simultaneously record fluorescence signals and gating currents, demonstrating that the voltage-dependent movement of the S4 segment in domain IV is a late step in the activation process after the S4 segments in domains I–III have moved. They further showed that the S4 segment of domain III most likely moves at the most hyperpolarized potentials and that the S4 segments in domains I and II move at more depolarized potentials. Chanda et al. [44] used the same approach to provide direct evidence for coupling interactions between the voltage sensors. Their results indicate that movement of all four voltage sensors is coupled to varying extents, with energetic linkage between the voltage sensors in domains I and IV. The technology has been continually improved in various ways. Sonnleitner et al. [45] used total internal reflection fluorescence microscopy, which allowed them to measure the movement of single voltage-gated Shaker potassium channels rather than the movements of large ensembles of proteins. Asamoah et al. [46] utilized a novel fluorescent probe (Di-1-ANEPIA) to record dynamic changes in the electric field during the gating process of the Shaker potassium channel. Cohen et al. [47] developed a novel fluorescent probe (aminophenoxazone maleimide), which made it possible to track the motions of the side chains to which the probe was attached. These approaches have provided very detailed mechanistic and structural information about the movements of specific regions of ion channels, and they are uniquely suited to expression in Xenopus oocytes. 1.5.3 Studies of Human Disease Mutations
The oocyte expression system has been extensively used to characterize the effects of ion channel mutations that cause human diseases. An example of this use is the study of mutant voltage-gated sodium channels that cause diseases of the musculoskeletal, cardiovascular and nervous system. Mutations in the SCN4A gene encoding the Nav1.4 skeletal muscle sodium channel cause three neuromuscular diseases, periodic paralysis, paramyotonia congenita and the potassium-ag-
19
20
1 Expression of Ion Channels in Xenopus Oocytes
gravated myotonias [48, 49]. Mutations in the SCN5A gene encoding the Nav1.5 cardiac channel cause long QT type 3, which predisposes to ventricular tachycardia (torsades de pointes), and Brugada syndrome, which is manifested as ventricular fibrillation [50]. Mutations in any of three neuronal sodium channel genes cause generalized epilepsy with febrile seizures plus (GEFS+). The mutations have been identified in SCN1A encoding Nav1.1 [51–53], SCN2A encoding Nav1.2 [54] and SCN1B encoding the b1 subunit [55, 56]. These mutations have been analyzed using a variety of different expression systems, each of which has certain advantages and disadvantages. Most studies of the mutations in the skeletal muscle sodium channel have been carried out using transfected HEK or tsA201 cells [57, 58], although some studies have been carried out using oocytes [59]. However, neither expression system is a very good model for skeletal muscle fibers, in which the mutant channels are expressed in vivo. In fact, Cannon et al. [60] used theoretical reconstructions to demonstrate that the integrity of the muscle cell T-tubule system is required to produce myotonia. Similarly, mutations in the cardiac sodium channel have been studied using both oocytes [61–63] and mammalian cells [63, 64], with the same reservation that neither system is a good model for cardiac myocytes. Papadatos et al. [65] solved this problem by constructing mice in which the mouse Scn5a gene encoding the Nav1.5 cardiac sodium channel was disrupted, which made it possible to study both the electrophysiological properties of the ventricular myocytes and the electrocardiographic characteristics of the mice. Similar studies have been carried out to analyze the effects of mutations causing GEFS+ using both oocytes [66–69] and mammalian cells [70, 71]. The results in the two different systems are sometimes comparable and sometimes different. For example, using the oocyte expression system, Spampanato et al. [66, 67] demonstrated that R1648H dramatically accelerates recovery from inactivation, W1204R shifts the voltage-dependence of activation and inactivation in the negative direction, and T875M enhances slow inactivation. These results suggest that multiple different alterations in sodium channel function can lead to a similar seizure phenotype. Lossin et al. [70] examined the same three mutations using an HEK cell expression system and obtained different results. They observed a marked increase in persistent current for R1648H and a slight increase in persistent current for T875M and W1204R, and they hypothesized that the epileptic phenotype resulted from the persistent current in all cases. It is not known which alterations reflect the actual effects of the mutations in neuronal cells in vivo. These discrepancies emphasize the necessity to examine the effects of diseasecausing mutations in the cell types in which they are normally expressed in vivo. There are certain instances in which the oocyte expression system is particularly well suited for the analysis of a disease-causing mutation. One mutation that causes GEFS+ is D1866Y, which alters an evolutionarily conserved aspartate residue in the C-terminal cytoplasmic domain of the sodium channel a subunit [72]. This mutation decreases modulation of the a subunit by b1, which normally causes a negative shift in the voltage-dependence of inactivation in oocytes. There is less of a shift with the mutant channel, resulting in a 10 mV difference between
References
the wild-type and mutant channels in the presence of b1. This shift increases the magnitude of the window current, which results in more persistent current during a voltage ramp. Computational analysis suggests that neurons expressing the mutant channels would fire an action potential with a shorter onset delay in response to a threshold current injection, and that they would fire multiple action potentials with a shorter inter-spike interval at a higher input stimulus. The results suggest that the D1866Y mutation weakens the interaction between the a and b1 subunits, demonstrating a novel molecular mechanism leading to seizure susceptibility. The use of oocytes made it possible to quantitatively assess the effects of b1 by injecting different ratios of RNA encoding the a and b1 subunits, which is very difficult to accomplish using a mammalian cell expression system.
1.6 Conclusions
In summary, Xenopus oocytes have been widely used as a heterologous expression system for the study of ion channels. Most channels can be expressed in a variety of different cell types, each of which has its own advantages and disadvantages. Oocytes are particularly well suited for studying many different samples, such as multiple mutations or the effects of different compositions of subunits. In addition, they are excellent for correlating structure with function using a combination of molecular biological and electrophysiological techniques, some of which have been developed specifically for oocytes. Finally, oocytes represent the only heterologous system in which some channels have been expressed. On the other hand, oocytes are not the native cells in which the channels are normally expressed, and this caveat must be remembered when interpreting the results.
Acknowledgments
Work in the author’s laboratory is supported by grants from the NIH (NS48336), the National Multiple Sclerosis Society (RG3405) and The McKnight Endowment Fund for Neuroscience (34653).
References 1 Barnard E. A, Miledi R., Sumikawa K., Translation of exogenous messenger RNA coding for nicotinic acetylcholine receptors produces functional receptors in Xenopus oocytes, Proc. R. Soc. London 1982, 215, 241–246. 2 Gundersen C. B., Miledi R., Parker I., Messenger RNA from human brain induces drug- and voltage- operated chan-
nels in Xenopus oocytes, Nature 1984, 308, 421–424. 3 Miledi R., Parker I., Sumikawa K., Properties of acetylcholine receptors translated by cat muscle mRNA in Xenopus oocytes, EMBO J. 1982, 1, 1307–1312. 4 Methfessel C.,Witzemann V., Takahashi T., Mishina M., Numa S., Sakmann B., Patch clamp measurements on
21
22
1 Expression of Ion Channels in Xenopus Oocytes
5
6
7
8
9
10
11
12
13
14
15
16
Xenopus laevis oocytes: currents through endogenous channels and implanted acetylcholine receptor and sodium channels, Pflügers Arch. 1986, 407, 577–588. Thornhill W. B., Levinson S. R., Biosynthesis of electroplax sodium channels in Electrophorus electrocytes and Xenopus oocytes, Biochemistry 1987, 26, 4381– 4388. Thornhill W. B., Levinson S. R., Biosynthesis of electroplax sodium channels, Ann. N. Y. Acad. Sci. 1986, 479, 356–363. Lübbert H., Hoffman B. J., Snutch T. P.,VanDyke T., Levine A. J., Hartig P. R., Lester H. A., Davidson N., cDNA cloning of a serotonin 5-HT1C receptor by electrophysiological assays of mRNAinjected Xenopus oocytes, Proc. Natl. Acad. Sci. USA 1987, 84, 4332–4336. Julius D., MacDermott A. B., Axel R., Jessell T. M., Molecular characterization of a functional cDNA encoding the serotonin 1 c receptor, Science 1988, 241, 558– 564. Weber W-M., Ion currents of Xenopus laevis oocytes: state of the art, Biochim. Biophys. Acta 1999, 1421, 213–233. Taglialatela M., Toro L., Stefani E., Novel voltage clamp to record small, fast currents from ion channels expressed in Xenopus oocytes, Biophys. J. 1992, 61, 78–82. Stefani E., Bezanilla F., Cut-open oocyte voltage-clamp technique, Methods Enzymol. 1998, 293, 300–318. Goldin A. L., Maintenance of Xenopus laevis and oocyte injection, Methods Enzymol. 1992, 207, 266–279. Goldin A. L., Sumikawa K., Preparation of RNA for injection into Xenopus oocytes, Methods Enzymol. 1992, 207, 279– 297. Stühmer W., Electrophysiologic recordings from Xenopus oocytes, Methods Enzymol. 1998, 293, 280–300. Schreibmayer W., Lester H.A., Dascal N.,Voltage clamping of Xenopus laevis oocytes utilizing agarose-cushion electrodes, Pflügers Arch. 1994, 426, 453–458. Stühmer W., Methfessel C., Sakmann B., Noda M., Numa S., Patch clamp characterization of sodium channels expres-
17
18
19
20
21
22
23
24
25
26
27
sed from rat brain cDNA, Eur. Biophys. J. 1987, 14, 131–138. Yang X-C., Sachs F., Characterization of stretch-activated ion channels in Xenopus oocytes, J. Physiol.(London) 1990, 431, 103–122. Goldin, A. L., Expression of ion channels by injection of mRNA into Xenopus oocytes, Methods Cell Biol. 1991, 36, 487– 509. Shih, T. M., Smith, R. D., Toro, L., Goldin, A. L., High-level expression and detection of ion channels in Xenopus oocytes, Methods Enzymol. 1998, 293, 529–556. Schmidt J. W., Rossie S., Catterall W. A., A large intracellular pool of inactive Na channel alpha subunits in developing rat brain, Proc. Natl. Acad. Sci. USA 1985, 82, 4847–4851. Colman A., Translation of eukaryotic messenger RNA in Xenopus oocytes, 1984, 1, 271–302. Zhou W., Chung I., Liu Z., Goldin A. L., Dong K., A voltage-gated calciumselective channel encoded by a sodium channel-like gene, Neuron 2004, 42, 101– 112. Liu Z., Chung I., Dong K, Alternative splicing of the BSC1 gene generates tissue-specific isoforms in the German cockroach, Insect Biochem. Mol. Biol. 2001, 31, 703–713. Salkoff L., Butler A., Wei A., Scavarda N., Giffen K., Ifune C., Goodman, R., Mandel G. Genomic organization and deduced amino acid sequence of a putative sodium channel gene in Drosophila, Science 1987, 237, 744–748. Stühmer W., Conti F., Suzuki H., Wang X., Noda M.,Yahagi N., Kubo H., Numa, S. Structural parts involved in activation and inactivation of the sodium channel, Nature 1989, 339, 597–603. Papazian D. M., Timpe L.C., Jan Y.N., Jan L.Y., Alteration of voltage-dependence of Shaker potassium channel by mutations in the S4 sequence, Nature 1991, 349, 305–310. Hoshi T., Zagotta W. N., Aldrich R.W., Biophysical and molecular mechanisms of Shaker potassium channel inactivation, Science 1990, 250, 533–538.
References 28 Zagotta W. N., Hoshi T., Aldrich R.W., Restoration of inactivation in mutants of Shaker potassium channels by a peptide derived from ShB, Science 1990, 250, 568–571. 29 Patton D. E.,West J. W., Catterall W. A., Goldin, A. L., Amino acid residues required for fast sodium channel inactivation. Charge neutralizations and deletions in the III-IV linker. Proc. Natl. Acad. Sci. USA 1992, 89, 10905–10909. 30 West J.W., Patton D. E., Scheuer T., Wang Y., Goldin A. L, Catterall W. A., A cluster of hydrophobic amino acid residues required for fast Na+ channel inactivation, Proc. Natl. Acad. Sci. USA 1992, 89, 10910–10914. 31 Yellen G., Jurman M. E., Abramson T., MacKinnon R., Mutations affecting internal TEA blockade identify the probable pore-forming region of a K+ channel, Science 1991, 251, 939–942. 32 MacKinnon R., Miller C., Mutant potassium channels with altered binding of charybdotoxin, a pore-blocking inhibitor, Science 1989, 245, 1382–1385. 33 MacKinnon R.,Yellen G., Mutations affecting TEA blockade and ion permeation in voltage-activated K+ channels, Science 1990, 250, 276–279. 34 MacKinnon R., Heginbotham L., Abramson T., Mapping the receptor site for a pore-blocking potassium channel inhibitor, Neuron 1990, 5, 767–771. 35 Yool A. J., Schwarz T. L., Alteration of ionic selectivity of a K+ channel by mutation of the H5 region, Nature 1991, 349, 700–704. 36 Hartmann H. A., Kirsch G. E., Drewe J. A., Taglialatela M., Joho R. H., Brown, A. M., Exchange of conduction pathways between two related K+ channels, Science 1991, 251, 942–944. 37 Javitch, J. A., Fu, D., Chen, J., Karlin, A., Mapping the binding-site crevice of the dopamine D2 receptor by the substituted-cysteine accessibility method, Neuron 1995, 14, 825–831. 38 Cha, A., Snyder, G. E., Selvin, P. R., Bezanilla, F., Atomic scale movement of the voltage-sensing region in a potassium channel measured via spectroscopy, Nature 1999, 402, 809–813.
39 Mannuzzu, L. M., Isacoff, E. Y. Independence and cooperativity in rearrangements of a potassium channel voltage sensor revealed by single subunit fluorescence, J. Gen. Physiol. 2000, 115, 257– 268. 40 Gandhi, C. S., Clark, E., Loots, E., Pralle, A., Isacoff, E. Y., The orientation and molecular movement of a K+ channel voltage-sensing domain, Neuron 2003, 40, 515–525. 41 Jiang,Y., Lee, A., Chen, J., Ruta,V., Cadene, M., Chait, B. T., MacKinnon, R., X-ray structure of a voltage-dependent K+ channel, Nature 2003, 423, 33–41. 42 Jiang,Y., Ruta,V., Chen, J., Lee, A., MacKinnon, R., The principle of gating charge movement in a voltage-dependent K+ channel, Nature 2003, 423, 42–48. 43 Chanda, B., Bezanilla, F., Tracking voltage-dependent conformational changes in skeletal muscle sodium channel during activation, J. Gen. Physiol. 2002, 120, 629–645. 44 Chanda, B., Asamoah, O. K., Bezanilla, F., Coupling interactions between voltage sensors of the sodium channel as revealed by site-specific measurements, J. Gen. Physiol. 2004, 123, 217–230. 45 Sonnleitner, A., Mannuzzu, L. M., Terakawa, S., Isacoff, E. Y., Structural rearrangements in single ion channels detected optically in living cells, Proc. Natl. Acad. Sci. USA 2002, 99, 12759– 12764. 46 Asamoah, O. K., Wuskell, J. P., Loew, L. M., Bezanilla, F. A., Fluoremetric approach to local electric field measurements in a voltage-gated ion channel, Neuron 2003, 37, 85–97. 47 Cohen, B. E., Pralle, A.,Yao, X. J., Swaminath, G., Gandhi, C. S., Jan, Y. N., Kobilka, B. K., Isacoff, E. Y. et al., A fluorescent probe designed for studying protein conformational change, Proc. Natl. Acad. Sci. USA 2005, 102, 965–970. 48 Cannon, S. C., From mutation to myotonia in sodium channel disorders. Neuromusc. Disord. 1997, 7, 241–249. 49 Cannon, S. C., Spectrum of sodium channel disturbances in the nondystro-
23
24
1 Expression of Ion Channels in Xenopus Oocytes
50
51
52
53
54
55
56
57
phic myotonias and periodic paralyses, Kidney Int. 2000, 57, 772–779. Balser, J. R., Inherited sodium channelopathies: models for acquired arrhythmias? Am. J. Physiol. Heart Circ. Physiol. 2002, 282, H1175–H1180. Escayg, A., MacDonald, B. T., Meisler, M. H., Baulac, S., Huberfeld, G., An-Gourfinkel, I., Brice, A., LeGuern, E. et al., Mutations of SCN1A, encoding a neuronal sodium channel, in two families with GEFS+2, Nat. Genet. 2000, 24, 343–345. Escayg, A., Heils, A., MacDonald, B. T., Haug, K., Sander, T., Meisler, M. H., A novel SCN1A mutation associated with generalized epilepsy with febrile seizures plus and prevalence of variants in patients with epilepsy, Am. J. Hum. Genet. 2001, 68, 866–873. Wallace, R. H., Scheffer, I. E., Barnett, S., Richards, M., Dibbens, L., Desai, R. R., Lerman-Sadie, T., Lev, D. et al., Neuronal sodium-channel a1-subunit mutations in generalized epilepsy with febrile seizures plus, Am. J. Hum. Genet. 2001, 68, 859–865. Sugawara, T., Tsurubuchi,Y., Agarwala, K. L., Ito, M., Fukuma, G., Mazaki-Miyazaki, E., Nagafuji, H., Noda, M. et al., A missense mutation of the Na+ channel aII subunit gene Nav1.2 in a patient with febrile and afebrile seizures causes channel dysfunction, Proc. Natl. Acad. Sci. USA 2001, 98, 6384– 6389. Wallace, R. H., Wang, D. W., Singh, R., Scheffer, I. E., George, A. L., Jr., Phillips, H. A., Saar, K., Reis, A. et al., Febrile seizures and generalized epilepsy associated with a mutation in the Na+channel b1 subunit gene SCN1B, Nat. Genet. 1998, 19, 366–370. Wallace, R. H., Scheffer, I. E., Parasivam, G., Barnett, S.,Wallace, G. B., Sutherland, G. R., Berkovic, S. F., Mulley, J. C., Generalized epilepsy with febrile seizures plus: mutation of the sodium channel subunit SCN1B, Neurology 2002, 58, 1426–1429. Bendahhou, S., Cummins, T. R., Hahn, A. F., Langlois, S., Waxman, S. G., Ptácek, L. J., A double mutation in fami-
58
59
60
61
62
63
64
65
lies with periodic paralysis defines new aspects of sodium channel slow inactivation, J.Clin.Invest. 2000, 106, 431–438. Struyk, A. F., Scoggan, K. A., Bulman, D. E., Cannon, S. C., The human skeletal muscle Na channel mutation R669H associated with hypokalemic periodic paralysis enhances slow inactivation, J.Neurosci. 2000, 20, 8610–8617. Featherstone, D. E., Fujimoto, E., Ruben, P. C., A defect in skeletal muscle sodium channel deactivation exacerbates hyperexcitability in human paramyotonia congenita, J. Physiol. (London) 1998, 506, 627–638. Cannon, S. C., Brown, R. H., Jr., Corey, D. P., Theoretical reconstruction of myotonia and paralysis caused by incomplete inactivation of sodium channels, Biophys. J. 1993, 65, 270–288. Chen, Q., Kirsch, G. E., Zhang, D., Brugada, R., Brugada, J., Brugada, P., Potenza, D., Moya, A. et al., Genetic basis and molecular mechanism for idiopathic ventricular fibrillation, Nature 1998, 392, 293–296. Rook, M. B., Alshinawi, C. B., Groenewegen,W. A., van Gelder, I. C., van Ginneken, A. C. G., Jongsma, H. J., Mannens, M. M. A. M.,Wilde, A. A. M., Human SCN5A gene mutations alter cardiac sodium channel kinetics and are associated with the Brugada syndrome, Cardiovasc. Res. 1999, 44, 507–517. Baroudi, G., Carbonneau, E., Pouliot, V., Chahine, M. SCN5A mutation (T1620M) causing Brugada syndrome exhibits different phenotypes when expressed in Xenopus oocytes and mammalian cells, FEBS Lett. 2000, 467, 12–16. Abriel, H., Cabo, C.,Wehrens, X. H. T., Rivolta, I., Motoike, H. K., Memmi, M., Napolitano, C., Priori, S. G. et al., Novel arrhythmogenic mechanism revealed by a long-QT syndrome mutation in the cardiac Na+ channel, Circ. Res. 2001, 88, 740–745. Papadatos, G. A., Wallerstein, P. M. R., Head, C. E. G., Ratcliff, R., Brady, P. A., Benndorf, K, Saumarez, R. C., Trezise, A. E. O. et al., Slowed conduction and ventricular tachycardia after targeted disruption of the cardiac sodium
References
66
67
68
69
channel gene Scn5a, Proc. Natl. Acad. Sci. USA 2002, 99, 6210–6215. Spampanato, J., Escayg, A., Meisler, M. H., Goldin, A. L., Functional effects of two voltage-gated sodium channel mutations that cause generalized epilepsy with febrile seizures plus type 2, J. Neurosci. 2001, 21, 7481–7490. Spampanato, J., Escayg, A., Meisler, M. H., Goldin, A. L., The generalized epilepsy with febrile seizures plus type 2 mutation W1204R alters voltage-dependent gating of Nav1.1 sodium channels, Neuroscience 2003, 116, 37–48. Alekov, A. K., Rahman, M. M., Mitrovic, N., Lehmann-Horn, F., Lerche, H., A sodium channel mutation causing epilepsy in man exhibits defects in fast inactivation and activation in vitro, J. Physiol.(London) 2000, 529, 533–539. Alekov, A. K., Rahman, M., Mitrovic, N., Lehmann-Horn, F., Lerche, H., En-
hanced inactivation and acceleration of activation of the sodium channel associated with epilepsy in man, Eur. J. Neurosci. 2001, 13, 2171–2176. 70 Lossin, C.,Wang, D. W., Rhodes, T. H., Vanoye, C. G., George, A. L., Jr., Molecular basis of an inherited epilepsy, Neuron 2002, 34, 877–884. 71 Lossin, C., Rhodes, T. H., Desai, R. R., Vanoye, C. G.,Wang, D., Carniciu, S., Devinsky, O., George, A. L., Jr., Epilepsy-associated dysfunction in the voltage-gated neuronal sodium channel SCN1A, J. Neurosci. 2003, 23, 11289– 11295. 72 Spampanato, J., Kearney, J. A., de Haan, G., McEwen, D. P., Escayg, A., Aradi, I., MacDonald, B. T., Levin, S. I. et al., A novel epilepsy mutation in the sodium channel SCN1A identifies a cytoplasmic domain for b subunit interaction, J. Neurosci. 2004, 24, 10022–10034.
25
27
2 Molecular Biology Techniques for Structure – Function Studies of Ion Channels Louisa Stevens, Andrew J. Powell, and Dennis Wray
2.1 Introduction
Recombinant DNA manipulation and expression techniques have been used to extensively characterise the amino acid residues and domains that influence specific ion channel functions. Structure–function studies have been carried out to investigate all aspects of ion channel function, including channel gating, ligand binding, channel inactivation/desensitisation, voltage-dependence, ion selectivity, multimeric assembly, channel modulation by intracellular pathways or accessory subunits, membrane trafficking and toxin/drug binding sites. Over the last ten years, a plethora of molecular biology techniques and tools have been described to manipulate cDNA sequences in order to generate expression constructs for protein structure–function studies. In this chapter, our aim is to review some of these techniques and to describe how they have been applied to ion channels. Rather than reproduce a basic molecular biology book, we refer readers unfamiliar with recombinant DNA manipulation to molecular biology textbooks (e. g. Watson et al. [1]) and for practical details for specific techniques to Sambrook et al. [2]. A typical ion channel structure–function study will initially involve an analysis of the amino acid sequence of the channel of interest to identify regions that may be involved in the channel property under investigation. For example, studies to identify residues involved in ligand binding may focus on residues within an extracellular domain that are conserved between channel family members that respond to the same agonist or that are not conserved in family members that do not respond to the agonist. Once candidate domains have been identified, the role of these domains can be investigated experimentally through the generation of cDNAs for chimeric channels, where the candidate domain from the agonist-responsive channel is replaced with the corresponding domain from the non-agonist-responsive channel. By functionally characterising the agonist responses of these “domain-swap” chimeric channels in a recombinant expression system the specific regions that are involved in agonist sensitivity can be identified. These studies can then be followed up by using site-directed mutagenesis to introduce single amino acid substitutions of specific candidate residues within that domain Expression and Analysis of Recombinant Ion Channels. Edited by Jeffrey J. Clare and Derek J. Trezise Copyright # 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim ISBN: 3-527-31209-9
28
2 Molecular Biology Techniques for Structure – Function Studies of Ion Channels
to pinpoint the key determinants involved. For example, characterisation of chimeric GluR5-GluR6 channels provided data supporting a critical role for the extracellular S2 domain in determining sensitivity to specific agonists [3]. Subsequent single amino acid mutagenesis within the S2 domain identified key amino acids involved in conferring sensitivity to receptor ligands such as AMPA, domoate and kainite. The methods that we will describe are those relating to the generation of cDNA clones for chimeras, amino acid insertions, deletions and substitutions, the addition of tags to enable channel characterisation and the generation of channel subunit concatamers to study homomeric or heteromeric channel assembly. Firstly, however, we will introduce the basic molecular biology methods that can be used to transfer full length or partial cDNA clones between different DNA constructs. The techniques described include the use of restriction enzymes, insertion of short linkers, creation of constructs by polymerase chain reaction (PCR) and the overlap extension method, and site-directed mutagenesis. Our aim is to pass on information about techniques which we ourselves have found useful in ion channel research.
2.2 Methods for cDNA Subcloning
The ability to carry out a detailed structure–function study on a particular channel requires the availability of a cDNA clone for that channel, in a plasmid construct appropriate for the expression system to be utilised for the functional characterisation experiments. The specialised techniques for de novo cDNA cloning are not covered here, but instead we describe techniques for sub-cloning cDNAs into alternate plasmid vectors. Historically, restriction enzymes, the basic tools of molecular biology, have been used to transfer cDNA fragments from one vector to another. Recently, however, several new techniques have emerged to expedite cDNA sub-cloning without the use of restriction enzymes [4]. These techniques are based on the use of cDNA amplification by PCR or the use of site-specific DNA recombination, each of which are also covered in this section. 2.2.1 Conventional Sub-cloning Using Restriction Enzymes and DNA Ligase
Restriction endonucleases cleave double stranded DNA at specific short sequences called restriction sites. Digestions of plasmid vectors with appropriate restriction enzymes allows the isolation of the required fragments, i. e. those encompassing the channel cDNA, which can be subsequently re-ligated into an alternate plasmid vector containing the desired regulatory sequences, for example, for recombinant expression (see Chapter 4). Over 500 restriction endonucleases have been isolated from several hundred bacterial strains and made commercially available. These include enzymes that recognise and cleave at 4-base, 6-base and 8-base sequences
2.2 Methods for cDNA Subcloning
in double-stranded DNA. Usually 6- and 8-base cutters are used for sub-cloning, since their recognition sequences are less likely to occur frequently in a cDNA sequence or plasmid construct backbone. Many enzymes cut leaving specific 5' or 3' single-strand DNA overhangs (“sticky ends”) while others leave no overhang (“blunt ends”). The advantage of using enzymes with single strand overhangs is that specific ligations can be carried out to enable precise assembly of constructs. On the other hand, blunt-end cutting enzymes are useful because any blunt ended fragment can be ligated with another blunt ended fragment produced by a different enzyme. Unique restriction enzyme recognition sites in channel cDNAs and in plasmid vector DNA sequences can be found using GCG software, other commercially available DNA analysis software packages, or on-line programs. The manufacturers always supply full information about reaction conditions and protocols. Ideally restriction enzymes should not be used at incubation temperatures above 37 8C because of DNA degradation; shopping around with different companies does help as some sell forms of the same enzyme with different incubation temperatures. Once the required restriction digest fragments have been isolated, following separation by agarose gel electrophoresis, in our hands we find it is best not to try to cut with two different enzymes at the same time (even if the manufacturer’s instructions allow it), as the efficiency of the enzymes may be compromised. Before the ligation step, it is best to dephosphorylate the vector (using shrimp alkaline phosphatase or antarctic phosphatase) to prevent self-ligation. The appropriate fragments can then be ligated using T4 DNA ligase and the ligated DNA transformed into E.coli bacteria. E.coli strains such as XL1-Blue (Stratagene) or DH10B (Invitrogen) can be used successfully in many cases; however strains that either decrease plasmid recombination (SURE, Stratagene or STBL2, Invitrogen) or decrease plasmid copy number (ABLE-C or ABLE-K, Stratagene) can be useful to sub-clone channel cDNAs that are difficult to propagate due to high GC content or long repetitive sequences (e. g. NaV or CaV channel cDNAs, see Chapter 4 for further details). Large clones, like calcium channels, do not always ligate easily, and it may be necessary to cut the clone into smaller fragments and then subclone these one at a time. The plasmid DNA can be extracted from individual bacterial colonies, correct clones identified by restriction digest analysis and sequenced to confirm the integrity of the new construct. It is sometimes necessary to insert a very short linker sequence into a clone or a vector. For instance, it may be required to insert a short tag into a sequence so that the resulting protein can be recognised, e. g. by antibodies directed against the tag, such as the FLAG epitope. Another example where a short linker may need to be inserted is in order to alter the multiple cloning site in a vector to enable the easy insertion of a cDNA using different restriction enzymes from those already present in the multiple cloning site of an existing vector. For this technique, two matched oligonucleotides (i. e. a sense oligonucleotide and corresponding antisense oligonucleotide) are designed so that when the pair are annealed they correspond to the required linker with the necessary sticky ends for the existing restriction sites. This is best illustrated by an example (Fig. 2.1), where we used this
29
30
2 Molecular Biology Techniques for Structure – Function Studies of Ion Channels
Fig. 2.1 Modification of multiple cloning site (MCS) in a vector by insertion of a synthetic oligonucleotide linker. The figure shows, on the left, part of the MCS of a vector, which is cut with BamHI and HindIII, removing a SnaBI site. The linear vector is then dephosphorylated to prevent religation. On the right, two synthetic oligonucleotides are phosphorylated with polynucleotide kinase (PNK) and annealed to form a double-stranded linker.
The annealed linker contains matching BamHI and HindIII sticky ends, as well as the sequences for the new restriction sites to be introduced (XbaI, EcoRI and NotI). After ligation of the linker into the linear vector (bottom of figure), the resulting vector contains XbaI, EcoRI and NotI restriction sites instead of SnaBI. (This figure also appears with the color plates.)
method to modify the multiple cloning site in pGEM-HE vector [5]. We needed to replace the SnaBI restriction site (between BamHI and HindIII) with XbaI, EcoRI and NotI sites to produce a new multiple cloning site; this modified vector was then successfully used to express the channel in Xenopus oocytes for voltageclamp recording (Fig. 2.2) [5]. For this example, matched oligonucleotides were designed containing the XbaI, EcoRI and NotI restriction sites, together with sticky overhangs at each end carefully designed to allow the linker to be ligated into the vector using BamHI and HindIII at either end; the exact details of these oligonucleotides are shown in Fig. 2.1. Synthetic oligonucleotides are not normally synthesised in the phosphorylated form; phosphorylation at the 5' end needs to be carried out to enable subsequent ligation. The annealed double-stranded oligonucleotide linker was then ligated (with T4 ligase) into the vector, which had been similarly cut with BamHI and HindIII.
2.2 Methods for cDNA Subcloning Fig. 2.2 Functional expression of the CaV3.1 calcium channel in Xenopus oocytes. (A) A sample recording is shown for a voltage step from –80 mV to –30 mV. (B) The current–voltage relation is shown for the mean of 12 cells.
2.2.2 PCR-based cDNA Sub-cloning
It is often necessary to subclone a cDNA construct or section thereof into a vector. However, when no suitable restriction sites are available in the cDNA construct, it is easily possible to create new restriction sites at either end of the construct using primers with overhangs and PCR. For this, primers are designed partly complementary to the construct to be inserted (around 20 bases), but with additional bases as overhangs containing the restriction sites (around 10 bases) (Fig. 2.3). Following amplification, the PCR product is restriction digested with enzymes corresponding to the sites in the overhangs and ligated into the digested vector as described in Section 2.2.1. An advantage of the PCR-based cloning approach is that additional sequences can be introduced, for example, translation initiation signals (Kozak sequence, see Chapter 4.2) or epitope tags to enable channel detection or purification (see Section 2.5 and Chapter 9). The use of thermostable DNA polymerases with proof-reading activity is recommended when using PCR amplification to generate cDNA fragments for sub-cloning, since the resulting cDNA is less likely to contain mutations introduced during the polymerization. Many DNA polymerases exhibit 3' to 5' proof-reading exonuclease activity that allows the enzyme to check each nucleotide during DNA synthesis and excise mismatched nucleotides in the 3' to 5' direction. A number of thermostable proof-reading DNA polymerases are commercially available, for example, Pfu polymerase (Stratagene), Pfx polymerase (Invitrogen) and Vent polymerase (New England BioLabs).
31
32
2 Molecular Biology Techniques for Structure – Function Studies of Ion Channels
Fig. 2.3 Creating constructs using PCR. The figure shows the construction of an N-terminal GST fusion protein attached to the Nterminal residues (codons 2–181) of the Kv2.1 potassium channel. The starting point was to PCR amplify a fragment of Kv2.1 (right-hand side of figure) using primers with overhangs containing restriction sites (EcoRI
and XhoI) (note that extra bases are added to allow the enzymes to cut efficiently). The PCR product was then cut with these enzymes. The pGex-4T-3 vector, which contains the GST tag (left-hand side of figure), was cut with the same enzymes, dephosphorylated, and ligated with the PCR product to produce the required fusion protein (rKv2.1N2–181).
An alternative PCR-based approach, that removes the requirement for restriction digests and ligation, takes advantage of the fact that Taq polymerase adds a single 3' adenine base to the end of the resulting DNA fragment [6, 7]. Several companies produce PCR fragment cloning kits that enable ligation into vectors which are provided as linear DNA with single 3'-thymidine overhang (TA cloning kit, Invitrogen, The T-Easy system, Promega). In many cases, the vectors contain the coding sequence for b-galactosidase which is interrupted by successful cloning of the PCR fragment. This gives white (rather than blue) colonies when transformed E.coli are plated onto agar plates with X-Gal, which would turn blue if broken down by b-galactosidase. Thus, clones that contain the vector and inserted PCR fragment are identified as white colonies. Since the TA sub-cloning approach does not determine the direction that the PCR product is inserted, it is necessary to analyse the resulting clones to determine the orientation of the cDNA in relation to the regulatory sequences contained in the vector. The TA vectors contain
2.2 Methods for cDNA Subcloning
restriction sites either side of the cloning site, to enable subsequent cDNA subcloning as required. An important consideration when TA subcloning PCR fragments is that Taq polymerase lacks proof-reading activity and is therefore prone to make errors which can introduce mutations. On the other hand, proof-reading polymerases such as Pfu produce blunt ended PCR products and therefore cannot be directly sub-cloned; instead a common approach is to create the PCR fragment with Pfu, and then subsequently incubate with Taq polymerase (72 8C for 10 min), which adds adenine overhangs at each blunt end. This product can then be cloned using the TA cloning systems. Alternatively, a system that allows high efficiency cloning of either blunt ended or 3'-A overhang PCR products has also been developed. The TOPO cloning system (Invitrogen) utilises a novel approach, whereby each end of the linear cloning vector provided is covalently linked to a single topoisomerase I enzyme [8]. Topoisomerase I catalyzes the ligation of the PCR product into the vector, releasing the enzyme from the construct. The ligated DNAs are transformed into E.coli and positive clones identified in a manner similar to the other methods described. A range of TOPO cloning vectors are available to allow the generation of cDNA constructs for a number of purposes. This system has also been adapted to allow directional cDNA cloning, through the introduction of a short specific sequence at the 5'end of the PCR primer used to initiate polymerization of the sense-strand of the cDNA (Directional TOPO Kit, Invitrogen). As an example of TA subcloning, this technique was used when cloning the human EAG2 potassium channel from a cDNA library [9]. Primers were designed to amplify the channel from a human foetal brain cDNA library using Pfu polymerase, and the PCR product was subcloned into pGEM-T-Easy (Promega) after treatment with Taq polymerase to yield 3'A overhangs. The insert was then transferred to the Xenopus oocyte expression vector pGEMHE using EcoRI restriction sites, and complementary RNA prepared for injection into oocytes. A clone with the hEAG2 cDNA in the correct orientation was identified, the channel expressed in oocytes and characterised by two-electrode voltage clamp electrophysiology [9]. Figure 2.4 shows examples of currents observed by voltage-clamp in oocytes and the resulting current–voltage relation for the newly cloned hEAG2 potassium channel. 2.2.3 Sub-cloning cDNA through Site-specific Recombination
Sub-cloning using restriction enzymes and ligase or PCR-mediated modification and amplification both require isolation, by agarose gel electrophoresis, and subsequent purification of the cDNA restriction fragment or PCR product. A further complication with this approach is that sub-cloning strategies can become rather complicated as it is often difficult to find suitable restriction sites that are either absent or occur infrequently within the target cDNA. Cloning systems that involve site-specific recombination avoid these problems by removing the need to use restriction enzymes and to isolate and purify DNA fragments, since recombination
33
34
2 Molecular Biology Techniques for Structure – Function Studies of Ion Channels Fig. 2.4 Functional expression of hEAG2 potassium channel in Xenopus oocytes. (A) An example of a current family is shown during a series of depolarising steps from –80 mV to +70 mV. (B) The current–voltage relation is shown for the mean of 17 cells, normalised to the current at +70 mV.
between complementary DNA sequences can take place directly between the parental circular plasmid DNAs. One such system, the Gateway Technology cloning system (Invitrogen) provides a straightforward mechanism for sub-cloning cDNA inserts between different cloning and expression vectors without the use of restriction enzymes and ligases. The system utilises well-characterised site-specific sequences involved in recombination and crossover in lambda bacteriophage. Lambda recombination occurs between site-specific attachment (att) sites [10–12]. These att sites contain the binding sequences for the recombination proteins, and the recombination reactions are catalyzed by a mixture of enzymes that bind to these specific sequences, bringing the target sites together, cleaving and covalently attaching the DNA. In this system, attP sites recombine with attB sites to give attL and attR sites (and vice versa as the reaction can be driven in either direction), giving a high specificity to the reaction. The experimental steps in the process are as follows. The first step is to create a PCR product of the desired insert/construct using primers with overhangs of attB1 and attB2 at the 5' and 3' ends of the construct (Fig. 2.5). The PCR product (i. e. the construct flanked by attB sites), is then reacted with the “Donor vector” (which already contains attP sites) to produce an “Entry clone” that now contains the construct flanked by attL sites (Fig. 2.5); this reaction is called the BP reaction. After transformation and selection with kanamycin, the Entry clone can now be used to subclone into a whole range of expression vectors. This step involves reaction of the Entry clone with a “Destination vector” (containing attR sites) to produce the required expression clone (with attB sites), as shown in Fig. 2.5, the LR
2.2 Methods for cDNA Subcloning
Fig. 2.5 GatewayTM cloning technique. attB sites are introduced upstream and downstream of the cDNA to be sub-cloned by PCR (top left), using primers with attB sequence overhangs. The PCR product is then reacted with a “Donor vector” (left) which contains attP sites, to create an “Entry clone” in which the cDNA sequence is flanked by attL sites. This reaction (“BP ClonaseTM reaction”) is cat-
alyzed by integrase and integration host factor (IHF), and the product selected with kanamycin. The cDNA from the Entry clone can then be integrated into a range of different “Destination vectors”, containing attR sites, in LR ClonaseTM reactions (right), yielding the required expression clone. The LR reaction is catalysed by integrase, excisionase and IHF and the product selected with ampicillin.
reaction. The expression clone is then ready for use after selection with ampicillin. Gateway-mediated subcloning allows the straightforward generation of a range of destination vectors that can be used for expression studies. Modification of the sequences between the flanking att-sites and the channel open reading frame within the cDNA insert, to ensure maintenance of the open reading frame through the attB site formed post-recombination, will allow the use of vectors that can introduce either N- or C- terminal tags as required. A range of destination vectors are commercially available that allow expression in bacteria, yeast, baculovirus and mammalian cells. The Gateway system is becoming more widely used; for instance Obrdlik et al. [13] used the system in their study of binding partners for Arabidopsis potassium channels (KAT1, AKT1 and AKT2). The Gateway site-specific recombination approach for cDNA sub-cloning is amenable for robotic automation. Once an Entry vector has been generated with
35
36
2 Molecular Biology Techniques for Structure – Function Studies of Ion Channels
the required channel cDNA flanked by attL sites, the processes required for Gateway mediated sub-cloning into multiple destination vectors are readily automated (the ClonaseTM reactions, bacterial transformation, DNA mini-prep analysis and sequencing), allowing the parallel generation and validation of multiple channel cDNA constructs for multiple purposes [14]. The application of automation is particularly beneficial when the Entry vector is designed such that it allows in-frame recombination into Destination vectors. Using this approach a panel of constructs can be generated to investigate the utility of different N- or C-terminus tags in expression studies, for example, for structural work (see Chapter 9).
2.3 Generation of Chimeric Channel cDNAs
Chimeric cDNA sequences can be generated using several techniques. Restriction enzymes can be used if appropriate sites are fortuitously available within the channel cDNA at positions that allow replacement of the domain of interest. This can provide a relatively straightforward and rapid method to create a range of constructs, including those for deletion and insertion mutants as well as domainswap chimeras. Often, however, restriction enzyme sites are not conveniently located within the channel cDNA to allow generation of the optimal chimera, deletion or insertion mutants needed to investigate the role of a specific domain in channel function. Thus, the chimeras generated are dictated by the position of the available restriction sites rather than by scientific consideration of the putative domain boundaries within the channel. To overcome this problem several methods have been developed over recent years that can be used to generate “seamless” chimeras. These approaches avoid the use of restriction sites through the use of specifically designed oligonucleotide primers and PCR amplification of the modified cDNA construct. In this section we will also give examples of the utility of various approaches in creating channel chimeras, insertion and deletion mutants. 2.3.1 Use of Restriction Enzymes to Generate Chimeric Channel cDNAs
Where restriction sites are conveniently available at the boundary of the domain to be replaced, these can be used to generate the chimeric channel cDNA, provided that ligation of the chosen restriction sites retains the open reading frame to produce the chimeric protein. An example of the use of restriction sites to create a chimeric channel is shown in Fig. 2.6. Restriction enzymes were used to create chimeras between rat and human forms of the voltage-gated potassium channel Kv2.1 [15]. This was done in order to locate the molecular domains that underlie the differences in activation kinetics between the two versions of the channel. As shown in Fig. 2.6, both rat and human Kv2.1 were digested with the enzymes BssHII and Bsp1407I (TypeII restriction endonucleases). The appropriate DNA fragments were isolated by purifi-
2.3 Generation of Chimeric Channel cDNAs
Fig. 2.6 Example of the use of restriction enzymes to create a chimera. A chimera was constructed between rat and human forms of the Kv2.1 potassium channel as shown. Restriction enzymes BssHII and Bsp1407I were used to cut the rat (gray) and human (striped) forms of the channel in their respective vectors. The required DNA fragments were isolated and ligated to form the chimeric cDNA with residues 108 to 528 of the rat channel replaced by human (rKv2.1h108–528).
37
38
2 Molecular Biology Techniques for Structure – Function Studies of Ion Channels
Fig. 2.7 Activation times of Kv2.1 channels. Activation times (time for current to rise from 10 % to 90 % of maximum current, t10–90 %) versus test potential for: (A) chimera rKv2.1h108–528 (residues 108–528 of rat replaced by human), (B) chimera rKv2.1h741– 853 (residues 741–853 of rat replaced by human). Human wild type (&, n = 4), rat wild type (*, n = 10), and chimera ~, n = 7).
cation from agarose gels and ligated in order to create the required chimeric channel with residues 108–528 of the rat channel replaced by corresponding residues from human channel (rKv2.1h108–528) [15]. Characterisation of the chimera demonstrated that it retained the fast activation kinetics typical of the rat wild type channel [15] (Fig. 2.7A), suggesting that residues 108–528 do not contribute to the differing activation kinetics between rat and human Kv2.1. Another chimera with residues 741–853 of rat Kv2.1 replaced by human Kv2.1 (rKv2.1h741–853) was created using the same technique (restriction enzymes BsmI and NotI). In this case, by contrast, the activation kinetics were similar to human Kv2.1 (Fig. 2.7B), showing that residues between amino acids
2.3 Generation of Chimeric Channel cDNAs
741–853 contribute to determining the rate of activation for the two potassium channels. 2.3.2 PCR-mediated Overlap Extension for Chimera Generation
Overlap extension PCR can be used to create chimeric, insertion or deletion mutants in specific regions by “seamlessly” fusing the required cDNA fragments [16, 17]. This powerful method is very useful for the precise creation of chimeric cDNAs when useful restriction sites are not available for the restriction-ligation method. The technique involves PCR amplification of fragments that encompass the required domains from each of the “parent” cDNAs. Assembly of these PCR fragments is achieved via combining the isolated fragments in a further PCR amplification; correct assembly of the fragments is enabled by the presence of complementary sequences (overhangs) incorporated within the primers used for the initial PCR. The example shown in Fig. 2.8 demonstrates the use of this method to construct a cDNA with a deletion of the C-terminus of the rat Kv2.1 potassium channel; in this example, residues 412 to 853 were deleted [18]. The initial step was to produce two separate PCR products for each part of the required construct, using primers with complementary DNA sequence at the join (i. e. just after codon 412 in one fragment, and just before the stop codon in the other fragment). In the second step, the two PCR products were then combined in a further PCR reaction. Joining of the two fragments is made possible by the overlaps which were introduced via the overhangs in the previous step, each fragment constituting a ‘megaprimer‘. The resulting DNA, constituting the fusion of the two initial PCR products, was then further amplified through polymerization initiated from two primers (1 and 4 in Fig. 2.8) complementary to the extreme ends of the fusion product. The final step involved cutting the PCR product at any nearby restriction sites (at sites outside the region being swapped, ApaI and NotI in this example) (Fig. 2.8), followed by ligation (with T4 DNA ligase) into the original clone, which was also cut with the same enzymes. This cDNA construct was used to express the deletion in COS-7 cells, followed by purification of the protein and electron microscope single particle studies [18]. Stepwise PCR amplification and overlap extension of two, three or more cDNA fragments can be carried out to create chimeras with specific fusion boundaries [19]. Thus, the technique can also be used to substitute domains within internal regions of a protein. A second example (Fig. 2.9) shows an extension of the method to construct a chimera between rat and human forms of the Kv2.1 potassium channel [15]. In this example amino acids 741 to 795 within the C-terminal activation domain of rat Kv2.1 were replaced by the corresponding residues in the human channel, therefore overlap extension of three cDNA sequences was required. As before, the initial step was to create PCR products with complementary sequence at the joins (Fig. 2.9). In the second and third steps, the PCR products were joined together by further PCR reactions, again facilitated by the comple-
39
40
2 Molecular Biology Techniques for Structure – Function Studies of Ion Channels
Fig. 2.8 Example of a deletion by the overlap extension PCR method. In this example, the overlap extension method was used to make a deletion of the C-terminal domain (codons 412–853) of the rat Kv2.1 potassium channel. The first step (“first round PCR”) was to PCR amplify part of the Kv2.1 channel that was to be included (up to codon 411, in black) along with a fragment of the pMT3 vector (in gray) using a primer (3) to introduce a stop codon.
Primers are designed with overhangs as shown. The two PCR products were used in a further PCR reaction (“second round PCR”) to produce a fragment containing restriction sites (ApaI and NotI). After cutting the fragment with these enzymes, the final product was ligated into the similarly cut wild type channel cDNA vector to generate the truncated Kv2.1 channel cDNA.
2.3 Generation of Chimeric Channel cDNAs
Fig. 2.9 Example of a chimera made by the overlap extension PCR method. The example shows the construction of a chimera between rat and human Kv2.1 potassium channels with codons 741–795 of the rat channel replaced by corresponding residues of the human channel. For the first round PCR, products were made for rat Kv2.1 sections (black) and the human Kv2.1 section (gray) that is to
be replaced to form the chimera. Primers are again used with overhangs as shown. The second round PCR joins two of the fragments, and the third fragment is added in the third round PCR. The final PCR product is digested with restriction enzymes (Bsp1407I and NotI) and sub-cloned into the similarly cut wild type rat Kv2.1 cDNA construct.
41
42
2 Molecular Biology Techniques for Structure – Function Studies of Ion Channels
mentary sequences introduced by the primers overlying the joins in the initial PCRs (Fig. 2.9). Digestion of the final PCR product is required, followed by ligation into the similarly digested wild type rat Kv2.1 clone. This chimera, with residues 741–795 of the rat Kv2.1 channel replaced by corresponding residues in the human channel (rKv2.1h741–795), was used in expression studies in oocytes to determine the effect of these residues on the activation kinetics of the channel [15]. The results shown in Fig. 2.10 show that these residues alone did not affect the activation kinetics of the rat channel, as the chimeric channel retained fast activation kinetics similar to wild type rat Kv2.1.
Fig. 2.10 Examples of data obtained using constructs made by overlap extension. Mean activation times (t10–90 %) are shown for chimera rKv2.1h741–795 (residues 741–795 of rat replaced by human). Human wild type (&, n = 5), rat wild type (*, n = 4), and chimera (~, n = 6).
Successful use of the overlap extension technique relies on careful primer design to ensure that the correct chimeric product will be expressed from the cDNA. It is advisable first to assemble the DNA sequence for the required construct in silico (using one of the available DNA sequence analysis programs) and to translate the sequence to make sure the new reading frame expresses the expected chimeric channel protein. For each PCR fragment, the overhangs in the primers should be 10 bases and the template-specific sequence for the fragment about 20 bases. Where possible, primers should be designed such that they have the same calculated melting temperature (for the template-specific part of the primer); 60 8C is optimal to allow the annealing temperature in the PCR reaction to be set at 55 8C. Large fragments (<3 kb) do not join very well to small fragments; it is worth trying to keep the length of the PCR fragments similar to help the stability of the PCR when it is annealed. It is possible to carry out the sequential PCR steps to produce the final PCR product in one reaction containing all the primers and cDNA template mix for all of the fragments to be fused. However, we have found that better yields are obtained if the PCR reactions are carried out in separate steps and the PCR products isolated on an agarose gel and checked for size prior to the next step. Increasing the amounts of DNA template or polymerase can be tried if the overlap extension is not initially successful. In addition, adding DMSO at a final concentration of 5 % v/v may also help, particularly when GC-rich cDNA sequences are being amplified. Proof reading polymerases (e. g. Pfu, Pfx or Vent)
2.4 Site-directed Mutagenesis
should again be used to minimize the possibility of introducing unwanted sequence errors during the PCR amplification and, to ensure errors have not been introduced, the entire sequence of the resulting chimeric cDNA insert should be confirmed by sequencing. For the final step, restriction enzymes should be chosen so that at least 500 bases are removed, making it easy to detect correct cutting of the final fragment when run on a gel. 2.3.3 PCR-mediated Integration or Replacement of cDNA Fragments
Another powerful PCR-based approach applicable for the generation of channel chimeras has recently been described by Geiser et al. [20]. This technique is an adaptation of the Stratagene QuikChange site-directed mutagenesis method [21, 22]) which is covered in the next section, and so we will return again to a discussion of this useful approach after the Quik-Change method has been described.
2.4 Site-directed Mutagenesis
Once a specific ion channel domain that influences a particular channel function has been identified through the characterisation of chimeric channel mutants, the role of individual amino acids within the domain can be characterised through the introduction of specific small (single, double or more) amino acid deletions, insertions or substitutions (“site-directed mutagenesis”). Generally, amino acid substitutions are preferred, since the role of specific amino acid side chains can be investigated while avoiding gross changes to the polypeptide backbone. The influence of charged, neutral or bulky amino acids in mediating a specific channel function can be determined through replacement with residues with either the opposite or intermediate characteristics. The most common technique for site-directed mutagenesis, commercialised by Stratagene (QuikChangeTM Mutagenesis), can be used quickly and easily to introduce mutations into a cDNA construct, and is illustrated in Fig. 2.11. To generate the mutation, this technique uses two primers that are each complementary to the opposite strands of the plasmid DNA template but contain the appropriate base substitutions to give rise to the desired mutation. After separation of the double stranded circular DNA at high temperature, primers are annealed at the target site at lower temperature and the template extended with Pfu polymerase, producing the required mutant strands. The procedure is repeated in a thermocycler some 25 times. It is worth noting that, although PCR-like processing is used, this is not strictly a PCR reaction because exponential amplification cannot occur. Next, the DNA products are treated with the enzyme DpnI, an endonuclease which cleaves dam-methylated DNA. Only DNA that has been previously isolated from E. coli will be dam-methylated and so the original parental (i. e. wild type) template will be digested, while the synthesised mutant product will remain. The mutant is
43
44
2 Molecular Biology Techniques for Structure – Function Studies of Ion Channels
then transformed into E. coli (which repairs a nick in the mutant strand, Fig. 2.11), and the mutated plasmid DNA extracted from several individual colonies and sequenced to confirm that the mutagenesis has been successful. As usual, primer design is crucial for successful results. The desired mutation should be in the middle of the primers to aid accurate annealing; primers should be between 25 and 45 bases long, and the theoretical annealing temperature should be greater than or equal to 78 8C, and they should have a minimal GC content of
Fig. 2.11 Site directed mutagenesis. Primers containing the required mutant sequence (in gray) are annealed to the cDNA construct (in black) and extended by thermocycling using Pfu DNA polymerase. The enzyme DpnI is added to digest the original dam-methylated plasmid template leaving the required mutant construct. The resulting mutated DNA is transformed into E. Coli (which repairs the nicks shown), and clones verified by sequencing.
2.4 Site-directed Mutagenesis
40 %. A control reaction should be carried out alongside the mutant, with the control mix omitting the polymerase. Upon transformation of control and mutant reaction products, colonies should be present for mutant but not for control (since the DpnI reaction should have removed the control wild type clones). In order to decrease the risk of unwanted mutations being introduced by the polymerase, it can be beneficial to sub-clone a short section of cDNA containing the site-directed mutation(s) back into the original wild type clone using appropriate restriction sites close to the mutations. In any case, sequence validation of the resulting cDNA clone is essential. 2.4.1 Examples of the Use of Site-directed Mutagenesis
The technique of site-directed mutagenesis has been widely used. One example is in the creation of silent mutations (i. e. mutations that alter bases but not the corresponding amino acid). Introduction of silent mutations can be useful in order to produce restriction sites that make subsequent sub-cloning easier. This sitedirected mutagenesis technique was used to introduce Bsu36I and BsmI sites into the human Kv2.1 channel [15]. The rat form of this channel already contained these restriction sites at homologous positions, and so introduction of the same sites enabled us to create the required chimeras between rat and human channels using these restriction enzymes. There are many examples in the literature describing the construction of site-specific mutations to meet the requirements of each individual study [e. g. 23–26]. For example, studies on Kir channels have focused on the role of native cysteine residues on channel function and membrane trafficking [25, 26]. Studies on P2X receptors to investigate the extracellular domain structures involved in ATP-binding, channel gating and modulation have utilised site-directed mutagenesis in order to mutate conserved positive residues [27–29], aromatic residues [28, 30], cysteines (to investigate disulfide bonding, [31, 32]), histidines (to investigate Zn and pH modulation, [33]) and proline residues (to investigate secondary structure, [34]). In contrast to studies where specific residues are substituted with others on the basis of specific expectations within the study, some mutagenesis approaches for investigation of protein structure and function involve the systematic replacement of residues in the domain of the channel under study by specific amino acids, followed by characterisation of the mutant’s functions. One such approach is alanine-scanning mutagenesis, where the sequential residues within the domain under investigation are each replaced by an alanine residue to generate a panel of mutant cDNAs for functional characterisation. Other frequently used scanning mutagenesis approaches involve replacement of single amino acid residues with cysteine, lysine, proline, tryptophan or histidine. Each approach enables determination of the role of specific amino acid properties in the function of the channel domain being studied. For example, mutation to alanine removes the amino acid side chain and substitutes a small hydrophobic group, which would be expected to interact favorably with membrane lipids or hydrophobic regions [35]. When ala-
45
46
2 Molecular Biology Techniques for Structure – Function Studies of Ion Channels
nine scanning was carried out on S1, S2, S3 and S4 regions of a potassium channel, periodic effects were observed on channel gating, consistent with a-helical structures of these regions and the clustering of gating effects on faces of the helices, leading to a model for the packing of these helices [36]. The technique has also been widely used to determine toxin and drug binding sites, either through mutagenesis of the channel to determine the key determinants of a channel binding site, such as that used to define the drug binding site on the hERG channel [37], or through mutagenesis of a toxin to identify the epitope on the toxin involved in binding, such as that carried out to identify key residues on o-atracotoxin-Hv1 a required for selectivity against calcium channels [38]. Alanine-scanning mutagenesis of the rat brain NaV1.2 channel identified specic amino acid residues in the IVS6 transmembrane domain involved in binding the local anesthetic, etidocaine, as several alanine substitutions significantly reduced voltageand frequency-dependent block [39] by the compound. Cysteine-scanning mutagenesis is particularly beneficial in studies of structure–function relationships in membrane proteins. The introduced cysteines can be covalently modified by charged, hydrophilic, sulfhydryl reagents to probe the accessibility of the substituted cysteines in transmembrane domains. The effects of the covalently bound reagent on channel function can then be characterised. This approach is termed the substituted cysteine accessibility method (SCAM, [40]) and has been used to study the dynamics of channel function in potassium channels [41, 42], calcium channels. [43], nicotinic acetylcholine receptors [44–46], P2X receptors [47–49] and GABA-A receptors [50, 51], amongst others. Typical sulfhydryl reagents used for these studies are parachloromercuribenzensulfonate (PCMBS) or derivatives of methane thiosulfonate (MTS), either positively charged methane thiosulfonate ethylammonium (MTSEA) and methane thiosulfonateethyltrimethylammonium (MTSET), negatively charged methane thiosulfonateethylsulfonate (MTSES) or neutrally charged methyl methane thiosulfonate (MTSM). In some studies, cysteine binding reagents that are not membrane permeable are applied in the extracellular solution, and the effects of the reagent will only be noted if the introduced cysteine residue is exposed and accessible to the extracellular solution. These cysteine accessibility studies have proved extremely useful in investigating ion channel structure and function, since they provide a method whereby dynamic changes in structure during channel activation or inactivation can be analysed. For example, the approach was used to demonstrate that the S4 transmembrane region of the prototypical Shaker voltage-gated potassium channel moves outwards upon membrane depolarisation, confirming the role of this region as the voltage sensor [52, 53]. Cysteine substitutions were introduced into the S4 domain, the mutants expressed in Xenopus oocytes and their sensitivity to extracellularly applied sulfhydryl reagents characterised by two-electrode voltage-clamp electrophysiology. Upon membrane depolarisation, movement of the S4 domain during channel gating exposes introduced cysteines (that were inaccessible prior to depolarisation) to the sulfhydryl reagent added in the external solution. Covalent modification of the exposed cysteine residues was evident through the appearance
2.4 Site-directed Mutagenesis
of cumulative current block upon repetitive activation of the channel. The data demonstrated that, upon depolarisation, the S4 transmembrane segment undergoes a conformational change that leads to channel opening. Similar results have been obtained for calcium channels using cysteine-scanning mutagenesis techniques [43]. Cysteine mutants were generated using QuikChangeTM site-directed mutagenesis at positions V263, A265, L266, A268, F269, and V271 within the S4 segment of domain I in a chimeric CaV1.2/CaV3.1 calcium channel. The cysteine-binding reagent PCMBS was applied extracellularly to each of the mutants expressed in oocytes and depolarizing pulses applied. As can be seen in Fig. 2.12, the introduced cysteines in mutants V263C, A265C, L266C, and A268C reacted with the PCMBS to give a reduction in the recorded current, with mutants F269C and V271C remaining unaffected. This shows that, upon depolarisation, movement of the S4 transmembrane domain S4 exposes residues 263–268 to the extracellular solution, while residues 269 and 271 remain buried in the membrane. Further experiments demonstrated that movement of S4 is voltage dependent because PCMBS gave a voltage-dependent block of CaV3.1 current, indicating outward movement of the S4 region upon depolarization. The differing charge on various cysteine binding reagents can be exploited in order to probe the influence of side chain charge on channel properties. For example, substituted cysteine accessibility mutagenesis identified a residue within the P2X2 ATP-gated channel (I67) that gives normal function when the mutant channel was exposed to neutral or positive methanethiosulfonates while exposure to negative MTSES decreased agonist potency, demonstrating that the maintenance of positive charge close to I67 (residues K69 and K71) is required for ATP binding and channel activation [29], and introduction of negative charge at residue 67 disrupts this. On the other hand, substituted cysteines that are accessible to modification by a neutral methanethiosulfonate were identified at positions within the first and second transmembrane domains of this channel; introduction of positive and negative charges at these positions using MTSET or MTSES has allowed investigation of the ion permeation pathway and the aqueous environment around the intracellular and extracellular ends of the transmembrane domains [29, 47, 48, 54]. Cysteine scanning mutagenesis has also been used without cysteine modifying reagents to investigate the proximity of residues through disulfide formation between engineered cysteines. This approach was used to address the subunit arrangement of homomeric P2X2 and P2X4 receptors and heteromeric P2X2/3 receptors [55]. Engineered cysteines in the outer ends of the first transmembrane domain of one subunit and the second transmembrane domain of the second subunit demonstrated the proximity of these domains in the heteromeric channel through their ability to form a disulfide bond. Using specific combinations of cysteine-substituted P2X2 and P2X3 mutants and measuring the DTT-sensitive reduction of channel function (caused by the formation of disulfide bonds between the cysteines) demonstrated that the heteromeric trimer formed by P2X2 and P2X3 receptor subunits consists of two P2X3 subunits and one P2X2 subunit. An alternative method of determining the accessibility of mutated residues is to carry out histidine scanning mutagenesis. Substituted histidine mutants are used
47
48
2 Molecular Biology Techniques for Structure – Function Studies of Ion Channels
2.4 Site-directed Mutagenesis
in combination with acidic buffers to investigate accessibility to hydrogen ion of mutated residues [56]. Changes in the solvent accessibility of the introduced histidines that accompany conformation changes in the channel voltage sensor can be detected as pH-dependent changes in the gating currents, due to the protonation of the exposed histidine. The advantages of histidine scanning mutagenesis are that protons are accessible to more confined spaces within the protein (compared to the more bulky sulfhydryl reagents), protonation rates are faster than channel gating rate constants so labelling is effectively instantaneous, and replacement of basic residues (such as the lysines and arginines in the S4 voltage sensor of voltage-gated channels) with histidine is less disruptive than neutralisation by cysteine because the native charge is maintained when protonated. Scanning mutations can be made instead to introduce tryptophan [57] or lysine [58], which have more bulky side chains and so, in principle, allow the detection of functional effects of residues that may be missed by the milder mutations introduced by alanine scanning. Also, tryptophan has a hydrophobic side chain while lysine has a hydrophillic one, so these residues can be used to discriminate between the requirement for hydrophobic or hydrophilic surfaces within a channel domain. For tryptophan scanning mutagenesis it is assumed that side chains involved in protein packing would be sensitive to substitution by tryptophan, whereas those exposed to the lipid membrane would be relatively tolerant to this mutation. In studies on the transmembrane domains of voltage-gated potassium channels [59–62] and ligand-gated channels [57, 63–66], this approach has yielded a pattern whereby every third or fourth residue exhibits sensitivity to an introduced tryptophan – suggesting an a-helical structure with a significant lipid-exposed surface. Hydrophillic lysine mutations can be used to determine the orientation of residue side chains towards core hydrophobic or surface hydrophillic environments. These mutations disrupt subunit assembly at overlapping hydrophobic regions and so may be useful in identifying such regions of overlap between subunits [67]. Lysine-scanning mutagenesis has also been used to characterise hydrophillic surfaces involved in channel–toxin [58] interactions. Proline scanning, though not widely used, has also given some interesting results for ion channels. This residue introduces a kink in the peptide chain, and studies using proline scanning have identified channel gating regions, for example, in an inward rectifier potassium channel [68].
3 Fig. 2.12 Cysteine scanning mutagenesis of the S4 segment of domain I of a calcium channel. (A) The effect of application of PCMBS (100 µM, indicated by the bars) on normalised currents for cysteine mutants V263C (n = 3), A265C (n = 4), L266C (n = 3), A268C (n = 3), F269C (n = 4), and V271C (n = 3) during repetitive depolarisations to +10 mV from a holding potential of –80 mV. (B) Membrane topology of domain I showing the positions of the residues mutated to cysteine (shaded). Movement outwards during depolarization is as far as A268.
49
50
2 Molecular Biology Techniques for Structure – Function Studies of Ion Channels
2.4.2 Modification of the QuikChange Method for the Replacement of cDNA Fragments
The QuikChange technique has been adapted to allow the construction of chimeras [20]. The approach involves two main steps. Firstly, a PCR product must be generated for the region to be integrated into a channel cDNA sequence. For this, primers need to be designed with overhangs at each end of the region to be inserted, such that the overhangs are complementary to the sequences at the intended sites of integration to the channel cDNA. Secondly, following isolation and purification of the PCR product, it is then mixed with the channel cDNA in its plasmid vector, and then thermocycling with Pfu is applied as for the Quikchange method described above. During this process, the primers anneal to the channel cDNA at the chosen insertion sites, determined by the PCR overhangs, and extension occurs from the 5' end of the product overhang along the circular channel cDNA plasmid until polymerization reaches the 3' end of the PCR insert, so integrating the PCR insert into the channel cDNA plasmid. As in the QuikChange method, the methylated parent plasmid is then digested with DpnI, and the reaction product is transformed into bacteria. Colonies containing the chimeric cDNA plasmid with the insertion or replacement are identified through restriction digest or PCR analysis, and the chimeric cDNA confirmed by sequencing. This approach was used to generate P2X receptor chimeras in order to define the determinants of differences in ATP analog sensitivity between the P2X2 and P2X4 receptor subtypes [69]. Functional chimeras were generated in which the extracellular domain of the P2X2 subtype was replaced with the P2X4 extracellular domain. Characterisation of the ligand potencies and receptor desensitisation rates of these chimeras defined the regions of the channels that determine ATP analogue sensitivity.
2.5 Epitope-tagged Channels and Fusion Partners
Structure–function studies of ion channels are often facilitated through the use of tagged ion channel protein or fusion of the channel to a reporter protein. The use of tags or fusions can be applied to monitor channel expression, to localise the channel at the subcellular level, to purify the channel (see Chapter 9) and for analysis of channel topology, dynamics and interactions. Tagging involves the addition of a short peptide sequence to the wild type ion channel polypeptide, which can be used along with specific antibodies or affinity reagents to detect or purify the channel [70]. Examples of frequently used affinity tags are a 10 amino acid fragment of the c-myc protein, an 8 amino acid peptide based on the enterokinase cleavage termed FLAG, a 9 amino acid peptide from the haemaglutinin HA1 protein of human influenza virus or a hexa-His polypeptide. Such peptide tags are usually introduced either at the N- or C-termini of the channel where they are less likely to interfere with the function of the channel; however in some cases, they
2.5 Epitope-tagged Channels and Fusion Partners
are placed within a specific intrapolypeptide domain, such as extracellular or intracellular loops to allow detection of specific channel functions. For example a HA tag was placed in the extracellular S1-S2 loop of the hERG potassium channel in order to detect channel trafficking to the plasma membrane [71]. Channel fusions to larger reporter polypeptides, such as green fluorescent protein, have also been used to analyse sub-cellular localisation of ion channels (see for example Refs. [72–74]). There are several approaches that can be used to generate cDNA that introduce short tags into a channel protein. One technique uses two matched oligonucleotides in a similar manner to the approach used to introduce new restriction digest sites described in Section 2.2.1. The oligonucleotide pair contains the coding sequence for the required tag flanked by a unique restriction site that can be used to insert the short sequence into the open reading frame of the channel cDNA at the required position (in this case the oligonucleotide pair is flanked by the same restriction site overhangs at either end). However, this method is generally limited due to the requirement for restriction sites at appropriate positions within the cDNA. In these cases, tags can be incorporated using PCR-based approaches. Another approach involves the introduction of a tag by placing the channel cDNA into a vector which already contains the necessary tag. The subcloning can be carried out using the available restriction sites in the vector together with matching restriction sites at either side of the channel cDNA insert. For this, the required restriction sites can be introduced into the channel cDNA by the PCR method described in Section 2.2.2, using PCR with primer overhangs containing the restriction sites. Careful primer design is required to ensure that the product is in the correct frame (translation of the required fusion/tagged protein from the resulting cDNA sequence is essential). For instance, this technique was used [15] to sub-clone the N-terminal region (residues 2–181) of a KV2.1 potassium channel into a pGex vector (Amersham Biosciences) which contains a glutathione-S transferase (GST) sequence, such that expression leads to a GST-tagged fusion protein. The GST-N-terminal KV2.1 fusion protein was purified utilising the GSTag and used in studies to show binding with the radiolabelled C-terminal region of the KV2.1 channel [15]. The PCR primers were designed such that only the N-terminus-encoding fragment of the cDNA was amplified and the 3'primer introduced an in-frame stop codon (Fig. 2.3). The 5'primer introduced a flanking EcoRI restriction site, while the 3'primer introduced a XhoI site, to allow in-frame subcloning of the PCR product into the similarly digested pGex vector. The overlap extension technique described in Section 2.3.2 can also be used to create tagged proteins, for instance to create cDNA constructs to express ion channels fused with GFP or its variants, YFP (yellow) or CFP (cyan). Here, the tag or fusion partner cDNA and the channel cDNA are first amplified by PCR using primers containing overhangs corresponding to the overlapping sequence for the intended fusion site. The PCR products are joined by further PCR, made possible by the primer overhangs, and the final PCR product is then cut with restriction enzymes and ligated back into the wild type channel. Putting both N- and C- terminal fluorescent tags on the Kv2.1 potassium channel by this technique enabled
51
52
2 Molecular Biology Techniques for Structure – Function Studies of Ion Channels
Fig. 2.13 Example of data obtained after insertion of a tag using overlap extension. Current-voltage curve for rKv2.1 wild type (!, n = 4), and the chimera rKv2.1N-YFP-C-CFP (*, n = 5) with fluorescent tags on both the N terminus (YFP) and C terminus (CFP).
the use of FRET experiments to investigate movement of the intracellular N- and C-terminal regions of the expressed channel during channel gating (work in progress, [75]). The tags themselves did not affect the electrophysiological properties of the channel (Fig. 2.13).
2.6 Channel Subunit Concatamers
Another aspect of ion channel structure–function analysis that merits discussion is the analysis of homomeric and heteromeric subunit assembly and stoichiometry. Many classes of ion channel consist of multi-subunit complexes, which can be either homomeric or heteromeric. Examples of functional homomeric channel complexes are the P2X1 receptor channel [76], the Shaker potassium channel [77] and the a7 nicotinic acetylcholine receptor channel [78]. Heteromeric examples are the P2X2/3 channel [55], the epithelial sodium channel (ENaC) [79] or the GABA-A receptor [80]. Structure–function studies, including mutagenesis, have helped to resolve the stoichiometry of subunit expression required to form heteromeric channels, and have identified specific domains involved in channel multimerisation. However, an additional approach that has proved useful to investigate the subunit composition required for functional channel formation is the generation and characterisation of channel concatamers, whereby two or more subunits are joined together to make a single recombinant protein. Usually consecutive subunits within these concatamers are separated through the inclusion of short, flexible peptide sequences (linkers); a short polyglutamine peptide (5–8 amino acids) is
2.7 Concluding Remarks
frequently used [79, 81, 82]. Studies using subunit concatamers can enable characterisation of heteromeric channels while avoiding interference from homomeric channels that would be present in co-expression studies using the individual subunits, for instance to allow studies of inter-subunit binding sites [83, 84]. A prerequisite for successful use of concatameric channels is that the N- and C-termini of the subunits are either both extracellular or both intracellular. For channel subunits with extracellular N-termini and signal peptide sequence, the signal sequence should be removed from the downstream subunit(s) to favor replication of the correct transmembrane topology in the concatameric channel [86]. The channel concatamers cDNAs can be generated using several of the techniques described in this chapter. For example, restriction sites can be utilised, along with a synthetic DNA linker encoding a short peptide linker, to couple two channel open reading frames, in a manner similar to that described to introduce a short peptide tag [85]. Alternatively, site-directed mutagenesis (preferably employing silent mutations) can be used to introduce appropriate restriction sites at the 3' and 5' ends of the open reading frames of the two subunits to be ligated. For example, this approach was used to generate a concatamer of three P2X2 subunits, and allowed characterisation of an intersubunit zinc binding site [83]. Overlap extension PCR can also be used to generate the concatameric channel cDNA when distinct subunits are to be coupled. The requirement for distinct PCR primer sequences at the 5' and 3' ends of the open reading frames to be coupled precludes the use of this approach to link identical subunits. As discussed previously, the overlap extension PCR technique removes the necessity to incorporate restriction enzyme sites into the concatameric open reading frame yielding “seamless” cDNA constructs. Several studies have highlighted the need to accompany functional characterisation of channel concatamers with biochemical validation, to ensure that the integrity of the engineered concatamer is maintained when expressed. For instance, biochemical characterisation of concatameric P2X1 channels demonstrated that unexpected monomer (or dimer) byproducts were readily expressed and could account for the functional responses observed rather than the concatamer itself [82]. Also, experiments with nicotinic acetylcholine receptor subunits showed that subunit concatamers can form functional channels through the unexpected incorporation of tandem-expressed subunits and that these were only revealed through analysis of the expressed channels using protein biochemical techniques (sucrose gradient sedimentation) [81, 85].
2.7 Concluding Remarks
Extensive structure-function data, generated by molecular biology techniques, described in this chapter, has already produced much vital information about ion channels. Recent progress in determining the three-dimensional structures of several ion channels has allowed models to be developed that account for multiple
53
54
2 Molecular Biology Techniques for Structure – Function Studies of Ion Channels
aspects of channel function, including activation, inactivation, ligand binding, subunit interactions, toxin or drug binding and channel regulation. Generation of structure–function data to support these models will continue to be required in order to resolve the specific channel domains and individual residues involved in particular aspects of ion channel function. We hope that the information presented in this chapter will guide investigators in their selection of molecular biology approaches for making ion channel cDNA constructs for structure–function studies.
References 1 Watson, J. D., Gilman, M.,Witkowski, J., Zoller, M., Recombinant DNA, Scientific American Books, 2nd edn., 1992. 2 Sambrook, J., Fritsch, E. F., Maniatis, T., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory Press, New York, 2nd edn., 1989. 3 Swanson, G. T., Gereau IV, R. W., Green, T., Heinemann, S.F., Identification of amino acid residues that control functional behaviour in GluR5 and GluR6 kainate receptors, Neuron 1997, 19, 913–926. 4 Lu, Q., Seamless cloning and gene fusion, Trends Biotechnol. 2005, 23,199–207. 5 Li, J., Stevens, L., Klugbauer, N., Wray, D., Roles of molecular regions in determining differences between voltage dependence of activation of CaV3.1 and CaV1.2 calcium channels, J. Biol. Chem. 2004, 279, 26858–26867. 6 Clark, J. M., Novel non-templated nucleotide addition reactions catalyzed by procaryotic and eucaryotic DNA polymerises, Nucleic Acids Res. 1998, 16, 9677– 9686. 7 Mead, D., Pey, N. K., Herrnstadt, C., Marcil, R. A., Smith, L. M., A universal method for the direct cloning of PCR amplified nucleic acid, Biotechnology 1991, 9, 657–663. 8 Shuman, S., Novel approach to molecular cloning and polynucleotide synthesis using vaccinia DNA topoisomerase, J. Biol. Chem. 1994, 269, 32678–32684. 9 Ju, M.,Wray, D., Molecular identification and characterisation of the human eag2 potassium channel, FEBS Lett. 2002, 524, 204–210.
10 Weisberg, R. A., Landy, A., Site-specific recombination in phage lambda, in Lambda II, Cold Spring Harbor Press, New York, 1983, pp. 211–250. 11 Landy, A., Dynamic, structural, and regulatory aspects of lambda site-specific recombination, Annu. Rev. Biochem. 1989, 58, 913–49. 12 Hartley, J. L., Temple, G. F., Brasch, M. A., DNA Cloning using in vitro sitespecific recombination, Genome Res. 2000, 10, 1788–1795. 13 Obrdlik, P. et al, K+ channel interactions detected by a genetic system optimized for systematic studies of membrane protein interactions, Proc. Natl. Acad. Sci. USA 2004, 101, 12242–12247. 14 Luan, C-H. et al., High-throughput expression of C.elegans proteins, Genome Res. 2004, 14, 2102–2110. 15 Ju, M., Stevens, L., Leadbitter, E., Wray, D., The Roles of N- and C-terminal determinants in the activation of the Kv2.1 potassium channel, J. Biol. Chem. 2003, 278, 12769–12778. 16 Horton, R. M., Hunt, H. D., Ho, S. N., Pullen, J. K., Pease, L. R., Engineering hybrid genes without the use of restriction enzymes: gene splicing by overlap extension, Gene 1989, 77, 61–68. 17 Ho, S.N., Hunt, H.D., Horton, R.M., Pullen, J.K., & Pease, L.R. Site-directed mutagenesis by overlap extension using the polymerase chain reaction, Gene 1989, 77, 51–59. 18 Stevens, L., Structure and function of potassium and calcium channels, PhD Thesis, University of Leeds, 2005. 19 Shevchuk, N. A., Bryksin, A. V., Nusinovich,Y. A., Cabello, F. C., Suther-
References
20
21
22
23
24
25
26
27
28
land, M., Ladisch, S., Construction of long DNA molecules using long PCR-based fusion of several fragments simultaneously, Nucleic Acids Res. 2004, 32, e19. Geiser, M., Cebe, R., Drewello, D., Schmitz, R., Integration of PCR fragments at any specific site within cloning vectors without the use of restriction enzymes and DNA ligase, BioTechniques 2001, 31(1), 88–92. Weiner, M. P., Costa, G. L., Schoettlin, W., Cline, J., Mathur, E., Bauer, J. B., Site-directed mutagenesis of doublestranded DNA by the polymerase chain reaction, Gene 1994, 151,119–123. Wang, W. Malcolm, B. A., Two stage PCR protocol allowing introduction of multiple mutations, deletions and insertions using QuikChangeTM site-directed mutagenesis. BioTechniques 1999, 26, 680–682. Wilson, G. G., O’Neill, C. A., Sivaprasadarao, A., Findlay, J. B.,Wray, D., Modulation by protein kinase A of a cloned rat brain potassium channel expressed in Xenopus oocytes, Pflugers Arch. 1994, 428, 186–193. Wilson, G. G., Sivaprasadarao, A., Findlay, J. B., Wray, D., Changes in activation gating of IsK potassium currents brought about by mutations in the transmembrane sequence, FEBS Lett. 1994, 353, 251–254. Bannister, J. P.,Young, B. A., Sivaprasadarao, A., Wray, D., Conserved extracellular cysteine residues in the inwardly rectifying potassium channel Kir2.3 are required for function but not expression in the membrane, FEBS Lett. 1999, 458, 393–399. Bannister, J.P.,Young, B.A., Main, M.J., Sivaprasadarao, A., Wray, D. The effects of oxidizing and cysteine-reactive reagents on the inward rectifier potassium channels Kir2.3 and Kir1.1. Pflugers Arch. 1999, 438, 868–878. Ennion, S., Hagan, S., Evans, R. J., The role of positively charged amino acids in ATP recognition by human P2X1 receptors, J. Biol. Chem. 2000, 275, 29361– 29367. Roberts, J. A., Evans, R. J., ATP binding at human P2X1 receptors: contribution
29
30
31
32
33
34
35
36
37
38
of aromatic and basic amino acids revealed using mutagenesis and partial agonists, J. Biol. Chem. 2004, 279, 9043– 9055. Jiang, L-H., Rassendren, F., Surprenant, A., North, R.A., Identification of amino acid residues contributing to the ATP-binding site of a purinergic P2X receptor, J. Biol. Chem. 2000, 275, 34190–34196. Nakazawa, K., Ojima, H., Ohno,Y., A highly conserved tryptophan residue indispensable for cloned rat neuronal P2X receptor activation, Neurosci. Lett. 2002, 324, 141–144. Clyne, J. D.,Wang, L-F., Hume, R. I., Mutational analysis of the conserved residues of the rat P2X2 receptor, J. Neurosci. 2002, 22, 3873–3880. Ennion, S. J., Evans, R. J., Conserved cysteine residues in the extracellular loop of the human P2X1 receptor form disulfide bonds and are involved in receptor trafficking to the cell surface, Mol. Pharmacol. 2002, 61, 303–311. Clyne, J. D., LaPointe, L. D., Hume, R. I., The role of histidine residues in modulation of the rat P2X2 purinoceptor by zinc and pH, J. Physiol. 2002, 539, 347–359. Roberts, J. A., Evans, R. J., Mutagenesis studies of conserved proline residues of human P2X1 receptors for ATP indicate that proline 272 contributes to channel function, J. Neurochem. 2005, Epub ahead of print. Morrison, K. L.,Weiss, G. A., Combinatorial alanine-scanning, Curr. Opin. Chem. Biol. 2001, 5, 302–307. Li-Smerin,Y., Hackos, D. H., Swartz, K. J., a-Helical structural slements within the voltage-sensing domains of a K+ Channel, J. Gen. Physiol. 2000, 115, 33– 50. Perry, M., de Groot, M. J., Helliwell, R., Leishman, D., Tristani-Firouzi, M., Sanguinetti, M. C., Mitcheson, J., Structural determinants of HERG channel block by clofilium and ibutilide, Mol. Pharmacol. 2004, 66, 240–249. Tedford, H.W., Gilles, N., Menez, A., Doering, C. J., Zamponi, G.W., King, G. F., Scanning mutagenesis of o-Atraco-
55
56
2 Molecular Biology Techniques for Structure – Function Studies of Ion Channels
39
40
41
42
43
44
45
46
47
48
toxin-Hv1 a reveals a spatially restricted epitope that confers selective activity against insect calcium channels, J. Biol. Chem. 2004, 279, 44133–44140. Ragsdale, D. S., McPhee, J. C., Scheuer, T., Catterall,W. A., Molecular determinants of state-dependent block of Na+ channels by local anesthetics, Science 1994, 265, 1724–1728. Karlin, A., Akabas, M. H., Substitutedcysteine accessibility method, Methods Enzymol. 1998, 293, 123–145. Milligan, C. J., Wray, D., Local movement in the S2 region of the voltage-gated potassium channel hKv2.1 studied using cysteine mutagenesis, Biophys. J. 2000, 78, 1852–1861. Pascual, J. M., Shieh, C. C., Kirsch, G. E., Brown, A. M., Multiple residues specify external tetraethylammonium blokkade in voltage-gated potassium channels, Biophys. J. 1995, 69, 428–434. Li, J., Stevens, L., Wray, D., Molecular regions underlying the activation of lowand high-voltage activating calcium channels, Eur. Biophys. J. 2005, DOI: 10.1007/ s00249–005-0487–7 [Epub ahead of print]. Akabas, M. H., Stauffer, D. A., Xu, M., Karlin, A., Acetylcholine receptor channel structure probed in cysteine-substitution mutants, Science 1992, 258, 307–310. Akabas, M. H., Karlin, A., Identification of acetylcholine receptor channel-lining residues in the M1 segment of the a-subunit, Biochemistry 1995, 34,12496–12500. Akabas, M. H., Kaufmann, C., Archdeacon, P., Karlin, A., Identification of acetylcholine receptor channel-lining residues in the entire M2 segment of the asubunit, Neuron 1994, 13, 919–927. Rassendren, F., Buell, G., Newbolt, A., North, R. A., Surprenant, A., Identification of amino acid residues contributing to the pore of a P2X receptor, EMBO J. 1997, 16, 3446–3454. Egan, T. M., Haines,W.R.,Voigt, M. M., A domain contributing to the ion channel of ATP-gated P2X2 receptors identified by the substituted cysteine accessibility method, J. Neurosci. 1998, 18, 2350– 2359.
49 Haines,W. R.,Voigt, M. M., Migita, K., Torres, G. E., Egan, T. M., On the contribution of the first transmembrane domain to whole-cell current through an ATP-gated ionotropic P2X receptor, J. Neurosci. 2001, 21, 5885–5892. 50 Xu, M., Covey, D. F., Akabas, M. H., Interaction of picrotoxin with GABA(A) receptor channel-lining residues probed in cysteine mutants, Biophys. J. 1995, 69, 1858–1867. 51 Xu, M., Akabas, M. H., Amino acids lining the channel of the g-aminobutyric acid type A receptor identified by cysteine substitution, J. Biol. Chem. 1993, 268, 21505–21508. 52 Yusaf, S. P., Wray, D., Sivaprasadarao, A., Measurement of the movement of the S4 segment during the activation of a voltage-gated potassium channel, Pflugers Arch. 1996, 433, 91–97. 53 Larsson, H. P., Baker, O. S., Dhillon, D. S., Isacoff, E.Y., Transmembrane movement of the Shaker K+ channel S4, Neuron 1996, 16, 387–397. 54 Jiang, L-H., Rassendren, F., Spelta,V., Surprenant, A., North, R.A., Amino acid residues involved in gating identified in the first membrane-spanning domain of the rat P2X2 receptor, J. Biol. Chem. 2001, 276,14902–14908. 55 Jiang, L-H., Kim, M., Spelta,V., Bo, X., Surprenant, A., North, R.A., Subunit arrangement in P2X receptors, J. Neurosci. 2003, 23, 8903–8910. 56 Starace, D.M., Bezanilla, F., Histidine scanning mutagenesis of the basic residues of the S4 segment of the Shaker K+ channel, J. Gen. Physiol. 2001, 117, 469– 490. 57 Silberberg, S. D., Chang, T. H., Swartz, K. J., Secondary structure and gating rearrangements of transmembrane segments in rat P2X4 receptor channels, J. Gen. Physiol. 2005, 125, 347– 359. 58 Li-Smerin,Y., Swartz, K. J., Helical structure of the COOH terminus of S3 and its contribution to the gating modifier toxin receptor in voltage-gated ion channels, J. Gen. Physiol. 2001, 117, 205– 218.
References 59 Monks, S. A., Needleman, D. J., Miller, C., Helical structure and packing orientation of the S2 segment in the Shaker K+ channel, J. Gen. Physiol. 1999, 113, 415– 423. 60 Choe, S., Stevens, C. F., Sullivan, J. M., Three distinct structural environments of a transmembrane domain in the inwardly rectifying potassium channel ROMK1 defined by perturbation, Proc. Natl. Acad. Sci USA 1995, 92, 12046–12049. 61 Hong, K. H., Miller, C., The lipid– protein interface of a Shaker K+ channel, J. Gen. Physiol. 2000, 115, 51–58. 62 Li-Smerin,Y., Hackos D. H., Swartz, K. J., A localized interaction surface for voltage-sensing domains on the pore domain of a K+ channel, Neuron 2000, 25, 411–423. 63 Jenkins, A., Andreason, A., Trudell, J. R., Harrison, N. L., Tryptophan scanning mutagenesis in TM4 of the GABA(A) receptor a1 subunit: implications for modulation by inhaled anaesthetics and ion channel structure, Neuropharmacology 2002, 43, 669–678. 64 Guzman, G. R., Santiago, J., Riccardo, A., Marti-Arbona, R., Rojas, L. V., Lasalde-Dominicci, J. A., Tryptophan scanning mutagenesis in the aM3 transmembrane domain of the Torpedo californica acetylcholine receptor: functional and structural implications, Mol. Pharamacol. 2003, 61, 12243–12250. 65 Pamchenko,V. A., Glasser, C. R., Mayer, M.L., Structural similarities between glutamate receptor channels and K+ channels examined by scanning mutagenesis, J. Gen. Physiol. 2001, 117, 345– 360. 66 Tamamizu, S., Guzman, G. R., Santiago, J., Rojas, L.V., McNamee, M.G., Lasalde-Dominicci, J. A., Functional effects of periodic tryptophan substitutions in the aM4 transmembrane domain of the Torpedo californica nicotinic acetylcholine receptor, Biochemistry. 2000, 39, 4666–4673. 67 Sine, S. M.,Wang, H. L., Bren N., Lysine scanning mutagenesis delineates structural model of the nicotinic receptor
68
69
70 71
72
73
74
75
76
77
ligand binding domain, J. Biol. Chem. 2002, 277, 29210–29223. Jin, T., Peng, L., Mirshahi, T., Rohacs, T., Chan, K.W., Sanchez, R., Logothetis, D.E., Identification of critical residues controlling G protein-gated inwardly rectifying K+ channel activity through interactions with the bg subunits of G proteins, Mol. Cell. 2002, 10, 469–481. He, M-.L., Zemkova, H., Stojilkovic, S. S., Dependence of purinergic P2X receptor activity on ectodomain structure, J. Biol. Chem. 2003, 278, 10182–10188. Jarvik, J. W., Telmer, C. A., Epitope tagging, Annu. Rev. Gen. 1998, 32, 601–618. Ficker, E., Dennis, A. T., Wang, L., Brown, A. M., Role of the cytosolic chaperones Hsp70 and Hsp90 in maturation of the cardiac potassium channel hERG, Circulation Res. 2003, 92, E87–E100. Grabner, M., Dirksen, R.T., Beam, K. G., Tagging with green fluorescent protein reveals a distinct subcellular distribution of L-type and non-L-type Ca2+ channels expressed in dysgenic myotubes, Proc. Natl. Acad. Sci. USA. 1998, 95,1903–1908. Makhina, E. N., Nichols, C. G., Independent trafficking of K-ATP channel subunits to the plasma membrane, J. Biol. Chem. 1998, 273, 3369–3374. Grailhe, R., de Carvalho, L. P., Paas, Y., Le Poupon, C., Soudant, M., Bregestovski, P., Changeux, J. P., Corringer P. J., Distinct subcellular targeting of fluorescent nicotinic a3b4 and serotoninergic 5-HT3A receptors in hippocampal neurons, Eur. J. Neuroscience. 2004, 19, 855–862. Kobrinsky, E., Stevens, L., Bentil, S., Wray, D., Soldatov, N.M., Molecular rearrangements associated with the voltage gating of the KV2.1 channel, FASEB J. 2004, 18(A226), Suppl. S. Nicke, A., Baumert, H.G., Rettinger, J., Eichele, A., Lambrecht, G., Mutschler, E., Schmalzing, G., P2X1 and P2X3 receptors form stable trimers – a novel structural motif of ligand-gated ion channels, EMBO J. 1998, 17, 3016–3028. MacKinnon R., Determination of the subunit stoichiometry of a voltage-activa-
57
58
2 Molecular Biology Techniques for Structure – Function Studies of Ion Channels
78
79
80
81
ted potassium channel, Nature 1991, 350, 232–235. Drisdel, R.C., Green,W. N., Neuronal a-bungarotoxin receptors are a7 subunit homomers, J. Neurosci. 2000, 20, 133– 139. Firsov, D., Gautschi, I, Merillat, A. M., Rossier, B. C., Schild, L., The heterotetrameric architecture of the epithelial sodium channel (ENaC), EMBO J. 1998, 17, 344–352. McKernan, R. M.,Whiting, P.J., Which GABAA-receptor subtypes really occur in the brain? Trends Neurosci. 1996, 19, 139– 143. Groot-Kormelink, P. J., Broadbent, S. D., Boorman, J. P., Sivilotti, L.G., Incomplete incorporation of tandem subunits in recombinant neuronal nicotinic receptors, J. Gen. Physiol. 2004, 123, 697– 708.
82 Nicke, A., Rettinger, J., Schmalzing, G., Monomeric and dimeric by-products are the principal functional elements of higher order P2X1 concatamers, Mol. Pharmacol. 2003, 63, 243–252. 83 Nagaya, N., Tittle, R. K., Saar, N., Dellal, S. S., Hume, R. I., An intersubunit zinc binding site in rat P2X(2) receptors, J. Biol. Chem. 2005, 280, 25982– 25993. 84 Yuill, K., Ashmole, I., Stanfield, P. R., The selectivity filter of the tandem pore potassium channel TASK-1 and its pHsensitivity and ionic selectivity, Pflugers Arch. Eur. J. Physiol. 2004, 448, 63–69. 85 Zhou Y., Nelson, M. E, Kuryatov, A., Choi, C., Cooper, J., Lindstrom, J., Human alpha4 beta2 acetylcholine receptors formed from linked subunits, J. Neurosci. 2003, 23, 9004–9015.
59
3 Unnatural Amino Acids as Probes of Ion Channel Structure – Function and Pharmacology Paul B. Bennett, Niki Zacharias, John B. Nicholas, Sue Dee Sahba, Ashutosh Kulkarni, and Mark Nowak
3.1 Introduction
Ion channel proteins are the fundamental electrical signaling molecules of living systems. Among other things they function as electrical field effect transistors that permit human beings to perform complex reasoning and such purposed actions as gene cloning, aircraft design and space travel. Protein structure, function and synthesis are complex processes programmed in the genetic code. A gene (sequence of DNA archived in human chromosomes) is transcribed into an mRNA which is read by the ribosomes whose task it is to assemble appropriate amino acids according to the code. The amino acids are each shuttled to the ribosomal complex by an amino acid-specific (transfer) tRNA. Proteins consist of an assortment of the 20 natural amino acids. Site directed mutagenesis, the purposeful substitution of a codon specifying an alternative amino acid at a specific position in the protein, has been widely employed to probe the role of amino acids in proteins both for structure–function and to elucidate drug binding to protein receptors (see Chapter 2). Although extremely powerful for elucidating some aspects of these relationships, conventional site-directed mutagenesis is limited to substitution of the original amino acid with one of the other 19 naturally occurring amino acids. Binding of a drug to an ion channel protein requires a very specific three-dimensional arrangement between the shape and chemical functionality of the drug and the amino acids in the protein binding site. Multiple, rather subtle types of interactions can occur, including hydrogen bonding and dipole attractions. Although the natural amino acids are used to perturb protein structure and function, they provide a limited range of structural and chemical diversity. As such, it is not possible with conventional mutagenesis to subtly or specifically probe the role of side chains or the protein carbonyl backbone. More recently, methods have been developed to permit incorporation of amino acids that do not occur in nature (unnatural amino acids) [1–4]. For example, it is possible to site-specifically incorporate any number of unnatural amino acids into almost any protein through the approach termed nonsense suppression. The terminology of nonsense suppression Expression and Analysis of Recombinant Ion Channels. Edited by Jeffrey J. Clare and Derek J. Trezise Copyright # 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim ISBN: 3-527-31209-9
60
3 Unnatural Amino Acids as Probes of Ion Channel Structure – Function and Pharmacology
comes from the field of microbial genetics. A nonsense mutation in nature is a base change that converts a codon within the gene sequence into a stop codon. In microbes, a suppressor is a second mutation that restores a function lost by a primary nonsense mutation. In the technique of nonsense suppression, a non-native nonsense (stop) codon is inserted into a cDNA and is reverted or suppressed by introduction of a novel amino acid-tRNA that recognizes the stop codon. This approach has enormous potential and permits systematic structure–function analyses with precision far beyond conventional site-directed mutagenesis. Nonsense suppression methodology has enabled a much wider range of protein structure– function studies with more detailed information [5–7]. Natural amino acid variants with subtle perturbations in side chain chemistry as well as probes such as spin labels, FRET (fluorescence resonance energy transfer) pairs or fluorescent groups can be incorporated [6, 8–11]. Using this approach insights can be gained that are not possible using conventional site-directed mutagenesis (see below). There are a number of excellent reviews on this subject [6, 8–12]. The goal of this chapter is to focus first on methodological issues that a potential user must recognize; and secondly, on recent applications of the methodology to illustrate general strategies and specific information obtainable using unnatural amino acids.
3.2 Unnatural Amino Acid Mutagenesis Methodology
The basic approach to incorporation of an unnatural amino acid is outlined in Fig. 3.1. First, a stop codon (TAG, “amber”) is incorporated into the recombinant cDNA at the position coding for the amino acid of interest using conventional site-directed mutagenesis (see Chapter 2). From this cDNA a UAG-containing mRNA is generated, either by in vitro transcription or within cells. Secondly, a tRNA that recognizes the amber stop codon is prepared and chemically acylated with the desired unnatural amino acid. The DNA encoding the gene of interest or the in vitro transcribed mRNA and the tRNA carrying the unnatural amino acid are then added to an appropriate expression system. Several groups have contributed important steps in the development of these methods [1–3, 13–18]. Methodological issues focus primarily on the tRNA. When considering nonsense suppression, care must be taken to ensure that the tRNA is orthogonal in the chosen expression system. This means that the exogenous suppressor tRNA must not be recognized by the native aminoacyl tRNA synthetases (RS), the enzymes that charge tRNAs with their cognate amino acids. Unless this is achieved, the suppressor tRNA, once it has delivered its unnatural amino acid to a protein, will be charged with a ‘natural’ amino acid and return to the protein synthesis cycle. This will produce a mixture of proteins with natural and unnatural amino acids and would be of limited value. Another methodological issue that must be addressed when using nonsense suppression is the fidelity of the system. To test this at least two control experi-
Fig. 3.1 The fundamental protocol for incorporating unnatural amino acids through nonsense suppression, showing an “in vivo” translation system with electrophysiology readout. A special nonsense codon is introduced into the cDNA of interest at the position of interest. A tRNA that recognizes
only the nonsense codon is created with the unnatural amino acid appended. The gene of interest (cDNA or in vitro transcribed mRNA) and the tRNA are introduced into a cell where the protein is expressed and detected using electrophysiology. (This figure also appears with the color plates.)
3.2 Unnatural Amino Acid Mutagenesis Methodology 61
62
3 Unnatural Amino Acids as Probes of Ion Channel Structure – Function and Pharmacology
ments are performed. In one control experiment, UAG-mRNA only is added to the expression system. This control tests if functional protein will be synthesized without suppressor tRNA present and if truncated protein will interfere with the assay. A functional protein could be synthesized if an endogenous tRNA misreads the UAG codon (read through) [2]. It is also possible for the ribosome to stop translation when it reaches the UAG codon, creating a truncated protein that could interfere with a functional assay. Another control experiment to confirm the fidelity of the system is to compare the function of wild-type protein expressed using nonsense suppression (UAG mRNA and tRNA acylated with the wild-type amino acid) and protein expressed using wild-type mRNA. Because one has placed the natural wild type amino acid back into the protein at its correct position, the protein expressed using nonsense suppression or wild-type mRNA should function identically [2]. We rarely see expression of protein using UAGmRNA, only in our in vivo nonsense suppression experiments in Xenopus oocytes, and the channels/receptors display indistinguishable characteristics when expressed using nonsense suppression or mRNA. Figure 3.2 illustrates some of the possible side chain modifications and their use.
Fig. 3.2 Unnatural amino acid mutations that probe H-Bonds, cation–p, and p–p interactions. Replacement of a serine by an alanine removes an –OH and the potential for hydrogen bonding; substitution of threonine sterically alters the –OH; replacement of the peptide backbone amide with an ester in hydroxythreonine impacts hydrogen bonding by the
backbone and the –OH side chain. Substitution of phenylalanine (Phe) for tyrosine (Tyr) removes an H-bonding group. Additions of fluorine (F) deactivate the ring and affect p–p or cation–p interactions. Cyclohexyl-alanine replaces an aromatic ring with an aliphatic ring to probe hydrophobic interactions without greatly altering side chain volume.
3.2 Unnatural Amino Acid Mutagenesis Methodology
The originally described yeast-derived suppressor tRNA [1] is not useful in some expression systems such as Xenopus oocytes because the endogenous aminoacyl tRNA synthetases (RS) reacylate the tRNA. Using established rules of RSs– tRNA recognition, we generated a new suppressor tRNA [13] by taking advantage of the fact that Tetrahymena thermophila has a nonstandard genetic code: TAG is not a stop codon but instead codes for glutamine. Thus, a naturally occurring suppressor tRNA exists and a slight variant of this (THG73) is a much improved suppressor in the Xenopus oocyte system [19]. A limitation of the nonsense suppression methodology is that the aminoacyl tRNA is consumed in the process and is not regenerated. The synthetic methods for generating the acylated tRNA reagent are well known, but still require a significant effort. Furthermore the suppression process is approximately 15–20 % as efficient as the natural process [2]. As such it requires excess aminoacyl tRNA for each molecule of protein synthesized. In order to sustain the presence of the altered protein, this reagent must be delivered in large excess or continually delivered. A solution to these problems is to synthesize sufficient quantities of aminoacyl tRNA required to generate the protein of interest in an expression system. For example, Xenopus oocytes can be injected with nanogram quantities of tRNA or in vitro assays can be scaled up. However, for assays with limited sensitivity such as NMR it can still be very difficult to synthesize enough protein, particularly membrane proteins. This limitation may be a primary reason limiting the use of nonsense suppression methodology. In principle, novel RNA synthases (RS) can be evolved and selected to recognize a given unnatural amino acid and thus overcome the requirement for the large quantities of exogenous suppressor aminoacyl-tRNA discussed above. Methods have been developed to generate orthogonal tRNA-RS pairs specific for a given unnatural amino acid [20–23]. In this approach, a novel orthogonal RS catalyses the acylation of the suppressor tRNA with the unnatural amino acid. Importantly, the suppressor tRNA can be re-acylated, avoiding the need to deliver large quantities of exogenous aminoacylated suppressor tRNA. To generate orthogonal tRNA-RS pairs directed evolution is employed [21, 22]. With directed evolution, all possible mutations are introduced at key residues in the RS important for recognition of the cognate amino acid. The mutant RS library is then screened for aminoacylation of only the corresponding suppressor tRNA (and not the endogenous tRNAs of the host expression system) with only the desired unnatural amino acid (and not any natural amino acids). Using this approach, specific orthogonal tRNA-RS pairs have been developed for a variety of unnatural amino acids and milligram quantities of unnatural amino acid protein mutants have been obtained in E. coli, yeast and mammalian expression systems [23–25]. The limitation of this approach is that a suppressor tRNA-RS pair must be developed for each unnatural amino acid and, therefore it is not generally applicable.
63
64
3 Unnatural Amino Acids as Probes of Ion Channel Structure – Function and Pharmacology
3.3 Unnatural Amino Acid Mutagenesis for Ion Channel Studies
An alternative strategy is to use a highly sensitive assay where only very small amounts of protein are required. Ion channels are complex, integral membrane proteins that lower the energy barrier for ion translocation across the hydrophobic lipid bilayer. They are highly dynamic proteins whose function (gating) and pharmacology depend on the channel conformational state. For the most part these proteins are not readily amenable to high-resolution structural methods such as X-ray crystallography and NMR (see Chapter 9). Because these methods provide only limited or no temporal information on the protein dynamics, they provide a description of one particular protein conformational state. Thus ion channels are good candidates for studies using unnatural amino acids as these methods provide information on structure and function. This approach can be readily adopted with ion channel proteins because there are exquisitely sensitive methods for measuring protein function. Ion channels can be easily expressed in a number of systems including HEK and CHO mammalian cells (Chapter 4) or in Xenopus oocytes (Chapter 1) which are a well-established general vehicle for heterologous expression and characterization of ion channel proteins [26, 27]. Using whole cell two-electrode voltageclamp electrophysiology in oocytes, as little as 10–15 M of protein can be detected. Using the patch-clamp method of single channel recording a single molecule can be analyzed. Xenopus oocytes are injected with UAG-mRNA for the ion channel of interest and the suppressor tRNA. Usually, after 24 to 48 h the cells have synthesized the ion channel and have embedded it into their cell membrane. The ion channels can then be studied using electrophysiology methods. The generality of the nonsense suppression method has been established with a large number of different unnatural amino acids (including those shown in Fig. 3.2) incorporated at a comparable number of sites in dozens of proteins. Scientists at Neurion Pharmaceuticals and the California Institute of Technology have successfully incorporated over 85 different amino and hydroxy acids (including 15 of the 20 natural amino acids) into at least 14 different receptors or ion channels (e. g. the M2 muscarinic acetylcholine (ACh) receptor, the nicotinic ACh receptor a1, a2, a4, and b1 subunits, inward rectifier and G-protein coupled potassium channels Kir1.1 (ROMK1), Kir2.1, GIRK1, GIRK4, the 5-HT3A receptor subunit, the MOD1 receptor, the NR2A subunit of the NMDA receptor, delayed rectifier (Kv and Shaker) and hERG potassium channels, CFTR, and the GAT1 GABA transporter) for a total of 454 combinations of amino acids/sites [5, 6, 28]. It has been generally observed that hydrophobic amino acids are incorporated more efficiently than charged ones [5]. Proteins are generally made up of l-amino acids (the levorotatory optical isomer). d-amino acids are not compatible with nonsense suppression, nor are b-amino acids [29]. Nevertheless, some fairly elaborate unnatural amino acids have been employed [5]. For example, a biotin derivative was incorporated consecutively in several positions in an ion channel to identify surface-exposed residues in the channel [30]. We have incorporated caged amino acids, unnatural amino acids that mimic ligands of ion channels, amino acids
3.4 Structure–Function Example Studies
that when exposed to light rearrange and break the backbone of the ion channel, and several hydroxy acids [31–36]. In addition to introducing amino acids with unnatural side chains, it is also possible to insert a-hydroxy acids in place of a-amino acids [37]. This mutation replaces the backbone amide bond with an ester, thus replacing amide NH with an ester O, eliminating a hydrogen bonding donor. In addition, the adjacent backbone carbonyl is a weaker hydrogen bond acceptor in the ester than in the amide. a-Hydroxy acid substitutions are useful as probes of secondary structure, because they disrupt hydrogen bonding that stabilizes a-helices and b-sheets. More important for our work, drugs often hydrogen bond to backbone carbonyls that are not already involved in hydrogen bonds with neighboring amino acids [28]. This will be described further in the hERG section of this chapter.
3.4 Structure–Function Example Studies 3.4.1 Nicotinic Acetylcholine Receptor
Studies employing conventional site-directed mutagenesis often suggest a particular role for a given amino acid in drug binding, yet the evidence is incomplete and does not lend itself to a strict physical chemical interpretation for a mechanism. This is due to the limited number of natural amino acids which precludes a detailed investigation of the relevant interactions between the protein and the drug. Additional systematic subtle changes, made possible by nonsense suppression, can provide the definitive evidence for a mechanism. For instance, in the nicotinic acetylcholine receptor (nAChR), early photo-affinity labeling studies identified many of the amino acids that constitute the binding site for acetylcholine (ACh, the agonist for the receptor) [38–40]. However, the exact amino acids that bind ACh and the specific binding interactions were not known. Recently, using nonsense suppression with unnatural amino acids, it was demonstrated that aTrp149 in the mouse muscle nAChR interacts with the quaternary ammonium of acetylcholine in a cation–p interaction [35, 41]. A cation–p bond is the noncovalent interaction that occurs between the p electrons of a conjugated system (like an aromatic ring) and a cation. In proteins, a cation–p interaction can occur between a cation and the aromatic rings of tyrosine, phenylalanine, neutral histidine, or tryptophan [42, 43]. The cation–p interaction in nAChR was determined by incorporating the unnatural amino acids 4-F-Trp (addition of fluorine in the 4 position of the tryptophan ring), 4,5-Trp, etc. into the ACh binding site [41]. Progressive fluorination decreases the ability of the tryptophan ring to interact with a cation while having minimal effect on the sterics of the amino acid (Fig. 3.3). A linear relationship was observed between the theoretically predicted change in the cation–p interaction energy upon successive fluorination and the experimentally observed change in receptor affinity for ACh binding.
65
66
3 Unnatural Amino Acids as Probes of Ion Channel Structure – Function and Pharmacology
DDG = 0.6 ln (KD-mutant /KD-WT)
(1)
This finding indicates that, upon binding, the quaternary ammonium of ACh makes van der Waals contact with the indole side chain of aTrp149, which allows us to infer the localization of the binding between ACh and Trp149 [41] to within 0.5 Å.
Fig. 3.3 Correlation between the change in EC50 and calculated binding energy in nicotinic acetylcholine receptor binding pocket. Tryptophan 149 was systematically replaced with fluorinated (F) tryptophan (Trp, 1–4 F) unnatural amino acids. Each added F further deactivates the Trp p electron cloud, resulting
in a decreased binding energy. Binding energy was calculated in the gas phase and the absolute values will be scaled down by the presence of water and other factors but the trend is expected to remain the same. (This figure also appears with the color plates.)
In marked contrast, substitution of fluorinated Trp residues at a149 did not appreciably affect the interaction of nicotine with the mouse nAChR. Instead, it was determined that nicotine interacts with the muscle nAChR via a hydrogen bond between the positively charged nitrogen in the pyrrolidine ring of nicotine and the backbone carbonyl of Trp a149. This was demonstrated by insertion of the unnatural amino acid a-OH-threonine into a150. This changes the a149 backbone amide bond to an ester, weakening the H-bonding capability of the carbonyl. This interaction had been previously suggested by modeling studies on the ACh binding protein [36]. Previous to these findings, ACh and nicotine were thought to be represented by a single pharmacophore. These studies clearly demonstrate that unnatural amino acid mutagenesis can be used to determine distinct ligand binding interactions, even between molecules with similar structures.
3.4 Structure–Function Example Studies
3.4.2 Drug Interactions with the hERG Voltage-gated Potassium Ion Channel
In order to implement this approach and focus it on a practical problem of importance to the medical community and the pharmaceutical industry, we are determining the structural requirements for the interactions of various agents with the hERG potassium channel. hERG potassium ion channels govern the repolarization phase of human ventricular action potentials [44, 45]. Many drugs and other xenobiotic agents or their metabolites can inhibit hERG potassium channels and lead to cardiac arrhythmias and sudden death [46–49]. As a consequence, tremendous effort has gone toward creating novel drugs that do not possess hERG channel interactions. Avoiding unintended blockade of the hERG potassium ion channel is a costly and time-consuming challenge for the pharmaceutical industry and health care in general. Present discovery programs often rely upon brute force screening efforts that attempt to empirically eliminate hERG channel activity. These efforts commonly rely on assays using only the wild-type hERG channel. From IC50 data in electrophysiology or radioligand binding/displacement studies medicinal chemists attempt to elucidate the hERG binding interactions and eliminate them from the molecule. However, we have discovered that there are multiple binding modes and without knowledge of the precise manner in which a given molecule binds to hERG, it may be difficult to synthesize a molecule that binds to the target but not to hERG [28]. We have employed natural and unnatural amino acid mutagenesis to reveal the structural determinants governing drug interactions with hERG. We believe with an understanding of how and where a compound is binding to hERG, medicinal chemists will then have the ability to design out hERG blockade for that particular compound. Figure 3.2 shows some of the natural and unnatural amino acids incorporated into hERG for that purpose. Previous studies have employed natural amino acid mutagenesis, e. g. alanine scanning to probe drug binding [50–52]. These experimental mutation studies demonstrated that the likely hERG binding region for most drugs is the cavity formed by the S6 helices under the selectivity filter [51, 53]. These studies identified two amino acids, Tyr652 and Phe656 putatively involved in drug binding, and the authors suggested the nature of interactions (cation–p, p–p and hydrophobic) based on the amino acid side chains involved [50–52]. Figure 3.4 shows homology models of hERG based on the bacterial channels KcsA (closed) and KvAP (open conformation) crystal structures [51, 54]. Studies have shown that drugs such as MK-499 bind within the large pore volume lined by the S6 helices that serves as the binding pocket for drugs that block hERG [51, 53]. From mutation data and models such as these, we are able to determine the amino acids likely to be involved in hERG block. Our work has focused on the roles of Thr623, Ser624, Tyr652, and Phe656, and the backbone carbonyl of Leu622. By substituting other amino acids at the positions 622, 623, 624, 652, and 656, we can probe in detail how these amino acids participate in the drug binding pocket. The amino acids that are substituted were chosen to provide the most information while not perturbing the overall structure or function of the protein.
67
68
3 Unnatural Amino Acids as Probes of Ion Channel Structure – Function and Pharmacology
Fig. 3.4 Homology models of hERG closed (left) and open ion conducting (right) states. Only the S5-P-S6 segments are shown. Many drugs enter the open channel and bind to residues along the S6 segment. (This figure also appears with the color plates.)
Our mutational studies of more than 60 drugs that block hERG show that they all bind to two or more of these five key residues within the hERG channel [28]. Each of these amino acids projects a side chain functionality into the hERG pore to which drug molecules can bind. For example, Tyr652 can bind drugs through cation–p or p–p interactions with the aromatic ring, or through hydrogen bonding with the 4' hydroxy group [42]. By making subtle but informative mutations to Tyr652 we can determine how it is binding drugs. For example, if we mutate Tyr652 to Phe using site-directed mutagenesis, we eliminate the hydrogen bonding ability. If a drug’s binding is decreased by this mutation, we can conclude that hydrogen bonding is important to this drug’s binding to hERG. We can use similar mutations to test for cation–p or p–p interactions. We have made a number of subtle pore mutations that probe the steric, hydrophobic and electronic features that determine drug binding at amino acid positions Thr623, Ser624, Tyr652, and Phe656. We have also explored carbonyl backbone hydrogen bonding interactions by mutating the peptide amide bond at Leu622 to an ester linkage. We expressed the mutated channels in Xenopus oocytes and measured K+ currents by using two-electrode voltage-clamp methods while perfusing the oocytes with various known hERG blockers. By comparing the change in hERG currents between the WT channel and the mutant channels, we determined the energetics of the specific interactions that govern drug binding (see Eq. (1) above). We typically generate data for a total of 11 mutants at these 4 amino acid positions. This data set, which we call a Mutant Activity Panel (MAPTM) is then plotted as shown
3.4 Structure–Function Example Studies
in Fig. 3.5. Our data indicate that the channel binding interactions vary greatly, even for structurally related hERG blockers. Figure 3.5 shows the hERG MAP for astemizole. The figure shows the structure of the molecule and the positions of the amino acid mutations on the S6 helix of hERG. The bar graph shows the change in binding energy, relative to WT, for each of the 11 mutant channels. From the data we can see that mutation of Ser624 to Ala greatly reduces drug block. The Ser624Ala mutation eliminates the hydrogen bonding ability of serine, thus indicating that hydrogen bonding at this position is important for hERG block by astemizole. The progressive decrease in drug binding when we mutate Phe656 to 4-F-Phe, and then 3,5-F2-Phe indicates that astemizole makes a cation–p or p–p interaction with Phe656. We have generated the hERG MAPs of more than 60 pharmaceuticals using Xenopus oocytes. Consequently, we have developed an extensive database of compound structure–activity relationships (SAR). We have determined the binding geometries of a variety of compounds with the hERG channel. For example, risperidone and haloperidol showed cation–p interactions at Tyr652, whereas amperozide, fluphenazine, and triflupromazine showed cation–p interactions at
Figure 3.5 hERG MAP astemizole: A hERG MAP elucidates the nature and relative importance of specific drug–channel interactions. The right hand bar graph shows the change in astemizole binding energy (kcal mol–1) at each position of the channel when that position is altered. These changes in binding energy may be interpreted in terms of atomic level interactions such as hydrogen-bonding, cation–p, hydrophobic, and ion pairing. Each hERG mutant is designed to identify a specific
noncovalent binding interaction with the channel. Each compound displays a unique hERG MAP signature. In this example, changes at serine 624 suggest H-bond interactions with the compound. Progressive changes in binding as fluorine (F) is added to phenylalanine at position 652 indicate cation– p or p–p aromatic interactions. The chemical structure of astemizole is shown. (This figure also appears with the color plates.)
69
70
3 Unnatural Amino Acids as Probes of Ion Channel Structure – Function and Pharmacology
Phe656. Most of the drugs also exhibited hydrogen bonding interactions with the backbone of Leu622 and the side chain of Ser624. Figure 3.6 highlights the unique patterns of different interactions seen with different compounds. For example amperozide and pimozide rely on hydrogen bonding at Leu622 carbonyl (623OH-Thr mutation), whereas haloperidol and astemizole do not. Only pimozide hydrogen bonds to the tyrosine hydroxy group, the other drugs do not. The substitution of cyclohexyl-alanine at position 656 (substitution with a nonaromatic ring) greatly affected amperozide, but not pimozide block. This information enables rational modification of these molecules. These insights cannot be obtained from IC50 measurements against wild-type hERG only, as is now commonly done.
Fig. 3.6 Differentiation of binding by hERG blockers. The mutation at 623 affects H-bonding. Both pimozide and amperozide interact at this site, whereas block by haloperidol was unaffected. Conversion of a Tyr to Phe at position 652 removes an H-bonding site on the ring. Cyclohexyl-alanine is a nonaromatic ring substituted for Phe at position 656. This has no effect on pimozide block but greatly affects amperozide.
Not only is it possible to manipulate amino acid side channels, but also one of the more remarkable possibilities of this approach is the possibility to insert a-hydroxy acids in place of a-amino acids, replacing the amide peptide linkage with a backbone ester bond [34, 55]. The ribosome can apparently mediate ester formation and the nonsense suppression methodology is well suited to this tactic. In fact, in the in vivo nonsense suppression methodology we have often observed that a-hydroxy acids are more efficiently incorporated than analogous a-amino
3.4 Structure–Function Example Studies
acids [33]. This observation, coupled with the facts that the synthetic protocols associated with a-hydroxy acids are generally simpler [33, 56] and the resulting hydroxyacyl tRNA is more stable than an aminoacyl tRNA, makes a-hydroxy acids very appealing for many types of nonsense suppression experiments. An a-hydroxy acid substitution can be placed at many sites in a protein without being disruptive. An a-hydroxy acid substitution makes the carbonyl a weaker hydrogenbond acceptor. The weakening of the carbonyl as an acceptor was determined to have a larger effect (0.89 kcal mol–1) than the deletion of the NH (0.72 kcal mol–1) [34]. We have used a-hydroxy acids as a means to probe the role of H-bonds in drug binding in the hERG pore region. A backbone ester cannot form hydrogen bonds of the sort found in an a-helix. Figure 3.7 documents the distribution of compounds that utilize H-bonding at the 622/623 carbonyl. Some compounds hydrogen bond while others do not. Only with this type of information can meaningful changes in a molecule to eradicate hERG interactions be made.
Fig. 3.7 Role of peptide backbone carbonyl at Thr623 in drug– hERG interactions. Bars show relative change in binding affinity (DDG) for compounds. Positive ÄÄG indicates decreased affinity relative to WT hERG. A DDG of 1 kcal mol–1 corresponds to an ~5-fold decrease in IC50. Some compounds interact with this position strongly (e. g. pimozide) while others do not (e. g. astemizole).
71
72
3 Unnatural Amino Acids as Probes of Ion Channel Structure – Function and Pharmacology
We clustered these compounds into distinct classes based on their binding interactions with the hERG channel. We then made pharmacophore models for each cluster. Our data demonstrate that different molecules bind to the hERG channel in a variety of ways, and there is no universal hERG binding pharmacophore. We are using this knowledge about hERG–drug interactions to synthetically eliminate the hERG binding ability of new therapeutic compounds.
3.5 Other Uses of Unnatural Amino Acids as Probes of Protein Structure and Function
A promising application of the nonsense suppression methodology is the site-specific incorporation of biophysical probes such as fluorescent groups or spin labels. Several groups have incorporated side chains with fluorescent reporter groups [7, 11, 57, 58]. Fluorescent groups can allow static and dynamic investigations of protein function in several ways. Chollet et al. [59, 60] incorporated an unnatural amino acid based on the nitrobenzoxadiazole (NBD) fluorophore into the neurokinin-2 receptor using the in vivo protocol and Xenopus oocyte expression. The neurokinin-2 receptor is a G-protein-coupled receptor that binds tachykinin. Fluorescence from the NBD group could be monitored in the membranes from ~10 oocytes [59]. Fluorescence resonance energy transfer between the fluorescent agonist and the labelled receptor was used to obtain distance information relating the location of the tachykinin agonist-binding site to selected residues in the receptor [59, 60] Similar studies could be carried out to investigate ligand–binding site interactions of ion channels. Spin labels are another potentially useful probe that could be incorporated by nonsense suppression [7]. Another strategy is to use nonsense suppression to incorporate side chains that permit post-translational modifications, such as phosphorylation or glycosylation [11, 61–63]. This could provide a useful adjunct to existing methods for evaluating the significance of these important processes. Incorporation of amino acids with photoreactive side chains into proteins is an especially useful application of the nonsense suppression methodology in neurobiology [64]. Applications have involved ‘caged’ side chains where a heteroatom is protected. After incorporation of the unnatural amino acid into the protein, photolysis removes the protective group and reveals the previously caged functionality. Photo-decaging also offers the potential for time-resolved studies in which the photolysis is the triggering event. This aspect of using caged unnatural amino acids was demonstrated using a caged tyrosine incorporated into several locations at the agonist-binding site of the nAChR [31]. Caged tyrosine, serine and threonine, hold promise as tools for intracellular cell signaling research. These amino acids are targets for protein kinases. Phosphorylation of these amino acids plays a role in the regulation of a number of proteins and signaling pathways. Kinase dependent modulation of ion channels plays a role in synaptic plasticity associated with learning and memory. Caged tyrosine was used to study the phosphorylation of Kir2.1 (inward rectifier K+ channel) in Xenopus oocytes [65]. When tyrosine kinases were active, flash deca-
3.6 Conclusions
ging of a caged tyrosine at position 242 in Kir2.1 led to decreased K+ currents and 15–26 % decrease in capacitance, implying net membrane endocytosis. It was discovered that decaging initiated two kinase dependent pathways, one involving direct modulation of the channel and the other involving endocytosis of the channel through a clathrin mediated mechanism [65]. This example illustrates how nonsense suppression can be used to deduce signaling pathways and provide new insights into pathways of biological signalling.
3.6 Conclusions
The nonsense suppression for unnatural amino acid incorporation into proteins is an important new tool for protein structure–function studies. Important scientific problems have been addressed, producing knowledge that is complementary to other methods and in some cases impossible to gain with conventional methods. The approach has been used to determine specific interactions governing drug–receptor interactions at the level of individual bonds and side chains [28]. It can be used to incorporate bioprobes to ascertain protein function in real time [5, 11]. It is possible to utilize a second suppressor tRNA to allow for incorporation of two different unnatural amino acids to evolve more sophisticated assays (e. g. FRET pairs) [66–69]. Much of the work discussed in this chapter was carried out using Xenopus oocytes which are a robust system industrialized at Neurion Pharmaceuticals. We are currently pursuing the routine incorporation of unnatural amino acids into proteins expressed in mammalian cells [27]. In addition, some groups are extending the nonsense suppression technique to use a four-base codon instead of the amber codon to incorporate unnatural amino acids [70–72]. In this chapter we have briefly discussed several methodological issues that must be considered including: mRNA read through, orthogonality of the suppressor tRNA, the problem of introducing large quantities of exogenous tRNA into the system. These issues have now been overcome in different expression systems including Xenopus oocytes, wheat germ lysate, and E coli [2, 73–75]. We have verified, using electrophysiology, a sensitive measure of protein function, that protein modified through nonsense suppression is functionally indistinguishable from protein expressed conventionally [2]. Such studies help validate unnatural amino acid mutagenesis as an important new tool for protein structure function studies. An existing challenge is the quantity of protein that can be generated and assayed. This is not a problem when studying ion channels by electrophysiology, but is a limitation in some other cases. The approach has been used to determined specific interactions governing drug–receptor interactions. It can be used to incorporate bioprobes to ascertain protein function in real time [5, 11].
73
74
3 Unnatural Amino Acids as Probes of Ion Channel Structure – Function and Pharmacology
Acknowledgements
Much of the pioneering work that made some of these studies possible was done at Caltech in the groups of Prof. Henry Lester, Division of Biology and Prof. Dennis Dougherty, Division of Chemistry. We gratefully acknowledge their experimental and intellectual contributions. We are also grateful for the experimental contributions of the Neurion scientists including Alisha Goodwin, David Paisner, Elisha Mackey, and Heinte Lesso. References 1 C. J. Noren, S. J. Anthony-Cahill, M. C. Griffith, P.G. Schultz, A general-method for site-specific incorporation of unnatural amino acids into proteins, Science 1989, 244, 182–188. 2 M. W. Nowak, J. P. Gallivan, S. K. Silverman, C. G. Labarca, D. A. Dougherty, H.A. Lester, In vivo incorporation of unnatural amino acids into ion channels in Xenopus oocyte expression system, Methods Enzymol. 1998, 293, 504–529. 3 T. G. Heckler, L. H. Chang,Y. Zama, T. Naka, M. S. Chorghade, S. M Hecht, T4 RNA ligase mediated preparation of novel “chemically misacylated” tRNAPheS, Biochemistry 1984, 23, 1468–1473. 4 J. D. Bain, C. G. Glabe, T. A. Dix, A. R. Chamberlin, Biosynthetic site-specific incorporation of a non-natural amino acid into a polypeptide, J. Am. Chem. Soc. 1989, 111, 8013–8014. 5 D. L. Beene, D. A. Dougherty, H. A. Lester, Unnatural amino acid mutagenesis in mapping ion channel function, Curr. Opin. Neurobiol. 2003, 13, 264–270. 6 D. A. Dougherty, Unnatural amino acids as probes of protein structure and function, Curr. Opin. Chem. Biol. 2000, 4, 645–652. 7 V. W. Cornish, D. R. Benson, C. A. Altenbach, K. Hideg, W. L. Hubbell, P. G Schultz, Site-specific incorporation of biophysical probes into proteins, Proc. Natl. Acad. Sci. U S A 1994, 91, 2910– 2914. 8 L. E. Steward, A. R. Chamberlin, Protein engineering with nonstandard
9
10
11
12
13
14
15
16
17
amino acids. Methods Mol. Biol. 1998, 77, 325–354. D. Mendel,V. W. Cornish, P.G. Schultz, Site-directed mutagenesis with an expanded genetic code, Annu. Rev. Biophys. Biomol. Struct. 1995, 24, 435– 462. L. Wang, P. G. Schultz, Expanding the genetic code, Angew. Chem. Int. Ed. Engl. 2005, 44, 34–66. T. A. Cropp, P.G. Schultz, An expanding genetic code, Trends Genet. 2004, 20, 625–630. T. Hohsaka, M. Sisido, Incorporation of non-natural amino acids into proteins, Curr. Opin. Chem. Biol. 2002, 6, 809–815. M. W. Nowak et al., Nicotinic receptor binding site probed with unnatural amino acid incorporation in intact cells, Science 1995, 268, 439–442. J. D. Bain, D. A Wacker, E. E Kuo, R. A Chamberlin, Site-specific incorporation of non-natural residues into peptides: effect of residue structure on suppression and translation efficiencies, Tetrahedron 1991, 47, 2389–2400. J. D. Bain, E. S. Diala, C. G. Glabe, D. A. Wacker, M. H. Lyttle, T. A. Dix, A. R. Chamberlin, Site-specific incorporation of nonnatural residues during in vitro protein biosynthesis with semisynthetic aminoacyl-tRNAs, Biochemistry 1991, 30, 5411–5421. S. A. Robertson, J. A. Ellman, P. G. Schultz, A general and efficient route for chemical aminoacylation of transfer RNAs, J. Am. Chem. Soc. 1991, 113, 2722–2729. F. A. Laski, R. Belagaje, U. L. RajBhandary, P. A. Sharp, An amber
References
18
19
20
21
22
23
24
25
26
27
suppressor tRNA gene derived by sitespecific mutagenesis: cloning and function in mammalian cells, Proc. Natl. Acad. Sci. U S A 1982, 79, 5813–5817. C. Kohrer, L. Xie, S. Kellerer, U. Varshney, U. L. RajBhandary, Import of amber and ochre suppressor tRNAs into mammalian cells: a general approach to site-specific insertion of amino acid analogues into proteins, Proc. Natl. Acad. Sci. U S A 2001, 98, 14310–14315. M. E. Saks et al., An engineered Tetrahymena tRNAGln for in vivo incorporation of unnatural amino acids into proteins by nonsense suppression, J. Biol. Chem. 1996, 271, 23169–23175. F. Hamano-Takaku et al., A mutant Escherichia coli tyrosyl-tRNA synthetase utilizes the unnatural amino acid azatyrosine more efficiently than tyrosine, J. Biol. Chem. 2000, 275, 40324–40328. D. Kiga et al., An engineered Escherichia coli tyrosyl-tRNA synthetase for site-specific incorporation of an unnatural amino acid into proteins in eukaryotic translation and its application in a wheat germ cell-free system, Proc. Natl. Acad. Sci. U S A 2002, 99, 9715–9720. D. R. Liu, P. G. Schultz, Progress toward the evolution of an organism with an expanded genetic code, Proc. Natl. Acad. Sci. U S A 1999, 96, 4780–4785. S. W. Santoro, L. Wang, B. Herberich, D. S. King, P. G. Schultz, An efficient system for the evolution of aminoacyltRNA synthetase specificity, Nat. Biotechnol. 2002, 20, 1044–1048. Z. Zhang, L. Alfonta, F. Tian, B. Bursulaya, S. Uryu, D. S. King, P. G. Schultz, Selective incorporation of 5-hydroxytryptophan into proteins in mammalian cells, Proc. Natl. Acad. Sci. U S A 2004, 101, 8882–8887. J. W. Chin, T. A. Cropp, S. Chu, E. Meggers, P. G. Schultz, Progress toward an expanded eukaryotic genetic code, Chem. Biol. 2003, 10, 511–519. H. A. Lester, Heterologous expression of excitability proteins: route to more specific drugs? Science 1988, 241, 1057–1063. S. L. Monahan, H. A. Lester, D. A. Dougherty, Site-specific incorporation of unnatural amino acids into receptors
28
29
30
31
32
33
34
35
36
37
expressed in mammalian cells, Chem. Biol. 2003, 10, 573–580. M. W. Nowak et al., hERG Mutant Panel for Lead Optimization of Compunds with hERG Liability, Abstracts of Papers of the American Chemical Society 2005, 229. L. M. Dedkova, N. E. Fahmi, S. Y. Golovine, S. M. Hecht, Enhanced D-amino acid incorporation into protein by modified ribosomes, J. Am. Chem. Soc. 2003, 125, 6616–6617. J. P. Gallivan, H. A. Lester, D. A. Dougherty, Site-specific incorporation of biotinylated amino acids to identify surface-exposed residues in integral membrane proteins, Chem. Biol. 1997, 4, 739–749. J. P. Miller, S. K. Silverman, P. M. England, D. A. Dougherty, H. A. Lester, Flash decaging of tyrosine sidechains in an ion channel, Neuron 1998, 20, 619–624. P. M. England, H. A. Lester, N. Davidson, D. A. Dougherty, Site-specific, photochemical proteolysis applied to ion channels in vivo, Proc. Natl. Acad. Sci. U S A 1997, 94, 11025–11030. P. M. England, H. A. Lester, D. A. Dougherty, Mapping disulfide connectivity using backbone ester hydrolysis, Biochemistry 1999, 38, 14409–14415. P. M. England,Y. Zhang, D. A. Dougherty, H. A. Lester, Backbone mutations in transmembrane domains of a ligandgated ion channel: implications of the mechanism of gating, Cell 1999, 96, 89– 98. L. Li , W. Zhong, N. Zacharias, C. Gibbs, H. A. Lester, D. A. Dougherty, The tethered agonist approach to mapping ion channel proteins–toward a structural model for the agonist binding site of the nicotinic acetylcholine receptor, Chem. Biol. 2001, 8, 47–58. A. L. Cashin, E. J. Petersson, H. A. Lester, D. A. Dougherty, Using physical chemistry to differentiate nicotinic from cholinergic agonists at the nicotinic acetylcholine receptor, J. Am. Chem. Soc. 2005, 127, 350–356. J. T. Koh,V. W. Cornish, P. G. Schultz, An experimental approach to evaluating
75
76
3 Unnatural Amino Acids as Probes of Ion Channel Structure – Function and Pharmacology
38
39
40
41
42
43
44
45
46
47
48
the role of backbone interactions in proteins using unnatural amino acid mutagenesis, Biochemistry 1997, 36, 11314– 11322. P. N. Kao, A. J. Dwork, R. R. J. Kaldany, M. L. Silver, J. Wideman, S. Stein, A. Karlin, Identification of the a subunit half-cystine specifically labeled by an affinity reagent for the acetylcholine receptor binding site, J. Biol. Chem. 1984, 259, 11662–11665. A. Karlin, Chemical modification of the active site of the acetylcholine receptor, J. Gen. Physiol. 1969, 54, s245–s264. H. R. Arias, Topology of ligand binding sites on the nicotinic acetylcholine receptor, Brain Res. Brain Res. Rev. 1997, 25, 133–191. W. Zhong, J. Gallivan,Y. Zhang, L. Li, H. Lester, D. A. Dougherty, From ab initio quantum mechanics to molecular neurobiology: A cation-p binding site in the nicotinic receptor, Proc. Natl. Acad. Sci. USA 1998, 95, 12088–12093. D. A.Dougherty, Cation–p interactions in chemistry and biology: A new view of benzene, Phe, Tyr, and Trp, Science 1996, 271, 163–168. N. Zacharias, D. A. Dougherty, Cation–p interactions in ligand recognition and catalysis, Trends Pharmacol. Sci. 2002, 23, 281–287. J. Warmke, B. Ganetzky, A Family of Potassium Channel Genes Related to eag in Drosophila and Mammals, PNAS 1994, 91, 3438–3442. M. C. Sanguinetti, C. Jiang, M. E. Curran, M.T. Keating, A mechanistic link between an inherited and an acquired cardiac arrhythmia: HERG encodes the IKr potassium channel, Cell 1995, 81, 299–307. R. R. Fenichel et al., Drug-induced torsades de pointes and implications for drug development, J. Cardiovasc. Electrophysiol. 2004, 15, 475–495. J. I. Vanderberg, B. D. Walker, T. J. Campell, HERG K+ channels: friend and foe, TIPS 2001, 22, 240–246. J. S. Mitcheson, M. D. Perry, Molecular determinants of high-affinity drug binding to HERG channels, Curr. Opin. Drug Discov. Dev. 2003, 6, 667–674.
49 M. T. Keating, M. C. Sanguinetti, Molecular and cellular mechanisms of cardiac arrhythmias, Cell 2001, 104, 569– 580. 50 J. A. Sanchez-Chapula, R. A. NavarroPolanco, C. Culberson, J. Chen, M. C. Sanguinetti, Molecular Determinants of Voltage-dependent Human Ether-aGo-Go Related Gene (HERG) K+ Channel Block, J. Biol. Chem. 2002, 277, 23587–23595. 51 J. S. Mitcheson, J. Chen, M. Lin, C. Culberson, M. C. Sanguinetti, A structural basis for drug-induced long QT syndrome, Proc. Natl. Acad. Sci. U S A 2000, 97, 12329–12333. 52 D. Fernandez, A. Ghanta, G. W. Kauffman, M. C. Sanguinetti, Physicochemical features of the HERG channel drug binding site, J. Biol. Chem. 2004, 279, 10120–10127. 53 M. C. Sanguinetti, J. S. Mitcheson, Predicting drug-hERG channel interactions that cause acquired long QT syndrome, Trends Pharmacol. Sci. 2005, 26, 119–124. 54 R. A. Pearlstein et al., Characterization of HERG potassium channel inhibition using CoMSiA 3D QSAR and homology modeling approaches, Bioorg. Med. Chem. Lett. 2003, 13, 1829–1835. 55 T. Lu, A.Y. Ting, J. Mainland, L. Y. Jan, P. G.Schultz, J. Yang, Probing ion permeation and gating in a K+ channel with backbone mutations in the selectivity filter, Nat. Neurosci. 2001, 4, 239–246. 56 P. M. England, H. A. Lester, D. A. Dougherty, Incorporation of esters into proteins: Improved synthesis of hydroxyacyl tRNAs, Tetrahedron Lett. 1999, 40, 6189–6192. 57 L. E. Steward, C. S. Collins, M. A. Gillmore, J. E. Carlson, J. B. A. Ross, A. R. Chamberlain, In vitro site-specific incorporation of fluorescent probes in b-galactosidase, J. Am. Chem. Soc. 1997, 119, 6–11. 58 A. V. Karginov, M. Lodder, S. M. Hecht, Facile characterization of translation initiation via nonsense codon suppression, Nucleic Acids Res. 1999, 27, 3283–3290.
References 59 G. Turcatti et al., Probing the structure and function of the tachykinin neurokinin-2 receptor through biosynthetic incorporation of fluorescent amino acids at specific sites, J. Biol. Chem. 1996, 271, 19991–19998. 60 G. Turcatti, K. Nemeth, M. D. Edgerton, J. Knowles, H. Vogel, A. Chollet, Fluorescent labeling of NK2 receptor at specific sites in vivo and fluorescence energy transfer analysis of NK2 ligand-receptor complexes, Receptors Channels 1997, 5, 201–207. 61 N. E. Fahmi, S. Golovine, B. Wang , S. M. Hecht, Studies toward the site specific incorporation of sugars into proteins: synthesis of glycosylated aminoacyl-tRNAs, Carbohydr. Res. 2001, 330, 149–164. 62 D. M. Rothman, E. J. Petersson, M. E. Vazquez, G. S. Brandt, D. A. Dougherty, B. Imperiali, Caged phosphoproteins, J. Am. Chem. Soc. 2005, 127, 846– 847. 63 Z. Zhang et al., A new strategy for the synthesis of glycoproteins, Science 2004, 303, 371–373. 64 E. J. Petersson, G. S. Brandt, N. M. Zacharias, D. A. Dougherty, H. A. Lester, Caging proteins through unnatural amino acid mutagenesis, Methods Enzymol. 2003, 360, 258–273. 65 Y. Tong et al., Tyrosine decaging leads to substantial membrane trafficking during modulation of an inward rectifier potassium channel, J. Gen. Physiol. 2001, 117, 103–118. 66 J. P. Capone, P. A. Sharp, U. L. RajBhandary, Amber, ochre and opal suppressor tRNA genes derived from a human serine tRNA gene, Embo J. 1985, 4, 213–221. 67 J. P. Capone, J. M. Sedivy, P. A. Sharp, U. L. RajBhandary, Introduction of UAG, UAA, and UGA nonsense mutations at a specific site in the Escherichia coli chloramphenicol acetyltransferase gene: use in measurement of amber, ochre, and opal suppression in mamma-
68
69
70
71
72
73
74
75
lian cells, Mol. Cell Biol. 1986, 6, 3059– 3067. C. Kohrer, J. H. Yoo, M. Bennett, J. Schaack , U. L. RajBhandary, A Possible Approach to Site-Specific Insertion of Two Different Unnatural Amino Acids into Proteins in Mammalian Cells via Nonsense Suppression, Chem. Biol. 2003, 10, 1095–1102. C. Kohrer, E. L. Sullivan, U. L. RajBhandary, Complete set of orthogonal 21st aminoacyl-tRNA synthetase-amber, ochre and opal suppressor tRNA pairs: concomitant suppression of three different termination codons in an mRNA in mammalian cells, Nucleic Acids Res. 2004, 32, 6200–6211. T. Hohsaka, M. Sisido, Incorporation of nonnatural amino acids into proteins by using five-base codon-anticodon pairs, Nucleic Acids Symp. Ser. 2000, 99–100. M. Sisido, Extension of biochemical functions by the introduction of nonnatural amino acids and artificial nucleic acid analogs, Nucleic Acids Symp. Ser. 2000, 275–276. J. C. Anderson, N. Wu, S. W. Santoro, V. Lakshman, D. S. King, P. G. Schultz, An expanded genetic code with a functional quadruplet codon, Proc. Natl. Acad. Sci. U S A 2004, 101, 7566– 7571. J. Ellman, D. Mendel, S. AnthonyCahill, C. J. Noren, P.G. Schultz, Biosynthetic method for introducing unnatural amino acids site-specifically into proteins, Methods Enzymol. 1991, 202, 301–336. D. R. Liu, T. J. Magliery, M. Pastrnak, P. G. Schultz, Engineering a tRNA and aminoacyl-tRNA synthetase for the sitespecific incorporation of unnatural amino acids into proteins in vivo, Proc. Natl. Acad. Sci. U S A 1997, 94, 10092– 10097. D. R. Liu, T. J. Magliery, P.G. Schultz, Characterization of an ‘orthogonal’ suppressor tRNA derived from E. coli tRNA2(Gln), Chem. Biol. 1997, 4, 685– 691.
77
79
4 Functional Expression of Ion Channels in Mammalian Systems Jeff J. Clare
4.1 Introduction
The expression of recombinant ion channels in heterologous systems is an invaluable tool for detailed analysis of their molecular and biophysical properties. This powerful approach compliments the analysis of endogenous channels in their native tissues by providing an opportunity to study and manipulate them in a defined and electrically inactive cell environment. This is advantageous for a wide range of studies, particularly for detailed definition of biophysical and pharmacological properties, for analysing structure–function relationships, and for characterising the role of individual subtypes and/or component subunits. An expression system that has been extensively used in this way is based on oocytes of the South African clawed frog, Xenopus laevis. This system has the advantage of being easy to manipulate while giving robust expression and stable recordings for a broad range of recombinant ion channels in an environment that is largely free of endogenous electrical activity (see Chapter 1). However, despite the utility of Xenopus oocytes for ion channel functional analysis, certain disadvantages are inherent. The maintenance of frogs and preparation of oocytes requires additional procedures and equipment beyond that usually found in a standard research laboratory. Oocyte quality is critical for success and this can be subject to wide variation due to seasonal and other factors. More importantly, mammalian channels can exhibit biophysical properties that are somewhat unexpected [1, 2], possibly due to an absence of regulatory, accessory and other factors that normally interact with the channels in their native environment. Similarly, pharmacological properties can be altered in oocytes. Typically, lower drug potencies are observed and this has been ascribed to a reduction of the free intracellular concentration of drug due to absorption by the large amounts of membrane and yolk particles found inside the oocyte [3]. Many of these drawbacks can be avoided, at least partially, by the use of mammalian cells as an expression host for studying recombinant mammalian ion channels. The aim of this chapter is therefore to review approaches that are currently available for heterologous ion channel expression in mammalian systems. For reExpression and Analysis of Recombinant Ion Channels. Edited by Jeffrey J. Clare and Derek J. Trezise Copyright # 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim ISBN: 3-527-31209-9
80
4 Functional Expression of Ion Channels in Mammalian Systems
search applications, expression of the channel in a functional form is normally a prerequisite. Similarly, although alternative formats exist (e. g. radioligand binding, chapter 7), the most reliable read-outs for ion channel drug screening are those based, either directly or indirectly, on functional activity of the channel in a cell-based assay. However, this can often present significant challenges given the huge diversity and organisational complexity found within the ion channel superfamily. Ion channel subunits generally contain multiple transmembrane spanning segments and functional channels are usually complex multi-subunit proteins. Correct membrane localisation and orientation, as well as appropriate subunit composition and stoichiometry, are often vital for faithful reproduction of the pharmacological and biophysical properties of the native channel. In addition, ion channel cDNAs can be difficult to manipulate due to high instability and frequently cause cytotoxicity to the host cell when over-expressed. Finally, even for well expressed channels, the generation of reagents that are sufficiently robust for use in a high throughput screening environment can be extremely problematic. In this chapter a number of strategies that can be used to address these challenges will be described. A number of case studies will be presented that highlight some of the theoretical and practical considerations for obtaining robust expression of functionally active channels.
4.2 cDNA Cloning and Manipulation
cDNA instability can be a fairly common phenomenon with certain ion channels, which can make cloning and manipulation using standard techniques difficult. This is presumably due to the presence of sequences that are either toxic or recombinogenic in E.coli (e. g. direct repeats, Z DNA, etc.) causing negative selection pressure and resulting in a strong orientation bias and/or few viable clones. Clones that do arise are frequently found to contain mutations ranging from large rearrangements and deletions to single base changes that cause mis-sense or frame shift mutations. Certain channel subfamilies, e. g. voltage-gated sodium (NaV) channels and ATP-binding cassette (ABC) transporters such as CFTR, sulfonylurea receptors, seem to be particularly susceptible to this, but the problem can also occur more sporadically in other subfamilies, e. g. voltage-gated calcium channels (CaV). These problems can be minimised using a variety of approaches, either separately or, if necessary, in combination. The use of “directional” cloning strategies (i. e. where insertion of the DNA fragment in the required orientation is forced by the use of incompatible restriction sites) is recommended wherever possible. Vectors that are “transcriptionally silent” (i. e. containing no E.coli promoter sequences, cyptic or otherwise, in the vicinity of the cloning site) and/or that replicate at low copy number in E.coli (e. g. pBR322-based) are also useful. In addition, E.coli host strains are commercially available that allow propagation of standard ColE1-based vectors at reduced copy number (ABLE, Stratagene). The use of commercially available recombination-deficient E.coli host strains (SURE, Stratagene,
4.3 Choice of Host Cell Background
STABL2, Invitrogen) is also recommended. Finally, propagation of E.coli clones at reduced temperature (e. g. 30 8C) is often found essential for problematic channels – the colonies may take several days to appear and will generally be much smaller than normal. Despite taking some or all of the above precautions, for channels that are known to be unstable, it is essential to rigorously validate final constructs by complete DNA sequencing and it frequently proves necessary to correct mutations by one or more rounds of site-directed mutagenesis. In extreme cases, it may also be necessary to resequence the channel-encoding region each time a new batch of DNA is prepared. Finally, for optimal expression, it is common to engineer the region upstream of the start codon to contain sequences compatible with efficient initiation of translation (Fig. 4.1). The consensus sequence associated with genes that are efficiently expressed in mammalian cells, identified by Kozak [4], is often used, and this can normally be inserted during construction of the expression vector.
4.3 Choice of Host Cell Background
As indicated in the introduction to this chapter, the choice of host cell background is often critical for efficient expression as well as for faithful reproduction of the properties of the native channel. Two major concerns are the existence of endogenous channels in the host cell that may be similar to the channel of interest and mask its expression, and the presence or absence of accessory factors that may normally modify the properties or expression of the channel in its native environment. Clearly, these parameters will vary between different cell types and are dependent on the nature of the channel of interest. Thus, it is advisable to screen a range of potential cell lines for their suitability as hosts before embarking on an expression project. This can be done by functional screening, preferably using the assay format that will ultimately be used for analysing the channel of interest, or alternatively by molecular screening, for example by quantitative mRNA analysis (e. g. Taqman). The latter approach is often more convenient and is useful as a primary screen in that the endogenous expression of wide range of channels and accessory proteins can be screened simultaneously (e. g. using reverse Taqman). However, this requires a reasonable level of background knowledge of the channel family and its likely modulatory or interacting partners, which may not always be available, especially for more novel channels. In addition, the detection of mRNA for a particular channel or accessory factor does not necessarily mean that the active protein is expressed, and functional assays are usually necessary as a secondary screen to establish this. Although pre-screening of potential host cell lines may indicate a lack of confounding background activities, it is worth remembering that it is formally possible for normally silent endogenous channel proteins to become up-regulated during the process of generating an expression reagent, perhaps in response to over expression of the target channel. Thus, it is prudent to validate expression of the
81
82
4 Functional Expression of Ion Channels in Mammalian Systems
Fig. 4.1 Quantitative comparison of different expression constructs and screening of stable clones using automated high throughput electrophysiology. (A) Comparison of representative hERG K+ channel expressing CHO cell lines that were generated using different types of expression construct. A and D = nonoptimised mRNA leader sequence, B = optimised leader, C = optimised leader in IRES expression vector. Each point represents peak current (left panel) or percentage of cells expressing (right panel) averaged from ~300
cells measured by IonWorks high throughput electrophysiology. (B) Screening of stable clones for functional hERG channel expression. 24 different clonal CHO cell lines generated with construct C (above), were analysed by Ionworks electrophysiology. The graphs show a comparison, for each clone, of the percentage of cells giving usable seals (>80 MO, bottom), percentage cells expressing currents above background (middle) and mean current amplitude (top). Data are averaged from 64 cells for each clone.
4.3 Choice of Host Cell Background
target protein in the final reagent by a sequence dependent method, for example by immunological validation, if selective antibodies are available, or by a quantitative PCR (Taqman). In a drug discovery setting another factor that should be considered when choosing a host background is the physical characteristics of the cells, which must be compatible with the intended assay format. Thus, electrophysiological assays depend on the formation of high resistance electrical seals with the plasma membrane, and microtitre plate-based assays (see Chapters 7 and 8) often require good adherence of cells to the plastic substrate. It is worth stating that these qualities can deteriorate in cells over-expressing ion channels compared to untransfected parent cells, especially over prolonged passage. A variant of the HEK293 cell line that has been engineered to be more adherent to plastic surfaces by expressing the macrophage scavenger receptor is available (Invitrogen). A variety of mammalian cells lines have been successfully used as hosts for recombinant ion channel expression. Standard hosts that are in general use for heterologous expression have also been successfully used for ion channels, including CHO, COS7, CV1, HEK293 etc. As indicated above, variants of these lines which are designed for specific applications have also been engineered and their use will be highlighted throughout this chapter. These commonly used hosts are relatively (though not completely) free of background conductances and are suitable for many channel types (e. g. see Refs. [5, 6]). However, it is worth noting that different isolates can vary in this respect and may behave differently in different laboratories, so it is still advisable to functionally screen for background currents of interest. In situations where there is relatively little background knowledge of the channel and its potential accessory factors, or where expression proves problematic in “standard hosts”, it is often worth searching for less common immortalised lines that are derived from tissues that natively express the channel of interest. For example, various neuroblastoma cell lines have proved useful as hosts for neuronal ion channel expression, such as SH-SY5Y, ND7–32 , NG108 [7–9]. In practise, the precise reason(s) for this have not been fully explored but it is likely that the intracellular environment and processing machinery are more favorable in a host cell derived from the tissue where the channel is normally expressed. Other factors that can be important probably include the presence of cognate accessory proteins and better tolerance to a wider range of resting membrane potential than nonexcitable cells. An example of this is provided by the sensory neuron specific NaV1.8 Na+ channel. This is only poorly expressed in commonly used host cells (e. g. HEK293) but is more efficiently expressed in an immortalised dorsal root ganglion-derived cell line (ND7–23, Fig. 4.2a). One possible explanation is that this relates to the accessory b3 subunit since this is endogenously expressed in ND7–23 cells but, not in HEK293 (Fig. 4.2c). In support of this, it was found that the recombinant b3 subunit enhances the level of NaV1.8 specific currents when co-expressed in HEK293 cells (Fig. 4.2b). Furthermore, this effect is also seen in ND7–23 cells, suggesting that the effect of endogenous b3 in these cells can be enhanced by over-expression of the recombinant subunit.
83
84
4 Functional Expression of Ion Channels in Mammalian Systems
Fig. 4.2 Effect of cell background and auxiliary subunits on efficiency of ion channel expression. (A) Comparison showing representative current traces recorded from HEK293 and ND7–23 cell lines stably expressing the NaV1.8 channel. Peak Na+ currents in HEK293 stable clones were smaller compared to ND7–23 and declined during passage or following freeze–thawing of cells. (B) Transient co-expression of the NaVb3 subunit enhances NaV1.8 expression in HEK293 cells.
Bars represent the mean peak currents obtained. (C) NaVb3 subunit mRNA is endogenously expressed in ND7–23 but not in HEK293 cells. RT-PCR analysis of cell extracts using subtype selective primers is shown. Lanes labelled “-” indicate –ve controls where reverse transcriptase was omitted; +ve control reactions, using the cloned b subunit cDNAs as template, are shown in the right hand lanes. Reproduced with permission from Ref. [89].
4.4 Post-translational Processing of Heterologous Expressed Ion Channels
As illustrated above, it is possible to engineer host cell lines to make them more favorable for ion channel expression by over-expressing accessory factors that may be required for efficient expression. It may also be desirable to generate cell lines that express reporter genes (e. g. Aequorin [10], halide-sensitive YFP [11]) for use as generic hosts for ion channel assays. An additional approach to engineering cell lines to make them more favorable as hosts is to manipulate their electrical properties by introducing additional channels. This is particularly useful when generating reagents for microtitre plate-based functional assays in which the readout is dependent on voltage-sensitive dyes, but can also apply to assays based on Ca2+ binding fluorescent dyes. A good example of this is shown in Fig. 4.3, which illustrates the expression of the R-type voltage-gated calcium channel in non-neuronal cells (HEK293). In these cells only small Ca2+ responses can be detected in fluorescence imaging plate reader (FLIPR, Molecular Devices) assays when they are depolarised, even though good sized currents can be detected electrophysiologically. This is apparently due to the resting potential of HEK293 cells which is more positive than that typically found in neurons (e. g. –10 to –20 mV compared with –60 to –80 mV) resulting in the majority of channels being inactivated. However, it is possible to increase the magnitude of depolarisation-induced Ca2+ responses by manipulating the resting potential by co-expressing a constitutively active K+ channel, KCNK2 (TREK-1). This leads to a more negative resting potential, resulting in more Ca2+ channels being available to open when the cells are depolarised. This dual expression system can also be used in reverse to measure activity of KCNK2 in FLIPR assays using the Ca2+ channel as a reporter.
4.4 Post-translational Processing of Heterologous Expressed Ion Channels
Since, by nature, they are complex multimeric transmembrane proteins, ion channels present particular challenges for efficient heterologous expression. In order to obtain functional activity a number of post-translational steps are required to ensure that channel proteins are correctly folded, processed, assembled and transported to the appropriate membrane compartment [ for review see Ref. [12]]. Since, each of these steps requires interaction of the heterologous channel protein with homologous host factors, there is potential for incorrect processing at every stage. In addition, abnormally high levels of expression can potentially overwhelm the capacity of the endogenous processing machinery, again leading to unprocessed or aberrant channel protein. Similarly, for any given channel, a particular processing step may require specific factors that might not be endogenous to the cell type being used (see below for examples), again leading to aberrant, nonfunctional protein. Processing “bottlenecks” such as these can result in the aberrant channel protein either accumulating and then aggregating within the cell [13], or alternatively being targeted for degradation [14, 15]. In extreme cases both of these outcomes can be difficult to diagnose, since the former can lead to cytotoxicity and low viability (see discussion below), and the latter can appear as a total failure
85
86
4 Functional Expression of Ion Channels in Mammalian Systems
4.4 Post-translational Processing of Heterologous Expressed Ion Channels
of expression unless this is being monitored by measuring mRNA levels. In less extreme cases a low level of the mature functional protein may be detected, but the amount produced is likely to be sensitive to both the level and rate at which expression is being driven. Paradoxically, attempting to improve yields in this situation by increasing expression, for example with the use of stronger promoters, may actually be counterproductive [16] since this could exacerbate blockage of the processing pathway. Instead, the use of titratable (e. g. viral) and/or inducible systems is recommended in order to try to optimise the balance between the total protein expressed and the proportion that is correctly processed and functional (see Sections 4.6.1 and 4.7.3). An alternative approach for increasing the proportion of mature channel protein is to augment the level of processing factors that may be present only in limiting amounts or absent altogether. As discussed previously, an empirical way of achieving this is to identify and use an alternative host cell type that is more related to the tissue in which the native channel is expressed. However, another approach is to overproduce factors that are likely to be, or are known to be, involved in channel maturation by co-expression of their cloned cDNA. Proteins with this activity broadly fall into two categories: those that are thought to play a general role in quality control and promoting protein folding and transport within most cells (e. g. chaperones), and those with more specialised trafficking roles for particular types of ion channel. Chaperones act by binding to nascent proteins in order to promote correct folding and targeting. It is known that proteins with large regions of exposed hydrophilic regions are toxic to cells, possibly by promoting membrane damage [17], and indeed this is believed to contribute to the pathogenic mechanism of certain “viral” proteins (e. g. prions [14, 15]). It is likely that the production of large amounts of misfolded recombinant membrane proteins can also cause cytotoxicity by similar mechanisms. Thus, co-expression of factors with chaperone-like activity should be beneficial, not only by increasing the proportion of mature channel protein produced, but also by counteracting this toxic effect. Examples of chaperones that have been shown to boost the surface expression of various recombinant ion channels (e. g. Kv, nAChR, HERG, CFTR, NaV) include calnexin, heat shock proteins (e. g. Hsp70, Hsp90) and fibroblast growth factor homologous factor 2B (FHF2) [18–23]. However, as well as their beneficial
3 Fig. 4.3 Manipulation of the host cell background to optimise functional expression of ion channels. (A) Co-expression of the TREK1 K+ channel is required in order to hyperpolarise the resting potential of HEK293 cells so that functional R-type CaV channels can be detected in a FLIPR-based fluorescence assay. Only when the TREK1 K+ channel is coexpressed, in this case using a BacMam viral vector, can robust Ca2+ influx (measured by an increase in fluorescence of the Ca2+ binding dye Fluo4) be induced by depolarisation (addi-
tion of KCl) of cells stably expressing the R-type CaV channel (CaV2.3 and b3 subunits). B) Similar functional responses can be observed when all three subunits (CaV2.2, b3 and TREK1) are delivered using BacMam. Little or no Ca2+ influx is seen in the absence of TREK1 virus. (C) The magnitude of Ca2+ influx (change in fluorescence) can be manipulated by varying the amount of TREK1 virus added. (X, no TREK1 virus added; ~, 56106 plaque forming units (pfu) ml–1 of TREK1 virus added, &, 107 pfu ml–1 ; ^, 26107 pfu ml–1.)
87
88
4 Functional Expression of Ion Channels in Mammalian Systems
effects, it must be remembered that, in some circumstances, binding of chaperones can actually target proteins for degradation [22]. In addition to chaperones, certain cytoskeletal scaffold proteins which interact with channel proteins at the cell surface can also promote surface localisation of the channel [24]. As well as proteins that have general chaperone-like activity within the cell, a number of channel-specific maturation factors have been identified. Examples include the PACS proteins, annexin II light chain, and KChap which promote surface localisation of Trp, Kv and NaV1.8 channels respectively [25–27]. In addition, numerous other channel-associated proteins are known that have dual functions, both acting as channel-specific modulatory subunits as well as enhancing channel trafficking. Examples include the b subunits of Kv, NaV (see Fig. 4.2B) and CaV channels [28–30], a2d subunits of CaV channels [31], calmodulin [32, 33] and stargazin [34] (which promote trafficking of IK/SK and AMPA channels respectively). Some of these are thought to act by masking specific ER retention motifs present in the channel protein. There is evidence for these retention signals in a wide range of channels, including Kir, SK/IK, Kv, Eag, HERG, NMDA, kainate, nAChR [35–44], and it is thought that these motifs are exposed in the monomeric subunit and become inaccessible upon assembly of the mature multimeric channel (see for example Ref. [44]). Thus, for optimal expression of any given channel, it is worth co-expressing any known channel-specific accessory subunits or interacting factors, particularly in cases where known ER retention signals are present. Another strategy that can be used to increase the amount of functional channel protein found at the cell surface is to lower the growth temperature at which the cells are maintained. This has been shown to give higher levels of surface expression for a number of different channel types. For several of these examples (e. g. CFTR, P2X7, HERG, NaV1.5 [45–49]) the effect was first noted with variants of the channel that are defective for trafficking. Incubation of cells expressing these mutants at reduced temperature (e. g. 25–30 8C) was found to rescue the defect. This appears to be a general phenomenon since it has also been observed with nonmutated channels (e. g. ROMK1, nAChR’s, KAT1 [50–55], and HERG – see Fig. 4.4 and 6.2). There are several possible explanations for this effect, for instance, lower temperatures could result in increased steady-state levels of transcript or synthesis of channel protein, reduced protein turnover, improved protein folding, assembly or transport. The best studied example is probably the CFTRD508 mutant that is the most prevalent cause of cystic fibrosis. Recent studies indicate that the deleted residue normally has a major contribution to the proper folding of the channel [56, 57]. Thus, at physiological temperature (37 8C) folding of the mutant is impaired and the protein is targeted for degradation, probably due to binding of the Hsp70/CHIP chaperone complex [22]. However, at lower temperature (30 8C) folding kinetics are more favorable, protein degradation is greatly reduced, and the mutant protein can then be detected at the cell surface. A similar mechanism probably accounts for the increased surface expression of nonmutant channels at lower temperature. Consistent with this, reduced protein degradation has been reported in several cases (ROMK1, a4b2nAChR [50, 53] and HERG – see Fig. 4.4b). Evidence suggests that degradation can occur via mechanisms that are both proxi-
4.4 Post-translational Processing of Heterologous Expressed Ion Channels
Fig. 4.4 Effect of reduced cell culture temperature on ion channel expression. (A) Confocal images of stable HERG-expressing CHO cells grown at different temperatures, following immunocytochemical staining with a HERG-specific antibody. The level of HERG immunoreactivity is significantly increased in cells grown at 27 8C and 30 8C compared to 37 8C. No immunoreactivity is seen in untransfected CHO cells (CHO-wt). The increase in total HERG protein at 30 8C seen here is also mirrored by an increase in surface-localised functional protein as measured by IonWorks electrophysiology
(see Fig. 6.2). Note that, in the HERG-expressing cells, at 37 8C and 30 8C immunoreactivity is uniform and granular throughout the cytoplasm whereas at 27 8C there is an accumulation in giant lysosome-like bodies. (B) Quantitative analysis of HERG immunoreactivity by flow cytometry confirms the increases seen at lower growth temperatures, as indicated by the rightward shift in peak fluorescence (Xaxis) observed within the populations of cells grown at 27 8C and 30 8C compared to 37 8C. (This figure also appears with the color plates.)
89
90
4 Functional Expression of Ion Channels in Mammalian Systems
mal (proteosomes) and distal (endocytic recycling) to the secretory pathway ([58], Fig. 4.4A). Thus, maintenance of cell cultures at lower temperatures would be expected to improve functional expression of any channel that is prone to misfolding when over-expressed.
4.5 Cytotoxicity
Many ion channels are poorly tolerated when over-expressed in mammalian cells, leading to poor growth and viability, and low levels of expression which tend to decline during cell passage. Usually this becomes evident during stable cell line generation when only a low number of viable clones can be selected, with few of those that do arise giving measurable levels of expression. Among these clones only a low proportion of cells actually express and there are large cell-to-cell variations. This is a hallmark of toxicity induced by heterologous protein expression, but often the situation can be at least partially rescued by the use of appropriate expression vectors (e. g. IRES vectors, see Section 4.7.1). Other strategies to address this problem include the use of transient or inducible expression systems (see Sections 4.6 and 4.7.3). However, it may also be possible to improve the situation by attempting to tackle the root cause(s) of the problem. As discussed above, one possible reason for toxicity is saturation of the host processing machinery – this may lead to deficiencies in the processing of essential host proteins, with consequent toxic effects, as well as the accumulation of large amounts of misfolded protein which may have membrane damaging effects. In this case co-expression of chaperones or channel accessory subunits may be beneficial. Another possible mechanism for toxicity is that functional activity of the channel itself has cytotoxic consequences for the host cell. Channels that are permeable to calcium may be particularly susceptible to this (e. g. NMDA-NR2 a and NR2 b [59]), since calcium signalling regulates many fundamental processes within the cell and is normally tightly regulated. Clearly, over-expression of large conductance calcium-permeable channels, or those that normally have a role in calcium homeostasis, is more likely to cause this type of problem. A potential solution to this problem is to use known channel blockers during reagent generation (e. g. during selection of stable clones) and also during routine maintenance of the recombinant cells [60]. In the light of this it is worth bearing in mind that aminoglycoside antibiotics, which are often used for selection during stable cell line generation (e. g. neomycin, geneticin), have been found to have inhibitory activity at several types of ion channel (e. g. TRPV1, CaV, NMDA [61–63]). This may be beneficial during cell line generation for toxic channels. On the other hand, it has also been found the aminoglycoside antibiotics can also potentiate channel activity (e. g. NMDA-NR2 b [63]), which may actually exacerbate toxicity.
4.6 Transient Expression Systems
4.6 Transient Expression Systems
Transient expression systems are commonly used in ion channel studies for a wide variety of applications. They are particularly valuable where rapid and/or high throughput analysis is required, for example in alanine scanning, cysteine scanning, site-directed or random mutagenesis studies and for the evaluation of expression constructs, channel variants and subunit combinations. Transient expression provides a flexible way of titrating the level of expression, controlling stoichiometry of multi-subunit channels and introducing reporters or interacting factors that may be required. In a drug screening environment, where many different ion channel assays may be run in parallel, transient expression can be more convenient than stable expression since this reduces cell culture demands by removing the need to continuously maintain multiple cell lines in culture. As discussed already, transient systems are also useful where toxicity is known to be a problem. Several alternative approaches for transient expression are available and each has different advantages and disadvantages. 4.6.1 ‘‘Standard’’ Transient Expression
“Standard” transient expression approaches usually involve the treatment of cells with chemical agents that promote the uptake and internalisation of naked DNA. Different types of agent are available, and these work with varying efficiencies on different cell types [64]. Among the most commonly used for ion channels are calcium phosphate [65] and various cationic lipid reagents which are available from commercial suppliers (e. g. Lipofectamine, Invitrogen). Although the latter come with detailed protocols, there are particular considerations for ion channels. For example, in electrophysiology experiments an appropriate balance is needed between the transfection rate (i. e. proportion of cells expressing) and the health of the cells following transfection. Thus, in order to achieve the best results for any given channel it may be worth optimising the transfection conditions (amount and time of reagent exposure, recovery time etc.). An alternative methodology to chemical reagents sometimes used for transient expression of ion channels is electroporation [66], though this tends to be less efficient. This is because the magnitude of the electrical pulse that can be given to promote DNA uptake is limited by deleterious effects on cell viability and once again a balance between transfection efficiency and cell health has to be struck. The efficiency of transient transfection and gene expression is highly dependent on the cell type and expression vector used. HEK293 and COS cells are among the more competent for DNA uptake and are commonly used for ion channel expression, though other cell types can also be used (e. g. CHO). Variants of HEK293 are available which enable plasmid replication when using vectors containing the origin of replication from SV40 due to the expression of SV40 T antigen (HEK293T, ATCC) [67, 68]. This “episomal” vector system should give enhanced expression
91
92
4 Functional Expression of Ion Channels in Mammalian Systems
due to the increased copy number of the channel cDNA and possibly due to other activities of T-antigen. Another episomal vector system uses an analogous transacting DNA replication factor (EBNA1) from Epstein Barr virus (EBV) which enables replication of vectors containing the replication origin from EBV (oriP) [69]. Expression may also be boosted via a reported EBNA1-dependent enhancer like activity of oriP [70]. Different variations of this system have been developed where EBNA1 is encoded either by the expression vector (e. g. pCEP4, Invitrogen) or by the host cells (HEK293E, Invitrogen). The former has the advantage of being compatible with any host cell type, whereas the latter may be more convenient since a smaller vector can be used which may aid insertion of the channel encoding cDNA. Streamlined oriP-containing episomal vectors of this type have been developed that contain optimised promoters and that have been shown to give very high levels of expression [71]. 4.6.2 Viral Expression Systems
Standard transient transfection is the most rapid means of mammalian cell expression. However, the efficiency is highly dependent on the host cell background and many cell types that are of interest for ion channel expression can be relatively resistant to transfection methodologies (e. g. neuroblastoma cell lines). A potential solution to this problem is to make use of virus-based systems for gene delivery, and two examples that have been successfully applied to the heterologous expression of ion channels and membrane-bound receptors in mammalian cells will be discussed here. The first system is derived from the single-stranded RNA virus, Semliki Forest Virus (SFV), and the second is based on the insect cell baculovirus, Autographa California nuclear polyhedrosis virus (AcMNPV). The SFV expression system comprises two plasmid vectors which together contain a cDNA copy of the viral genome [72]. The expression vector contains the nonstructural genes, the strong SFV 26S promoter for expression of the heterologous gene and a viral packaging signal. The helper vector encodes the structural proteins, including the capsid and envelope genes, but no packaging signal. A stock of recombinant viral particles is produced by first generating single-stranded helper and expression vector RNAs using in vitro transcription and then co-transfecting these into baby hamster kidney (BHK) cells. These particles can then be used to efficiently infect a broad range of mammalian cell lines (including BHK, COS, HEK293), as well as primary cells, and are capable of giving rapid, high-level expression. However, no virus progeny is produced because the recombinant particles contain no helper RNA and are therefore replication deficient. This system has been used extensively to express G-protein coupled receptors (GPCRs) [73] often giving good expression levels and correctly folded protein, as determined by ligand binding assays and in some cases by coupling to G-protein signalling pathways. The potential for ion channel expression has also been explored and several channels have been expressed, including 5HT3, P2X1, 2 and 4 [74], GABAa [75] and Kv [76]. Functional activity was demonstrated for 5HT3, P2X receptors and GABAa.
4.6 Transient Expression Systems
Recombinant baculoviruses have been routinely used for efficient protein expression in insect cells for many years. However, although baculovirus virions are able to enter mammalian cells, no viral expression can normally be detected. Recently baculovirus-based “BacMam” vectors have been engineered to allow expression in mammalian cells by insertion of promoters that are active in mammalian hosts (for a review see Ref. [77]). The gene of interest is inserted into a transfer vector downstream of the mammalian promoter (e. g. CMV) in which, similar to the baculovirus system, it is flanked by regions that promote insertion into the baculovirus genome, either by homologous recombination in insect cells or by transposon-mediated recombination in E.coli cells (see Fig. 4.5). A stock of virus particles is generated by propagation of the recombinant virus genome in insect cells and this can then be used to deliver the gene for transient expression in a range of different mammalian cell types, including many of those commonly used for recombinant protein production. The BacMam transfer vector commonly used also contains a selectable marker conferring resistance to the antibiotic G418, thus allowing the additional option of selecting stable cell lines from virus-transduced cells [78]. The BacMam system works particularly well with certain cell types including HEK293, and the human osteosarcoma derived cell line, U2OS in which transduction and expression rates are high [79, 80]. It has been used with good success for membrane protein expression, including a large range of GPCRs and ion channels [81, 82]. In common with other transient systems it offers advantages for expression of toxic channels and for multi-subunit and multi-channel expression [81]. For example, the NMDA-NR2 b glutamate receptor, which requires expression of both the NR2 b and NR1 subunits for functional activity, is extremely toxic when expressed in non-neuronal mammalian cells [59]. In the author’s laboratory, repeated attempts to generate NR2b-expressing stable cell lines were unsuccessful, even using IRES vectors (see Section 4.7.1) and even in the presence of a channel blocker (ketamine). However, by generating separate BacMam viruses for each subunit, and transducing a mixture of these into HEK293 cells, robust glutamate-induced calcium influx can be observed that is not present in mock-transduced cells (Fig. 4.6). The magnitude of the response is proportional to the total amount of virus added and the stoichiometry of the two subunits can be readily investigated and optimised by varying the dose of virus for each subunit. An example of multi-channel expression using BacMam viruses is shown in Fig. 4.3. In addition to viruses for two different subunits of the R-type calcium channel (CaV2.3, a2d), a virus encoding the TREK-1 K+ channel was also introduced, in order to hyperpolarise the resting potential of the host HEK293 cells and thus prevent inactivation of the R-type channel. In this way, robust calcium influx can be measured when the TREK-1 virus is added, but not in its absence, and the magnitude of the response can be varied by altering the amount of TREK-1 virus added. In summary, the BacMam system is a versatile tool for the robust analysis of many ion channels and is particularly suitable for toxic or multi-subunit channels. It allows flexible control over subunit or channel stoichiometry simply by varying the dose of the corresponding viral constructs used for transduction. It is also a
93
94
4 Functional Expression of Ion Channels in Mammalian Systems
Fig. 4.5 The BacMam expression system. (A) Map of the pFastBacMam1 shuttle vector which used to generate recombinant BacMam viruses. The gene of interest is inserted downstream of the CMV promoter where it is flanked by Tn7 inverted repeats that direct site-specific transposition into the viral gen-
ome when transfected into the appropriate E.coli host. (B) Workflow for the generation of recombinant BacMam virus stock and transduction into mammalian cells for expression. Reproduced with permission from Ref. [77]. (This figure also appears with the color plates.)
4.6 Transient Expression Systems
Fig. 4.6 Expression of a cytotoxic channel using the BacMam system. (A) Functional expression of the NMDA-NR2 b receptor using BacMam. Glutamate-induced Ca2+ influx, as measured by increased fluorescence of the Ca2+ binding dye Fluo4, can be observed in HEK293 cells transduced with a 1 : 5 mixture of BacMam viruses expressing the NMDA-
NR1A and NR2 b subunits, respectively. The arrow indicates the point of glutamate addition. (B) The magnitude of the peak response is dependent on the total amount of virus added, as measured by MOI (multiplicity of infection – the number of virus particles added per cell).
convenient means of introducing additional factors into existing ion channel stable cell lines, for example chaperones or reporter genes that might be required for use in alternative assay formats (e. g. aequorin, halide sensitive YFP). Little or no cytopathic effects of virus transduction are observed and cells can normally be used for electrophysiology assays the day after transduction. BacMam viruses are also safe since they are unable to replicate in mammalian cells and the budded form of the virus is noninfectious for the natural insect host [77].
95
96
4 Functional Expression of Ion Channels in Mammalian Systems
4.7 Stable Expression of Ion Channels
For many applications stable expression of the target ion channel may be desirable, for example for detailed biophysical studies or extended compound screening campaigns. Unlike transient systems, this avoids the need to continually carry out transfections or transductions, and there is no requirement for repeated re-synthesis of vector DNA or virus reagents. In theory, stable expression would also be expected to be more reproducible than transient expression. Nevertheless, a certain degree of day-to-day and cell-to-cell variation of expression in stable cell lines is liable to occur and this can be considerable for some ion channels. In practise, the worst stable cell lines can be more variable than some transient systems, e. g. BacMam viruses, which are capable of giving perfectly acceptable reproducibility. 4.7.1 Bicistronic Expression Systems
The generation of robust ion channel expressing stable cell lines can be problematic. As discussed already, since many ion channels are poorly tolerated when over-expressed, difficulties can arise due to poor growth and loss of expression during passage, even when selection is maintained. Often this is accompanied by large cell-to-cell variability in expression level even within clonally isolated lines, which can be a particular problem for single-cell format electrophysiology assays. A related problem during cell line generation is that, following transfection and selection, only a low proportion of clones actually express measurable levels of channel activity. This situation can be improved by using vectors where expression of the channel is closely coupled to that of the selectable marker. The best way of achieving this is to use a viral internal ribosome entry site (IRES) which promotes translation of both proteins from a single bicistronic mRNA [83]. The IRES element is placed between the two coding regions, with the channel upstream and the selection marker immediately downstream (Fig. 4.7). Translation of the upstream channel is initiated, as normal, by signals in the 5' untranslated region, whereas translation of the downstream selection marker is initiated by binding of ribosomes to the IRES element. Thus, both proteins are produced from a single transcript with the advantage that expression of the two is very tightly linked. This is unlike “standard” vectors where the selection marker and gene of interest are in separate expression cassettes and it is not uncommon for expression of the two to become separated, either during clonal selection or subsequent maintenance of the cell line, giving rise to the problems described above. This is especially true with toxic genes where the growth disadvantage conferred on expressing cells is an additional selection pressure for this to occur. Thus, an additional benefit with IRES vectors is that expression should remain relatively stable over extended periods as long as selection is maintained (Fig. 4.8). The advantages of IRES vectors for stable cell line generation become particularly apparent when using high-throughput electrophysiology assays. Automated
4.7 Stable Expression of Ion Channels
Fig. 4.7 Bicistronic expression vectors for stable ion channel expression. (A) Using an IRES element the expression of the ion channel of interest is co-translationally linked to the selection marker (neoR), thus avoiding loss of expression and instability problems that can occur with conventional expression vectors due to independent segregation dur-
ing generation and propagation of the cells. (B) Plasmid map of a typical bicistronic expression vector, pCIN5L, that can be used to generate stable cell lines. The ion channel cDNA is inserted upstream of the IRES element between the Not I and Nhe I restriction sites. (IVS, intervening sequence or intron).
patch-clamp instruments are planar array instruments (see Chapter 6), and place great demands on the quality of the cell line being used since they are single cell assays in which the cells are randomly selected (though this problem is now being addressed by the latest generation instruments, e. g. IonWorks Quattro). Thus, a very high proportion of cells in the population that express currents of usable size is essential in order to avoid low success rates and unacceptably high costs resulting from compound wastage and reduced throughput. On the other hand, these instruments provide invaluable tools for functional screening during cell line generation. They enable a vast increase in the number of clones that can be screened compared to that previously possible using conventional patch-clamp (Fig. 4.2B). Using a combination of IRES vectors and screening by high throughput automated electrophysiological screening, very high quality stable cell lines can readily be obtained with a variety of different ion channels, which typically express peak currents in the nA range in greater than 90–95 % of the cell population (Fig. 4.9). High throughput patch-clamp instruments also add further precision to the
97
98
4 Functional Expression of Ion Channels in Mammalian Systems
Fig. 4.8 Quantitative analysis of multiple subunit expression using automated high throughput electrophysiology. (A) Population histogram of current amplitudes in individual cells (n=3180) co-expressing KCNQ1 and the accessory subunit minK. The fitted line is a 2 peak gaussian fit indicating the presence of two populations – nonexpressors (currents <0.5 A, mean = 0.33 nA), and expressors (currents >0.5 A, mean = 1.05 nA). The ex-
pressors comprise >80 % of the cell population. (B) Stability of expression over passage showing percentage of cells expressing (triangles) and mean peak current in expressing cells (circles). Each point represents the mean of >250 cells. Kinetic analysis of the currents (see Fig. 6.5) indicates that virtually every cell expresses the minK subunit as well as KCNQ1.
4.7 Stable Expression of Ion Channels
Fig. 4.9 Detailed evaluation of ion channel stable cells lines using automated high throughput electrophysiology. HEK293 cells were transfected with an expression vector for human NaV1.7 and multiple stable clones were selected, expanded and then screened for expression of Na+ currents using IonWorks. The best expressing clone was identified and then characterised further. (A) Repre-
sentative inward Na+ current evoked by depolarisation to –10mV, showing blockade by tetrodotoxin (TTX, 300 nM). Mean peak currents (B) and distribution of peak current (C) measured from 270 cells are shown. (D) Greater than 80 % of cells gave usable seals (>80 MO) and expressed currents above background.
screening and functional validation of ion channel reagents by providing the capability for statistically analysing expression in large numbers of individual cells. This is highly advantageous for monitoring the often very subtle effects of auxiliary subunits or for carrying out detailed comparative studies, for example, of different expression constructs; vectors, host cells etc. (see Fig. 4.1, 4.8, 4.9 and 6.5). IRES-containing vectors carrying the CMV promoter to drive transcription of the bicistronic mRNA and the IRES element derived from Encephalomyocarditis virus (EMCV) are commercially available (BD Biosciences Clontech). Versions with different variants of the EMCV IRES are also available. In the original pCIN vector
99
100
4 Functional Expression of Ion Channels in Mammalian Systems
(and similar vectors, e. g. pCIN3 [84]) the efficiency of expression of the downstream neomycin-resistant selection marker is relatively low since the distance between the neomycin phosphotransferase initiation codon and the IRES element is nonoptimal (i. e. 35bp less than is found between the IRES element and the major site of initiation of the viral polyprotein in the native EMCV virus) [85]. This is likely to be advantageous for many applications because a relatively low efficiency of translation of neomycin phosphotransferase should favor the selection of clones expressing high levels of the bicistronic mRNA, therefore giving relatively high levels of the upstream ion channel gene also. However, for reasons already discussed, this is not necessarily optimal for functional expression of ion channels and an alternative vector, pCIN5, that gives lower expression is also available [85, BD Biosciences Clontech]. In this vector the initiation codon of neomycin phosphotransferase has been engineered to be in precisely the same position with respect to the IRES as in the downstream ORF of the native EMCV virus. This is likely to give more efficient expression of the selectable marker resulting in lower levels of bicistronic mRNA and correspondingly lower levels of channel expression from the upstream cistron. The pCIN5 vector has been used successfully for a variety of different channels (see Fig. 4.2, 4.4, 4.8, and 4.9 and also Refs. [7, 83–88]). 4.7.2 Stable Expression of Multiple Subunits
As well as those with different IRES variants, vectors with different selection markers are available [85, BD Biosciences Clontech] and have been used successfully for a variety of multi-subunit and multi-channel examples [87, Fig. 4.8]. It is possible to successfully co-express up to three subunits or channels using these different vectors with different selection markers for each. However, most cell types grow relatively poorly under such extreme selection pressure in the presence of three antibiotics. Thus, for channels containing more than three subunits, and also where channel expression itself has deleterious effects on host cells, a different strategy is sometimes required. One possibility is to link two of the required subunits on the same expression vector, either as separate expression cassettes or via an IRES element. As previously discussed, a concern with using two separate expression cassettes on the same vector is that expression of the two subunits could become segregated during cell line selection or maintenance. Similarly, a concern with linking the two subunits via an IRES element is that, unless arranged in a tricistronic configuration (see below), the selection marker must be expressed as a separate expression cassette, again with the risk that it can become separated from expression of the channel subunits. Another potential problem with the latter approach is that, because expression of cDNAs downstream of the IRES element is considerably lower than that of cDNAs positioned upstream, the stoichiometry of subunits expressed may be inappropriate. Nevertheless, this system has been used successfully for functional expression of the P2X2/3 channel [89]. Another application where this arrangement has been successful is a variation where the downstream cistron is a reporter that can be used for fluorescence-
4.7 Stable Expression of Ion Channels
activated cell sorting (FACS), e. g. green fluorescent protein (GFP). In this case, GFP fluorescence can be used as a more convenient surrogate for monitoring expression of the upstream gene of interest and for selecting a subpopulation of transfected cells that are enriched for expression [90]. Obtaining an acceptable stoichiometry of subunits is also a consideration when using tricistronic expression constructs. Since this arrangement involves the expression of three different cDNAs (e. g. two channel subunits plus selection marker) linked using two IRES elements, the expression of the cDNA at the 3'end will be very low indeed. Stability of expression with tricistronic constructs is also a potential issue due to intrinsic instability of such lengthy transcripts, especially for larger channel subunits e. g. NaV or CaV a subunits. In addition, unless IRES elements from different sources (i. e. having different sequences) are used, there is also the potential for deletions and rearrangements to occur by homologous recombination. Despite these caveats there are reported examples of successful expression using tricistronic constructs [85, 89, 91]. Another interesting strategy for multi-subunit channels that may be applicable to some channel types is the expression of tandemly concatenated subunits. This approach has been shown to be feasible in a number of instances [92] and is particularly useful for investigating or fixing the stoichiometry and assembly of complex channels, since covalently linking subunits predetermines their composition and arrangement in the mature protein. 4.7.3 Inducible Expression
While the use of IRES vectors has proven effective for the selection of stable cell lines for a wide variety of ion channel types, sometimes expression of the channel can be so cytotoxic that the growth rate of antibiotic resistant clones is prohibitively slow or, in extreme cases, no clones actually survive antibiotic selection. As reviewed above, one option is to use transient expression to address this problem but, if stable expression is required, an alternative strategy is to use inducible expression. As with transient expression, inducible systems offer the possibility of regulating the level of channel expression, in this case by titration of the inducer. Several inducible different systems have been described, but probably those most commonly used for ion channels make use of the bacterial tetracycline resistance operon. Alternative versions are available depending on whether regulation occurs via a derepression or transactivation mechanism. In the original system, expression of the gene of interest is driven by a mini CMV promoter containing a tet-repressor binding site [93, BD Biosciences Clontech]. This is activated by a fusion protein which is a hybrid between the tet repressor and the activation domain of the VP16 protein from Herpes Simplex virus. When tetracycline is added this hybrid tet/VP16 repressor protein no longer binds the promoter and no expression of the gene of interest can occur. This “Tet-off ” system has been used for several ion channels including NMDA-NR2 a [94], NaV1.7 and NaV1.8 [95]. A “Tet-on” variation of this has been developed which uses a mutant tet repressor/VP16 hybrid
101
102
4 Functional Expression of Ion Channels in Mammalian Systems
activator that binds to the promoter only when tetracycline is present [96]. An ion channel example using this system is NR2 b [97]. The other tetracycline regulated system (“T-REx”, Invitrogen) uses a wild-type tet repressor protein (i. e. not fused to VP16) which normally binds to tet operator sequences within the CMV promoter silencing expression of the downstream gene of interest. When tetracycline is added the repressor protein no longer binds and expression of the gene of interest is derepressed [98]. A potential disadvantage of this derepression approach, compared to transactivation, is that production of the tet repressor needs to be very efficient in order to effectively silence the powerful CMV promoter. Thus, basal levels of expression can be relatively high, which may be a problem for highly toxic channels. Nevertheless, this system has been successfully used for several ion channels including TrpM2, CaV3.2 and Kv2.1 [99–101]. On the other hand the level of expression when fully induced is higher with the derepression system since this uses a stronger CMV promoter which does not rely on the activity of a hybrid viral transactivator. Other types of inducible system that have been used for ion channels involve the use of transactivating steroid hormone receptors. One of these is based on the insect hormone, ecdysone, and uses a heteromeric receptor that is functional in mammalian cells [102]. This receptor is a heteromer between the mammalian retinoid X receptor (RXR) and the insect ecdysone receptor (EcR) that binds to an engineered promoter containing specific recognition sequences and tightly represses
Fig. 4.10 Generation and analysis of stable clones with inducible channel expression. Expression screening of 16 HERG-GeneSwitch clones by flow cytometry using a HERG-specific antibody. Clones were generated by transfection of a pGENE-HERG vector into HEK293 cells expressing the GeneSwitch regulatory protein (pSwitch) and selection with zeocin. For each clone expression in both
non-inducing (filled bars) and inducing conditions (mifepristone added, open bars) is shown. Clones 9 and 16 showed substantial increases in HERG immunoreactivity following induction, though the induced level of expression was lower than that seen in 4 representative clones generated with an expression vector that uses the CMV promoter (pCIN5, lanes shown at far left).
Acknowledgements
expression of the downstream gene. In the presence of synthetic ecdysone analogue inducers (muristerone A or ponasterone A) the conformation of the receptor is altered and transactivation of the downstream gene then occurs. A similar system, “GeneSwitch” (Invitrogen), uses a receptor that is a hybrid between the DNA binding domain from the yeast Gal4 transactivator, the ligand binding domain from the human progesterone receptor and the activation domain from the NFkb transcription factor (p65AD). This binds to an engineered GAL4-adenovirus E1 b promoter upstream of the gene of interest. In the absence of the inducer, mifepristone, the hybrid GeneSwitch receptor represses the promoter but when mifepristone is added it binds to the receptor and induces a conformational change that causes activation of expression. An example of an ion channel expressed using this system is shown in Fig. 4.10. Both the ecdysone and the GeneSwitch systems should give relatively low basal expression levels since the receptors are able to repress promoter activity in the absence of inducer, so may be suitable for the expression of highly toxic channels. For example, the ecdysone system has been used to express the cytotoxic NMDA-NR2 a and NR2 b receptors [103, 104]. However, the fully induced level of expression may not be as high as with vectors that use the CMV promoter (see Fig. 4.10).
4.8 Summary
In conclusion, this chapter has attempted to highlight some of the many potential difficulties that functional expression of heterologous ion channels in mammalian cells can present. These challenges can be magnified even further within a drug discovery setting, due to the stringent demands for robust, highly reproducible expression over a long term and on a large scale. Nevertheless, as has been outlined, a variety of approaches to address these problems are available, each with their own set of advantages and disadvantages. The choice of expression system will ultimately be governed by the nature of the channel of interest and by the final assay format that is required. However, it is often not possible to predict with any certainty how successful a particular system will be and in many cases, particularly for more novel channels, a combination of approaches may be necessary. Thus, it is often worth exploring several different strategies in parallel in order to develop a final expression reagent that is most appropriate for the particular channel-assay combination(s) required.
Acknowledgements
The author would like to acknowledge numerous colleagues at GSK who have given support and input, particularly Mark Chen, Yuhua Chen, Pat Condreay, Dave Grose, Bruce Hamilton, Andy Powell, Andy Randall, Steve Rees, Mike Romanos, Derek Trezise and Simon Tate.
103
104
4 Functional Expression of Ion Channels in Mammalian Systems
References 1 G. Seebohm, C. Lerche, A. E Busch, A. Bachmann, Dependence of I(Ks) biophysical properties on the expression system, Pflugers Arch. 2001, 442, 891–895. 2 A. Shcherbatko, F. Ono, G. Mandel, P. Brehm,Voltage-dependent sodium channel function is regulated through membrane mechanics, Biophys. J. 1999, 77, 1945–1959. 3 H. Witchel, J. T. Milnes, J. S. Mitcheson, J. C. Hancox, Troubleshooting problems with in vitro screening of drugs for QT interval prolongation usung HERG K+ channels expressed in mammalian cell lines and Xenopus oocytes, J. Pharmacol. Toxicol. Methods 2002, 48, 65– 80. 4 M. Kozak, The scanning model for translation: an update, J. Cell. Biol. 1989, 108, 229–241. 5 P. H. Lalik, D. S. Krafte,W. A. Volberg, R. B. Ciccarelli, Characterisation of endogenous sodium channel gene expressed in Chinese hamster ovary cells, Am. J. Physiol. 1993, 264, C803–C809. 6 G. J. Stephens, K. M. Page, J. R. Burley, N. S. Berrow, A. C. Dolphin, Functional expression of rat brain cloned a1 e calcium channels in COS7 cells, Pflugers Arch. 1997, 433, 523–532. 7 V. H. John et al., Heterologus expression and functional analysis of rat NaV1.8 (SNS) Voltage-gated sodium channels in the dorsal root ganglion neuroblastoma cell line ND7–23, Neuropharmacol 2004, 46, 425–438. 8 J. Chemin, A. Monteil, S. Dubel, J. Nargeot, P. Lory, The alpha1I T-type calcium channel exhibits faster gating properties when overexpressed in neuroblastoma/glioma NG 108–15 cells, Eur. J. Neurosci. 2001, 14, 1678–86. 9 T. Ljungstrom, M. Grunnet, B. S. Jensen, S. P. Olesen, Functional coupling between heterologously expressed dopamine D(2) receptors and KCNQ channels, Pflugers Arch. 2003, 446, 684–94. 10 V. J. Dupriez, K. Maes, E. Le Poul, E. Burgeon, M. Detheux, Aequorinbased functional assays for G-proteincoupled receptors, ion channels, and
11
12 13
14 15
16
17
18
19
20
21
tyrosine kinase receptors, Receptors Channels 2002, 8, 319–30. L. J. V. Galietta, M. F. Springsteel, E. Masahiro, E. J. Niedzinski, K. By, M. J. Haddadin, M. J. Kurth, M. H. Nantz, A. S. Verkman, Novel CFTR Chloride Channel Activators Identified by Screening of Combinatorial Libraries Based on Flavone and Benzoquino.lizinium Lead Compounds, J. Biol. Chem. 2001, 276, 19723–19728. C. Deutsch, The birth of a channel, Neuron 2003, 40, 265–276. J. Johnston, C. L. Ward, R. R. Kopito, Aggresomes: A cellular response to misfolded proteins, J. Cell Biol. 1999, 143, 1883–1898. C. M. Dobson, Protein folding and misfolding, Nature 2003, 426, 884–890 A. L. Goldberg, Protein degradation and protection against misfolded or damaged proteins, Nature 2003, 426, 895–899. M. K. Higgins, M. Demir, C. G. Tate, Calnexin co-expression and the use of weaker promoters increase expression of correctly assembled Shaker potassium channel in insect cells, Biochim. Biophys. Acta 2003, 1610, 124–132. J. I. Kourie, C. L. Henry, Ion channel formation and membrane linked pathologies of misfolded hydrophobic proteins: the role of dangerous unchaperoned molecules, Clin. Exp. Pharmacol. Physiol. 2002, 29, 741–753. L. N. Manganas, J. S Trimmer, Calnexin regulates mammalian Kv1 channel trafficking, Biochem. Biophys. Res. Commun. 2004, 322, 577–584. W. Chang, M. S Gelman, J. M Prives, Calnexin-dependent Enhancement of Nicotinic Acetylcholine Receptor Assembly and Surface Expression, J. Biol. Chem. 1997, 46, 28925–28932. S. H. Keller, J. Lindstrom, P. Taylor, Involvement of the chaperone protein calnexin and the acetylocholine receptor beta-subunit in the assembly and cell surface expression of the receptor, J. Biol. Chem. 1996, 37, 22871–22877. E. Ficker, A. T Dennis, L. Wang, A. M Brown, Role of the Cytosolic Chaperones Hsp70 and Hsp90 in Maturation of
References
22 23
24
25
26
27
28
29
30
31
32
the Cardiac Potassium Channel hERG, Circ. Res. AHA 2003, 92, e87–e100. M. D. Amaral, CFTR and Chaperones, J. Mol. Neurosci. 2003, 23, 0895–8696. E. K. Wittmack, A. M Rush, M. J Craner, M. Goldfarb, S. G Waxman, S. D Dib-Hajj, Fibroblast growth factor homologous factor 2B: association with NaV1.6 and selective colocalization at nodes of Ranvier of dorsal root axons, J. Neurosci. 2004, 30, 6765–6775. C. Altier et al., Trafficking of L-type Calcium Channels Mediated by the Postsynaptic Scaffolding Protein AKAP79, J. Biol. Chem. 2002, 277(37), 33598– 33603. M. Köttgen et al., Trafficking of TRPP2 by PACS proteins represents a novel mechanism of ion channel regulation, EMBO J. 2005, 24(4), 705–716. K. Okuse, M. Malik-Hall, M. D Baker, W.Y Poon, H. Kong, M. V. Chao, J. N. Wood, Annexin II light chain regulates sensory neuron-specific sodium channel expression, Nature 2002, 6, 417(6889), 653–656. Y. A. Kuryshev, T. I Gudz, A. M Brown, B. A Wible, KChAP as a chaperone for specific K (+) channels, Am. J. Physiol. Cell. Physiol. 2000, 278(5), C863–C864. O. T. Pongs et al., Functional and molecular aspects of voltage-gated K + channel beta subunits, Ann. N.Y. Acad. Sci. 1999, 868, 344–355. L. L. Isom, Sodium channel beta subunits: anything but auxiliary, Neuroscientist 2001, 1, 42–54. M. W. Richards, A. J. Butcher, A. C. Dolphin, Ca2+ channel ß-subunits: structural insights AID our understanding, Trends Pharm. Sci. 2005, 12, 0165– 6147. N. Klugbauer, E. Marais, F. Hofmann, Calcium channel alpha2delta subinuts: differential expression, function, and drug binding, J. Bioenerg. Biomembr. 2003, 35(6), 639–647. W. J. Joiner, R. Khanna, L. C. Schlichter, L. K. Kaczmarek, Calmodulin Regulates Assembly and Trafficking of SK4/ IK1 Ca2+-activated K+ Channels, J. Biol. Chem. 2001, 276(41), 37980–37985.
33 W-S. Lee, T. J Ngo-Anh, A. BrueningWright, A. Maylie, J. P. Adelman, Small Conductance Ca2-activated K+ Channels and Calmodulin, J. Biol. Chem. 2003, 278(28), 25940–25946. 34 W. Vandenberghe, R. A. Nicoll, D. S. Bredt, Stargazin is an AMPA receptor auxiliary subunit, PNAS. 2005, 102(2), 485–490. 35 D. Ma, N. Zerangue,Y-F. Lin, A. Collins, M. Yu,Y. N. Jan, L. Y. Jan, Role of ER Export Signals in Controlling Surface Potassium Channel Numbers, Science 2001, 291. 36 M. M. Zarei, M. Eghbali, A. Alioua, M. Song, H. G. Knaus, E. Stefani, L. Toro, An endoplasmic reticulum trafficking signal prevents surface expression of a Voltage- and Ca2(+)- activated K+ channel splice variant, Proc. Natl. Acad. Sci. USA 2004, 101(27), 10072– 10077. 37 R. Roncarti, I. Decimo, G Fumagalli, Assembly and trafficking of human small conductance Ca2(+)- activated K+ channel SK3 are governed by different molecular domains, Mol. Cell. Neurosci. 2005, 28(2), 314–325. 38 H. M. Jones, K. L. Hamilton, G. D. Papworth, C. A. Syme, S. C. Watkins, N. A. Bradbury, D. C. Devor, Role of the NH2 terminus in the assembly and trafficking of the intermediate conductance Ca2(+)activated K+ channel hIK1, J. Biol. Chem. 2004, 279(15),15531–15540. 39 D. Li, K. Takimoto, E. S. Levitan, Surface expression of Kv1 channels is governed by a C-terminal motif, J. Biol. Chem. 2000, 275(16), 11597–11602. 40 M. Jenke, A. Sánchez, F. Monje, W. Stühmer, R. M. Weseloh, L. A. Pardo, C-terminal domains implicated in the functional surface expression of potassium channels, EMBO 2003, 22(3), 395–403. 41 A. Akhavan, R. Atanasiu, A. Shrier, Identification of a COOH-terminal segment involved maturation and stability of human ether-a-go-go-related gene potassium channels, J. Biol. Chem. 2003, 278(41), 40105–40112. 42 D. B. Scott, I. Michailidis,Y. Mu, D. Logothetis, M. D. Ehlers, Endocyto-
105
106
4 Functional Expression of Ion Channels in Mammalian Systems
43
44
45
46
47
48
49
50
51
sis and Degradative Sorting of NDMA Receptors by Conserved Membrane-Proximal Signals, J. Neurosci. 2004, 24(32), 706–7109. Z. Ren, N. J. Riley, E. P. Garcia, J. M. Sanders, G. T. Swanson, J. Marshall, Multiple Trafficking Signals Regulate Kainate Receptor KA2 Subunit Surface Expression, J. Neurosci. 2003, 23(16), 6608–6616. J-M.Wang, L. Zhang,Y. Yao, N. Viroonchatapan, E. Rothe, Z-Z. Wang, A transmembrane motif governs the surface trafficking of nicotinic acetylcholine receptors, Nature 2002, 5, No. 10 10.1038/nn198. G. M. Denning, M. P. Anderson, J. F. Amara, J. Marshall, A. E. Smith, M. J. Welsh, Processing of mutant cystic fibrosis transmembrane conductance regulator is temperature-sensitive, Nature 1992, 27,358(6389), 761–764. L. C. Denlinger et al., Mutation of a Dibasic Amino Acid Motif Within the C Terminus if the P2X7 Nucleotide Receptor Results in Trafficking Defects and Impaired Function, J. Immunol. 2003, 171, 1304–1311. Z. Zhou, Q. Gong, C. T. January, Correction of Defective Protein Trafficking of a Mutant hERG Potassium Channel in Human Long QT Syndrome, J. Biol. Chem. 1999, 274(44), 31123–31126. A. Paulussen, A. Raes, G. Matthijs, D. J. Snyders, N. Cohen, J. Aerssens, A Novel Mutation (T65P) in the PAS Domain of the Human Potassium Channel hERG Results in the Long QT Syndrome by Trafficking Deficiency, J. Biol. Chem. 2002, 277(50), 48610–48616. C. R. Valdivia, M. J. Ackerman, D. J. Tester, T. Wada, J. McCormack, B. Ye, J. C. Makielski, A novel SCN5A arrhythmia mutation, M1766L, with expression defect rescued by mexiletine, Cardiovasc. Res. 2002, 55(2), 279–289. M. Brejon, S. Le Maout, P. A. Welling, J. Merot, Processing and Transport of ROMK1 Channel is Temperature-Sensitive, Biochem. Biophy. Res. Commun. 1999, 261, 364–371. K. Schroeder, J. Wu, L. Zhao, R. J. Lukas, Regulation by cycloheximide and
52
53
54
55
56
57
58
59
60
lowered temperature of cell-surface a7nicotinic acetylcholine receptor expression on transfected SH-EP1 cells, J. Neurochem. 2003, 85, 581–591. M. E. F. Nelson, F. Wang, A. Kuryatov, C. H Choi,V. Gerzanich, J. Lindstrom, Functional Properties of Human Nicotinic AChRs Expressed by IMR-32 Neuroblastoma Cells Resemble Thise of a3â4 AChRs Expressed in Permanently Transfected HEK Cells, J. Gen. Physiol. 2001, 118, 563–582. S. T. Cooper, P. C. Harkness, E. R. Baker, N. S. Millar, Up-regulation of Cell-surface a4b2 Neuronal Nicotinic Receptors by Lower Temperature and Expression of Chimeric Subunits, J. Biol. Chem. 1999, 17, 27145–27152. M. E. Nelson, A. Kuryatov, C. H. Choi, Y. Zhou, J. Lindstrom, Alternate Stoichiometries of a4b2 Nicotinic Acetylcholine Receptors, Mol. Pharm. 2003, 63(2), 2035–1038020. I. Szabò, A. Negro, P. M. Downey, M. Zoratti, F. I. Schiavo, G. M. Giacometti, Temperature-Dependent Functional Expression of a Plant K+ Channel in Mammalian Cells, Biochem. Biophys. Res. Commun, 2000, 274, 130–135. K. Du , M. Sharma, G. L. Lukas, The DeltaF508 cystic fibrosis mutation impairs domain-domain interactions and arrests post-translational folding of CFTR, Nat. Struct. Mol. Biol. 2005, 12(1), 17–25. P. H. Thibodeau, C. A. Brautigam, M. Machius, P. J. Thomas, Side chain and backbone contributions of Phe508 to CFTR folding, Nat. Struct. Mol. Biol. 2005, 12(1), 10–6. M. Gentzsch et al., Endocytic Trafficking Routes of Wild Type and ÄF508 Cystic Fibrosis Transmembrane Conductance Regulator, Mol. Biol. Cell 2004, 15, 2684– 2696. N. J. Anegawa, D. R. Lynch, T. A. Verdoorn, D. B. Pritchett. Transfection of N-methyl-D-aspartate receptors in a nonneuronal cell line leads to cell death, J. Neurochem. 1995, 64(5), 2004–2012. N. J. Anegawa, R. P. Guttman, E. R. Grant, R. Anand, J. Lindstrom, D. R. Lynch, N-Methyl-D-aspartate receptor
References
61
62
63
64
65
66
67
68
69
70
mediated toxicity in non-neuronal cell lines: characterization using fluorescent measures of cell viability and reactive oxygen species production, Brain Res. Mol. Brain. Res. 2000, 77(2), 163–175. M. Raisinghani, L. S. Premkumar, Block of native and cloned vanilloid receptor (TRPVI) by aminoglycoside antibiotics, J.Pain 2004, 0304–3959. D. Dobrev, U. Ravens, Therapeutically relevant concentrations of neomycin selectively inhibit P-type Ca2+ channels in rat striatum, Eur. J. Pharmacol. 2003, 461(2–3), 105–11. T. Masuko, T. Kuno, K. Kashiwagi, T. Kusama, K. Williams, K. Igarashi, Stimulatory and Inhibitory Properties of Aminoglycoside Antibiotics at N-MethylD-Asparate Receptors, J. Pharm. Exp. Ther. 1999, 290(3),1026–1033. A. Colosimo et al., Transfer and expression of foreign genes in mammalian cells, Biotechniques 2000, 2, 314–318, 320–322, 324 passim. C. Chen, H. Okayama, High-efficiency transformation of mammalian cells by plasmid DNA, Mol. Cell Biol. 1987, 7, 2745–2752. J. Gehl, Electroporation: theory and methods, perspectives for drug delivery, gene therapy and research, Acta. Physiol. Scand. 2003, 177(4), 437–447. RE. B. DuBridge, P. Tang, H. C. Hsia, P. M. Leong, J. H. Miller, M. P. Calos, Analysis of mutation in human cells by using an Epstein-Barr virus shuttle system, Mol. Cell. Biol. 1987, 1, 379–387. P. Mellon,V. Parker,Y. Gluzman, T. Maniatis, Identification of DNA sequences required for transcription of the human alpha 1-globin gene in a new SV40 host-vector system, Cell. 1981, 2(1), 279–288. C. R. Sclimenti, M. P. Calos, EpsteinBarr virus vectors for gene expression and transfer, Curr. Opin. Biotechnol. 1998, 9, 476–479. B. Sugden, N. Warren, A promoter of Epstein-Barr virus that can function during latent infection can be transactivated by EBNA-1, a viral protein required for viral DNA replication during latent infection, J. Virol. 1989, 63(6), 2644–2649.
71 Y. Durocher, S. Perret, A. Kamen, High-level and high-throughput recombinant protein production by transient transfection of suspension-growing human 293-EBNA1 cells, Nucleic Acids Res. 2001, 30(2), e9. 72 P. Liljeström, H. Garoff, A New Generation of Animal Cell Expression Vectors Based on the Forest Vorus Replicon, Bio/ Tech. 1991, 9. 73 K. Lundstrom, Semliki Forest virus vectors for rapid and high-level expression of integral membrane proteins, Biochim. Biophys. Acta. 2003, 90–96. 74 K. Lundstrom, A. Michel, H. Blasey, A. R. Bernard, R. Hovius, H. Vogel, Surprenant A. Expression of ligand-gated ion channels with the Semliki Forest virus expression system, J. Recept. Signal Transduct Res. 1997, 17(1–3), 115–126. 75 G. H. Gorrie,Y. Vallis, A. Stephenson, J. Whitfield, B. Browning, T. G. Smart, S. J. Moss, Assembly of GABBA receptors composed of alpha1 and beta2 subunits in both cultured neurons and fibroblasts, J. Neurosci. 1997, 17(17), 6587– 6596. 76 O. Shamotienko, S. Akhtar, C. Sidera, F. A. Meunier, B. Ink, M. Weir, J. O. Dolly, Recreation of Neuronal Kv1 Channel Oligomers by Expression in Mammalian Cells using Semliki Forest Virus, Biochemistry. 1999, 38, 16766–16776. 77 T. A. Kost, J. P. Condreay, Recombinant baculoviruses as mammalian cell genedelivery vectors, Trends Biotechnol. 2002, 20(4). 78 J. P. Condreay, S. M.Witherspoon, W. C. Clay , T. A. Kost, Transient and stable gene expression in mammalian cells transduced with a recombinant baculovirus vector, Proc. Natl. Acad. Sci. USA. 1999, 96, 127–132. 79 W. C. Clay, J. P. Condreay, L. B. Moore, S. L. Weaver, M. A.Watson, T. A. Kost, J. J. Lorenz, Recombinant baculoviruses used to study estrogen receptor function in human osteosarcoma cells, Assay Drug Dev. Technol. 2003, 1(6), 801–810. 80 R. Ames, P. Nuthulaganti, J. Fornwald, U. Shabon, H. van-der-Keyl, N. Elshourbagy, Heterologous expression of G protein-coupled receptors in
107
108
4 Functional Expression of Ion Channels in Mammalian Systems
81
82
83
84
85 86
87
88
89
U-2 OS osteosarcoma cells, Receptors Channels. 2004, 10(3–4), 117–124. J. L. Pfohl, J. F. Worley 3rd, J. P. Condreay, G. An, C. J. Apolito, T. A. Kost, J. F. Truax, Titration of KATP channel expression in mammalian cells utilizing recombinant baculovirus transduction, Receptors Channels. 2000, 8(2), 99–111. R. Ames et al.. BacMam recombinant baculoviruses in G protein-coupled receptor drug discovery, Receptors Channels. 2004, 10(3–4),99–107. S. K. Jang, H. G. Krausslich, M. J. Nicklin, G. M. Duke, A. C. Palmenberg, E. Wimmer, A segment of the 5' nontranslated region of encephalomyocarditis virus RNA directs internal entry of ribosomes during in vitro translation, J. Virol. 1988, 62(8), 2636–2643. S. Rees, J. Coote, J. Stables, S. Goodson, S. Harris, M. G. Lee, Bicistronic Vector for the Creation of Stable Mammalian Cell Lines that Predisposes all Antibiotic-Resistant Cells to Express Recombinant Protein, Biotechniques 1996, 20(1), 102–110. S. Rees. Y-H. Chen, T. J. Dale, M. A. Romanos, W. R. J. Whitaker, X. M. Xie, J. J. Clare, Cloning, distribution and functional analysis of the type III sodium channel from human brain, Eur. J. Neurosci. 2000, 12, 4281–4289. M. J. Main, J. E. Cryan, J. R. B. Dupere, B. Cox, J. J. Clare, S. A. Burbidge, Modulation of KCNQ2/3 Potassium Channels by the Novel Anticonvulsant Retigabine, Mol. Pharm. 2000, 58, 253– 262. S. A. Burbidge, T. J. Dale, A. J. Powell, W. R. J. Whittaker, X. M. Xie, M. A. Romanos, J. J. Clare, Molecular cloning, distribution and functional analysis of the NaV1.6. Vol.tage-gated sodium channel from human brain, Mol. Brain. Res. 2002, 103, 80–90. V. H. John et al., Heterologous expression and functional analysis of rat NaV1.8 (SNS) voltage-gated sodium channels in the dorsal root ganglion neurobastoma cell line ND7–23, NeuroPharm. 2004, 46, 425–438.
90 E. Kawashima et al., A novel and efficient method for the stable expression of heteromeric ion channels in mammalian cells, Receptors Channels 1998, 5(2), 53– 60. 91 D. A. Persons et al., Enforced expression of the GATA-2 transcription factor blocks normal hematopoiesis, Blood 1999, 93(2), 488–499. 92 J. Zhu, M. L. Musco, M. J. Grace, Three-color flow cytomentry analysis of tricistronic expression of eBFP, eGFP, and eYFP using EMCV-IRES linkages, Cytometry 1999, 37(1), 51–59. 93 F. Minier, E. Sigel, Techniques: Use of concatenated subunits for the study of ligand-gated ion channels, Trends Pharm. Sci. 2004, 25(9). 94 M. Gossen, H. Bujard, Tight control of gene expression in mammalian cells by tetracycline-responsive promoters, Proc. Natl. Acad. Sci. USA 1992, 89, 5547– 5551. 95 S. Renard, C. Drouet-Pétré, M. Partiseti, S. Z. Langer, D. Graham, F. Besnard, Development of an inducible NMDA receptor stable cell line with an intracellular Ca2+ reporter, Eur. J. Pharm. 1999, 366, 319–328. 96 I. Akibu et al., Stable Expression and Characterization of Human PN1 and PN3 Sodium Channels, Receptors Channels 2003, 9, 291–299. 97 M. Gossen, S. Freundlieb, G. Bender, G. Muller, W. Hillen, H. Bujard, Transcriptional activation by tetracyclines in mammalian cells, Science 1995, 268(5218), 1766–1769. 98 A. Rossignoli, M.G. Giribaldi, S Corazza, Establishment of a CHO cell line expressing the NMDA NR1a/NR2 b subunits for functional analysis of receptor ligands, presented at The Society for Biomolecular Screening Conference, 2002. 99 F. Yao, T. Svensjo, T. Winkler, M. Lu, C. Eriksson, E. Eriksson, Tetracycline repressor, tetR, rather than the tetRmammalian cell transcription factor fusion derivatives, regulates inducible gene expression in mammalian cells, Hum. Gene Ther. 1998, 9(13), 1939–1950. 100 A-L. Perraud et al., ADP-ribose gating of the calcium-permeable LTRPC2 chan-
References nel revealed by Nudix motif homology, Nature 2001, 411. 101 M. Xia et al., Generation and Characterization of a Cell Line with Inducible Expression of CaV3.2 (T-Type) Channels, Assay Drug. Dev. Tech. 2003, 1(5). 102 J. G. Trapani, S. J. Korn, Control of ion channel expression for patch clamp recordings using an inducible expression system in mammalian cell lines, BMC Neurosci. 2003, 4, 15. 103 D. No, T. P. Yao, R. M. Evans, Ecdysoneinducible gene expression in mamma-
lian cells and transgenic mice, Proc. Natl. Acad. Sci. USA. 1996, 93(8), 3346–3351. 104 D. Kurkó, P. Dezsõ , A.Boros, S. Kolok, L. Fodor, J. Nagy, Z Szombathelyi, Inducible expression and pharmacological characterization of recombinant rat NR1a/NR2A NMDA receptors, Neurochem. Int. 2005, 46, 369–379. 105 J. Nagy, A. Boros, P. Dezsõ, S. Kolok, L. Fodor, Inducible expresión and pharmacology of recombinant NMDA receptors, composed of rat NR1a/NR2B subunits, Neurochem. Int. 2003, 43, 19–29.
109
111
5 Analysis of Electrophysiological Data Michael Pusch
5.1 Overview
This chapter outlines electrophysiological methods for extracting biophysical parameters that describe two fundamental properties of ion channels: gating and permeation. The Introduction provides a broad overview of the general concepts of ion channel biophysics and the text a review of the kind of information that can be extracted from electrophysiological recordings. The later sections introduce several methods for the analysis of electrophysiological experiments on heterologously expressed ion channels. Many parts are explicit and can be directly applied “at the bench”. Other, more advanced topics (gating current measurements, single-channel kinetic analysis) are touched upon only superficially since their application requires further background that can be found in the references. A number of simple and useful mathematical equations are included which should assist the reader in the design, execution, and analysis of electrophysiological experiments. Readers interested in further theoretical background to the concepts outlined in this chapter are referred to Ref. [1] and references therein.
5.2 Introduction
The function of ion channels is to rapidly pass – in a passive but selective manner –a large number of ions across biological membranes. This electrogenicity is exploited by excitable cells to quickly change the transmembrane voltage allowing, for example, the conduction of the action potential and the postsynaptic electrical response to chemical neurotransmission. Other cells and organelles exploit the large capacity of charge transport for ion homeostasis and transepithelial transport. For the researcher the electrogenicity is interesting and useful because it allows the measurement of ion channel functioning in “real-time”. It is possible to monitor the action of ion channels both in vivo or in appropriate simplified in vitro systems like brain slices. Intracellular electrodes directly measure the membrane Expression and Analysis of Recombinant Ion Channels. Edited by Jeffrey J. Clare and Derek J. Trezise Copyright # 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim ISBN: 3-527-31209-9
112
5 Analysis of Electrophysiological Data
voltage of individual cells while extracellular electrodes monitor cellular electrical activity indirectly. However, the recording of the physiological voltage provides little information regarding the biophysical parameters of the underlying channels, for two main reasons. First, all cells possess a complement of different ion channels and other electrogenic transporters that make it difficult to tease apart their respective contribution to the electrical response. Second, the physiological recordings are current-clamp measurements in which the membrane voltage is “freely floating” and the concentration of neurotransmitters or other ligands is uncontrolled. In order to reliably define a physical parameter of an ion channel (ligand affinity, slope of the voltage-dependence etc.) these key variables have to be fixed, or “clamped”. A further important step is to isolate, as far as possible, a single type of ion channel. In physiological preparations this can be partially achieved using appropriate solutions and blocking agents. Heterologous systems are even better in this respect, even if they carry the risk that some physiologically important molecular component is missing. From the perspective described above the application and interpretation of the voltage-clamp measurements by Hodgkin and Huxley [2] revolutionized the approach to ion channel analysis. Today’s description and interpretation of ion channel function still draws heavily on the principles embodied in this work. One of the most important concepts for ion channels is the “gate”. A gate can be either open or shut (closed). There is no intermediate, half-open gate. At the time of Hodgkin and Huxley, there was no direct evidence for this as practically nothing was known at the level of the single channel. The assumption of a simple open or closed gate allowed a convenient description, as a two-state Markov process associated with a linear differential equation at a fixed voltage or ligand concentration. For the classical “m-gate”, the activation gate of the voltage-gated Na+ channel, this equation reads dm
t dt
m
t
1
m
t
where m(t) is the probability of the gate being open at time t. The opening and closing rate constants, a and b, respectively, are voltage-dependent and represent a model for the underlying molecular rearrangement leading to gate opening. A verification of the open–closed dichotomy of most ion channels became possible with the patch clamp technique [3, 4] that allowed the real-time visualization of single channel opening and closing in almost all types of animal, plant, and bacterial cells (for simplicity here, the existence of “subconductance” states, “flickering”, and other probably ubiquitous complicated single-channel behavior is ignored). Of course, the description of gating as a two-state process is an idealization, opening is not instantaneous. Also, microscopically, a single “open” state does not exist, but rather an almost infinite number of possible molecular configurations that are macroscopically lumped together into “the open” state, because functionally, they are almost indistinguishable from each other. The transitions from closed to open (and vice versa) appear “instantaneously” on single channel
5.3 Expression Systems and Related Recording Techniques
recordings, but in reality take several 100 ns. However, despite the availability of several ion channel structures (see for example Refs. [5–7]) and large computing power it is not yet possible to explore channel gating by molecular dynamics. Even ion permeation that occurs on a much faster time scale cannot be fully simulated, even though some theoretical progress has been made, especially for K+ channels (see for example Ref. [8] and Chapter 10). Quantitative functional measurements are still essential for a detailed insight into the mechanisms of ion channels. It can be expected that many more structural data for ion channels will be available in the near future. The structures will guide computational studies and rational mutagenesis in order to understand the mechanisms of function at a molecular model, to obtain high affinity ligands, and possibly to exploit ion channels as molecular devices in applied technological systems. Computational predictions and structure-based hypotheses have to be tested experimentally with functional data. The present chapter aims to provide an aid to designing, analyzing and interpreting such measurements.
5.3 Expression Systems and Related Recording Techniques
Each type of heterologous expression system determines a range of possible recording techniques. The most popular expression systems are Xenopus oocytes and mammalian cell lines, like HEK293 or CHO cells. A more rarely used system is the incorporation of relatively crude vesicles or purified proteins into planar lipid bilayers. 5.3.1 Expression in Xenopus Oocytes
The expression in Xenopus oocytes represents an extremely versatile system that allows the application of many different electrophysiological and biochemical methods [see Chapter 1 and Refs. 9–11]. Normally, in vitro transcribed cRNA is microinjected but expression can also be achieved using nuclear injection of eukaryotic expression plasmids. The oocyte system is popular because electrophysiological recordings can be easily performed by nonexperts employing the two-electrode voltage-clamp technique (TEV) [9]. This method allows a relatively high throughput compared to patch-clamp techniques and is thus often used, for example, for drug screening. A commonly underestimated problem of the TEV technique that is relevant also for qualitative measurements concerns the error introduced by the so-called series resistance (see for example Ref. [12]). The series resistance is caused by a finite conductivity of the oocyte cytoplasm, leading to a voltage drop within the cytoplasm and thus to a voltage error (see Fig. 5.1). Typical values of the series resistance are of the order of 0.5–1 kO. Thus a current of 10 µA will cause a voltage error of the order of 5–10 mV, a value that cannot always be neglected.
113
114
5 Analysis of Electrophysiological Data Fig. 5.1 The intracellular series resistance in Xenopus oocytes. Current flowing through the interior of the oocyte leads to a voltage drop caused by the finite resistance of the cytoplasm.
Furthermore, even when the series resistance error is accounted for, the TEV technique has a limited time resolution of the order of almost 1 ms in most realworld applications. The apparent time resolution can be enhanced but the oocyte nevertheless provides a nonideal space-clamp. “Fast” kinetic parameters that are derived from TEV measurements are therefore seldom comparable to the same parameters measured with the patch clamp technique. Another disadvantage of whole oocytes is that their cytoplasmic content cannot be controlled. This may lead to a significant variability of measurements from different oocytes if the channel properties depend on the cytosolic composition. Also one would often like to change, or at least to fix, the intracellular solution. Furthermore, in the case of large expression the intracellular ion concentrations can be significantly altered by the voltage-clamp measurements. For example it is very difficult to handle a large expression (>10 µA) of the Cl– selective muscle channel CLC-1, because its kinetics depends strongly on the intracellular Cl– concentration. The disadvantages described above are partially overcome by the “cut-open” oocyte technique [13]. With this method only a small part of the surface area of the oocyte is clamped and the intracellular solution can be exchanged. However, the method is low throughput, necessitates considerable skill, and perfusion of the interior solution is very slow. Thus, this method finds a narrow range of special applications. A general problem with the expression of Ca2+-permeable channels or channels that depend on intracellular Ca2+ is that Xenopus oocytes endogenously express a large Ca2+ dependent Cl– current, ICl– (Ca2+) [14,15]. With maximal stimulation ICl– (Ca2+) can reach several tens of µA of current. Thus activation of Ca2+ permeable channels that leads to an influx of Ca2+ will inevitably activate this endogenous current and confound the measurements. It is also practically impossible to manipulate the intracellular Ca2+ concentration in order to study its effect on expressed channels. Nevertheless, the endogenous current can be exploited as a Ca2+ sensor to test for a possible Ca2+ permeability and also to test if the activation of (expressed) receptors and/or G-proteins results in an increase in the intracellular Ca2+ concentration (see for example Ref. [16]). One other advantage of the oocyte system is that several cRNAs coding for different subunits of an ion channel or other interacting proteins can be co-injected
5.3 Expression Systems and Related Recording Techniques
at defined proportions. For example dominant heterozygous genotypes of channelopathies can be simulated by a one to one co-expression of WT and mutant subunits, and possible dominant negative effects can be quantified (see for example Ref. [17]). Co-expression of different proteins can also be achieved in transfected cells. However, with the oocyte injection it is easier to precisely control the relative expression of each protein. Finally, electrical data recorded with the TEV can be correlated for the same oocyte with the surface expression of the expressed protein, using for example an introduced extracellular epitope [18, 19]. This is of particular importance for channelopathies because many disease-causing mutations are pathogenic because they effectively reduce or enhance the plasma membrane expression of the channel (see for example Refs. [18, 20]). The patch clamp technique can also be applied to Xenopus oocytes after the vitelline membrane has been removed [10]. Recordings can be performed in the cellattached, the inside out and the outside out configuration (see Ref. [4] for a description of these methods). The electrical properties of the obtainable seal are exceptional – seal resistances >100 GO can be achieved, which allow very high resolution recordings. The size of the patch pipette range from very small to “giant” [21], allowing single-channel or macroscopic recordings. Rapid solution exchanges can be applied to excised patches allowing a precise investigation of, for example, transmitter activated channels (see for example Ref. [22]). The excellent electrical properties of the cell-free patch-clamp configuration represent a significant advantage over whole cell recordings of small cells that can suffer from limited time-resolution due to the access (series) resistance (see below). 5.3.2 Expression in Mammalian Cells
Another popular expression system is “transfected” mammalian cell lines like HEK, CHO or many others (see Chapter 4 and Ref. [23]). The expression of one or more proteins is induced by the introduction of the DNA in an appropriate eukaryotic (often mammalian) expression vector by various chemical or physical methods. Cells can be either transiently or stably transfected. Stable transfection generally requires the integration of one or more copies of the expression construct into the genome and is initially more labor intensive than transient transfection. It is, however, convenient for long term studies on a particular channel or for drug screening where large numbers of cells may be required. Many molecular biological methods exist that increase transfection efficiency and the level of expression. Expression can also be induced with several different kinds of viruses [24]. The patch-clamp technique is the method of choice for studying the function of channels expressed in these small cells. All configurations (cell attached, whole cell, inside out, outside out) can be applied but the whole cell configuration is the most straightforward and widely used. Indeed, several different technical approaches have been taken to automate the whole cell patch-clamp for high throughput drug screening [see Chapter 6 and Ref. [25]]. Several factors have to be
115
116
5 Analysis of Electrophysiological Data
considered for the analysis of whole cell data. The time resolution in voltage-jump experiments is limited by the time that is necessary to charge the membrane capacitance, Cm , across the access resistance, Ra , given by t = Cm Ra Typical values of Cm = 20 pF, Ra = 5 MO yield a charging time constant of 0.1 ms. This is adequate for most applications but can lead to problems for very fast kinetics observed for example in voltage-gated Na+ or Cl– channels [26, 27]. The access resistance leads also to an error in the voltage-reading similar to the series resistance problem in the two-electrode voltage-clamp. For a membrane current Im the voltage error amounts to DV = Im Ra which for typical values Ra = 5 MO, Im = 1 nA amounts to 5 mV and becomes worse for larger currents and/or access resistances. In voltage-jump experiments with large currents and fast kinetics the two kinds of errors described combine to create a complex dynamic error that can render certain measurements uninterpretable. Most amplifiers provide an access (series) resistance compensation that compensates both kinds of errors based on an estimate of Cm and Ra. Since the compensation involves positive feedback elements it increases the noise and is prone to oscillations. Care must therefore be taken with its application (see Ref. [28]). Similar to the oocyte system cell-free patches allow a much better voltage-clamp and also faster solution changes. However, because most mammalian cells are quite small, “macroscopic” recordings are more difficult to achieve in excised patches since large pipette diameters are poorly tolerated. 5.3.3 Leak and Capacitance Subtraction
In heterologous expression systems unwanted currents can arise either from true leak caused by the recording electrodes or from endogenous ion channels and transporters. These have to be carefully avoided using appropriate solutions and protocols. The subtraction of currents remaining after application of a specific blocker, if available, at a saturating concentration, is a very good but often tedious method. For ligand-gated channels the subtraction of currents at zero concentration of ligand is obviously a good method, because the spontaneous open probability is very small in most cases. For voltage-gated channels studied by step-protocols the responses are additionally distorted by the capacitive transients. These can be assumed to be linear, meaning that their size is proportional to the voltage step but independent of the voltage from which the pulse is delivered. Thus smaller steps applied in a voltage range where channels are closed or steps to the reversal potential can be used to subtract capacitive transients after appropriate scaling (see for example Refs. [29,30]). The most commonly used protocol for the subtrac-
5.4 Macroscopic Recordings
tion of linear leak and capacity currents when measuring voltage-gated sodium and potassium channels is called the “P/4 method” [29], that is a standard feature of most data acquisition programs. For this method four small voltage pulses with a quarter of the size of the main voltage pulse are delivered before or after the main pulse. The small pulses are subthreshold and elicit exclusively leak and capacitive currents. The response to the four quarter-sized pulses is summed and subtracted from the main response. Linear components are therefore practically completely subtracted.
5.4 Macroscopic Recordings
In the following sections it is assumed that the ion channel of interest is expressed in a heterologous system and represents the major contribution to the total membrane conductance. The methodologies to extract various biophysical parameters that are useful for a characterization of ion channels are explained. Single channel measurements, if recorded at a sufficient bandwidth, contain, in principle, more information than macroscopic ensemble measurements. However, they are significantly more technically demanding and the very small single channel currents for many ion channels renders their analysis virtually impossible. In addition, fast kinetics are difficult to measure at the single channel level because of the lower signal to noise ratio that has to be compensated by heavier filtering. Thus, for many applications macroscopic recordings represent the only practical approach. The most important relation regarding macroscopic currents is given by I=Nip
(1)
that describes the total current, I, through a homogenous population of N independent channels. A basic assumption is that the channel under investigation possesses a single open state with current level i, and is without subconductance states. Of course this is an oversimplification for many channels. Without this assumption, however, a practical interpretation of macroscopic currents is almost impossible, because it is very hard to tease apart different conductance levels of a single channel from macroscopic recordings. The parameter p represents the open probability of the channel, that is the time- and/or voltage- and/or ligand-dependent probability of the channel being in a conducting state with an associated current, i. The single parameter p in Eq. (1) summarizes the combined action of all gates of the channels. Often it is useful to think of the gates as independent devices. For example the voltage-gated Na+ channel of Hodgkin and Huxley has 3 mgates and one h-gate, all independent from each other such that the parameter p is equivalent to p = m3 h. While the independence of different gates is seldom realistic, it is very useful conceptually.
117
118
5 Analysis of Electrophysiological Data
Eq. (1) incarnates one of the dogmas of ion channel biophysics: permeation through the open channel, characterized by the parameter i, is independent of channel gating, characterized by the parameter p. This is a very useful conceptual distinction, although in real life it breaks down immediately: the occupancy of the pore by permeating ions generally stabilizes the protein structure and thereby influences gating. However, such effects are generally relatively small with some exceptions. For example, in CLC type Cl–-channels, permeation and gating are strongly coupled [31, 32]. On the other hand, gating has practically no influence on permeation through the open pore, because, by definition “open” implies an open gate. Any influence of closed gates on ion occupancy of pore binding sites vanishes rapidly after opening the gate because the two processes occur on vastly different time scales. This assumption becomes questionable only if a rapid “flicker type” gate is present that opens and closes on a time scale more similar to that of ion conduction. For example in KvLQT1 (KCNQ1) K+ channels a rapid flicker type gate seems to be present that leads to drastic effects of Rb+ ions on the macroscopic current amplitude [33]. In this case it is difficult to distinguish between an effect of Rb+ on permeation or gating because the concept of a “gate” becomes questionable. 5.4.1 Analysis of Pore Properties – Permeation
Two basic parameters are important when considering permeation properties. One is ion selectivity and the other conductance. Ion selectivity is the ability to favor one ion over another and is expressed as the “permeability” ratio of the two ions, for example PK/PNa for potassium and sodium. It is, in practice, determined from the reversal potential measured in mixtures of the two ions. The simplest situation is when the ions are present at equal concentrations, one on one side of the membrane and the other on the other side – so called bi-ionic conditions. We consider the example of a cationic channel measured with 150 mM NaCl intracellular and 150 mM KCl extracellular. Then the reversal potential, Erev, is given by Erev = RT/(zF) ln (PK/PNa)
(2)
where R is the gas constant, T the absolute temperature, F the Faraday constant and z the valence (z = 1 for the example above). At room temperature the factor RT/F amounts to ~25 mV, a value that is very useful to remember for electrophysiologists. Inverting Eq. (2) yields PK/PNa = ezFErev/(RT) Thus, for example, Erev = 25 mV indicates an e-fold higher permeability of K+ versus Na+ (e ~ 2.718). Bi-ionic conditions are preferable but are not easily achieved in some experimental systems. For example in TEV recordings from oocytes the intracellular solution cannot be changed. In this case, the difference of the reversal potential that arises by changing from one extracellular solution to an-
5.4 Macroscopic Recordings
other is measured. To obtain a permeability ratio, the Goldman–Hodgkin–Katz equation is used Erev = RT/F ln((PK [K] ext + PNa [Na] ext)/(PK [K] int + PNa [Na] int))
(3)
where [K] ext is the extracellular K+ concentration and similarly for Na+ (for anions the sign of the reversal potential has to be inverted). We consider the case where the extracellular concentrations of Na+ and K+ are changed from [Na] 0 to [Na] 1 and from [K] 0 to [K] 1, respectively, and assume that the measured reversal potential changes from E0 to E1. From Eq. (3) it follows that the permeability ratio is given by PK /PNa = ([Na] 1 – [Na] 0 ef)/([K] 0 ef – [K] 1) where f = F(E1–E0)/(RT) ~ (E1–E0)/(25 mV). The assumption is that the intracellular concentrations do not change. When ions of different valence are compared (for example Na+ and Cl– or Na+ and Ca2+) the equations change slightly [1] but the basic type of measurement remains the same. One important and often overlooked problem of reversal potential measurements is the presence of liquid junction potentials that invariably arise when a solution is exchanged for another. The liquid junction potential is caused by the different mobility of different ions and is most pronounced when small inorganic mobile ions (for example Na+, Cl–) are exchanged by large organic quite immobile ions (for example NMDG+, gluconate–; see Ref. [34] for how to determine and correct for liquid junction potentials). When the Cl– concentration is changed care must be taken because most reference electrodes are Ag/AgCl electrodes that must be shielded from the solution exchange, for example by agar bridges. It may be difficult to determine the reversal potential because the current flowing through the channel is small, such that endogenous background conductances or a leak conductance dominate the effective reversal potential. One reason for this might be that the gates are closed at the expected reversal potential. This is especially a problem for voltage-gated channels. In this case so-called tail-current analysis can be applied to determine the reversal potential and the shape of the single channel current–voltage relationship, as illustrated in Fig. 5.2. The currents shown in Fig. 5.2c were simulated based on the two-state scheme shown in Fig. 5.2a using the pulse-protocol illustrated in Fig. 5.2b. Opening and closing rate constants are exponentially voltage-dependent such that the channel closes at negative voltages and opens maximally at positive voltages. The channel was assumed to have a linear single-channel current–voltage relationship (i–V) with an imposed reversal potential of Erev = –70 mV. However, from the steady state current–voltage relationship, obtained at the end of the variable pulse (see box in Fig. 5.2c, squares in Fig. 5.2d), the reversal potential cannot be obtained, because the open probability is too low. In contrast, the initial current, “immediately” after the end of the activating voltage-step (arrow in Fig. 5.2c), is measurable at these negative voltages. This “instantaneous tail current” and the resulting in-
119
120
5 Analysis of Electrophysiological Data Fig. 5.2 Tail current analysis. Macroscopic currents were simulated for the two state scheme shown in A. The reversal potential was –70 mV. From the holding potential of –80 mV a 0.2 s prepulse to 60 mV was followed by variable pulses ranging from –140 to 80 mV as illustrated in B. At negative voltages currents deactivate quickly such that the reversal potential cannot be reliably obtained from the steady-state currents (squares in D). The initial tail currents (circles in D) faithfully reproduce the linear single-channel current–voltage relationship and allow a precise determination of the reversal potential.
stantaneous current–voltage relationship (Fig. 5.2d, circles) reflect the shape of the single channel current–voltage relationship. This can be seen from the equation Itail(Vt) = N pend i (Vt)
(4)
where Itail is the instantaneous current at the tail-voltage,Vt, pend is the open-probability at the end of the activating prepulse, and i (Vt) is the single channel current at Vt. It is assumed that the open probability immediately after the voltage-step remains at the value it had at the end of the prepulse (pend). Then, the factor N pend is independent of the tail voltage, and Itail (Vt) is proportional to i (Vt). If the purpose is only to determine the reversal potential it is not very important to get exactly the initial current, and an average over a short stretch of currents shortly after the voltage jump can be calculated. For measuring the exact shape of the single channel i – V, care must be taken to determine the “correct” initial value. This may be complicated if the deactivation is fast and the voltage jump is associated
5.4 Macroscopic Recordings
with a large capacitance transient. In this case the time course of the deactivating current (after the capacitance transient) is fitted by a suitable function (for example an exponential function) that is then back-extrapolated to time “zero”. The determination of the permeability of a blocking ion can also be difficult. For example the muscle Cl– channel CLC-1 is blocked by iodide while iodide has a significant permeability with a permeability ratio of PI/PCl ~ 0.2. If extracellular Cl– is completely exchanged by iodide, however, CLC-1 is totally blocked and the reversal potential is dominated by endogenous background conductances, resulting in a wrong estimate of PI/PCl. The problem can be solved by partially exchanging extracellular Cl– for iodide (for example change from 100 mM Cl– to a solution containing 20 mM Cl– and 80 mM iodide) leaving enough Cl– to allow for a significant conductance. Eq. (3) can then be used again to quantify the permeability ratio. 5.4.2 Analysis of Fast Voltage-dependent Block – the Woodhull Model
Tail current analysis is useful to analyze voltage-dependent block by fast blockers. A commonly used model to describe voltage-dependent block is the Woodhull model [1, 35]. In this model it is assumed that the charged blocking particle enters the channel pore to a certain distance, and senses therefore part of the transmembrane electric field. Block is quantified by I
c I
0
1 c exp
zdVF=
RT 1 KD
0
(5)
Here I(c) is the current in presence of blocker at concentration c, KD(0) is the dissociation constant at zero voltage, z the valence of the blocking ion and d the “electrical” distance of the binding site from the bulk solution, that stands for the fraction of the electric field from the bulk solution to the binding site. Eq. (5) describes simultaneously the concentration and the voltage-dependence of block with just two parameters, KD(0) and d (see Fig. 5.3). Another way to describe the Woodhull model is in terms of an exponentially voltage-dependent dissociation constant (Fig. 5.3c). The simplicity of the Woodhull model makes it attractive and it is often a good initial model. Several assumptions are, however, seldom truly satisfied. Firstly, “blocking” ions are often permeable to some extent and may “punch through” at large voltages. Also, almost all ion channels have multi-ion pores in which the ions interact. A blocking ion could displace a permeable ion present at the blocking site within the pore. Such effects add to the intrinsic voltage dependence of block (described by d) and complicate the picture. The incorporation of such features into mechanistic models is beyond the scope of this chapter. See Ref. [1] for a comprehensive description of blocking mechanisms.
121
122
5 Analysis of Electrophysiological Data
Fig. 5.3 Illustration of the Woodhull model of channel block. A linear single-channel i – V and a zero reversal potential was assumed for the unblocked channel (solid line in A). The parameters of the block were KD(0) = 1 mM and zd = 0.4. Application of increasing amounts of blocker (0.5, 1, and 5 mM) produces the increasing voltage-dependent block
(dashed lines in A). The concentration dependence of block is illustrated in B for different voltages (–100, –30, 30, and +100 mV). The exponential voltage-dependence of the apparent KD, determined by fitting the curves in B with a simple 1 : 1 binding curve is illustrated in C.
5.4.3 Information on Gating Properties from Macroscopic Measurements
Macroscopic currents can be used to obtain an estimate about the open-probability. According to Eq. (1) the macroscopic current is proportional to the open probability, p, but further information or assumptions about the number of channels, N, and the single-channel current, i, are necessary to estimate p from the measured current, I. The number of channels can be assumed to be constant in a typical experiment, as long as its duration is relatively short and no particular maneuvers are undertaken to enhance or decrease protein turnover. Some ion channels can be drastically affected by co-expression and acute manipulation of co-expressed or endogenous regulating proteins, like the ubiquitin-protein ligase Nedd4 [36]. The number of channels can also be nonspecifically affected by agents that lead to a general retrieval of plasma membrane. For example treating Xenopus oocytes with phorbol esters, activators of protein kinase C, leads to an unspecific reduction of expressed conductances via a reduction of the plasma membrane surface [37]. Nevertheless, in most cases, N can be regarded as fixed. If measurements are performed at a fixed voltage, as in the case of ligand activated channels or in “isochronous tail-current” measurements for voltage-gated channels (see below) the single channel current, i, is also fixed. Otherwise, the shape of the singlechannel current–voltage relationship (i – V) has to be taken into account. For the
5.4 Macroscopic Recordings
purpose of extracting information about the open probability, the i – V is parametrized in a phenomenological manner, without necessarily interpreting the corresponding parameters mechanistically. In the absence of direct information the i – V is often assumed to be linear i (V ) = g (V – Erev) with a single-channel conductance, g, and a reversal potential, Erev. This assumption is particularly appropriate if the reversal potential is relatively close to 0 mV, because in this case the intrinsic “Goldman”-rectification [38] has little influence on the shape of the i – V. If the reversal potential is far from zero, and if the channel is highly selective for one ion species present in the solutions, the Goldman– Hodgkin–Katz equation is more appropriate to describe the i – V because it takes the concentrations of the permeable ion on the two sides of the membrane into account [1]: i
V K
exp
z
r 1 exp
z 1
(6)
Where K is a constant depending on the ionic concentrations, f = VF/(RT) and fr = ErevF/(RT). The nonlinear shape of the Goldman current–voltage relationship is illustrated in Fig. 5.4 for a monovalent cation with a reversal potential of – 60 mV. It is also not uncommon that some voltage-dependent block or strong rectification is present that has to be taken into account for the description of the i – V. Such a block can be phenomenologically described by a factor that is derived from the Woodhull model. For a Goldman-type rectification with additional block the i – V is described by i
V K
exp
z
r 1 1 exp
z 1 1 exp
V
V1 =V2
(7)
where V1 and V2 are empirical parameters describing the block; an example is shown in Fig. 5.4 (dashed line).
Figure 5.4 The Goldman–Hodgkin–Katz equation. The solid line is drawn according to Eq. (6) with Erev = –60 mV. The dashed line is drawn according to Eq. (7) with Erev = –60 mV and V1 = V2 = 50 mV.
123
124
5 Analysis of Electrophysiological Data
5.4.3.1 Equilibrium Properties – Voltage-gated Channels Two types of pulse protocols are most often used to extract the overall-open probability from macroscopic measurements. They are illustrated in Fig. 5.5, together with simulated currents based on the 2-state scheme of Fig. 5.2. In the direct pulse protocol (Fig. 5.5a and b) pulses are delivered to various potentials and the maximum current during the pulse is plotted versus voltage (Fig. 5.5c, squares). Usually, the open-probability of voltage-gated channels is parametrized with a Boltzmann distribution, here written in two different versions
popen
1 1 exp
V1=2
V=k
1 1 exp
zg
V1=2
VF=
RT
(8)
In both forms the voltage of half-maximal activation,V1/2, describes the voltage at which popen = 0.5. The steepness of the voltage dependence is described either by the so-called “slope-factor”, k (in mV), or the “apparent gating valence” zg (dimensionless). These two quantities are inversely related by k
RT zg F
Fig. 5.5 Determination of the open probability for voltage gated channels. Currents were simulated according to the 2-state scheme shown in Fig. 5.2A assuming a reversal potential of 0 mV and applying the pulse protocol shown in A. Steady state currents measured at the end of the variable-voltage pulse (see arrow in B) are plotted in C as squares. The tail currents at the beginning of the constant tail pulse to 60 mV are shown as circles.
5.4 Macroscopic Recordings
The Boltzmann equation derives from the statistical Boltzmann equilibrium that relates the ratio of the probability to be in one of two microscopic states, O and C that differ in free energy by a certain amount DG: pO exp
DG=
RT pC The Boltzmann distribution of voltage-gated channels (Eq. (8)) stems from a simple model for voltage-gated channels that assumes that the free energy difference between the open and the closed state is additively composed of an electrical term, determined by the electrical charge, denoted by QC and QO, respectively, and a purely chemical term, DG0, such that DG is given by DG DG0 V
QO
QC DG0 Vzg F
where zg is the apparent gating valence. The larger the charge difference between the closed and open state, the more sensitive is the channel to voltage, and the steeper the popen(V ) curve. To extract the gating component (p) from the permeation component (i) for currents obtained from the “direct” I – V (Fig. 5.5c, squares) the I – V is fitted by the product of the i – V term and the Boltzmann term: I
V K
exp
z
r 1 1 exp
z 1 1 exp
zg
V1=2
VF=
RT
(9)
Here, in Eq. (9), the Goldman–Hodgkin–Katz equation (Eq. 6) was used for a description of the i – V. The four parameters, K, fr,V1/2, and zg are obtained from a fit to the macroscopic data, while only the two parameters V1/2, and zg are relevant for gating. The tail-pulse protocol illustrated in Fig. 5.5a and b (see arrow) is often called “isochronous tail protocol” because the fixed tail pulse is applied after a fixed amount of time. The initial tail current is a measure of the open probability at the end of the (variable) pre-pulse (see Eq. (4)). As for the instantaneous I–V (see Section 5.4.1) a correct determination of the initial tail current may be hindered by the capacitive artifact. A careful back-extrapolation of the time course of the tail current to “time 0” may be necessary to obtain a reliable estimate of the initial tail current. The tail voltage should be chosen such that the relaxations are well resolved with the employed voltage-clamp technique. The resulting initial tail currents are then plotted versus the pre-pulse voltage (Fig. 5.5c, circles) and fitted by I
V
Imax 1 exp
zg
V1=2 VF=
RT
(10)
125
126
5 Analysis of Electrophysiological Data
Here, Imax , is the maximal current obtained at saturating voltages. It can be determined by normalization with the measured currents or it can be left as an independent parameter. The latter possibility is particularly appropriate if the employed voltage range is not sufficient to saturate channel gating. In this case the plateau of the Boltzmann distribution is not reached and currents can not be normalized by the maximally measured value. Sometimes it happens that currents do not tend to zero at voltages where the channel should close according to Eq. (8). This may be an intrinsic property of the gating mechanism of the channel or may represent an uncompensated leak component. If such a “residual” open probability is an intrinsic property, a description with a Boltzmann distribution (Eq. (8)) is, strictly speaking, not adequate. Nevertheless the shape of the popen(V) curve can often be described phenomenologically by a modified Boltzmann distribution I
V Imax pmin
1 pmin 1 exp
zg
V1=2 VF=
RT
Where pmin describes the minimal open probability reached at saturating voltages where channels are maximally closed. V1/2 is no longer the voltage at which popen = 0.5 but where popen = pmin + 0.5(1 – pmin). The isochronous tail-current protocol is, in principle, superior to the direct I – V because it is not influenced by the shape of the i – V. However, in certain cases a direct method has to be employed. For example, voltage-gated Na+ channels are governed by two main gating processes of opposite voltage dependence and one wants to determine separately their respective voltage dependence. Na+ channels inactivate with a voltage-dependent time-course after an activating voltage step. The steady state, isochronous tail current I – V would determine only the “window” current, the region where activation and inactivation gates are both open. To separate the two gates of the Na+ channel two different protocols are used to assess the voltage dependence of the activation and the inactivation gate, respectively. The “peak current” of the direct I – V is generally used as a measure of the activation gate (Fig. 5.6a and b). This is justified because the time constant of activation is considerably faster than that of inactivation. The inactivation is measured with a two-pulse protocol similar to the isochronous tail current protocol (Fig. 5.6c and d).
5.4.3.2 Equilibrium Properties – Ligand Gated Channels Conceptually, the determination of equilibrium properties of ligand activated channels is similar to that of voltage activated channels. The energy driving the conformational change is not supplied by the membrane voltage but by the chemical energy of ligand binding. Accordingly, the relevant intensive variable is the ligand concentration, here denoted by [L]. However, for ligand activated channels the allosteric action of the ligand is much more evident and explicit than is the voltage for voltage gated channels [39] (even though most quantitative models of
5.4 Macroscopic Recordings
Fig. 5.6 Activation and inactivation of voltage-gated sodium channels. Currents were recorded from tsA201 cells transfected with the cardiac sodium channel and measured using the whole cell configuration of the patch clamp technique. In A currents were elicited by 10 ms voltage steps from –80 to 50 mV (see inset). In B the peak currents are plotted versus the test voltage (symbols) superimposed with a fit of the equation I
V GNa
V
Erev
1 1 exp
zg
V1=2
as shown by the lines. C shows currents from a different cell evoked by a two-pulse protocol as shown in the inset. The response to the 100 ms long prepulse to voltages from –120 to –10 mV is not shown. The currents represent the response to the fixed tail pulse to – 10 mV that assays channel availability. The peak currents are plotted in D (symbols) together with a fit of Eq. (10). Note that activation (A, B) and inactivation (C, D) have an opposite voltage-dependence.
VF=
RT
voltage gated channels often have an allosteric character). Thus instead of assuming a scheme of the form (Scheme 1) where binding of a ligand directly opens the channel, the minimal scheme applied for ligand activated channels assumes that binding of the ligand favors opening but does not directly open the channel. This scheme is given by (Scheme 2) with a closed, unliganded state U, a liganded (bound) but closed state, B, and an open state, O [40]. Ligand binding occurs with second-order association rate kon
127
128
5 Analysis of Electrophysiological Data
and dissociation rate koff , while the allosteric transition is described by rate constants a and b. More complex schemes are necessary to include the possibility that unliganded channels are able to open [39]. Allosteric schemes and equations become even more complex if more than one binding site is present, the usual case in real life. Therefore, mostly for reasons of simplicity, the phenomenological description of ligand activated channels is often expressed in terms of the Hill equation popen
pmax n KD 1 L
(11)
Where KD is the apparent dissociation constant of the binding site(s) and n the Hill coefficient, an estimate of the number of binding sites [1]. For the simple Scheme 2, equilibrium properties can indeed be expressed in the form of Eq. (11) popen
1 1 1 KD 1 r r L
r=
1 r K 1 D L
(12)
Where r = a/b, KD = koff /kon , and the apparent affinity KD* = KD/(1 + r). The maximum open probability is pmax = r/(1 + r) = a/(a + b). Thus, even though the concentration dependence of the macroscopic current strictly follows a 1 : 1 binding isotherm, the measured apparent affinity can be very different from that of the true affinity of the ligand binding site. KD* is always smaller than KD and only if r is very small (a 5 b) is the apparent affinity equal to the true affinity. But in this case the ligand is not very effective (pmax 5 1). If r is very large, the ligand is very effective (pmax ~ 1) but the apparent affinity is much higher than the true affinity (KD* 5 KD). Since the absolute open probability is difficult to determine from macroscopic equilibrium measurements alone, additional information is necessary to determine true affinities and ligand efficacies. These can stem from kinetic macroscopic measurements, noise analysis, or single-channel analysis (see below). A fundamental difference between voltage gated channels and ligand gated channels is that the latter can be more or less efficiently activated by different types of ligands (for example certain glutamate receptors can be activated also by NMDA), while there is only one stimulus (voltage) for voltage gated channels. In terms of the simple model shown by Scheme 2, quantified by Eq. (12), different ligands have generally a different (true) affinity, and a different efficacy (pmax). For ligands that occupy the “same” binding sites the true number of binding sites is the same. The Hill coefficient in Eq. (11) may nevertheless be different, since Eq. (11) is an approximate phenomenological description of channel activation. Certain ligands might also counteract the effect of a more potent activator. Furthermore, certain receptors possess different kinds of binding sites. For example certain glutamate receptors need glycine as a co-factor for full activation by glutamate
5.4 Macroscopic Recordings
[41]. A good overview over equilibrium properties of ligand gated ion channels with further reference is given by [39]. Many ligand gated channels exhibit desensitization: currents decrease despite the continuous presence of ligand. The degree and kinetics of desensitization vary wildly between different channel types (see Ref. [42] and references therein). This phenomenon is conceptually similar to the “inactivation” of voltage gated channels. The presence of desensitization complicates the determination of activation properties. Experimentally, the most difficult problem, particularly if desensitization is an issue, is the fast application of the ligand (see Section 5.3.2).
5.4.3.3 Macroscopic Kinetics Channel kinetics can be evoked by a variety of stimuli. Sometimes, biophysical analysis alone is not sufficient to determine the physiological effect in a complex cellular system, as for example a cardiac myocyte, where numerous channel types contribute to the various phases of the action potential. In such cases, a “physiological” stimulation with an action potential waveform or other stimuli might reveal if, for example, a given mutation leads to action potential shortening (see for example Ref. [43]). However, such physiological stimuli are not well suited to uncovering the underlying mechanistic effect. As outlined in the Introduction, for this purpose clamping the relevant intensive physical parameter (voltage, ligand concentration, temperature, pressure, light intensity,) to a fixed value and performing jump experiments are more informative. Like practically all kinds of conformational changes of proteins, current relaxations of a homogenous population of channels induced by a step-wise change of an intensive parameter, can be described by the sum of a constant term (the steady-state current, I ?) and one or more exponential functions
I
t I1
n X
ai exp
t=i
(13)
i1
With amplitudes, ai , and time constants, ti (t is the time after the jump). The kinetics is thus determined by 2n+1 parameters. The time constants, ti , depend only on the actual value of the relevant physical parameter (the voltage or the ligand concentration after the jump), while the coefficients, ai, depend on the state occupancy before the jump. The exponential time dependence is a mathematical consequence from the Markov property of the conformational changes: once the channel undergoes a conformational change it loses “immediately” the memory of from what state it arrived (if there is more than one possibility to arrive in a certain state) and it has no memory of how long it has already been in a given state. One of the underlying assumptions is, of course, that there exists a finite set of definable, stable “states”. Actually, it is more scientific to turn the argument around: the experimental result that relaxation kinetics for most channels can be well described by Eq. (13) (with a reasonably small and reproducible number, n, of components) suggests that the Markov assumption is valid for ion channels. What is a
129
130
5 Analysis of Electrophysiological Data
reasonably small number, n? This is indeed a difficult question. In principle, a gating scheme with N states predicts exponential relaxation with N-1 components (for example a two-state system has single-exponential kinetics). In practice it is very difficult to reliably fit more than two or three exponential components. However, gating schemes often require more than four states. Indeed it is extremely difficult to define a “correct” gating scheme based on fitting of current relaxations. Often gating schemes that have a large number of states can be simplified with symmetry arguments and the number of free parameters can be reduced. A beautiful example are the Hodgkin–Huxley equations (see Ref. [1]), but also recent models of Shaker K+ channels have a relatively small number of parameters, despite a large number of states (see Refs. [44, 45]). Furthermore, current relaxations are often approximately single- or double exponential under certain conditions, even though a full kinetic model requires many states, because most components are negligible. For example the deactivation of Na+ channels at very negative voltages after a brief activating pulse can be well described by a single exponential, even though the general gating involves very many states. Often, kinetics of ion channels is fitted and time constants are determined phenomenologically without necessarily wanting to define a molecular mechanism of gating. In many circumstances, this is the only practical choice, because the underlying mechanism is too complex to be determined reliably from the measurements. One of the most difficult problems in curve fitting with exponentials is to separate components with time constants that differ less than, let us say three-fold. Extreme care has to be taken in such cases and reproducibility has to be tested extensively. Often data can be reasonably well described with the sum of two exponentials, even though the “true” mechanism would require at least three. In such cases, the time constants (and relative weights) of the exponential components determined from the double-exponential fit can be almost meaningless. If the true time course is distorted slightly, for example by voltage-clamp errors (see above), the kinetic analysis becomes even more difficult. Under certain conditions it is impossible to directly follow the kinetics of a process measuring the ionic current. For example, the channel may be closed quickly by one kind of gate while another gate is slowly changing its status. Another reason that renders impossible a direct measurement might be that the voltage is close to the reversal potential or that a strong block occurs. Also, a large capacitive artifact may obscure fast relaxations [27]. In these cases so-called “envelope” protocols can often be applied, as illustrated in Fig. 5.7 that shows the classical protocol to study the recovery from inactivation of the voltage gated Na+ channel or, completely analogously it illustrates the measurement of the recovery from desensitization of a ligand gated channel. It is insightful to explicitly consider the kinetics of the most simple, two state system, because often more complex schemes can also be simplified to it, allowing an easy quantitative description. (Scheme 3)
5.4 Macroscopic Recordings
Fig. 5.7 Envelope protocol to study recovery from inactivation. The pulse protocol is illustrated in A, current traces are shown in B. Currents during the recovery period are not shown. In C the peak current at the final test pulse is plotted versus the recovery time together with a single exponential fit. Currents were simulated with a simplified Hodgkin–Huxley model.
Opening occurs with rate-constant a, closing with rate constant b. These rate constants depend on the intensive physical parameters (voltage, ligand concentration,). The equilibrium open probability is p1
a ab
While the relaxation time constant is given by t
1 ab
Knowing, both p and t allows the determination of a and b: a p1 =t; b
1
p1 =t
Relaxations that start from a given value of open probability, p0, proceed in time as p
t p1
p0
p1 exp
t=t
The Del Castillo–Katz model for ligand gated channels (Scheme 2) can be reduced to an effective two-state system if the ligand binding/unbinding is much faster than the opening isomerization transition (described by a and b). In this case the receptor is always in equilibrium with the ligand (see Ref. [1]):
131
132
5 Analysis of Electrophysiological Data
(Scheme 4) Where the effective opening rate is given by aeff
1 a KD 1 L
Combining steady state-measurements (Eq. (12)) and relaxation measurements with step-changes in ligand concentration allows the determination of all three parameters (KD, a, b) of the model of Scheme 4: for jumps into zero concentration of ligand, the relaxation rate is t–1 = because aeff = 0 for [L] = 0. For jumps into saturating concentration of ligand the relaxation rate is t–1 = a + b because aeff = a for [L] 4 KD. These kinetic experiments thus provide estimates for a and b. From the equilibrium measurements the apparent affinity, KD* is determined (Eq. (12)) and using the values for a and b the true affinity, KD, can be calculated from KD = KD* (1 + a/b). This simple example illustrates how the combined use of equilibrium and kinetic measurements in addition to simplifying assumptions can be used to obtain quantitative information about a molecular mechanism. 5.4.4 Channel Block
All ion channels interact with a variety of smaller molecules (peptides, small organic molecules) that directly or indirectly reduce ion permeation. Such substances are called blockers or inhibitors and are the bread and butter of the pharmaceutical industry interested in ion channel targets. Blockers may directly block the pore and physically impede ion flow. Inhibitors may reduce current flow by stabilizing the closed state of a channel gate, for example, in which case the binding site may be far away from the ionic pore. Such inhibitors are often called “gating modifiers”. This distinction between these two kinds of modulators is actually not so strict – many pore blockers additionally alter the gating by binding more tightly to one or another gating state (state-dependent block). Such blockers may be useful tools to study the properties of channels [46, 47]. Many pore blockers exert a voltage-dependent block that can often be described by the Woodhull model (see Section 5.4.2). For most practical purposes channel block is quantified by the Hill equation I
B I
0
1 n B 1 KD
that quantifies the ratio of current in the presence of blocker at concentration [B] to the current in its absence with an apparent affinity KD and the Hill coefficient, n.
5.4 Macroscopic Recordings
5.4.5 Nonstationary Noise Analysis
The opening and closing of ion channels is a random process that renders current registrations “noisy”, in particular if few channels are present. The statistical properties of the noise can be used to infer some characteristics of the underlying elementary events. A prerequisite is that the channel-induced noise is significantly above the background noise of the recording system. This condition is, for example, generally not fulfilled in TEV recordings from Xenopus oocytes. The patchclamp technique, on the other hand, is exceptionally well suited for noise analysis. However, background noise may be large if the series resistance in whole cell recordings is highly compensated (see Section 5.3.2). For stationary noise analysis the system is recorded for a prolonged time at fixed external conditions (voltage, ligand concentration). The power spectrum is then fitted with the sum of Lorentzian functions. While this method can yield important information [48], nonstationary noise analysis is often faster and easier to perform. Under appropriate conditions, each of the elementary parameters of Eq. (1) can be determined assuming a single open conductance level. This should be verified with single channel analysis. For standard nonstationary noise analysis a step-protocol (that may be a voltagestep or stepwise change in ligand concentration) is applied repeatedly, with enough time passing between individual stimulations to ensure identical initial conditions for each step. Each current response, Ii (t), is recorded (i = 1, …, n) (Fig. 5.8a). From these recordings the mean can be calculated by
n 1X Ii
t n i1
(14)
This is now a much smoother curve than the individual traces (Fig. 5.8 b) and because of this it can be written a Nip
t where p(t) is the time course of the (“true”) open probability. The variance of the response, s2(t), for each time point, t, is given by s2
t
n 1X
Ii
t 2 n i1
(15)
the standard statistical definition (Fig. 5.8c). However, the most important “trick” in nonstationary noise analysis is to calculate the variance not as suggested by Eq. (15) but as the squared difference of consecutive records: s2
t
n 1 X
1 2
n
1
i1
Ii1
t
Ii
t2
(16)
133
134
5 Analysis of Electrophysiological Data
(Note the scaling by a factor of 1/2 in Eq. (16)). For a perfectly stable system the results obtained from Eq. (15) and Eq. (16) are identical. However, in reality, small drifts of the total current amplitude (“run-down”, “run-up”) or of the reversal potential or other parameters are practically unavoidable. These small drifts are well cancelled out using Eq. (16) while they artificially increase the variance when calculated by Eq. (15) and may render the noise analysis meaningless, particularly if the single channel conductance is small [49]. Meaningful results can be obtained even with run-down up to a factor of two or more. Since the variance is obtained from the differences of records, leak currents and capacity transients also cancel out (if they are associated with negligible inherent noise). That means that the individual records do not have to be, and should not be, leak-corrected for the application of Eq. (16), in contrast to the calculation of the mean (Eq. 14). A nice property of the variance is that independent noise source adds independently to it. Thus, total variance is given by s2tot s2channel s2background and the background variance can usually be assumed to be independent of voltage. It can thus be determined at a voltage where no ion current flows through the channels (for example at the reversal potential or in the absence of ligand for ligand activated channels or at a voltage where channels are closed for voltage gated channels). Having determined the variance and the mean it is now possible to proceed with the “variance-mean analysis”. For this we use the equality s2 Ni2 p
1
p
(17)
This fundamental equation [48, 49] can be understood intuitively: for p = 0 the variance is null because no current flows. Also for p = 1 (the maximum value it can attain) there is no fluctuation in the current because all channels are permanently open. The largest fluctuations are present for p = 0.5 when channels are half open and half closed on average. Note that (Eq. (17)) is independent of the kinetics of the fluctuation. However, the bandwidth of the recording system must be sufficiently large to resolve the fastest transitions. Combining Eq. (17) with Eq. (1) yields s2 iI
I 2 =N
(18)
that relates the two macroscopic quantities, s2, and mean current I, in a parabolic function that depends on two parameters, the single channel current i, and the number of channels, N, that can be determined by a least-squares fit (Fig. 5.8d). Often the whole traces are not plotted and fitted against each other but they are first binned in an appropriate manner [49] (see Fig. 5.8d). The two parameters, i and N, are best defined if a large interval of popen is covered by the relaxation. If only a very small interval of popen is sampled, the two parameters cannot be determined independently: a small variance can be caused by a small number of chan-
5.4 Macroscopic Recordings
nels and/or by a small or large popen and vice versa. It may be that the current excursion in the relaxation is substantial but that popen remains significantly smaller than 0.5. In this case the second term in Eq. (18) is negligible, the relationship is linear, and only the single channel current can be determined. If both, i and N, can be determined the absolute popen during the relaxation can be calculated from p
t
I
t Ni
Thus, using a simple experimental protocol (Fig. 5.8) allows a quite precise determination of fundamental channel parameters, without the need to perform single channel analysis.
Fig. 5.8 Nonstationary noise analysis. Currents were repeatedly evoked by a test pulse and individual responses are shown in A (currents were simulated with a simplified Hodgkin–Huxley model). The mean and the variance are shown in B and C, respectively. In D the (binned) variance is plotted versus the respective mean together with a fit of Eq. (18). The horizontal line in D marks the level of the background variance.
5.4.6 Gating Current Measurements in Voltage Gated Channels
Another way of obtaining additional information about molecular gating mechanisms is to measure the so-called “gating currents” associated with the molecular rearrangements of voltage gated cation channels [50]. These are transient currents, similar to capacitive currents, that reflect the movement of the gating charges within the electric field. Of course, any conformational rearrangement of
135
136
5 Analysis of Electrophysiological Data
the channel protein that is associated with charge redistribution gives rise to “transient” gating currents, even if they do not reflect the movement of a “voltage sensor”. However, in voltage gated K+, Na+ and Ca2+ channels the voltage-sensor movements clearly dominate the gating currents. In order to resolve gating currents that are of small magnitude it is necessary to eliminate the normal ionic currents by applying blockers or by eliminating permeant ions. However, it is necessary to ensure that such maneuvers of completely eliminating ion flow through the pore does not alter significantly the gating process itself, an often difficult task. It is beyond the scope of this chapter to describe the design and the analysis of gating current measurements (see Ref. [51] for review). However, the estimation of the total gating charge of a single voltage gated K+ channel provides a nice example of this approach [52]. The authors determined first the number of channels, N, in an inside out patch using nonstationary noise analysis (see Section 5.4.5). Then they replaced intracellular K+ with TEA+, a blocker of K+ channels, to eliminate the ionic currents and measured the total gating charge, Q, by integrating the gating currents. The ratio Q/N, the gating charge per channel, was about 12 elementary charges, consistent with more indirect measurements. Another nice result was obtained by Conti and Stühmer [53], who estimated the size of the charge of a single voltage sensor in Na+ channels. Voltage gated cation channels possess four voltage sensors that move more or less independently of each other. The movement of each sensor produces a spike-like tiny current. The ensemble of many sensors is the random superposition of many such spikes, filtered at the recording frequency. Nonstationary noise analysis yielded an elementary charge of individual spikes of about 2.3 elementary charges, a very reasonable value.
5.5 Single Channel Analysis
The possibility to observe and analyze the opening and closing of single ion channel molecules marked a revolution in ion channel research and remains one of the few techniques that allow a true single molecule measurement in real time [3, 4]. Single channel recordings can provide a wealth of information and numerous numerical methods for single channel analysis have been developed. The reader is referred to “Single channel Recording” by Sakmann and Neher for detailed information [54]. Here a very broad overview of a typical single channel analysis is provided. Recent papers utilizing more advanced techniques can also be found [55, 56]. 5.5.1 Amplitude Histogram Analysis
The first step of a single channel analysis is usually to construct an amplitude histogram. Already at this point one ever returning aspect of single channel analysis becomes important: adequate filtering. Of course the data should be filtered with
5.5 Single Channel Analysis
a good filter (for example an 8-pole Bessel filter) with a cut-off of at least at one half the sample frequencies to avoid aliasing [57]. In order to get, at least in principle, as much information as possible, the sampling rate must be sufficiently high. It is however useless to acquire data at a low signal-to-noise ratio. A dilemma is sometimes that one would like to see online the highly filtered data in order to get an immediate impression of its quality, but one also wants to acquire at a higher frequency for quantitative offline analysis. One possibility is to divert the signal after a primary anti-alias filter into two separately sampled channels. One signal is acquired after only the first filter, while in the second the current signal is subjected to further filtering for immediate inspection. This can be done on an oscilloscope, or, if the acquiring software allows the simultaneous sampling of two channels, the highly filtered signal can be acquired as a second input channel and visualized on the computer screen. For the construction of the amplitude histogram all sample points that fall in a given “current-bin” are counted, resulting in the number of events per bin that are then plotted versus the mid-point of the current bin. This is illustrated in Fig. 5.9. To each current level of the channel (“closed” and “open” in Fig. 5.9) corresponds a “peak” in the amplitude histogram. The peaks may not be well separated because noise is large (Fig. 5.9b). In this case the data can be digitally filtered by a Gaussian filter that has various convenient properties [57] (Fig. 5.9c). If the baseline is not stable the amplitude histogram becomes distorted. Excessive baseline drift can make the single channel analysis very difficult and must be corrected. Several analysis programs are available that allow baseline correction and many other features. Once an acceptable amplitude histogram has been constructed it is fitted with the sum of Gaussian functions, Gi, one for each peak, i H
I
X
Gi
I
i
X ai i
si
exp
I
i 2 2s2i
(19)
Where each Gaussian functions is characterized by a mean mi, a width, si , and amplitude, ai. The inclusion of the width, si , in the prefactor ai/si in the Gaussian fit (Eq. (19)) facilitates the calculation of the relative area, Ai, that is occupied by each Gaussian component: ai Ai 100% P aj j
The relative area, Ai, is a measure of the probability to dwell in the conductance state associated with mean mi. Often it happens that the membrane patch contains an unknown number, N, of identical channels, leading to equidistant peaks of the amplitude histogram at levels ni (n = 0, 1, …), where i is the amplitude of a single channel. Even though the absolute open probability cannot easily be determined in such a situation a useful parameter to evaluate effects of drug application or other maneuvers is the so-called “NpO”, i. e. the product of the (unknown) number
137
138
5 Analysis of Electrophysiological Data
Fig. 5.9 Amplitude histogram analysis. Currents were simulated based on a simple 2state scheme with an open conductance level of 1 pA and an added Gaussian noise of 0.4 pA SD (panel A). The baseline and the open conductance level are indicated by horizontal lines. The noisy trace in A seems to be almost useless. However, the amplitude histogram shown in B clearly shows two peaks at
the right amplitudes (0 pA and 1 pA), and the fit with the sum of two Gaussian functions (thick line in B) correctly predicts the amplitude and area of each conductance level. The trace shown in C is strongly filtered and the histogram in D shows two well separated peaks of correct amplitude and weight. The bin width used for the histograms was 10 f.A.
of channels and their open probability. From the histogram fit the “NpO” can be calculated as 1 P
NpO
j0 1 P
jAj Aj
j0
where the “areas” Aj are obtained from the Gaussian fits as described above, and A0 corresponds to the baseline. 5.5.2 Kinetic Single Channel Analysis
A comprehensive description of the kinetic analysis of single channel data is beyond the limits of this chapter and only general directions can be given. The strategic decisions that can be taken for kinetic analysis are outlined schematically in Fig. 5.10. The most direct way of analysis is depicted on the left of the figure and
5.5 Single Channel Analysis
Fig. 5.10 Flow diagram of strategies for kinetic single channel analysis.
consists of directly fitting a “hidden Markov model” to the “raw” data [58–60]. A Markov model is a kinetic scheme like those described above (for example, Scheme 2) with possibly many states of various conductance levels connected by rate constants. The rate constants and the conductance levels of the various states are the parameters fitted in this approach. The model is “hidden” for two reasons. First, several kinetic states may be associated with the same conductance. Transitions among these states are therefore not directly visible. Second, the noise can hide shortlived dwell times or low conductance states. One advantage of the hidden Markov model approach is that it takes the noise into account explicitly [59, 60]. Algorithmically, for the hidden Markov model the following question is raised: for a given set of parameters (these parameters include the rate constants of the model, the conductance level of each state, and parameters that describe the noise) what is the probability of observing the currents that have been measured? The parameters of the model and the characteristics of the noise are then
139
140
5 Analysis of Electrophysiological Data
adapted to maximize this probability. The calculation of this probability is a formidable task but efficient algorithms have been developed that allow the analysis of quite long data sets. One drawback of the method is that a reasonable kinetic model should a priori be known. The method can be applied for example to study the effect of mutations of an ion channel for which a kinetic scheme has been established previously. Another drawback is that the method is a kind of black box with little possibility of visual evaluation to check if the results are “reasonable”. A further serious problem may occur if the general properties of the noise are not adequately treated [60]. Thus, while the hidden Markov modeling can be a powerful tool, its use requires some experience and results should be checked with other methods. The more traditional methods of analysis do not work directly on the raw data trace but this is first “idealized” (Fig. 5.11). In the idealization process the events of channel opening and closing are detected either completely automatically or interactively using specially designed computer programs. The noisy raw data trace is thus substituted by the idealized smooth trace (Fig. 5.11) that can be easily represented as a list with two numbers for each entry in the list: the duration of each dwelling together with the corresponding current level. For idealization one must decide if a current fluctuation represents a transition to another conductance level or if it is just noise. As a criterion the 50 % level criterion is most often
Fig. 5.11 Dwell-time analysis. In A a short stretch of a simulated trace of a two-state scheme is shown. In B the corresponding idealized trace is displayed. The cumulative dwell-time histograms shown in C and D are based on a total of 408 events each and represent the relative frequency of events to be longer than a given duration. By definition the
cumulative distribution equals 1 for a duration of 0. A single exponential dwell-time distribution is represented by a straight line in the logarithmic scaling of Fig. 5.11. Other representations of dwell-time histograms are probably more common [62] but require a much larger number of events for a satisfactory graphical display.
5.5 Single Channel Analysis
employed [57]. To apply this criterion, the possible conductance levels are estimated first by the fitting of the amplitude histogram (see above). In addition, events are only accepted if they are of a minimal length. For a reliable assignment of the transitions data usually have to be filtered more than for the hidden Markov approach, at least if the signal to noise level is low. The basic problem in the idealization process is that short events are easily missed (the missed events problem) but short events can also be artifactually introduced if noise is excessive. The problem is double: missing a short closure not only leads to the loss of a closed event but it also lengthens the opening time of the event during which the closure occurred. Similarly, the artifactual introduction of a short closure not only alters the closed times but also shortens, and divides into two the underlying opening. To deal with this problem in generality is not easy. First, a reasonable cut-off is defined such that no events shorter than this are rigorously accepted and the resulting error in the final fitting procedure can then be compensated [56, 61]. For simplicity, we ignore here the missed events problem. The idealized trace can then be analyzed in several ways. A first step is to construct and inspect dwell-time histograms. Several kinds of binning procedures and histograms to construct have been proposed (see for example [62]). In Fig. 5.11 so-called cumulative dwell time histograms for the two conductance levels are displayed. These histograms can then be fitted with the sum exponential components in order to extract kinetic information from the single channel data. The construction and fitting of histograms is always a first step in data analysis if little is known about the underlying channel and if one wants to obtain an impression about its kinetic behavior, like for example an estimate of the number of open and closed states. Histogram fitting may also provide a purely empirical set of parameters whose variation under the influence of ligands or mutation can be studied. If a good working hypothesis for a kinetic Markov model for the channel is available it may instead be a good idea to fit directly the likelihood of the idealized channel trace. This approach is similar to the hidden Markov approach described above in that the full kinetic information including possible correlations are exploited. The maximum-likelihood fitting overcomes one of the biggest problems of histogram fitting: it is not clear how the time constants and coefficients extracted from closed and open time histograms have to be weighted in fitting a concrete Markov scheme. In the maximum-likelihood approach the rate constants defining the Markov scheme are directly optimized [56, 61]. Recordings obtained under different conditions can also be fitted simultaneously. This approach thus allows on the one hand an objective estimate of physical parameters similar to the hidden Markov fitting and on the other hand the results can be easily judged visually by comparing the predictions for all kinds of dwell-time histograms [56].
141
142
5 Analysis of Electrophysiological Data
5.6 Summary
This chapter provides a broad overview of current concepts and methods of analysis of electrophysiological data. Several of the methods are incorporated into the free analysis program written by the author that is available for download at http://www.ge.cnr.it/ICB/conti_moran_pusch/programs-pusch/software-mik.htm. The simulation program used to generate several of the figures can also be found there.
Acknowledgements
I thank Armando Carpaneto for critically reading the manuscript. The financial support by Telethon Italy (grant GGP04018) and the Italian Research Ministry (FIRB RBAU01PJMS) is gratefully acknowledged.
References 1 B. Hille, Ion Channels of Excitable Membranes, Sinauer, Sunderland, MA, 2001. 2 A. L. Hodgkin, A. F. Huxley, A quantitative description of membrane current and its application to conduction and excitation in nerve, J. Physiol. 1952, 117, 500–544. 3 E. Neher, B. Sakmann, Single-channel currents recorded from membrane of denervated frog muscle fibres, Nature 1976, 260, 799–802. 4 O. P. Hamill, A. Marty, E. Neher, B. Sakmann, F. J. Sigworth, Improved patch-clamp techniques for high-resolution current recording from cells and cell-free membrane patches, Pflugers. Arch. 1981, 391, 85–100. 5 D. A. Doyle et al., The structure of the potassium channel: molecular basis of K+ conduction and selectivity, Science 1998, 280, 69–77. 6 Y. Jiang et al., The open pore conformation of potassium channels, Nature 2002, 417, 523–526. 7 R. Dutzler, E. B. Campbell, M. Cadene, B. T. Chait, R. MacKinnon, X-ray structure of a ClC chloride channel at 3.0 Å reveals the molecular basis of anion selectivity, Nature 2002, 415, 287–294.
8 S. Bernèche, B. Roux, A microscopic view of ion conduction through the K+ channel, Proc. Natl. Acad.Sci. U. S. A. 2003, 100, 8644–8648. Epub 2003 Jul 01. 9 W. Stühmer, Electrophysiological recording from Xenopus oocytes, Methods Enzymol. 1992, 207, 319–339. 10 W. Stühmer, Electrophysiological recordings from Xenopus oocytes, Methods Enzymol. 1998, 293, 280–300. 11 M. F. Romero,Y. Kanai, H. Gunshin, M. A. Hediger, Expression cloning using Xenopus laevis oocytes, Methods Enzymol. 1998, 296, 17–52. 12 G. Nagel, T. Szellas, J. R. Riordan, T. Friedrich, K. Hartung, Non-specific activation of the epithelial sodium channel by the CFTR chloride channel, EMBO Rep. 2001, 2, 249–254. 13 E. Stefani, L. Toro, E. Perozo, F Bezanilla, Gating of Shaker K+ channels: I. Ionic and gating currents, Biophys. J. 1994, 66, 996–1010. 14 M. E. Barish, A transient calcium-dependent chloride current in the immature Xenopus oocyte, J. Physiol. 1983, 342, 309–325.
References 15 C. Hartzell, I. Putzier, J. Arreola, Calcium-activated chloride channels, Annu. Rev. Physiol. 2005, 67, 719–758. 16 T. M. Moriarty et al., Beta gamma subunits of GTP-binding proteins inhibit muscarinic receptor stimulation of phospholipase C, Proc. Natl. Acad. Sci. U. S. A. 1988, 85, 8865–8869. 17 M. Pusch, K. Steinmeyer, M. C. Koch, T. J. Jentsch, Mutations in dominant human myotonia congenita drastically alter the voltage dependence of the CIC-1 chloride channel, Neuron 1995, 15, 1455– 1463. 18 D. Firsov et al., Cell surface expression of the epithelial Na channel and a mutant causing Liddle syndrome: a quantitative approach, Proc. Natl. Acad. Sci. U. S. A. 1996, 93, 15370–15375. 19 N. Zerangue, B. Schwappach,Y. N. Jan, L.Y. Jan, A new ER trafficking signal regulates the subunit stoichiometry of plasma membrane K(ATP) channels, Neuron 1999, 22, 537–548. 20 M. Schwake, M. Pusch, T. Kharkovets, T. J. Jentsch, Surface expression and single channel properties of KCNQ2/ KCNQ3, M-type K+ channels involved in epilepsy, J. Biol. Chem. 2000, 275, 13343– 13348. 21 D. W. Hilgemann, C. C Lu, Giant membrane patches: improvements and applications, Methods Enzymol. 1998, 293, 267–280. 22 D. J. Maconochie, D. E. Knight, A method for making solution changes in the sub-millisecond range at the tip of a patch pipette, Pflugers. Arch. 1989, 414, 589–596. 23 R. J. Kaufman, Overview of vector design for mammalian gene expression, Methods. Mol. Biol. 1997, 62, 287–300. 24 G. P. Nolan, A. R. Shatzman, Expression vectors and delivery systems, Curr. Opin. Biotechnol. 1998, 9, 447–450. 25 P. B. Bennett, H. R. Guthrie, Trends in ion channel drug discovery: advances in screening technologies, Trends Biotechnol. 2003, 21, 563–569. 26 W. Stühmer et al., Structural parts involved in activation and inactivation of the sodium channel, Nature 1989, 339, 597–603.
27 A. Accardi, M. Pusch, Fast and slow gating relaxations in the muscle chloride channel CLC-1, J. Gen. Physiol. 2000, 116, 433–444. 28 F. J. Sigworth, in Single-channel Recording, B. Sakmanmn, E. Neher (Eds.), Plenum Press, New York, 1995, pp. 95–127. 29 F. Bezanilla, C. Armstrong, Inactivation of the sodium channel. I. Sodium current experiments, J. Gen. Physiol. 1977, 70, 549–566. 30 C. Saviane, F. Conti, M. Pusch, The muscle chloride channel ClC-1 has a double-barreled appearance that is differentially affected in dominant and recessive myotonia, J. Gen. Physiol. 1999, 113, 457–468. 31 M. Pusch, U. Ludewig, A. Rehfeldt, T. J. Jentsch, Gating of the voltage-dependent chloride channel CIC-0 by the permeant anion, Nature 1995, 373, 527– 531. 32 T-Y. Chen, Structure and function of CLC channels, Annu. Rev. Physiol. 2005, 67, 809–839. 33 M. Pusch, L. Bertorello, F. Conti, Gating and flickery block differentially affected by rubidium in homomeric KCNQ1 and heteromeric KCNQ1/ KCNE1 potassium channels, Biophys. J. 2000, 78, 211–226. 34 E. Neher, Correction for liquid junction potentials in patch clamp experiments, Methods Enzymol. 1992, 207, 123–131. 35 A. M. Woodhull, Ionic blockage of sodium channels in nerve, J. Gen. Physiol. 1973, 61, 687–708. 36 O. Staub et al., Regulation of stability and function of the epithelial Na+ channel (ENaC) by ubiquitination, EMBO J. 1997, 16, 6325–6336. 37 M. S. Awayda, Specific and nonspecific effects of protein kinase C on the epithelial Na+ channel, J. Gen. Physiol. 2000, 115, 559–570. 38 D. E. Goldman, Potential, impedance, and rectification in membranes, J. Gen. Physiol. 1943, 27, 37–60. 39 J. Krusek, Allostery and cooperativity in the interaction of drugs with ionic channel receptors, Physiol. Res. 2004, 53, 569– 579.
143
144
5 Analysis of Electrophysiological Data 40 J. Del Castillo, B Katz, Interaction at end-plate receptors between different choline derivatives, Proc. R. Soc. London, Ser. B. Biol. Sci. 1957, 146, 369–381. 41 J. D. Clements, G. L. Westbrook, Activation kinetics reveal the number of glutamate and glycine binding sites on the N-methyl-D-aspartate receptor, Neuron 1991, 7, 605–613. 42 J. P. Changeux, S. J Edelstein, Allosteric receptors after 30 years, Neuron 1998, 21, 959–980. 43 G. Berecki et al., HERG Channel (Dys)function Revealed by Dynamic Action Potential Clamp Technique, Biophys. J. 2005, 88, 566–578. 44 W. N. Zagotta, T. Hoshi, R. W. Aldrich, Shaker potassium channel gating. III: Evaluation of kinetic models for activation, J. Gen. Physiol. 1994, 103, 321–362. 45 J. Zheng, F. J. Sigworth, Selectivity Changes during Activation of Mutant Shaker Potassium Channels, J. Gen. Physiol. 1997, 110, 101–117. 46 E. D. Koch, B. M. Olivera, H. Terlau, F. Conti, The Binding of {kappa}-Conotoxin PVIIA and Fast C-Type Inactivation of Shaker K+ Channels are Mutually Exclusive, Biophys. J. 2004, 86, 191–209. 47 M. Pusch et al., Mechanism of block of single protopores of the Torpedo chloride channel ClC-0 by 2-(p-chlorophenoxy)butyric acid (CPB), J. Gen. Physiol. 2001, 118, 45–62. 48 F. Conti, Noise analysis and singlechannel recordings, Curr. Top. Membranes Transport 1984, 22, 371–405. 49 S. H. Heinemann, F. Conti, Nonstationary noise analysis and application to patch clamp recordings, Methods Enzymol. 1992, 207, 131–148. 50 C. M. Armstrong, F. Bezanilla, Currents related to movement of the gating particles of the sodium channels, Nature 1973, 242, 459–461. 51 F. Bezanilla, The voltage sensor in voltage-dependent ion channels, Physiol. Rev. 2000, 80, 555–592.
52 N. E. Schoppa, K. McCormack, M. A. Tanouye, F. J. Sigworth, The size of gating charge in wild-type and mutant Shaker potassium channels, Science 1992, 255, 1712–1715. 53 F. Conti, W. Stühmer, Quantal charge redistributions accompanying the structural transitions of sodium channels, Eur. Biophys. J. 1989, 17, 53–59. 54 Single-channel Recording, B. Sakmann, E. Neher (Eds.), Plenum Press, New York, 1995. 55 F. Qin, Restoration of single-channel currents using the segmental K-means method based on Hidden Markov Modeling, Biophys. J. 2004, 86, 1488–1501. 56 D. Colquhoun, C. J. Hatton, A. G. Hawkes, The quality of maximum likelihood estimates of ion channel rate constants, J. Physiol. (London) 2003, 547, 699–728. 57 D. Colquhoun, F. J. Sigworth, in Single-channel Recording, B. Sakmann, E. Neher (Eds.), Plenum Press, New York, 1995, pp. 483–585. 58 S. H. Chung, J. B. Moore, L. G. Xia, L. S. Premkumar, P. W. Gage, Characterization of single channel currents using digital signal processing techniques based on Hidden Markov Models, Philos. Trans. R. Soc. London, Ser. B Biol. Sci. 1990, 329, 265–285. 59 F. Qin, A. Auerbach, F. Sachs, Hidden Markov modeling for single channel kinetics with filtering and correlated noise, Biophys. J. 2000, 79, 1928–1944. 60 L. Venkataramanan, F. J. Sigworth, Applying hidden Markov models to the analysis of single ion channel activity, Biophys. J. 2002, 82, 1930–1942. 61 F. Qin, A. Auerbach, F. Sachs, Estimating single-channel kinetic parameters from idealized patch-clamp data containing missed events, Biophys. J. 1996, 70, 264–280. 62 F. J. Sigworth, S. M. Sine, Data transformations for improved display and fitting of single-channel dwell time histograms, Biophys. J. 1987, 52, 1047–1054.
145
6 Automated Planar Array Electrophysiology for Ion Channel Research Derek J. Trezise
6.1 Introduction
Electrophysiological methods have long been a cornerstone of biomedical science. Basic extracellular field potential recording, for example, has proved invaluable in numerous preclinical and clinical scenarios, as wide ranging as neural network analysis in invertebrates to human electrocardiomyography. Capacitance and impedance measurements, intracellular voltage monitoring, and short circuit current detection have also been extensively used. The patch clamp electrophysiology technique, pioneered in the late 1970s [1–3], allows the direct recording of ionic currents in either a patch or the entire plasmalemmal membrane of a cell. It is capable of resolving single channel gating events on the submillisecond time scale. This exquisite sensitivity and temporal resolution has truly revolutionized ion channel functional analysis, and patch clamp electrophysiology is now well established as the gold standard method. However, the technically demanding nature of this method has restricted the extent and speed at which data can be gathered, and thus almost 30 years on, its full potential has yet to be realized. The advent of automated planar array electrophysiology has begun to address this bottleneck [4, 5]. New commercially available systems offer operator de-skilling and, in some cases, quantal increases in throughput. This chapter introduces automated planar array electrophysiology methods and work practices and aims to highlight key considerations and challenges in the emerging field. Applications ranging from ion channel reagent validation, characterization and high throughput screening will be introduced. The reader is referred elsewhere for reviews that cover automated nonplanar array electrophysiology [5–7].
6.2 Overview of Planar Array Recording
Patch clamp recordings are generally made one at a time via an electrolyte filled glass pipette (1–2 mm tip diameter) positioned on the surface of the cell with the aid Expression and Analysis of Recombinant Ion Channels. Edited by Jeffrey J. Clare and Derek J. Trezise Copyright # 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim ISBN: 3-527-31209-9
146
6 Automated Planar Array Electrophysiology for Ion Channel Research
of microscopy and micromanipulation. Providing that a tight seal between the electrode tip and the cell membrane is formed, electrical access for current injection and recording can be obtained that can permit good voltage control and isolation of the biological current. Full details of this method can be found elsewhere [2, 3, 8, 9]. Fertig and co-workers [10, 11] from Nanion technologies provided the first detailed description of the replacement of the micropipette with a planar, microstructured quartz chip. Cell recording sites were created as small apertures (1 mm diameter, 3–5 MO) in the quartz substrate, by irradiation with accelerated gold ions followed by wet etching. A cell in suspension was attracted to the aperture by suction, and once a high electrical resistance seal had formed, further suction was applied to rupture the membrane to provide access to the cell interior. In this way whole cell recordings were made without cell visualization and pipette micromanipulation (see Fig. 6.1). By creating multiple recording sites in parallel on a chip, quantal increases in patch clamp throughput were promised. Another advantage was that the bioelectric properties of the planar substrates were such that extremely low electrical noise recordings could be achieved. The use of apertures in planar substrates now forms the basis for most commercially available higher throughput patch clamp systems. As of 2005, there are
Fig. 6.1 Schematic diagram of the planar patch clamp principle. In step 1, cells in suspension are attracted to the aperture via suction from beneath the substrate – the measured current evoked by the voltage step yields a hole resistance of 2–5 MO. Once a single cell finds the aperture (step 2) the resistance markedly increases (the ‘giga-ohm’ seal’). Further suction ruptures the mem-
brane to form the ‘whole-cell’ configuration, evident by the large increase in the capacitance spikes (step 3). After the internal solution has fully dialysed the cell, the biological signal (in this case an inward Na+ current) can be studied in the absence (step 4) and presence (step 5) of drug (in this case a Na+ channel blocker).
6.3 Experimental Methods and Design
four established multichannel systems for mammalian cells on the market. Three of these, PatchXpress7000A (Molecular Devices Corporation, www.moleculardevices.com/), Q-patch16 (Sophion Biosciences, www.sophion.dk/) and NPC-16p (Nanion technologies, www.nanion.de) use disposable chips with 16 recording sites whilst the fourth, IonWorksHT (Molecular Devices Corporation) has 384 sites per plate (the ‘Patch plate’). Whilst the precise details of the fabrication methods for these consumables have not been disclosed, it is known from patent literature that glass, polyimide/polyethylene terephthalate and silicon wafer substrates are used in conjunction with etching or laser drilling (see Ref. [5] for review). Seal rate (i. e. fraction of successful recordings), seal stability, and cost are key parameters for the consumable. The 16-channel devices have chips that form tight seals with cell membranes of 1–10 GO resistance and use suction from beneath the aperture to break into the cell for electrical access. In contrast, the IonWorksHT system achieves only 50–300 MO resistance seals to the substrate and uses a membrane permeabilising reagent (e. g. amphoterocin) for ‘perforatedpatch’ access [12]. Overall success rates for recording range from 25–95 % depending on the criteria, cell line and experimental design (see Section 6.5.4). Generally, long lasting recordings (>45 min) can be made with the planar array method since vibration in the recording system does not disturb the electrical seal between the aperture and cell, as occurs with the pipette and cell membrane in conventional electrophysiology. Capacitance and series resistance artifacts arising from the substrate and cell can be fully compensated for within the software of the PatchXpress, Q-patch16 and NPC-16p instruments, akin to conventional patch clamp, but not in IonWorksHT. The major trade off for these different properties is the cost per well of the consumables – the 16-channel systems work out at somewhere between $5–$10 per well whilst the IonWorks patch plates are 50c–$1 per well.
6.3 Experimental Methods and Design
Planar array electrophysiology differs fundamentally from conventional electrophysiology in many ways, perhaps the most important of which are the cell preparation, experimental design and data analysis methods. In conventional electrophysiology cells are plated on glass coverslips or Petri-dishes and visualized by microscopy. A trained electrophysiologist will increase his/her likelihood of success by selecting healthy looking cells with a ‘clean’ cell surface and good morphology. With planar array systems this is not possible – cells are placed in suspension and recorded from ‘blind’. Often the goals of an automated electrophysiology experiment may differ from those tackled with conventional methods, creating new challenges for protocol design and analysis. This is especially true in the pharmaceutical setting where very pragmatic approaches may be taken in high(er) throughput drug screening. The ability to record from greater and greater numbers of cells per se opens up possibilities of ion channel experiments that would
147
148
6 Automated Planar Array Electrophysiology for Ion Channel Research
not otherwise have been attempted. These and other considerations are dealt with in the following sections. 6.3.1 Cell Preparation
To maximize the probability of generating a ‘useful’ recording at a given aperture it is essential to obtain healthy, isolated cells in suspension at an appropriate density. Optimisation of the suspension method can have a major impact on final assay quality, and the key to consistent assays is the reproducibility of these cell preparation steps. Typically, using a CHO-cell line stably expressing the ion channel of interest, cells are first grown to 80 % confluence in cell culture media in a T75 flask and then washed with phosphat-buffered saline and incubated for 5 min in a cell dissociation solution (e. g. versene EDTA). The lifted cells are then centrifuged (4000 rpm for 2 min) and the cell pellet is washed again and resuspended in the external recording solution. A clean cell preparation minimizes the possibility of cell debris contacting the aperture and preventing a good recording. For an IonWorks experiment the suspension is gently triturated for 60 s with a Gilson pipette to obtain viable, dissociated cells. Final cell densities of (0.5–2)6106 cells ml –1 are optimal. Shorter trituation times appear to work better for the 16-channel systems. For HEK cells, a trypsin-based cell dissociation solution generally works better than versene and more trituration is required to avoid cells forming clumps. Common problems are either insufficient or excessive enzymatic digestion or trituration, which can detrimentally impact both seal rate and length of the recording. In most cases the cell preparation is improved by a short settling period (3–10 min) prior to use. Of course, the creation of good cell suspensions is of little use if the fraction of cells that exhibit the ionic current of interest is unacceptably low. In the extreme, certain transient transfection methods and host cell combinations may yield <5 % of expressing cells in recombinant systems (see Chapter 4). In this case, planar array recording can be disadvantageous compared to conventional electrophysiology where cell markers such as GFP can be used to guide selection [13]. Experiments with native cell preparations on dorsal root ganglion or hippocampal neurons may also be complicated by the presence of glial cells or astrocytes unless methods are applied to exclude these. In contrast, with good stable cell lines >95 % of cells can be made to express the channel of interest. For this reason these have been the major focus for most automated electrophysiology work. Indeed, the higher throughput systems like IonWorksHT are extremely useful tools for validating clonal cell lines per se by functional expression. Large numbers of clones (50– 100) can be grown and prepared for testing within the same day. Promising clones, based on expression criteria such as the fraction of ‘expressors’ and the median current amplitudes, can be rapidly identified and selected for more detailed profiling (see Chapter 4, Fig. 4.2). For some cell lines there is a strong relationship between the growth conditions and ion channel expression, highlighting the need for attention to detail in cell
6.3 Experimental Methods and Design
Fig. 6.2 Upregulation of hERG expression at low temperature, quantified by automated electrophysiology. The histograms show the cell population distributions for peak tail currents (measured at –40 mV) from a CHO cell line stably expressing hERG. Bin size 0.1 nA. Cells were cultured for 24 h at either 37 8C (A)
or 30 8C (B). The fraction of cells with currents >0.1 nA increased from 74.8% to 95.8% (P<0.001) by lowering the culture temperature, and the mean current amplitude increased from 0.28 nA to 0.93 nA (P < 0.001; n = 657 at 37 8C and 686 at 30 8C). M. X. Chen and D. J. Trezise, unpublished data.
culture. For example, hERG K+ channel stable cell lines appear to lose expression when the confluency rises above 90 %, presumably as a consequence of specific regulation on cell to cell contact or as part of the growth cycle [14,15]. Conversely, if the same cells are grown at 30 8C for 24 h prior to the experiment a marked increase in channel expression occurs (Fig. 6.2). From anecdotal observations different media, serum and even plasticware can also affect ion channel expression, although to date automated electrophysiology data has yet to be described. 6.3.2 Cell Sealing and Recording
A prerequisite for high-fidelity patch clamp is the high resistance seal that forms between the cell and the pipette tip, or the substrate in the case of planar electrophysiology. The microscopic processes that occur at this interface are not fully understood [10, 16, 17], but what is clear is that a clean contact surface is essential. Thus, the IonWorks patch plates come dry-packed and Aviva SealChips are stored in double distilled water to avoid contamination with dust or salts. It is equally important to ensure that all recording solutions that will be in contact with the patch wells are filtered (e. g. 0.22 mM). Microscopic particles can block the apertures and are a major cause of poor performance. Another common problem is blocking of
149
150
6 Automated Planar Array Electrophysiology for Ion Channel Research
the apertures by air bubbles, and it is strongly advised to degas solutions too. Generally, prior to addition of cells and in filtered and degassed ionic buffers, >95 % of holes on commercially supplied chips/plates are open (unblocked) and have acceptable resistances (1–5 MO). As with glass pipettes in conventional electrophysiology it appears that recording chips cannot be easily reused. Once positioned in the instrument, suction from beneath the substrate is applied to attract a cell to the aperture and help form the seal. Electro-positioning, whereby an electrical field is created to attract the polarized cell to the recording aperture, has been attempted but appears to offer no advantage [18, 19]. In systems such as PatchXpress and Q-patch the suction parameters are entirely user configurable and can be optimized depending on the cell type that is being used. For more fragile cells gentle suction ramps appear to work best whilst for sturdier cells rapid GO seals can be achieved with steeper ramps or steps (e. g. negative pressure ramp –10 to –32 mmHg at 1.6 mmHg s–1 for 25 s [20]). In a detailed analysis of the SealChips used with the PatchXpress device, Xu and coworkers [19] found that for CHO-Kv cells >1 GO seals could be obtained on >90 % occasions. 82 % of seals occurred within 15 s of the cell landing at the recording site. With HEK-hERG cells, both Dubin et al. [20] and Quinn [21] had lower success rates with GO seals on 50–60 % occasions. Depending on the cell type, >1 GO seals were obtained for 40–95 % of trials with a prototype of the Sophion Q-patch system [22]. For IonWorksHT, the suction parameters cannot be edited from the graphical user interface. Somewhat lower resistances are observed (100–300 MO) but >95 % seal rates can routinely be achieved [12, 23]. Taken together, these and other data illustrate that cell sealing methods to apertures in planar substrates are now well established and no longer represent the technical challenge they once were. To attain the whole cell recording configuration, further suction is applied to rupture the cell membrane in the aperture. Suction to each well/cell is controlled individually so that it can be disengaged immediately after the ‘whole cell’ configuration is achieved. Excessive suction can destroy the seal and recording and this is avoided by continuous monitoring of the cell capacitance coupled to a feedback circuit to the suction control. One common problem is ‘premature breakthrough’ where membrane rupture occurs before a high resistance membrane seal is achieved [20, 21]. Overall, whole cell recordings with membrane resistances >1 GO can be achieved for between 40–60 % of cells depending on the system and/or cell type [19–22, 24]. Whilst this is clearly impressive, since perhaps 6–10 whole recordings may be obtained on a 16-channel seal chip, there is scope for improvement here. It will be interesting to see whether this single cell suction method for whole cell access proves truly scalable with ever larger parallel arrays. ‘Perforated-patch’ clamp with membrane permeabilising agents such as amphoterocin, gramicidin or b-escin is arguably a more elegant solution commensurate to higher throughput planar array electrophysiology [12, 25, 26]. Cells can be treated homogeneously by exposure of the cell surface at the aperture to a fixed concentration of agent, removing any requirement for capacitance/suction feedback circuitry. Provided that sufficient time is allowed for permeabilisation, stable low access resistance pathways (<15 MO) can be consistently obtained. Indeed, in the
6.3 Experimental Methods and Design
authors laboratory and from published findings the success of gaining long term (>45 min) stable electrical access once a seal is achieved is >95 %. For some experiments, the low permeability of the ‘perforation’ to large species and ions (e. g. Ca2+) restricts flexibility in the study of certain ion channels i. e. Ca2+-activated K+ channels. However, incomplete dialysis of the cell is generally advantageous since it preserves key regulatory pathways and components in the cell, which leads to more stable current measurements. This is a major consideration when characterizing channels that rapidly run down (e. g. voltage-gated Ca2+ channels, KCNQ K+ channels) and when conducting pharmacological studies (see Section 6.3.4). Minimising potential errors in voltage command and control and isolating extraneous nonbiological ionic currents from the final measured signal are other prerequisites for high quality patch clamp recordings [3, 27]. Theoretically, the challenges of signal filtering, sampling frequency, leak and offset corrections, and capacitance and series resistance compensation are no different to conventional patch clamp. Many of the amplifiers used in planar array electrophysiology are very similar or identical to those found on a conventional electrophysiology system, and the correction algorithms applied in software are the same. For the systems that treat each cell individually, criteria for several membrane parameters can be configured (e. g. access resistance <15 MO, membrane resistance >500 MO) for quality assurance. If any parameters drift outside the acceptable range corrective action such as compensation readjustment or the application of further suction is triggered. If the problem persists the recording is terminated. In practice, with planar array recordings, membrane parameters and compensation comparable to conventional electrophysiology can be achieved. In the IonWorksHT system a fundamentally different philosophy to data capture is applied. Certain error sources are deemed insignificant and are ignored (e. g. capacitance transients) whilst for others, corrections are made across the entire plate for all cells rather than on a cell by cell basis. For many experiments demanding higher throughput, especially expression profiling and drug screening this pragmatic approach is perfectly acceptable. The initial voltage difference (Voff) between the head and ground Ag/AgCl electrodes in the open circuit solutions, for example, is measured in every well and then the median value from 384 is taken and applied to all wells. Provided that the electrodes are well chlorided so that the standard deviation of VOff across the plate is low (<3 mV) in the vast majority (>95 %) of the cells <10 mV error will occur. Cells with unacceptably large VOff errors (i. e. >15 mV) can be removed by post-hoc filtering. Leak correction is not performed by the traditional ‘P/4’ method in which a scaled signal 1/4 of the size (and the same length) of the test waveform is applied 4 times and summed for subtraction from the acquired signal. Rather, the current obtained from a fixed duration and size step signal (e. g. –10 mV, 40 ms) is scaled assuming totally linear leak prior to subtraction. In practice, in most cases <50 pA of poorly corrected leak is observed. Given that comparatively low seal resistances are generally obtained (100– 300 MO), and hence the relative contribution of leak is high, this method works remarkably well. The main exception to this is when working with leak-like channels (e. g. twin-pore K+ channels, Kirs) where discrimination of the biological sig-
151
152
6 Automated Planar Array Electrophysiology for Ion Channel Research
nal from the nonbiological leak can prove troublesome. Series resistance is not compensated for in the IonWorks system, and thus large ionic currents (>2 nA) should be studied with caution to avoid unacceptable voltage errors. 6.3.3 Drug Application
There are several key considerations in the design of drug application systems for automated electrophysiology. First, how much compound sample is likely to be available? In a pharmaceutical setting, or when peptides or toxins are being studied, often only a few mL (2–200) of test compound can be afforded. Designs that incorporate low volume (10–50 mL) wells have evolved for this reason (see e. g. Ref. [28]). Next, how rapidly and for how long do drugs need to be applied? Fluid exchange times of <30 ms are required to fully resolve fast-ligand gated ion channel events such as nAChR and NMDA receptor activation, otherwise receptor desensitization prematurely curtails the signal. Short application times (1–10 s) are also desirable to avoid longer term desensitization or tachyphylaxis. In contrast, long incubation times (2–20 min) may be required for small molecule ion channel blockers to fully equilibrate or when long voltage-command protocols are being studied (e. g. steady-state inactivation curves) The third consideration is how many different compounds or concentrations of the same compound need to be applied to the same cell? Cumulative concentration-response curves for single test compounds are often constructed with conventional electrophysiology methods by applying increasing concentrations of drug immediately after the previous concentration has reached equilibrium. The long recordings (>45 min) achievable with the patch clamp method provide this option. If different test compounds are to be studied on the same cell the ability to rapidly and fully washout the previous drug is important. For studying antagonists or modulators of ligand-gated channels it is necessary to preincubate and then co-apply with the reference agonist. In practice, there is often a strong trade off in speed between a desire to maximize the data that can be captured from a single cell versus the ensuing complex logistics of the experimental design and data analysis (see next section). This is compounded by the unpredictability of novel drug behavior (e. g. differences in association rates and washout times). Finally, in what format are the compounds of interest likely to be supplied? If only low numbers of compounds are to be profiled this is largely of academic interest. However, in almost every drug screening environment compounds are supplied on standard geometry microtitre plates (96 well, 384 well), for which the liquid handling of the automated electrophysiology system must be compatible. With these considerations in mind, different vendors have implemented either perfusion systems based on microfluidics, exchange methods in which the external buffer can be largely replaced, or static wells in which additional volume (drug) can be added followed by a mixing step. In the Sophion Q-patch, the consumable contains a sophisticated laminar flow channel system driven by a passive capillary pump – this requires <10 mL drug and claims liquid exchange times
6.3 Experimental Methods and Design
commensurate with the study of most ligand-gated ion channels (e. g. <100 ms). Washout is possible and multiple drug applications can be made to the same cell. The 16 recording channels are supplied with drugs via a 4-channel pipettor which aspirates from either a 96- or 384-well drug plate. The Nanion NPC-16p also uses microfluidics for drug application and receives low volume (<15 mL) samples from a pipetting arm. Fluid exchange times of <50 ms are claimed. Seal chips used in the PatchXpress instrument have wells of larger volume (up to 100 mL) – a single pipettor is used to first aspirate buffer from the recording well and then rapidly replace it with a drug containing solution – the time to 90 % exchange is several hundred milliseconds. All three systems use control software to schedule drug addition according to when cells have achieved pre-set membrane and signal quality assurance criteria. For IonWorksHT there is considerably less flexibility in the drug application system, which is based on a ‘mix and read’ method. Compounds are applied to the recording wells according to a predetermined schedule via a 12-channel fluidicshead, which aspirates from a 96- or 384- well drug plate. 3.5 mL of drug is added to the 7 mL of buffer in the well and mixed to create a 1 : 3 final dilution. Optimisation of the mixing conditions (i. e. number of mixes, mix volume, pipettor speed) is important and can negatively impact the observed pharmacology if done incorrectly. Currently it is not possible to make more than one drug application to a given cell, or to read during the drug addition time. In its current guise, this precludes the study of even slow ligand-gated channels since the minimum time between drug addition and data acquisition is >30 s The philosophical difference here is that with so many recordings on the plate (up to 384) it is possible to accumulate single (drug) point data on many cells for amalgamation (e. g. to construct concentration–response curves) more quickly and robustly than it is to apply multiple concentrations or drugs to individual cells. An inventive design feature is the 1 : 4 transfers from the drug plate to the patch plate in which each drug well is applied to 4 different patch wells. This approach circumvents any requirement to only add drugs to ‘good cells’. If >75 % of recordings are acceptable then probability theory states that >99 % of the compounds from a 96-well drug plate will see one or more good cells [12, 23]). It is only since the first generation planar array automated electrophysiology systems have been used in ‘real world’ drug screening that certain issues regarding compound adherence and adsorption to recording substrates, compound plates and pipetting systems have come to light. When comparing with conventional patch clamp data, an underestimation of the potency of certain compounds has been observed. This is especially true for highly lipohilic, ‘sticky’ drugs such as astemizole and terfenadine, for which 10–100-fold drop in potency for block of hERG channels has been observed [20, 23, 24, 29]. For PatchXpress, adding the compound three times to the recording well appears to resolve some of this discrepancy – whilst the exact reason for this is unclear it may be that compound in the sample addition first occupies binding sites on the substrate or plastic rather than the cell. Dispensing from glass-coated rather than polypropylene drug plates goes someway toward mitigating this and can increase compound potency 3–5-
153
154
6 Automated Planar Array Electrophysiology for Ion Channel Research
Fig. 6.3 Effect of compound plate type on inhibition of Kv7.1+minK K+ currents by L735281 [36] measured using IonWorksHT. (A) shows mean log concentration–response curves for L-735281 obtained when the drug was plated on either polypropylene- (.) or
glass- (o) coated 96-well plates. Note the 6fold increase in potency and shallowing of the Hill slope from 2.37 to 0.90 with the glasscoated plates. (B) shows the individual derived pIC50 values from 39 (glass) and 20 (plastic) concentration–response curves.
fold (see e. g. Ref. [21]; Molecular Devices technical note). This has been observed both on the IonWorks and PatchXpress platforms. As the technology becomes more widely adopted it will be interesting to see whether fewer complications are observed with the Q-patch system in which the laminar flow channels are made of glass. Carry-over of compounds on the pipette tips from one well or plate to the next is another pitfall to be aware of – with some systems that use fixed tips 100 % ethanol and water washes between drug additions are required to avoid this. Returning briefly to the challenge of fast drug application and the study of ligand-gated ion channels, it is fair to say that the existing platforms do not fully meet the requirements of ultrafast addition (<30 ms) coupled to significant parallelization. With a perfusion system and single cell recording, experimental design will always be complex if interactions between multiple concentrations of gatingligand and modulator/blocker are to be studied. This is compounded by the need to make ultrashort agonist applications with rapid washout. One interesting nonplanar array automation innovation in this area is the DynaflowTM microfluidic chip [30, 31], designed to integrate with the recording stage of a conventional patch clamp set up. This device is a multi-channel (currently 48) laminar flow system that creates an array of continuous, controlled liquid environments through which a cell can be ‘scanned’. The rise time when moving from one flow channel to the next can be a few milliseconds. Preloading of the chip to create the flow channels markedly simplifies experiments with multiple combinations of ligand and modulator. Integration of this technology with a parallel patch clamp system could provide a solution to most of the challenges of electrophysiology drug screening at fast ligand-gated ion channels.
6.3 Experimental Methods and Design
6.3.4 Experimental Design and Data Analysis
On the whole the experimental design and analysis methods deployed in the 16channel systems are very similar to those used in conventional electrophysiology, albeit on a larger scale. The acquisition and analysis softwares provide great flexibility and few constraints. For biophysical characterization of heterologously expressed ion channels it is relatively straightforward to script diverse voltage command protocols that can be concatenated. Once the experiment is started the instruments can gather data unattended. The same is true for simple drug addition protocols. In contrast, by virtue of the extent of the parallelization and the drug application philosophy, the IonWorksHT approach presents a new set of challenges for experimental design and analysis. These are a combination of many that are continually faced by the high throughput screening community with others more immediately recognizable to the classically trained electrophysiologist. This is best exemplified by the manner in which data are ‘quality controlled’. All patch clampers are used to applying good judgment as to whether data from a given cell are valid – cells can be rejected for many reasons such as the seal becoming unstable over time, the holding current or access resistance increasing, or the biological signal proving too small to measure reliably. In many cases this is done more subjectively than objectively. With IonWorksHT, data from >2500 cells can be gathered in a single day – far too many to go though one by one, even for the most dedicated researcher. Nevertheless, it is important to exclude poor recordings from subsequent analysis to avoid inappropriate conclusions. This is achieved by setting simple filters in the software such that, for example, any cells with seal resistances <50 MO or holding currents >50 pA are excluded. Another useful filter is on current amplitude to eliminate poorly expressing cells (i. e. peak current <100 pA). More complex ‘stability’ filters can be created such that differences between the pre-drug and post-drug membrane parameters are monitored, e. g. pre- minus post- holding current > or <50 pA. What may be less familiar are the methods used to monitor and control for time-dependent changes in the biological signal of interest, and/or effects of drug vehicles. Commonly, in conventional patch clamp an ionic current may be evoked repeatedly by a voltage step, for example, and monitored until it is stable. At this point the drug vehicle may be applied. Providing that only a small change in signal (e. g. <10 %) is observed, the operator moves on to applying test compounds to the cell. The contribution of time-dependent (not drug-induced) changes in the signal (if any) is controlled either by washout of the drug back to the original baseline, or in some cases by a correction factor from extrapolation of the signal vs. time plot. With IonWorksHT, long term repeated channel gating, multiple drug addition and washout are not possible and hence a different technique is required. In the author’s laboratory, two columns of the drug plate, corresponding to 1/6 of the total wells, are used for assay plate quality control. The first (low, column 11) contains the drug vehicle, usually 0.3 % final dimethylsulfoxide (DMSO) and con-
155
156
6 Automated Planar Array Electrophysiology for Ion Channel Research
trols any vehicle and time-dependent changes between the post- and pre-addition signals. The second (high, column 12) contains a supramaximal concentration of reference compound (e. g. a blocker of the ion channel under study) to ensure that the biological signal is not contaminated by other conductances or leak. At the end of the experiment, and after filtering of cells with poor membrane parameters (see above), a Z’ value can be calculated for the pre- and post-addition signals using the equation Z’=1– (3 (s.d.low + s.d.high)/(meanlow–meanhigh)) [32]. Z’ values are a universally accepted statistical measure of assay quality and reflect signal dynamic range and the data variation associated with the signal measurements. Assay plates can thus be accepted or rejected based on Z’ criteria – in our hands Z’ values >0.3 are suitable for single concentration screening, and for optimized assays values of 0.6–0.8 can be routinely achieved. For concentration–response curve analysis on IonWorksHT, data must be amalgamated from a sufficient number of cells from the plate to provide a precise quantitation of the pIC50 (or pEC50) value of the drug of interest. The drug plate format needs to account for this and the practicalities of liquid handling for generating compound plates. Of course, a major consideration is also cost, for which a goal in a pharmaceutical setting is to find the optimum balance between throughput and data quality. We originally tried ‘cross-plate’ dilutions whereby the highest concentration of 8 different test compounds was positioned in A1, B1, C1 through to H1, and serial dilutions were made across the plate. However, row-to-row carry over with certain potent “sticky” compounds occurred which proved difficult to eliminate through tip washing. By plating different compounds at the highest test concentration in row H (1–10), and diluting 1 : 3 up the plate, the sequencing of the fluidics head is such that potential carry over cannot occur between compound columns (i. e. the same dispensing needle sees only a single compound, and moves from low concentrations to higher ones). Moreover, 10 rather than 8 concentration–response curves comprised of data from up to 32 cells could be obtained, with columns 11 and 12 as controls. In practice, after filtering, 20–30 cells usually remain for inclusion in the four parameter equation logistic fitting for IC50. It is at this point that the balance between over-filtering (too few data included in the curve fit) versus under filtering (poor data is included which skews the curve fit) is most apparent. After filtering, we normalize the post: pre drug values to the high and low controls in columns 11 and 12, and then use all points, rather than the mean of the 0–4 cells obtained at each drug concentration, for curve fitting. In this way, each raw data point can be equally weighted. As with other screening methods, it is good practice to include a reference compound concentration–response curve for additional quality control for which if the pIC50 falls outside acceptable limits (e. g. >0.5 log units from a historical mean value) the plate is rejected. Another analysis challenge and opportunity with the higher throughput systems is kinetic determinations. Patch clamp electrophysiology provides detailed temporal information, which can be used to intepret conformational state transitions, mechanism of drug action and the function of regulatory subunits, as examples (see Chapter 5 and Ref. [33] for review). Typically, a kinetic is quantified by fit-
6.3 Experimental Methods and Design
Fig. 6.4 Compound plate format and curve fitting example for concentration–response curve analysis using IonWorksHT. A, shows a summary view from the software of the 96 well plate with ‘up-plate dilutions’ for 10 different compounds arrayed at the highest concentration in H1, H2 etc and diluted up the plate 1 : 3 (see text for further explanation). The values in each well indicate the number of cells that have ‘passed’ exclusion filters such as low seal resistance and low peak cur-
rent amplitude. Compound wells for which >50 % block of the signal occurred are shaded, such that active compounds can be readily visualized. The processed data for a single compound (column 9) is shown in B. The IC50 value is obtained by fitting a fourparameter logistic function to the data from all of the post filtered cells, in this case 29. One ‘outlier’ (circled) is detected by the curve fitting software and is excluded from the curve fit.
ting a component of the current waveform to an appropriate function (e. g. single or bi-exponential decay functions often well describe the tail currents observed when voltage-gated channels deactivate). However, this process usually requires some ad hoc interaction with the data by the analyst, which, for thousands of cells, would constitute a significant computational overhead. It should also be borne in mind that any ultrafast kinetic determination is likely to be imperfect given the absence of capacitance (and series resistance) compensation. Nevertheless, depending on the question in mind, pragmatic approximations to kinetic behavior can prove extremely valuable. This is best illustrated by example. Activation of Kv7.1 (KCNQ1) K+ channels is markedly slowed when the regulatory subunit minK is co-expressed [34, 35]. In that minK is not pore forming per se (it is a single trans-membrane domain protein) the best way to detect its presence in a given cell is to use the slower activation, compared to Kv7.1 alone, as a marker. In building stable cell lines for drug screening we confirmed the presence of minK by setting simple measures at a fixed time point (150 ms) after the gating step and at full activation (4 s). The ratio of these two values (I150 :IPeak) was markedly lower (<0.4) in the presence than in the absence of minK (>0.6), indicative of slower activation. The power of this approach is that we could rapidly and confidently determine, based on a population analysis (n>200 cells), what fraction of cells within the Kv7.1+minK stable cell line were not expressing the minK subunit (i. e. exhibited a high I150 :IPeak value). We found that <2 % of KCNQ1 expressing cells had rapid kinetics and were therefore low minK expressors (see Fig. 6.5).
157
158
6 Automated Planar Array Electrophysiology for Ion Channel Research
Fig. 6.5 Kinetic estimations to determine the presence of regulatory subunits in cell lines using the IonWorksHT electrophysiology platform. (A) shows data from a Kv7.1 CHO cell line and (B) shows data from a Kv7.1 line stably transfected with the minK subunit. Expression of minK markedly slowed the activation of Kv7.1
as measured ‘classically’ by fitting single exponential functions to individual cells (n=8) and also by using simple metrics and population statistics (see text for full explanation). Abscissae: ratio of current amplitude at 150 ms to the peak current. Ordinate: cell count bin size 0.01. The time/amplitude bar is 1 s and 0.5 nA.
As noted earlier, the IonWorks system cannot simultaneously voltage clamp and apply drugs. The 48-channel electronics (E-) head must move away from the wells, transiently unclamping the cells, whilst the fluidics (F-) head dispenses and mixes. For certain voltage-gated channels, the impact of this transient unclamping requires consideration in the experimental design. Channels that are strongly inactivated at unclamped membrane potentials (e. g. –40 to –5 mV) may need a long recovery period once clamping resumes. Certain drugs with strongly voltage-dependent binding may also block more potently during the unclamped (depolarized) phase and may only slowly unblock upon repolarization. Other compounds may require prolonged repeated channel gating (‘use-dependence’) to block fully, which is not possible during the mixing phase. Despite these challenges, with understanding and care it has proved possible to configure robust, meaningful assays for most members of the voltage-gated super family attempted so far.
6.4 Overall Success Rates and Throughput
As has been highlighted, there are a number of reasons why a given planar array recording may prove unsuccessful or yield unsatisfactory data. Blocked apertures, unhealthy cells, premature whole cell access, inappropriate or unstable mem-
6.5 Population Patch Clamp
brane parameters, small or unstable ionic currents, or slow drug washout can all, either individually or in combination, reduce the final output. Of course, the ‘success’ criteria themselves will be strongly dependent on the experimental objective. Entire plates, rather than individual cells might be failed based on Z’ statistics or the discrepant pharmacology of a standard compound in a drug screening mode. There is currently little published data on the overall attrition that these factors contribute, and less still on the overall throughput benefit afforded by the current systems. For sure, the ‘real world’ does not always match the vendor claims. For the PatchXpress system, overall success rates of 25–36 % were obtained for compound screening at the hERG K+ channel [20, 21, 24]. This particular assay comprised stringent QC criteria around membrane parameters, current stability and, in the Dubin study, the activity of an internal control compound on each cell. There was no compromise in the data quality compared to conventional electrophysiology and 3–4 point IC50 values were generated from each cell. The overall increase in throughput was 3- to 4-fold. For this type of study, the other 16-channel systems (Sophion Q patch, Nanion NPC-16p) are likely to offer similar efficiency gains. For screening cell lines, biophysical characterisation and for less demanding pharmacological protocols 5–10-fold increases may be obtainable. With IonWorksHT, 8 plate runs of 45 min each are easily achievable in a working day, corresponding to 86384 = 3072 attempted recordings. For most cell or compound profiling applications with high expressing cell lines, using success criteria of cells >50 MO, Ihold <50 pA, peak current > 200 pA and a plate Z' value of >0.3 for stability, useful data can be gathered on >300 cells per run, or >2400 cells per day ([12] and Trezise and Dale, unpublished data). If the application is screening cell clones for expression this equates to >100-fold efficiency gain compared to conventional patch clamp. For compound profiling, once the 1 : 4 transfers, data amalgamation across cells and control wells are accounted for, 640 single test compounds or 64 10-point concentration–response curves can be generated within a day. This corresponds to a throughput increase of 30–80-fold. Whilst certain detailed biophysical analyses may expose the limitations in working with low seal resistances and without capacitance or Rs compensation, for most screening applications this trade off for the quantal throughput increase is perfectly tolerable. Indeed, for the first time, this level of throughput is sufficient to support iterative structure–activity relationship screening by electrophysiology for a modern medicinal chemistry drug discovery program.
6.5 Population Patch Clamp
In an exciting new development in the field, Jan Hughes and Alan Finkel at Molecular Devices have shown that planar array electrophysiology methods can be applied to multiple cells in a single well, a principle termed ‘population patch clamp’ (PPC). In modified IonWorks plates with 64 apertures microfabricated in each well, robust ensemble ionic currents were observed with Kv1.5, NaV1.5 and hERG
159
160
6 Automated Planar Array Electrophysiology for Ion Channel Research
Fig. 6.6 Screening performance for a voltagegated Na+ channel assay using IonWorksHT to exemplify assay stability and quality control. Each panel shows data taken from 64 independent runs (assay plates), from which different variables were monitored. A, shows the number of blocked wells and the number of good seals (or recordings) from a possible 384. B, shows the median seal resistances at different time points in the experiment and C,
the average number of usable currents per plate and peak current amplitude. In D the Z’ value and pIC50 value for a standard compound added to each plate are shown. Plate 62 was the only failure based on the QC criteria of Z’ < 0.3 and standard pIC50 value within ±0.5 of the rolling mean value (i. e. 4.7 ± 0.5). T. J. Dale and D. J.Trezise, unpublished data.
expressing stable cell lines. By theory, with resistors in parallel, the inverse of the measured resistance across the entire well will simply be the sum of the inverse of the resistance of the individual apertures (1/Rmeasured = S 1/Rindividual). Provided that a high fraction of the apertures are occupied by cells (which display high resistance) the ‘short-circuit’ contribution of the unoccupied (low resistance) wells becomes insignificant. This relationship is modeled in Fig. 6.7. For robust leak correction seal resistances >80 MO are required, which corresponds to an aperture occupancy of >95 %, assuming a single cell resistance of 120 MO. The measured currents become the sum of the individual currents arising from each cell, and can be simply scaled (i. e. divided by 64) to provide quasi-average single cell currents. As far has been determined, ionic currents measured using this approach do not markedly differ biophysically or kinetically from single cell recordings. For certain applications in the screening arena, this simple idea of PPC offers enormous potential advantages over single cell automated electrophysiology. The
6.5 Population Patch Clamp
Fig. 6.7 Principle of population patch clamping with IonWorks Quattro. Recordings are made from a population of cells on planar substrates with multiple apertures in the same well. These behave as resistors in parallel (A). Assuming that any individual aperture is either occupied or not, with respective resistances of 120 MO and 10 MO, the equation in (B) describes the relationship between the
fraction of occupied holes and the measured resistance across the well. For these resistance parameters in the model >95 % occupancy is required for a final equivalent resistance of >75 MO (C), which is adequate for acceptable leak subtraction and voltage control. Remade from Molecular Devices Corporation with permission.
requirement to build cell lines in which >90 % of cells express sufficient channels of interest will become less stringent. As long as the mean current is measurable it will be of less importance if a higher fraction of cells are low expressors. Consistency of data from well to well should also be markedly improved, as the impact of cell to cell variability is minimized by data averaging. Indeed, with optimized assays a good signal should be obtained in every well, as is the case with the vast majority of plate readers (e.g fluorescence and luminescence). Compounds can now be added 1 : 1 to assay wells rather than 1 : 4 as in the original IonWorksHT platform yielding an immediate 4-fold increase in drug screening throughput. With high quality assays it may be possible to dispense with the pre-drug read altogether (since they will all be very similar) thus shortening plate read times significantly. Channel multiplexing may also be easier – by mixing cell lines that express ion channels with different phenotypes (e. g. a voltage-gated Na+ channel and voltage-gated K+ channel) two (independent) signals could be resolved in the same well, without the need to generate a single cell line that expressed both channels (see Chapter 4). This approach is very powerful for selectivity profiling, and further increases the ‘value’ from each well screened. Overall, PPC should bring automated electrophysiology ever closer to the true ‘mix and read’ screening paradigm that is so desirable in high throughput screening.
161
162
6 Automated Planar Array Electrophysiology for Ion Channel Research
6.6 Summary and Perspective
Automated planar array recording is now an established variant of patch clamp electrophysiology, offering operator de-skilling and higher throughput. Its primary use so far has been for rapid drug screening in the pharmaceutical and biotechnology sectors where it has proved truly enabling. Undoubtedly, as the capital and operational costs fall it will become more widely adopted in the academic community where more fundamental questions in the ion channel field will be addressed. As with any new technology, challenges and considerations arise with use, some foreseen and others completely unexpected. The major differences between automated planar array recording and conventional patch clamp lie in the cell preparation, drug application and experimental design and analysis. With understanding, thought and care it has proved possible in several drug screening laboratories to exploit the potential of the first generation commercially available instruments. A major area of current unmet need is for integrated fast drug application coupled to a quantal increase in throughput – this will hopefully be addressed in the next generation of instruments. Excitingly, population patch clamp, which extends the patch clamp method to a multicell recording paradigm, should bring further improvements in quality and throughput. In the medium term, automated planar array electrophysiology should help fulfil the remaining untapped potential of the patch clamp method, and accelerate the search for new ion channel therapeutics.
Acknowledgments
The author gratefully acknowledges the support and input of colleagues at GSK, notably Tim Dale and Claire Townsend, who have made significant contributions to the deployment of high throughput electrophysiology methods.
References 1 Neher E., Sakmann B., Single-channel currents recorded from membrane of denervated frog muscle fibres, Nature 1976, 260(5554), 799–802. 2 Hamill O. P., Marty A., Neher E., Sakmann B., Sigworth F. J., Improved patch-clamp techniques for high-resolution current recording from cells and cell-free membrane patches, Pflugers Arch. 1981, 391(2), 85–100. 3 Sakmann B., Neher E., Patch clamp techniques for studying ionic channels in excitable membranes, Annu Rev. Physiol. 1984, 46, 455–472. (Review.)
4 Sigworth F. J., Klemic K. G., Patch clamp on a chip, Biophys. J. 2002, 82(6), 2831–2832. 5 Wang X., Li M., Automated electrophysiology: high throughput of art, Assay Drug Dev. Technol. 2003, 1(5), 695–708. 6 Wood C.,Williams C., Waldron G. J., Patch clamping by numbers, Drug Discovery Today 2004, 9(10), 434–441. 7 Trezise, D. J., Patch clamp electrophysiology steps up a gear, Eur. Pharm. Rev. 2005, 2, 69–73.
References 8 Cahalan M., Neher E., Patch clamp techniques: an overview, Methods Enzymol. 1992, 207, 3–14. 9 Molleman, A., Patch Clamping: an Introductory Guide to Patch Clamp Electrophysiology, John Wiley & Sons Ltd, 1992. 10 Fertig N., Blick R. H., Behrends J. C., Whole cell patch clamp recording performed on a planar glass chip, Biophys. J. 2002, 82(6), 3056–3062. 11 Fertig N., George M., Klau M., Meyer C., Tilke A., Sobotta C., Blick R. H., Behrends J. C., Microstructured apertures in planar glass substrates for ion channel research, Receptors Channels 2003, 9(1), 29–40. 12 Schroeder K., Neagle B., Trezise D. J., Worley J., Ionworks HT: a new highthroughput electrophysiology measurement platform, J. Biomol. Screen. 2003, 8(1), 50–64. 13 Marshall J., Molloy R., Moss G. W., Howe J. R., Hughes T. E., The jellyfish green fluorescent protein: a new tool for studying ion channel expression and function, Neuron 1995, 14(2), 211– 215. 14 Crociani O., Guasti L., Balzi M., Becchetti A., Wanke E., Olivotto M., Wymore R. S., Arcangeli A., Cell cycle-dependent expression of HERG1 and HERG1B isoforms in tumor cells, J. Biol. Chem. 2003, 278(5), 2947–2955. 15 Cherubini A. et al., Human ether-a-gogo-related gene 1 channels are physically linked to {beta}1 integrins and modulate adhesion-dependent signaling, Mol. Biol. Cell 2005, 16(6), 2972– 2983. 16 Corey D. P., Stevens, C. F., Science and technology of patch recording electrodes, in Single Channel Recording, Sakmann, B., Neher, E. (Eds.), Plenum Press, New York 1983, pp. 53–86. 17 Opsahl L. R., Webb W.W., Lipid-glass adhesion in giga-sealed patch-clamped membranes, Biophys. J. 1994, 66(1), 75– 79. 18 Worley J., Li M., Location is key – recent progress in single-cell-based high-throughput assays, Drug Discovery Today 2001, 6(9), 454.
19 Xu J., Guia A., Rothwarf D., Huang M., Sithiphong K., Ouang J., Tao G., Wang X.,Wu L., A benchmark study with sealchip planar patch-clamp technology, Assay Drug Dev. Technol. 2003, 1(5), 675–684. 20 Dubin A. E. et al., Identifying modulators of hERG channel activity using the PatchXpress planar patch clamp, J. Biomol. Screen. 2005, 10(2), 168–81. 21 Quinn, C., Rapid ICE (Rapid Ion Channel Electrophysiology): an ion channel screening service using the PatchXpress 7000A system, Presentation at Molecular Devices Electrophysiology User’s Meeting, Long Beach CA, February 2005. 22 Kutchinsky J. et al., Characterization of potassium channel modulators with QPatch automated patch-clamp technology: system characteristics and performance, Assay Drug Dev. Technol. 2003, 1(5), 685–93. 23 Kiss L., Bennett P. B., Uebele V. N., Koblan K. S., Kane S. A., Neagle B., Schroeder K., High throughput ionchannel pharmacology: planar-arraybased voltage clamp, Assay Drug Dev. Technol. 2003, 1(2), 127–35. 24 Guo L., Guthrie H., Automated electrophysiology in the preclinical evaluation of drugs for potential QT prolongation, J. Pharmacol. Toxicol. Methods 2005, in press. 25 Finkelstein A., Holz R., Aqueous pores created in thin lipid membranes by the polyene antibiotics nystatin and amphotericin B, Membranes 1973, 2, 377–408. 26 Fan J. S., Palade P., Perforated patch recording with beta-escin, Pflugers Arch. 1998, 436(6), 1021–1023. 27 Armstrong C. M., Gilly W. F., Access resistance and space clamp problems associated with whole-cell patch clamping, Methods Enzymol. 1992, 207, 100– 122. 28 Bruggemann A., George M., Klau M., Beckler M., Steindl J., Behrends J. C., Fertig N., High quality ion channel analysis on a chip with the NPC technology, Assay Drug Dev. Technol. 2003, 1(5), 665–673.
163
164
6 Automated Planar Array Electrophysiology for Ion Channel Research 29 Tao H. et al., Automated tight seal electrophysiology for assessing the potential hERG liability of pharmaceutical compounds, Assay Drug Dev. Technol. 2004, 2(5), 497–506. 30 Sinclair J., Pihl J., Olofsson J., Karlsson M., Jardemark K., Chiu D. T., Orwar O., A cell-based bar code reader for high-throughput screening of ion channel-ligand interactions, Anal Chem. 2002, 74(24), 6133–6138. 31 Olofsson J., Pihl J., Sinclair J., Sahlin E., Karlsson M., Orwar O., A microfluidics approach to the problem of creating separate solution environments accessible from macroscopic volumes, Anal Chem. 2004, 76(17), 4968– 4976. 32 Zhang J. H., Chung T. D., Oldenburg K. R., A Simple Statistical Parameter for Use in Evaluation and Validation of High Throughput Screening As-
33
34
35
36
says, J. Biomol. Screen. 1999, 4(2), 67– 73. Hille, B., Ion Channels of Excitable Membranes, Sinauer Verlag, Sunderland MA, 2001. Barhanin J., Lesage F., Guillemare E., Fink M., Lazdunski M., Romey G., K(V)LQT1 and lsK (minK) proteins associate to form the I(Ks) cardiac potassium current, Nature 1996, 384(6604), 78–80. Sanguinetti M. C., Curran M. E., Zou A., Shen J., Spector P. S., Atkinson D. L., Keating M. T., Coassembly of K(V)LQT1 and minK (IsK) proteins to form cardiac I(Ks) potassium channel, Nature 1996, 384(6604), 80–83. Seebohm G., Chen J., Strutz N., Culberson C., Lerche C., Sanguinetti M. C., Molecular determinants of KCNQ1 channel block by a benzodiazepine, Mol. Pharmacol. 2003, 64(1), 70– 77.
165
7 Ion Flux and Ligand Binding Assays for Analysis of Ion Channels Georg C. Terstappen
7.1 Introduction
Ion channels are pore-forming membrane proteins which enable the rapid passage (flux) of ions across cell membranes. Their ion conductivity is often highly specific and has been used for their general classification into sodium, potassium, calcium, chloride and nonselective cation channels. As opposed to active transport by membrane pumps such as the Na+/K+ ATPase, ion channels allow only passive transport of ions along a concentration gradient. Their opening and closing (‘gating’) is regulated by a range of different stimuli including transmembrane voltage, ligand binding, mechanical stress and temperature. The first two stimuli are the most common and therefore these membrane proteins are broadly grouped into voltage-gated and ligand-gated ion channels. Ion channels are central to many biological and disease processes and are particularly important for regulating electrical properties of excitable cells such as neurons and myocytes. In many other cell types they contribute to important physiological processes such as hormonal secretion and blood pressure regulation. Although ion channels constitute a complex gene family one common feature is a pore-forming region which determines ion selectivity and mediates ion flux across cell membranes. The recent sequencing of the human genome has revealed around 400 pore-forming ion channel genes corresponding to about 1.3 % of the human genome [1]. These pore-forming ion channel subunits (a-subunits) contain a minimum of 2 trans-membrane domains, as in the case of the inward rectifying K+ channel Kir, and up to 24 transmembrane domains, as in the case of voltage-gated Na+ and Ca2+ channels. Some K+ channels contain two pore-forming regions in tandem. Additional complexity is generated since functional ion channels are often homo- or heteromeric protein complexes which can co-assemble with accessory (b- and further) subunits, thus creating a vast number of physiological ion channel complexes with different functions and pharmacology. A significant number of disease relevant ion channels have been identified and in conjunction with novel assay technologies [2] and mechanistic insights into channel function the development of selective and state-dependent drugs is on the horizon. Expression and Analysis of Recombinant Ion Channels. Edited by Jeffrey J. Clare and Derek J. Trezise Copyright # 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim ISBN: 3-527-31209-9
166
7 Ion Flux and Ligand Binding Assays for Analysis of Ion Channels
7.2 Ion Flux Assays
Activation of ion channels leads to a movement of charged molecular species across the cell membrane. This ion flux along a concentration gradient leads to changes in membrane potential that can be quantified by the Nernst equation (Fig. 7.1). If ions that pass through the channel under study can be radiolabeled, radioactive flux assays can be developed for functional analysis and screening of these ion channels. If such ions are not available, or the use of radioactive isotopes needs to be avoided, ions that pass through the channel can also be analyzed by atomic absorption spectrometry, a technique traditionally used for the detection of trace elements in environmental, biological and medical samples. In any case, a cellular system is necessary that either natively or recombinantly expresses the ion channel of interest. Typically, mammalian cells such as HEK293 or CHOK1 are employed for recombinant expression of ion channels although some channels need other cellular ‘backgrounds’ for proper functional expression [3] (see Chapter 4). Since functional ion flux assays represent a direct measure of channel activity, they are robust and insensitive to disturbances. Compared to electrophysiological methods which can be considered the ‘gold standard’ for functional analysis of ion channels, their temporal resolution is limited to the sec-
Fig. 7.1 In cellular systems activation of ion channels leads to ion flux along a concentration gradient and concomitant changes in transmembrane potential which can be quantified by the Nernst equation (shown on the right). If radioactively labeled ions are added or if K+ or Na+ ions are exchanged with Rb+ or Li+ (see text), changes in ion concentrations
can be determined by measurement of radioactivity or atomic absorption spectrometry, respectively. Both measurements represent the basis for the development of functional assays to monitor ion channel activation. Shown free intra- and extracellular ion concentrations have been taken from Ref. [12].
7.2 Ion Flux Assays
onds/minutes range and the membrane potential cannot be controlled precisely. Thus, these assays cannot be employed for screening of bona fide state-dependent ion channel modulators. 7.2.1 Radioactive Ion Flux Assays
The application of radioactive isotopes of ions that pass through the channel under study – and thus can serve as tracers for these ions in cellular assay systems – has long been used. Radioactive isotopes of the naturally conducting ion species, such as 22Na+ [4], 45 Ca2+ [5] and 36Cl– [6], can be employed as tracers as can other radioactive ion species which are conducted by the channel. For instance, 86Rb+ has been used for the study of potassium and nonselective cation channels [7] and 14C-guanidinium for analysis of sodium channels [4]. Based on transmembrane concentration gradient and ion conductivity (Fig. 7.1) influx of radiotracer is usually measured for sodium, calcium and chloride channels, whereas efflux is measured for potassium channels upon activation. Cells expressing the ion channel of interest are typically grown in standard cell culture compatible microplates. Voltage-gated channels are activated by adding a ‘depolarizing’ concentration of 650 mM KCl to the cell medium whereas other channels are, for instance, activated by adding an appropriate concentration of ligand. Measurement of radioactivity using standard equipment is either carried out in the cell supernatant, the cell lysate or both matrices by direct Cerenkov counting (e. g. 86Rb) or liquid scintillation counting (e. g. 45Ca). Measurements of both matrices allow calculation of the relative flux of radiotracer thus eliminating potential well-to-well differences in cell densities and tracer loading. A homogeneous radioactive ion flux assay format is possible using CytoStar-T scintillating technology (GE Healthcare) employing b-emitting isotopes. Based on the principles of scintillation proximity assay (SPA) technology (see Section 7.3) each well of a special microplate is coated at the bottom with scintillant that will only detect radioisotopes in close vicinity to it. Thus, if cells are cultured as monolayers on these plates and channels are activated in the presence of radiotracer, the influx or efflux can be measured as a change of light using any plate-based scintillation counter. With this technique, no wash steps and no separation of initially applied radiotracer are necessary thus making this assay format more amenable to the requirements of high throughput screening (HTS). A few examples of measuring influx of 14C-guanidinium for sodium channels [8], influx of 45 Ca2+ for ionotropic glutamate receptors [9] and efflux of 86Rb+ for potassium channels [10] have been described. A major advantage of radioactive ion flux assays is that no special apparatus is necessary if the laboratory is equipped for the measurement of radioactivity, which is usually the case in both academic and industrial environments. The main disadvantage of flux assays is the use of radioisotopes which is associated with significant costs, safety hazards and environmental (e. g. disposal) problems. This is the
167
168
7 Ion Flux and Ligand Binding Assays for Analysis of Ion Channels
main reason why radioactive ion flux assays, which were frequently used in the 1990s in the pharmaceutical industry, have largely been abandoned and replaced by nonradioactive alternatives such as nonradioactive ion flux assays based on atomic absorption spectrometry (Section 7.2.2) and fluorescence-based assays (see Chapter 8). 7.2.2 Nonradioactive Ion Flux Assays based on Atomic Absorption Spectrometry
Atomic absorption spectrometry (AAS) is a well established technology, traditionally used for the detection of trace elements in environmental, biological and medical samples, that uses thermal energy to generate free ground state atoms in a vapor phase which absorb light of a specific wavelength. In practice, atomization is achieved by spraying a sample containing the element to be measured into the flame of the burner of an atomic absorption spectrometer. Absorption of light, which is typically emitted by hollow cathode lamps, is measured with a photomultiplier (Fig. 7.2). Thus, an atomic absorption spectrometer can be imagined as a photometer where the cuvette is replaced by a burner generating the flame (”flame photometry”). The law of Lambert–Beer–Bouger applies and can be employed to determine the concentration of an element by measuring its absorption. In practice, however, this is usually done by comparing the light absorption of a sample with a standard curve obtained under identical experimental conditions.
Fig. 7.2 Schematic diagram of an atomic absorption spectrometer. For details refer to the text.
7.2.2.1 Nonradioactive Rubidium Efflux Assay An AAS-based rubidium efflux assay for functional analysis of potassium and nonselective cation channels was established in the 1990s [11]. Rubidium is an alkali metal with atomic number 37 and an ionic radius of 1.61 Å which is not present in eukaryotic cells. Its similarity to K+ leads to a high permeability in potassium and nonselective cation channels [12]. It can easily be detected by using atomic absorption spectrometry with a sensitivity (‘characteristic concentration’) of 0.11 mg l –1 measuring absorption at 780 nm.
7.2 Ion Flux Assays
In general, the experimental protocol for a nonradioactive Rb+ efflux assay consists of two parts, cell biology and physical determination of the tracer rubidium by AAS. First, cells expressing the ion channel under study are cultured in cell compatible microplates and loaded with rubidium by simply exchanging potassium in a cell compatible buffer solution with the same concentration of rubidium. This loading phase, which is usually finished within 2–4 h, can be inhibited by the cardiac glycoside oubain, pointing to the involvement of Na+/K+-ATPases in transporting Rb+ into the cells. Prior to starting efflux experiments it is necessary to remove excess Rb+ by a series of quick wash steps with buffer containing KCl. The frequency and buffer volumes used for these wash steps mainly depend on the cell type, cell density, microplate formats and washing devices employed and should be optimized on a case-by-case basis since appropriate removal of excessive Rb+ is essential in order to obtain good signal-to-background ratios. Activation of the ion channel under study leads to Rb+ efflux into the cell supernatant due to the established concentration gradient for this tracer ion (see also Fig. 7.1). For voltage-gated potassium channels activation can be achieved by adding a depolarizing concentration of KCl (typically 650 mM) to the cells and for ligandgated channels by adding an appropriate concentration of ligand. The incubation time with the channel activator has to be optimized empirically in order to achieve optimal efflux results but in most cases a period of 610 min is sufficient. When compounds are screened for channel blocking effects they should be added prior to channel activation (e. g. 10 min) because of kinetic considerations. Cell supernatants which contain the ‘effluxed’ Rb+ are removed and collected along with the cell lysates. Both of these Rb+-containing matrices can be stored at room temperature prior to AAS analysis which is not disturbed by cell debris. In principle, rubidium determinations can be carried out with any type of flame atomic absorption spectrometer and measurements should be carried out according to the instructions of the manufacturer. The recent development of an innovative AAS instrument for ion channel analysis (ICR 12000, Aurora Biomed Inc., Vancouver, Canada), featuring a sophisticated microsampling process utilizing 96or 384-well microplates and simultaneous measurements of 12 samples at a time, allows the measurement of up to 60 000 samples per day (http://www.aurorabiomed.com/ICR12000.htm), making the nonradioactive Rb+ efflux assay compatible with the throughput requirements of HTS in drug discovery. Typically, the relative amount of rubidium in the supernatant is calculated as [Rb in supernatant/Rb in supernatant + Rb in cell lysate] thus eliminating potential well-to-well differences in cell densities and Rb+ loading. This relative rubidium efflux is a robust and direct measure of ion channel activity and a 6twofold increase of Rb+ efflux upon channel activation over basal efflux levels is usually sufficient for the configuration of good quality HTS assays [13] since the standard deviations for rubidium measurements by AAS are low [11]. If sample throughput needs to be increased, under highly standardized experimental conditions it might be possible to measure rubidium only in the supernatant. The following assay protocol for the analysis of calcium-activated BKCa channels [14] stably expressed in mammalian CHO-K1 cells was successfully used for func-
169
170
7 Ion Flux and Ligand Binding Assays for Analysis of Ion Channels
tional selection of stable recombinant clones expressing this ion channel and subsequent screening for the identification of channel modulators. This protocol can serve as a basis for the development of such assays for other potassium and nonselective cation channels. Cells are grown at 37 8C in 96-well cell culture compatible microplates for 48 h to a final cell density of about 16104 cells per well in standard cell culture medium. After aspirating the medium, 0.2 ml cell buffer containing RbCl is added (5.4 mM RbCl, 150 mM NaCl, 2 mM CaCl2, 0.8 mM NaH2PO4, 1 mM MgCl2, 5 mM glucose, 25 mM HEPES, pH 7.4) and cells are incubated for 4 h at 37 8C. Cells are then quickly washed three times with buffer (same as above, but containing 5.4 mM KCl instead of RbCl) to remove extracellular Rb+. Subsequently, 0.2 ml buffer containing a saturating concentration of 25 µM of the Ca2+ specific ionophore A23187 (Fig. 7.3) is added to the cells in order to activate BKCa channels via a ‘molecular Ca2+ injection’ and after incubation for 10 min the supernatant is carefully removed and collected for rubidium measurements. Cells are lysed by the addition of 0.2 ml 1% Triton X-100 and also collected for rubidium determinations. AAS measurements are carried out with a flame atomic absorption spectrometer following the instructions of the manufacturer. The stimulated relative Rb+ efflux [Rb in supernatant/Rb in supernatant + Rb in cell lysate] with these recombinant cells amounts to 80 % (Fig. 7.4), which represents a five-fold increase over basal conditions. The specificity of the induced Rb+ efflux is further demonstrated by the use of the specific BKCa channel ligand iberiotoxin isolated from the scorpion Buthus tamulus [15] which blocks the channel in a concentration-dependent manner with an IC50 of 15 nM (Fig. 7.5). If this protocol is used for the analysis of other potassium or nonselective cation channels, channel activation and specificity analysis have to be adapted appropriately. In addition to the above mentioned example, a screening assay was developed for blockers of BKCa channels (Table 7.1) recombinantly expressed in HEK293 cells [16]. The diphenylurea analogue NS1608 was used to activate the channels
Fig. 7.3 Activation of BKCa channels recombinantly expressed in CHOK1 cells with increasing concentrations of the calcium-specific ionophore A23187 leads to increasing Rb+ efflux. For details refer to the text.
7.2 Ion Flux Assays Fig. 7.4 The Rb+ efflux induced with 25 mM of the ionophore A23187 is not observed in CHO-K1 cells which were used to generate the recombinant BKCa channel expressing cell line. For details refer to the text.
Fig. 7.5 Rb+ efflux induced with 25 mM of the ionophore A23187 in recombinant CHO-K1 cells expressing BKCa channels is inhibited in a concentration-dependent manner with the selective ligand iberiotoxin. For details refer to the text.
leading to a three- to four-fold Rb+ efflux which was completely blocked by the specific ligand iberiotoxin (IC50 = 12 nM). A pharmacological profile obtained with a series of known openers and blockers of BK channels compared very well with results obtained with a radioactive 86Rb efflux assay [16], demonstrating the utility of the nonradioactive Rb+ efflux assay for high throughput screening campaigns as well as SAR studies. SK3 channels (Table 7.1) are one of three members of small conductance calcium-activated potassium (SKCa) channels [17] which are all activated by submicromolar increases in intracellular Ca2+ concentration mediated by calmodulin [18]. SK3 channels were recombinantly expressed in HEK293 cells [19] and the above described assay protocol was employed, activating channels by increasing intracellular Ca2+ concentrations with thapsigargin, an inhibitor of endoplasmatic Ca-ATPase which leads to a release of Ca2+ from intracellular stores (Fig. 7.6). Calcium activation of SK3 channels led to a two- to three-fold Rb+ efflux which could
171
172
7 Ion Flux and Ligand Binding Assays for Analysis of Ion Channels Table 7.1 Published examples of recombinant ion channels that have been analyzed employing nonradioactive Rb+ efflux assay technology. Voltage-gated K+ channels
Ca2+-activated K+ channels
Ligand-gated nonselective cation channels
Kv1.1
SKCa (SK3)
Nondisclosed nonselective cation channel
Kv1.3 Kv1.4 Kv1.5 Kv7.2 (KCNQ2) Kv7.2/3 (KCNQ 2/3) Kv11 (hERG)
BKCa
Fig. 7.6 Recombinant HEK293 cells expressing SK3 channels are activated by thapsigargin, an inhibitor of endoplasmatic CaATPase which leads to release of Ca2+ from intracellular stores. Rb+ efflux as measure of channel activity is completely blocked by the SK channel selective ligand apamin. For details refer to the text.
be completely blocked with the specific SK channel ligand apamin, a peptide present in bee venom toxin. Nonradioactive Rb+ efflux assays were developed for several recombinantly expressed voltage-gated potassium channels (Table 7.1). In the case of Kv1.1 and Kv1.4 channels expressed in HEK293 cells, activation with 50 mM KCl led to a less than twofold Rb+ efflux in 10 min, which was blocked by the nonspecific potassium channel blocker TEA [11]. The relatively low KCl-induced Rb+ efflux which was attributed to low expression levels and/or inactivation properties of the
7.2 Ion Flux Assays
channels was not sufficient to configure a robust screening assay. More recently, an assay was described for Kv1.3 channels expressed in CHO-K1 cells (http:// www.aurorabiomed.com/New-Pro/CHO_Kv13_CellLine.pdf). Depolarization with 63 mM KCl led to a fourfold Rb+ efflux in 15 min which could be blocked with an IC50 value of 0.66 nM by Agitoxin-2, a peptide blocker isolated from the venom of the scorpion Leiurus quinquestriatus. Although no further details were disclosed, the generation of a Kv1.5 expressing cell line for the establishment of a Rb+ efflux assay was noted by Merck Research Laboratories at an ion channel conference in 2004 (http://www.aurorabiomed.com/retreat2004.htm). For Kv7.2 (KCNQ2) channels stably expressed in HEK293 cells stimulation with 50 mM KCl led to a fourfold Rb+ efflux [20]. Calculated Z’ factors [13] were 0.73 for a 96-well plate format and 0.6 for a 384-well format, respectively, indicating the high suitability for screening and SAR studies. The pharmacological profile of Kv7.2 defined by electrophysiology was faithfully reflected by the Rb+ efflux assay which allowed measuring 1000 data points per day in a 96-well plate format [20]. For the identification of heteromeric Kv7.2/3 (KCNQ2/3) channel (M-current) modulators a recombinant CHO-K1 cell line expressing these channels was employed [21]. Channels were activated with 20 mM KCl in the presence of the channel opener Way-1 and an average Z’ value of 0.81 for the 96-well format was calculated from a total of 20 experiments. A throughput of about 40 compounds per day for obtaining EC50 values (8-point curves) was achieved if AAS determinations of rubidium were only carried out in cell supernatants which gave results consistent with calculating relative Rb+ efflux by measuring rubidium contents in both supernatants and cell lysates. Specificity was demonstrated by using the known M-current blocker linopirdine which inhibited Rb+ efflux with an IC50 of 2.85 mM, a value in close agreement with results obtained from electrophysiological analysis. The voltage-gated potassium channel Kv11 (hERG) seems to be particularly susceptible to inhibition by many xenobiotics and drugs leading to potentially lethal arrhythmias [22]. In fact, several drugs have recently been withdrawn from the market due to hERG channel activity. Thus, in drug discovery hERG channel liability of novel compounds is a major concern. A nonradioactive Rb+ efflux assay was developed using hERG channels stably expressed in CHO-K1 cells [23]. Channels were activated by addition of 50 mM KCl for 10 min which resulted in an about two-fold Rb+ efflux. Although the signal-to-background ratio was relatively low, a Z’ value of 0.53 was calculated for a 96-well plate format thus meeting HTS standards. A pharmacological characterization employing a series of known hERG channel blockers (dofetilide, terfenadine, sertindole, astemizole, cisapride) showed the same rank order of potency as electrophysiology. Absolute IC50 values were 5–20-fold higher when compared to electrophysiological results obtained with mammalian cells but were similar to data in Xenopus oocytes. These results indicate the suitability of the Rb+ efflux assay for hERG compound profiling. A similar recombinant cell line was also employed by AstraZeneca who disclosed results at an ion channel conference in 2003 (http://www.aurorabiomed.com/ main-1.htm). In this case, a four-fold Rb+ efflux was measured in a 384-well plate format after 30 min incubation with 50 mM KCl. The calculated Z’ value was
173
174
7 Ion Flux and Ligand Binding Assays for Analysis of Ion Channels
60.5 utilizing the ICR 12000 atomic absorption spectrometer (Aurora Biomed, Vancouver, Canada). Studies on native ligand-gated nonselective cation channels (nicotinic acetyl choline receptors and purinergic P2X receptors) in PC12 cells have also been described [11]. The development and application of nonradioactive Rb+ efflux assays for such recombinantly expressed ion channels has, however, yet to be fully described. In a recent presentation from Amgen at an ion channel conference in 2003 a Rb+ efflux assay for a non-disclosed ligand-gated cation channel expressed in CHO-K1 cells was overviewed (http://www.aurorabiomed.com/main-1.htm). Exposing the recombinant cells to 3–10 mM of a nondisclosed agonist resulted in an about four-fold Rb+ efflux. This efflux was blocked by a nondisclosed antagonist with an IC50 of 344 nM which was in very good agreement with electrophysiological results, again demonstrating the reliability of such functional ion flux assays.
7.2.2.2 Nonradioactive Lithium Influx Assay Due to the high sensitivity of AAS for the determination of Li+ ions (0.035 mg l–1), influx experiments for screening of sodium channels which display a high conductivity for Li+ [12] should also be possible. Although no data have been published yet in scientific journals, at an ion channel conference in 2003 promising results obtained with SH-SY5Y cells were presented by AstraZeneca (www.aurorabiomed.com/main-1.htm). Cells were differentiated for 3–5 days with retinoic acid which induces the expression of tetrodotoxin-sensitive voltage-gated sodium channels. Cells were washed with buffer in which NaCl was replaced with choline chloride in order to remove free Na+. After incubation for 10 min at 37 8C in wash buffer, cells were treated with 5.4 mM KCl (basal) or 120 mM KCl (depolarization) in buffer containing LiCl for 15 min. After three wash steps at room temperature, cells were lysed with 1% Triton X-100 and cell lysates analyzed for Li+ concentrations with AAS. Under these conditions, a three-fold Li+ influx over basal levels was obtained which could be completely blocked by preincubation for 5 min with 1 mM tetrodotoxin. Results obtained with recombinantly expressed sodium channels are awaited.
7.2.2.3 Nonradioactive Chloride Influx Assay A more indirect application of AAS-based methods for analysis of ion channels was briefly noted for the investigation of chloride channels [24]. In this case, Cl– flux is measured after precipitating these ions with silver nitrate as AgCl and determining free silver by AAS. The utility of this indirect method remains to be established.
7.2.2.4 Conclusions Taken together, nonradioactive ion flux assays based on AAS have largely displaced radioactive flux assays in drug discovery [25]. These assays represent a direct measure of channel activity, are HTS compatible if special equipment is used
7.3 Ligand Binding Assays
and do not require radioactive isotopes. To date, their application has mainly been limited to the functional analysis and screening of potassium and nonselective cation channels. As compared to the ‘gold standard’ electrophysiology their temporal resolution is relatively low (seconds–minutes) and the transmembrane potential cannot be controlled precisely. Compared to other screening technologies such as fluorescence-based methods, these robust assays are less prone to identifying ‘false positive’ hits in drug screening programs and are thus highly reliable.
7.3 Ligand Binding Assays
Binding assays employing radiolabeled ligands have a long tradition in the study of receptors. For the investigation of ion channels, in particular for drug screening purposes, these assays were frequently used in the 1980s/1990s before more information-rich functional cell-based assays became available. The use of radiolabeled neurotoxins has revealed the existence of multiple binding sites for sodium channels [26] whereas the use of radiolabeled glibenclamide has been instrumental for the discovery of first and second generation sulfonylurea compounds (blockers of KATP channels) for treatment of type-2 diabetes [27]. More recently, radioligand binding assays have also been employed to investigate a potential hERG (Kv11) channel activity of novel compounds (see above) which might lead to potentially lethal arrhythmias [22]. The known hERG channel blockers dofetilide and MK499 were used in tritiated form and as a 35S analogue, respectively, to identify a hERG liability of compounds [28]. For the configuration of radioligand binding assays a radiolabeled ligand binding to the ion channel under study is necessary. Table 7.2 summarizes major ion channel radioligands that have been described to date. Furthermore, a source of the channel is also needed which can be organs, tissues or cells. Although binding assays can be performed with living cells cultured in microplates [3], usually cell membranes are prepared and utilized since they can be stored at –80 8C and are much easier to handle. In saturation experiments the affinity (Kd) of a radioactive ligand for the ion channel is determined whereas in competition (displacement) experiments the affinity of unlabeled ligands (Ki) is measured [29]. For screening purposes, which aim at the identification of novel chemical entities acting on ion channels, displacement experiments are carried out which can identify compounds that bind to the same site as the radioligand or sites allosterically coupled to it. Thus, such assays do not provide bona fide information about the effects of compounds on channel function. Usually the concentration of a compound necessary to displace 50 % of the radioligand (IC50 value) is determined as a measure of the compound’s affinity for the channel. If Ki values are necessary, these can be calculated from IC50 values by using the Cheng–Prusoff equation [30]. Despite their ease and compatibility with the requirements of HTS, such ligand binding assays suffer from the need for radioisotopes such as 3H and 125I and the associated costs, safety hazards and environmental (e. g. disposal) problems.
175
176
7 Ion Flux and Ligand Binding Assays for Analysis of Ion Channels Table 7.2 Major ion channel radioligands. Channel
Type /Current
Radioligand
Ref.
Cav 1
L
[3H]-isradipine [3H]-devapamil [3H]-diltiazem [3H]-DTZ323
37–39
Cav 2
P/Q N
[125I]-o-conotoxin GVIA [125I]-o-conotoxin MVIIC
40–42
[3H]-saxitoxin [3H]-batrachotoxin [125I]-scorpion toxins [3H]-tetrodotoxin [3H]-brevetoxin [3H]-PbTx-3
43–49
calcium
sodium Nav
potassium Kv 1
Shaker-related
[125I]-DT [125I]-BgK [125I]-DTX [125I]-HgTX1 [125I]-MgTx
50, 51
Kv 11
erg
[3H]-astemizole [3H]-dofetilide
52, 53
Kir 6
ATP-sensitive potassium channel
[3H]-glibenclamide [125I]-glibenclamide [125I]-A312110 [3H]-PKF217–744
54, 55
KCa 1
BK
[125I]-charybdotoxin [125I]-iberiotoxin [19F]-BMS204352
56–58
KCa2
SK
[125I]-apamin
59
nicotinic AChR
3
[ H]-nicotine [3H]-epibatidine [3H]-cytisine [3H]-MLA [3H]-bungarotoxin [3H]-tetracaine [3H]-TCP [3H]-ethidium [14C]-amobarbital [125I]-TID
60–63
7.3 Ligand Binding Assays Table 7.2 (continued) Channel
Type /Current
Radioligand
Ref.
glutamate
NMDA
[3H]-MK801
64
3
AMPA
[ H]-AMPA [3H]-LY395153 [3H]-Ro48–8587
65–67
Kainate
[3H]-kainic acid [3H]-NBQX
68, 69
[3H]-L-glutamate [3H]-CPP
70, 71
GABAA
[3H]-BIDN [3H]-muscimol [3H]-SR 95531 [3H]-flunitrazepam [3H]-zolpidem [3H]-Ro151788(flumazenil) [3H]-TBPS [3H]-Ro15–4513 [3H]-indiplon
72, 73
5HT3
[3H]-zacopride [3H]-BRL 43694 [3H]-GR65630 [3H]-LY278584
74
glycine
[3H]-strychnine
75
Although many fluorescent-labeled ligands for ion channels, in particular peptide ligands, are commercially available or could easily be synthesized, to date ligand binding assays for screening purposes have largely been limited to using radiolabels due to the often reduced affinity of fluorescent-labeled ligands. 7.3.1 Heterogeneous Binding Assays Employing Radioligands
Since ion channels are integral membrane proteins, a suitable ion channel containing membrane preparation has to be obtained. Nowadays, recombinant cell lines expressing the ion channel under study are often employed due to the high ion channel densities that can be achieved. After incubating the membrane preparation with a high affinity radioligand (^nM), which should exhibit a high specific radioactivity (630 Ci mmol–1), until equilibrium has been reached (typically minutes to hours), channel-bound radioligand is quickly separated from free radioligand, usually by filtration and washing. For screening assays typically glass
177
178
7 Ion Flux and Ligand Binding Assays for Analysis of Ion Channels
fiber filter-mounted 96- or 384-well plates are utilized which retain ion channelbound radioactivity on the filter. Subsequently, filter-bound radioactivity is measured using scintillation or g-counting, depending on the isotope used. If compounds are screened in displacement experiments they are usually added to the channel preparation during the incubation phase. Plotting bound radioactivity (e. g. bound cpm) against the concentration of displacing compound allows easy calculation of an IC50 value for this compound as measure of its affinity for the channel. The GraphPad Prism software (GraphPad Software Inc., San Diego) which was developed for analysis of radioligand binding is most useful for data analysis and visualization. The bee venom peptide toxin apamin is a high affinity ligand of small conductance calcium-activated potassium (SK) channels which are expressed in the cortex of rat brain [19]. The following protocol which was successfully applied for the characterization of SK channels can also serve as the basis for the configuration of other ligand binding assays with a filtration step for removing free ligand. All steps of tissue preparation were performed at 4 8C unless otherwise indicated. Cerebral cortex from rats was homogenized in 10 volumes of ice cold buffer solution [0.32 M sucrose, 5.4 mM KCl and 25 mM HEPES pH 7.0 supplemented with a protease inhibitor cocktail (CompleteTM, Roche)] using a glass/teflon homogenizer (1000 g min–1 for 12 cycles). The homogenate was centrifuged at 1000g for 10 min and the supernatant re-centrifuged at 48 000g for 20 min. The resultant pellet was washed twice with the above solution without sucrose. The final pellet was re-suspended in this buffer and the membrane suspension divided in aliquots and stored frozen at –80 8C until further use. [125I]-Apamin binding experiments were performed in 20 mM HEPES, 5.4 mM KCl, 0.2 % BSA (pH 7.4) using 200 pM radioligand (specific activity 81.4 TBq mmol–1) and 70 µg protein equivalent of rat cortex membrane preparations in a total volume of 500 µl per test tube. Nonspecific binding was determined in the presence of an excess of 1 µM unlabeled apamin. For displacement experiments the respective concentrations of compounds to be tested were added to the test tubes. After incubation for 60 min at 4 8C unbound radioligand was removed by rapid filtration through glass fiber filters (Whatman GF/C filters, pre-soaked in 0.3 % polyethyleneimine for 1 h) utilizing a cell harvester (Brandel) followed by three wash steps with ice-cold buffer. Filters were placed in scintillation vials with 3.5 ml scintillation cocktail (Filter CountTM, Packard) and bound radioactivity was measured using a b-counter (Packard). 7.3.2 Homogeneous Binding Assays Employing Radioligands
In order to avoid the separation of bound from free radioligand and associated necessary wash steps, a homogeneous assay format was developed which is more compatible with the requirements of HTS, largely due to reduced complexity of the experimental protocol (‘mix & measure’). This scintillation proximity assay (SPA) format [31] is based on solid microspheres (‘beads’) containing scintillant
7.3 Ligand Binding Assays
which are chemically modified at their surfaces to enable the coupling of molecules (Fig. 7.7). A commonly used bead type for applications with receptors and ion channels contains the lectin wheat germ agglutinin (WGA) at the surface which immobilizes membrane preparations by binding to glycosyl residues [32]. If a specific radioligand is added, it will bind to the ion channel contained in the immobilized membrane fraction and hence its emitted radiation will be in close enough proximity to activate the scintillant. The resulting emission of light around 400 nm (Fig. 7.7) can be measured with a scintillation counter. The energy released from unbound free radioligand is absorbed by the aqueous environment before it reaches the bead and hence does not activate the scintillant (Fig. 7.7). Since b-particles emitted by 3H and Auger electrons released from 125I have very short pathlengths in aqueous environments (<1 mm and about 17 mm), radioligands labeled with these isotopes are best suited for these assays. Due to their homogeneous format such assays can easily be automated and adapted to 384well and higher density microplate formats which makes it especially useful for HTS. One disadvantage of ‘homogeneity’ is assay interference of test compounds due to quenching, which leads to reduced scintillation counting efficiency and hence reduced assay signals. In particular, yellow colored compounds will lead to quenching of the blue light emitted from the beads. A ‘second generation’ type of beads (SPA Imaging Beads, GE Healthcare) which contain a different scintillant that produces a red-shifted signal has been developed in order to avoid such quenching problems. The protocol described below was routinely employed for the characterization of SK channels (Fig. 7.8) and serves as a basis for the development of other SPAs. Cell membranes were prepared from recombinant HEK293 cells stably expressing SK3 channels [18] by detaching cells cultured in 175 cm2 T-flasks by PBS/EDTA treatment, homogenization with a Polytron at full speed for 3 bursts of 10 s in 25 mM HEPES, pH 7.4 supplemented with 5.4 mM KCl (SPA buffer) and centrifugation for 30 min at 48 000 g. Membrane pellets were re-suspended in SPA buffer (1–2 ml per 175 cm2 T-flask) and membrane suspensions stored as aliquots at
Fig. 7.7 Schematic diagram of SPA technology. Left: Binding of radioligand to an ion channel immobilized at the bead surface leads to light emission via the scintillant incorporated in the beads. Right: Free radioligand is not in close enough proximity to the beads and hence does not stimulate light emission.
179
180
7 Ion Flux and Ligand Binding Assays for Analysis of Ion Channels
–80 8C until further use. Binding experiments were carried out in 96-well microplates in a final volume of 200 ml using 10 pM [125I]-apamin and 15 mg per well of SK3 channel containing membrane preparation. Each well also contains 1 mg of WGA-coated SPA beads (GE Healthcare). Nonspecific binding was determined in the presence of 1 mM unlabeled apamin. For displacement experiments the respective concentrations of compounds to be tested were added to the wells. After 15 min of incubation at room temperature with gentle shaking, the microplates were left to stand over night. Subsequently, bound radioactivity was measured with a b-counter (TopCount, Packard).
Fig. 7.8 [125I]-apamin binding to membranes prepared from recombinant HEK293 cells expressing SK3 channels analyzed employing an SPA format. (A) total binding (TB) and nonspecific binding (NSB) measured in the presence of 1 mM unlabeled apamin, (B) the concentration-dependent displacement of [125I]apamin binding to SK3 channels by the antidepressant drug fluoxetine.
7.3.3 Homogeneous Binding Assays Employing Fluorescent-Labeled Ligands and Fluorescence Polarization
Since bound and free ligands show differences in molecular rotation, binding assays employing fluorescent-labeled ligands can be configured using fluorescence polarization. This read-out system is a well established analytical technique in the field of diagnostics [33]. The principle of this method is based on the observation that excitation of a fluorophore with polarized light leads to the emission of light, which will retain the initial degree of polarization depending on the rotation that occurred during the fluorescence lifetime, typically on the nanosecond time scale
7.3 Ligand Binding Assays
[34]. At constant temperature and viscosity, the rotational relaxation time of a molecule is directly proportional to its molecular volume. Hence, if a fluorescentlabeled ligand binds to a macromolecular receptor the increase in molecular volume and concomitant decrease in rotation result in an increase in fluorescence polarization which can be measured. Advances in appropriate instrumentation over the last ten years have enabled the configuration of assays employing 96- and 384-well microplate formats. A fluorescence polarization reader can be imagined as a standard filter fluorometer with the addition of polarizing filters for the generation and detection of polarized fluorescence light. Samples are excited with polarized light and the emitted light is passed through both horizontal and vertical polarizing filters, prior to detection with a photomultiplier. Hence, the degree of polarization of the emitted light is determined in form of a ratiometric measurement, thus eliminating interferences such as ‘inner filter effects’. Although dimensionless numbers are the result of such measurements, the unit P (polarization unit) was introduced for convenience and fluorescence polarization readers usually present numbers as milli P (mP). Since fluorescence polarization is a homogeneous ‘mix and measure’ technology, it can easily be automated. For screening purposes assays are usually carried out in the form of competition (displacement) binding experiments (see above), determining the decrease in fluorescence polarization as a function of the concentration of competing compounds to be identified. Although significant improvements in the sensitivity of instrumentation have been achieved, high expression levels of receptors (61 pmol (mg protein) –1) and high ligand binding affinities (Kd ^ 10 nM) are still necessary in order to configure robust fluorescence polarization assays. To date binding assays employing fluorescence polarization have not been widely used for the analysis of ion channels [35]. The main reason for this is that fluorescent labels such as fluorescein, Bodypy, Texas RedTM, Oregon Green and Rhodamine RedTM are quite bulky chemical entities whose attachment to ligands often results in steric hindrance and concomitant reduction of binding affinity [36]. Moreover, coupling chemistry of such fluorescent labels for peptide ligands is quite advanced whereas considerable efforts might be necessary to create fluorescent derivatives of small organic molecules. In addition, since a substantial fraction of the fluorescent ligand needs to be bound in order to configure a robust fluorescence polarization assay, this might lead to significant ligand depletion and thus affect measured absolute IC50 values [35]. 7.3.4 Conclusions
For primary drug screening purposes aimed at identifying novel active chemical molecules, ligand binding assays have largely been abandoned in favor of functional cell-based assays (see above and Chapters 6 and 8). The main reason for this is the limited information content of such assays which allows no distinction between agonists and antagonists. Moreover, compounds identified in binding assays will typically reflect the mode of action of the ligand used. Thus, compounds
181
182
7 Ion Flux and Ligand Binding Assays for Analysis of Ion Channels
with novel mechanisms of channel modulation cannot be readily identified. A more technical limitation relates to the fact that for many ion channels there are no selective high-affinity ligands. Nowadays, ligand binding assays are more often employed as secondary assays in screening cascades in order to compare the molecular pharmacology of new compounds with known ligands of the channel. Furthermore, the mode of action of new compounds can be investigated if they can be labeled and used as ligands in binding assays themselves. This might also support the molecular and structural characterization of the ion channel under study since new active binding sites might be identified. The latter aspects may become increasingly important as more ion channel structures and novel modulators are discovered, and guarantee that ligand binding assays will remain an important tool for ion channel analysis.
Acknowledgements
The author would like to thank Dr. Renza Roncarati (Sienabiotech S.p.A., Discovery Research) for assistance in generating Table 7.2 and helpful discussions during the preparation of the manuscript.
References 1 J. C. Venter et al., The sequence of the human genome, Science 2001, 291, 1304–1351. 2 G. C. Terstappen, Ion channel screening technologies today, Drug Discovery Today: Technologies 2005, 2, 133–140. 3 C. Virginio, A. Giacometti, L. Aldegheri, J. M. Rimland, G. C. Terstappen, Pharmacological properties of rat alpha7 nicotinic receptors expressed in native and recombinant cell systems, Eur. J. Pharmacol. 2002, 445, 153–161. 4 M. Willow, E. A. Kuenzel, Catterall, Inhibition of voltage-sensitive sodium channels in neuroblastoma cells and synaptosomes by the anticonvulsant drugs diphenylhydantoin and carbamazepine, Mol. Pharm. 1984, 25, 228–234. 5 C. A. Rasmussen, J. L. Sutko, W. H. Barry. Effects of ryanodine and caffeine on contractility, membrane voltage, and calcium exchange in cultured heart cells, Circ Res. 1987, 60, 495–504. 6 K. G. Thampy, E. M. Barnes Jr., Gamma-aminobutyric acid-gated chlor-
7
8
9
10
ide channels in cultured cerebral neurons, J. Biol. Chem. 1984, 259, 1753– 1757. S. W. Weir, A. H. Weston, The effects of BRL 34915 and nicorandil on electrical and mechanical activity and on 86Rb efflux in rat blood vessels, Brit. J. Pharmacol. 1986, 88, 121–128. J. Denyer, J. Worley, B. Cox, G. Allenby, M Banks, HTS approaches to voltage-gated ion channel drug discovery, Drug Discovery Today 1998, 3, 323–332. A. Cushing, M. J. Price-Jones, R. Graves, A. J. Harris, K. T. Hughes, D. Bleakman, D. Lodge, Measurement of calcium flux through ionotropic glutamate receptors using Cytostar-T scintillating microplates, J. Neurosci. Methods 1999, 90, 33–36. J. W. Ford, E. B. Stevens, J. M. Treherne, J. Packer, M. Bushfield, Potassium channels: gene family, therapeutic relevance, high-throughput
References
11
12
13
14
15
16
17
18
19
screening technologies and drug discovery, Prog. Drug Res. 2002, 58, 133–68. G. C. Terstappen, Functional analysis of native and recombinant ion channels using a high-capacity nonradioactive rubidium efflux assay, Anal. Biochem. 1999, 272, 149–155. B. Hille, Ionic Channels of Excitable Membranes, Sinauer Associates, Sunderland, MA, 1992 J-H. Zhang, T. D. Y. Chung, K. R. Oldenburg, A simple statistical parameter for use in evaluation and validation of high throughput screening assays, J. Biomol. Screen. 1999, 4, 67–73. M. Garcia-Calvo, H-G. Knaus, O. B. McManus, K. M. Giangiacomo, G. Kaczorowski, M. L. Garcia, Purification and reconstitution of the highconductance, calcium-activated potassium channel from tracheal smooth muscle, J. Biol. Chem. 1994, 269, 676– 682. V. K. Gribkoff, et al., Effects of channel modulators on cloned large-conductance calcium-activated potassium channels, Mol. Pharmacol. 1996, 50, 206–217. A. S. Parihar, D. R. Groebe,V. E. Scott, J. Feng, X-F. Zhang, U. Warrior, M. Gopalakrishnan, C-C. Shieh, Functional analysis of large conductance Ca2+ activated K+ channels: ion flux studies by atomic absorption spectrometry, Assay Drug Dev. Technol. 2003, 1, 647–654. M. Kohler, B. Hirschberg, C. T. Bond, J. M. Kinzie, N. V. Marrion., J. Maylie, J. P. Adelman, Small-conductance Ca2+ activated K+ channels from mammalian brain, Science 1996, 273, 1709–1714. X. M. Xia, et al., Mechanism of Ca2+ gating in small conductance Ca2+-activated K+ channels, Nature 1998, 395, 503–507. G. C. Terstappen, A. Pellacani, L. Aldegheri, F. Graziani, C. Carignani, G. Pula, C. Virginio, The antidepressant fluoxetine blocks the human small conductance calciumactivated potassium channels SK1, SK2
20
21
22
23
24
25
26
27
28
and SK3, Neuroscience Lett. 2003, 346, 85–88. C. W. Scott, D. E. Wilkins, S. Trivedi, D. J. Crankshaw, A medium-throughput functional assay of KCNQ2 potassium channels using rubidium efflux and atomic absorption spectroscopy, Anal. Biochem. 2003, 319, 251–257. K. Wang, et al.,Validation of an atomic absorption rubidium ion efflux assay for KCNQ/M- channels using the Ion Channel Reader 8000, Assay Drug Dev. Technol. 2004, 2, 525–534. J. I. Vandenberg, B. W. Walker, T. J. Campbell, HERK K+ channels: friend and foe. Trends Pharmacol. Sci. 2001, 22, 240–246. W. Tang, J. Kang, X. Wu, D. Rampe, L. Wang, H. Shen, Z. Li, D. Dunnington, T. Garyantes, Development and evaluation of high throughput functional assay methods for hERG potassium channel, J. Biomol. Screen. 2001, 6, 325–331. L. Stankovich, D. Wicks, S. Despotovski, D. Liang, Atomic absorption spectroscopy in ion channel screening, Assay Drug Dev. Technol. 2004, 2, 569– 574. G. C. Terstappen, Nonradioactive Rb+ efflux assay and its applications in drug discovery and development, Assay Drug Dev. Technol. 2004, 2, 553–559. W. A. Catterall,V. Trainer, D. G. Baden, Molecular properties of the sodium channel: a receptor for multiple neurotoxins, Bull. Soc. Path. Exp. 1992, 85, 481–485. J. L. Challinor-Rogers, D. C. Kong, M. N. Iskander, G. A. McPherson, Structure-activity relationship of glibenclamide analogs: A comparison of potency as levcromakalim antagonists in rat aorta vs. affinity for [3H]-glibenclamide binding to membranes from rat cerebral cortex, J. Pharmacol. Exp. Ther. 1995, 273, 778–86. K. Finlayson, L. Turnbull, C. T. January, J. Sharkey, J. S. Kelly, [3H]Dofetilide binding to HERK in transfected membranes: A potential high throughput preclinical screen, Eur. J. Pharmacol. 2001, 430, 147–148.
183
184
7 Ion Flux and Ligand Binding Assays for Analysis of Ion Channels 29 D. B. Bylund, M. L. Toews, Radioligand binding methods: practical guide and tips, Am. J. Physiol. 1993, 265, L421– 429. 30 R. Cheng, G. Prusoff, Relationship between the inhibition constant (Ki) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction, Biochem. Pharmacol. 1973, 22, 3099–4006. 31 N. D. Cook, Scintillation proximity assay: a versatile high-throughput screening technology, Drug Discovery Today 1996, 1, 287–294. 32 F. Graziani, A. Aldegheri, G. C. Terstappen, High throughput scintillation proximity assay for the identification of FKBP-12 ligands, J. Biomol. Screen. 1999, 4, 3–7. 33 W. B. Dandliker,V. A. de Saussure, Fluorescence polarization in immunochemistry, Immunochemistry 1970, 7, 799–828. 34 P. Perrin, Polarisation de la lumière de fluorescence, J. Phys. Radium 1926, 7, 390–401. 35 M. Allen, J. Reeves, G. Mellor, High throughput fluorescence polarization: a homogeneous alternative to radioligand binding for cell surface receptors, J. Biomol. Screen. 2000, 5, 63–69. 36 P. Wu, M. Brasseur,W. Schindler, A high throughput STAT binding assay using fluorescence polarization, Anal. Biochem. 1997, 249, 29–36. 37 H. Glossmann, J. Striessnig, Molecular properties of calcium channels, Rev. Physiol. Biochem. Pharmacol. 1990, 114, 1–105. 38 A. Koschak, D. Reimer, I. Huber, M. Grabner, H. Glossmann, J. Engel, J. Striessnig, Cav1.3 subunits can form L-type calcium channels activating at negative voltages, J. Biol. Chem. 2001, 276, 22100–22106. 39 M. Hagiwara, S. Adachi-Akahne, T. Nagao, High-affinity binding of [3H]DTZ323 to the diltiazem-binding site of L-type Ca2+ channels, Eur. J. Pharmacol. 2003, 466, 63–71. 40 M. E. Williams, et al., Structure and functional expression of an o-cono-
41
42
43
44
45
46
47
48
49
toxin-sensitive human N-type calcium channel, Science 1992, 257, 389–395. P. Favreau, et al., A new omega-conotoxin that targets N-type voltage-sensitive calcium channels with unusual specificity, Biochemistry 2001, 40, 14567– 14575. R. J. Lewis, et al., Novel omega-conotoxins from Conus catus discriminate among neuronal calcium channel subtypes, J. Biol. Chem. 2000, 275, 35335– 35344. J. W. West, T. Scheuer, L. Maechler, W. A. Catterall, Efficient expression of rat brain type IIA Na+ channel subunits in a somatic cell line, Neuron 1992, 8, 59–70. J. C. Rogers,Y. Qu, T. N. Tanada, T. Scheuer, W. A. Catterall, Molecular determinants of high affinity binding of ù-scorpion toxin and sea anemone toxin in the S3-S4 extracellular loop in domain IV of the Na+ channel subunit, J. Biol. Chem. 1996, 271, 15950–15962. S. Cestèle,Y. Qu, J. C. Rogers, H. Rochat, T. Scheuer, W. A. Catterall,Voltage sensor-trapping: enhanced activation of sodium channels by o-scorpion toxin bound to the S3-S4 loop in domain II, Neuron 1998, 21, 919–931. R. S. Sheldon, N. J. Cannon, H. J. Duff, Binding of [3H]-batrachotoxinin A benzoate to specific sites on rat cardiac sodium channels, Mol. Pharmacol. 1986, 30, 617–623. M. Taouis, R. S. Sheldon, R. J. Hill, H. J. Duff, Cyclic AMP-dependent regulation of the number of [3H]-batrachotoxinin benzoate binding sites on rat cardiac myocytes, J. Biol. Chem. 1991, 266, 10300–10304. A. J. Bourdelais, S. Campbell, H. Jacocks, J. Naar, J. L. Wright, J. Carsi, D. G. Baden, Brevenal is a natural inhibitor of brevetoxin action in sodium channel receptor binding assays, Cell. Mol. Neurobiol. 2004, 24, 553–63. M. Yotsu-Yamashita, K. Nishimori, Y. Nitanai, M. Isemura, A. Sugimoto, T. Yasumoto, Binding properties of [3H]-PbTx-3 and [3H]-saxitoxin to brain membranes and to skeletal muscle
References
50
51
52
53
54
55
56
57
membranes of puffer fish Fugu pardalis and the primary structure of a voltagegated Na+ channel alpha-subunit (fMNa1) from skeletal muscle of F. pardalis, Biochem. Biophys. Res. Commun., 2000, 267, 403–412. O. Shamotienko, S. Akhtar, C. Sidera, F. A. Meunier, B. Ink, M. Weir, J. O. Dolly, Recreation of neuronal Kv1 channel oligomers by expression in mammalian cells using Semliki Forest virus, Biochemistry 1999, 38, 16766–16776. J. Racape, A. Lecoq, R. Romi-Lebrun, J. Liu, M. Kohler, M. L. Garcia, A. Menez, S. Gasparini, Characterization of a novel radiolabeled peptide selective for a subpopulation of voltagegated potassium channels in mammalian brain, J. Biol. Chem. 2002, 277, 3886–3893. P. J. Chiu, et al.,Validation of a [3H]-astemizole binding assay in HEK293 cells expressing HERG K+ channels, J. Pharmacol. Sci. 2004, 95, 311–319. K. Finlayson, A. J. Pennington, J. S. Kelly, [3H]-dofetilide binding in SHSY5Y and HEK293 cells expressing a HERG-like K+ channel, Eur. J. Pharmacol. 2001, 412, 203–212. R. Davis-Taber, et al., [125I]A-312110, a novel high-affinity 1,4-dihydropyridine ATP-sensitive K+ channel opener: characterization and pharmacology of binding, Mol. Pharmacol. 2003, 64, 143–153. P. W. Manley, C. Loffler-Walz, U. Russ, A. Hambrock, T. Moenius, U. Quast, Synthesis and characterization of a novel tritiated KATP channel opener with a benzopyran structure, Br. J. Pharmacol. 2001,133, 275–85. M. Hanner, R. Vianna-Jorge, A. Kamassah., W. A. Schmalhofer, H. G. Knaus, G. J. Kaczorowski, M. L. Garcia, The beta subunit of the high conductance calcium-activated potassium channel. Identification of residues involved in charybdotoxin binding, J. Biol. Chem. 1998, 273, 16289–16296. A. Koschak, R. O. Koch, J. Liu, G. J. Kaczorowski, P. H. Reinhart, M. L. Garcia, H. G. Knaus, [125I]-IberiotoxinD19Y/Y36F, the first selective, high spe-
58
59
60
61
62
63
64
65
cific activity radioligand for high-conductance calcium-activated potassium channels, Biochemistry 1997, 36, 1943– 1952. D. O. Kiesewetter, E. M. Jagoda, J. E. Starrett Jr,V. K. Gribkoff, P. Hewawasam, N. Srinivas, D. Salazar,W. C. Eckelman, Radiochemical synthesis and biodistribution of a novel maxi-K potassium channel opener, Nucl. Med. Biol. 2002, 29, 55–59. M. Hugues, D. Duval, P. Kitabgi, M. Lazdunski, J. P. Vincent, Preparation of a pure monoiodo derivative of the bee venom neurotoxin apamin and its binding properties to rat brain synaptosomes, J. Biol. Chem. 1982, 257, 2762–2769. D. J. Anderson, S.P Arneric, Nicotinic receptor binding of [3H]cytisine, [3H]-nicotine and [3H]-methylcarbamylcholine in rat brain, Eur. J. Pharmacol. 1994, 253, 261–267. D. Gnadisch, E. D. London, P. Terry, G. R. Hill, A. G. Mukhin, High affinity binding of [3H]-epibatidine to rat brain membranes, Neuroreport 1999, 10, 1631–1636. H. A. Navarro, D. Zhong, P. Abraham, H. Xu, F. I. Carroll, Synthesis and pharmacological characterization of [125I]-iodomethyllycaconitine ([125I]iodo-MLA). A new ligand for the alpha 7 nicotinic acetylcholine receptor, J. Med. Chem. 2000, 43, 142–145. H. R. Arias, J. R. Trudell, E. Z. Bayer, B. Hester, E. A. McCardy, M. P. Blanton, Noncompetitive antagonist binding sites in the torpedo nicotinic acetylcholine receptor ion channel. Structureactivity relationship studies using adamantane derivatives, Biochemistry 2003, 42, 7358–7370. F. Murray, J. Kennedy, P. H. Hutson, J. Elliot, I. Huscroft, K. Mohnen, M. G. Russell, S. Grimwood, Modulation of [3H]-MK-801 binding to NMDA receptors in vivo and in vitro, Eur. J. Pharmacol. 2000, 397, 263–270. L. Bunch, T. H. Johansen, H. Brauner-Osborne, T. B. Stensbol, T. N. Johansen, P. Krogsgaard-Larsen, U. Madsen, Synthesis and receptor
185
186
7 Ion Flux and Ligand Binding Assays for Analysis of Ion Channels
66
67
68
69
70
binding affinity of new selective GluR5 ligands, Bioorg. Med. Chem. 2001, 9, 875–879. A. M. Linden, H. Yu, H. Zarrinmayeh,W. J. Wheeler, P. Skolnick, Binding of an AMPA receptor potentiator ([3H]-LY395153) to native and recombinant AMPA receptors, Neuropharmacology 2001, 40, 1010–1018. V. Mutel, et al., Binding characteristics of a potent AMPA receptor antagonist [3H]Ro 48–8587 in rat brain, J. Neurochem. 1998, 71, 418–426. N. Crawford, T. K. Lang, D. S. Kerr, D. J. de Vries, High-affinity [3H] kainic acid binding to brain membranes: a reevaluation of ligand potency and selectivity, J. Pharmacol. Toxicol. Methods 1999, 42, 121–125. K. K. Dev,V. Petersen, T. Honore, J. M. Henley, Pharmacology and regional distribution of the binding of 6-[3H]nitro-7-sulphamoylbenzo[ f ]-quinoxaline-2,3-dione to rat brain, J. Neurochem. 1996, 67, 2609–2612. L. H. Martini, C. R. Souza, P. B. Marques, J. B. Calixto, R. A. Yunes, D. O. Souza, Compounds extracted from Phyllantus and Jatropha elliptica inhibit the binding of [3H]-glutamate and [3H]GMP-PNP in rat cerebral cortex membrane, Neurochem. Res. 2000, 25, 211– 215.
71 R. H. Porter, R. S. Briggs, P. J. Roberts, Modulation of [3H]3-((+-)-2-carboxypiperazin-4-yl)propyl-1-phosphonic acid ([3H]CPP) binding by ligands acting at the glycine and the polyamine sites of the rat brain NMDA receptor complex, Eur. J. Pharmacol. 1992, 227, 83–88. 72 J. J. Rauh, et al., Effects of [3H]-BIDN, a novel bicyclic dinitrile radioligand for GABA-gated chloride channels of insects and vertebrates, Br. J. Pharmacol. 1997, 121, 1496–1505. 73 S. K. Sullivan, R. E. Petroski, G. Verge, R. S. Gross, A. C. Foster, D. E. Grigoriadis, Characterization of the interaction of indiplon, a novel pyrazolopyrimidine sedative-hypnotic, with the GABAA receptor, J. Pharmacol. Exp. Ther. 2004, 311, 537–546. 74 S. C. Lummis, J. Baker, Radioligand binding and photoaffinity labelling studies show a direct interaction of phenothiazines at 5-HT3 receptors, Neuropharmacology 1997, 36, 665–70. 75 R. J. Vandenberg, C. R. French, P. H. Barry, J. Shine, P. R. Schofield, Antagonism of ligand-gated ion channel receptors: two domains of the glycine receptor alpha subunit form the strychnine-binding site, Proc. Natl. Acad. Sci. U S A. 1992, 89, 1765–1769.
187
8 Ion Channel Assays Based on Ion and Voltage-sensitive Fluorescent Probes Jesús E. González, Jennings Worley, and Fredrick Van Goor
8.1 Introduction
Ion channels are critical for physiological signaling and are the targets of several drugs. Most of these drugs were discovered using in vivo pharmacology models or directly from observation in humans and only later was it determined that their mechanism of action involved modulation of ion channel activity. Ion channels are widely recognized as “druggable” targets due to their modulation by a wide diversity of small molecules. Despite their promise, these targets have historically been difficult to pursue because of limited structural information, low expression levels, requirement of a membrane environment for proper folding and pharmacology, and limitations or absence of high-throughput screening methods. In the last 15 years drug discovery approaches have increasingly relied on high throughput methods to profile a larger number of candidate compounds in increasing numbers of in vitro assays aimed at providing insight into which molecules will be more likely to be efficacious, safe, and able to be administered in humans. Assays for primary targets, safety counter-screens, chemical properties, and in vitro metabolism are key components of modern discovery processes and approaches. Ion channel assays play as an important role and are particularly challenging as they generally require cellular expression. Fluorescence readouts are commonly use for cell-based assays because of their high sensitivity, compatibility with readily available instrumentation and microtiter plates, and the availability of a variety of probes and reagents [1]. The application of physiological indicators of intracellular calcium and membrane potential has made possible functional fluorescence based ion channels assays with the requisite throughput, sensitivity, and reliability required for large scale profiling. In this chapter we will review the range of fluorescence probes, approaches and concepts for assaying activity for sodium, calcium, potassium, and chloride channels, including those activated and regulated by ligands and voltage. Within each class of channels the utility and challenges will be discussed. Where appropriate, relevant reviews will be cited and illustrative examples will be given. Finally, we will Expression and Analysis of Recombinant Ion Channels. Edited by Jeffrey J. Clare and Derek J. Trezise Copyright # 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim ISBN: 3-527-31209-9
188
8 Ion Channel Assays Based on Ion and Voltage-sensitive Fluorescent Probes
provide examples and describe areas of where novel fluorescence assays are being developed, with an emphasis on drug discovery applications and new approaches for measuring ion channel trafficking.
8.2 Membrane Potential Probes
Fluorescent probes of cellular membrane potential were initially developed as physiological indicators and are divided into three major categories. The first includes fast electrochromic probes, such as Di-4-ANEPPS, that are capable of detecting microsecond voltage changes yet have low voltage sensitivity (~ 1–10 % DF/Fo per 100 mV)[2]. The second class is comprised of environment-sensitive dyes that distribute between cellular sites and extracellular solution according to the membrane potential. These probes have good sensitivity (1–100 % DF/Fo per 100 mV) yet relatively slow time resolution, requiring minutes to attain maximal fluorescence response [2]. In the last five years new redistribution dyes have been introduced by Molecular Devices that respond substantially faster, in tens of seconds instead of minutes [3, 4]. The third category involves probes based on fluorescence resonance energy transfer (FRET) readout of rapid transmembrane translocation of fluorescent hydrophobic ions [5, 6]. These probes have good sensitivity (20–150 % DF/Fo per 100 mV) and respond much more rapidly than redistribution dyes since the temporal response results from facile transmembrane redistribution of a mobile voltage-sensitive ionic dye and not slow diffusion across multiple membrane/water interfaces. Ion channels in cells throughout the body open and close in micro- to milliseconds to produce rapid, < 5 s, changes in membrane potential which are important for cell signaling and are not, in many cases, readily detected with slow redistribution dyes. For this reason most applications requiring the highest temporal resolution, such as neuronal signaling, require electrochromic or FRET dyes. Redistribution and FRET probes are generally used for most ion channel drug discovery applications because of their relatively high sensitivity and ease of use. 8.2.1 Redistribution Probes
Fluorescent redistribution probes are environment-sensitive charged dyes that equilibrate between intracellular hydrophobic sites and extracellular solution according to membrane potential. Fig. 8.1A is a schematic illustrating the redistribution mechanism and Fig. 8.1B shows a fluorescence micrograph of cells stained with the oxonol DiSBAC4(3). Generally, the fluorescence quantum yield is high when the probe binds to hydrophobic sites such as proteins and membranes and very low when in aqueous solution. Consequently positively charged probes such as cyanines and rhodamines result in bright fluorescence at negative membrane potentials and are relatively dim at less negative or positive potentials. The oppo-
8.2 Membrane Potential Probes
Fig. 8.1 Oxonol probes that operate via redistribution. (A) Schematic of voltage-sensitive redistribution mechanism. Negativelycharged oxonol molecules (small circles) distribute into a cell (large circles) according to the membrane potential Vm. The dye fluorescence is greatly enhanced, represented by the small filled circles, when bound to hydrophobic intracellular protein and membrane sites. At less negative potentials, represented
on the right panel, more dye accumulates in the cell and the total fluorescence increases. (B) A fluorescence micrograph of CHO-K1 cells stained with DiSBAC4(3) showing extensive intracellular staining. The extracellular solution is relatively dark because the quantum efficiency in aqueous solution is neglible. (C) Structures of bis-barbiturate trimethine oxonols.
site is true for negatively charged probes such as oxonol dyes, which make it more challenging to detect significant oxonol fluorescence from cells that hyperpolarize to very negative potentials. Anions translocate across the membranes and into cells more easily than cations because of the dipole that originates from the lipid carbonyl groups. The rate difference due to charge can be orders of magnitude, as has been shown for the isostructural borates and phosphonium ions [7]. This is an important reason why negatively charged probes, such as oxonols, are routinely used and positively charge probes are not. The structures of commonly used bisbarbiturate oxonol dyes are shown in Fig. 8.1C. One of the first fluorescent assays specifically designed for identifying ion channel modulators used the redistribution oxonol probe DiBAC4(3) to develop a higher throughput assay for identifying openers of KATP channels [8]. This work played a major role in the development of the fluorometric imaging plate reader (FLIPR) [9]. DiBAC4(3)’s high sensitivity to temperature and excitation and emission properties were key considerations in the original FLIPR design specifications. Key issues with DiBAC4(3) are temperature sensitivity, slow temporal response, interference from fluorescent compounds and relatively high false positive rate. Even at 37 8C, the time response often requires 15 min or more to reach the maximal fluorescence change. Despite these limitations, DiBAC4(3) has been broadly applied and has also been used with a microfluidic device, in
189
190
8 Ion Channel Assays Based on Ion and Voltage-sensitive Fluorescent Probes
combination with a cationic dye, to measure ion channel activity in human T lymphocytes [10]. Recently Molecular Devices have introduced a new FLIPR membrane potential (FMP) kit that includes a redistribution dye that offers advantages in temporal response and reduced fluorescence interference compared to DiBAC4(3). The probe has fluorescence properties similar to known thiobarbiturate oxonols and has redistribution kinetics much faster than DiBAC4(3) with maximal response achieved in approximately 10–15 s. The kit also includes a second quenching dye that reduces extracellular fluorescence and improves signal to background and potentially reduces the effect of some fluorescent compounds. Another FMP kit advantage for HTS applications is that the dyes can be added directly to cells and do not require additional washing steps, streamlining the process and resulting in greater throughput. 8.2.2 FRET Probes
Membrane potential sensors based on FRET are useful for high-throughput screening (HTS) of ion channel targets [11–13]. They are comprised of two fluorescent components. The first is a highly fluorescent, hydrophobic ion that binds to the plasma membrane and “senses” the transmembrane electric field. The ion sensor rapidly redistributes between two binding sites on opposite sides of the membrane, establishing a Nernstian equilibrium (10-fold concentration ratio for ~ 60 mV). In response to a membrane potential change, the hydrophobic ions electrodiffuse across the membrane and establish a new equilibrium corresponding to the new membrane potential. The voltage-dependent redistribution is converted into a ratiometric fluorescent readout with a second fluorescent molecule that binds specifically to one face of the plasma membrane and functions as a FRET partner to the mobile voltage-sensing ion. A schematic of the mechanism is shown in Fig. 8.2A. A variety of fluorescent membrane-bound molecules have been designed to function as voltage-sensitive FRET partners with different voltage sensitivities, temporal responses, and wavelengths. In the most commonly used configuration, the two fluorescent dye components are a ChloroCoumarin-labeled phospholipid (e. g. CC1-DMPE and CC2-DMPE) and a bis-(1,3-dialkylthiobarbituric acid) trimethine oxonol (DiSBACn(3)), where n corresponds to the number of carbon atoms in the n-alkyl group, shown in Fig. 8.2B. Both are very bright fluorophores, the properties are shown in Table 8.1. CC1/2-DMPE selectively partitions into the outer leaflet of the plasma membrane and acts as a fixed FRET donor to the mobile voltage-sensitive and negatively charged oxonol acceptor. CC1/2-DMPE does not cross the bilayer because of two negative charges from the coumarin and the phosphate groups. Cells have negative (inside) resting membrane potentials and under these conditions the majority of the negatively charge oxonols populate the relatively positive extracellular leaflet, resulting in efficient FRET (i. e. quenching of the coumarin donor and increase in the oxonol acceptor emission). As illustrated in Fig. 8.2A, depo-
8.2 Membrane Potential Probes Fig. 8.2 Voltage-sensitive FRET probes. (A) Schematic of voltagesensitive FRET mechanism. Fluorescent donor molecules bind selectively to the extracellular leaflet of the plasma membrane, represented by a circle with hatching. Negatively charged acceptor, bold circles, distribute across the plasma membrane according to the membrane potential in a Nernstian manner. At negative resting potentials the acceptors are predominantly at the extracellular surface and FRET is efficient, as shown on the left panel. Upon depolarization, the transmembrane acceptor equilibrium changes so that more oxonols are at the intracellular side. This causes a decrease in FRET and results in an increase and decrease in donor and acceptor fluorescence, respectively. (B) Structures of FRET donor CC1-DMPE and thiobarbiturate oxonols.
Table 8.1 Fluorescence and sensitivity properties of FRET membrane potential probes. kex (nm)
kem (nm)
Tc (ms)
Donor CC1/2-DMPE
405
460
na
Acceptors DiSBAC2(3) DiSBAC4(3) DiSBAC6(3) DiSBAC2(5) DiSBAC4(5) DiSBAC6(5)
540 540 540 640 640 640
560 560 560 660 660 660
500 20 2 50 2 0.40
e (M–1 cm–1)
} Q.Y.
Vm sensitivity %DR per mV
40 000
1.0
na
200 000 200 000 200 000 225 000 225 000 225 000
0.44 0.44 0.44 0.67 0.67 0.67
1–3 0.6–1 0.4–0.8 0.5–2 < 0.4 < 0.2
larization causes translocation of the oxonol to the inner surface of the plasma membrane and an increase in the mean donor and acceptor distance. Because FRET is very sensitive to donor–acceptor distance, this charge movement causes a simultaneous increase and decrease in the CC1/2-DMPE and oxonol fluorescence, respectively. The donor and acceptor fluorescence emission changes are reversed upon repolarization. The oxonol moves reversibly in the membrane with sub-second kinetics, allowing voltage-sensitive FRET to report fast voltage changes.
191
192
8 Ion Channel Assays Based on Ion and Voltage-sensitive Fluorescent Probes
Simultaneous patch-clamp and rapid optical recording have been used to demonstrate the speed, sensitivity and ratiometric nature of voltage-sensitive FRET in cells [5, 6]. The coumarin donor to oxonol acceptor fluorescence emission ratio is independent of the excitation intensity, the number of cells being detected, and the optical path length, providing fewer experimental artifacts compared to single wavelength probes. These dyes load well in about 20 min at room temperature and cells can be maintained for hours with the dyes without significantly degrading voltage responses. Analysis of data from a wide variety of cell types gives a sensitivity of about ~ 1–3 % ratio change per mV for DiSBAC2(3) over the relevant physiological range. Oxonols differ in their physical, optical, and voltage-sensing properties. DiSBAC2(3) is more water soluble and is left in the extracellular media during assay. DiSBAC2(3) is often the first choice of ion channel screening applications because of its ease of loading, assay stability, and sensitivity. With a response time constant of ~500 ms, a FRET assay using DiSBAC2(3) as acceptor is 2 orders of magnitude faster than DiBAC4(3) redistribution assays [14] and 10-fold faster than the FMP probes, making this dye well suited for liquid addition protocols which are often used to trigger membrane potential changes in HTS assays. FRET assays with even higher temporal resolution are possible by using more hydrophobic oxonols, such as DiSBAC6(3) [5]. These more hydrophobic oxonols require using excipients, for example Pluronic F-127, to assist cellular loading and are fast enough to measure millisecond voltage changes. In the case of hexyl substituted pentamethine oxonol the response time constant was measured at 400 ms [6]. A summary of fluorescence and voltage-sensing properties of various FRET membrane potential probes is given in Table 8.1. The recent development of rapid electrical stimulation methods has enabled FRET ion channel assays that utilize the millisecond probes (Huang et al., submitted). 8.2.3 Advantages and Limitations of Membrane Potential Probes
Ion channels pose unique challenges for functional assay development due to their variety and complex kinetics. Direct sensing of membrane potential is an attractive ion channel readout because it is sensitive, generic, and is applied in intact living cells. Fluorescent membrane potential assays can be as, or more, sensitive as other ion channel assays methods including isotopic flux assays and patch clamping while being more amenable to high-throughput profiling. Detection sensitivity of ion channel agonists can often be comparable to or better than voltage-clamp in whole cells, depending on channel density and background cellular conductances. Consistent with drug–receptor models where the assay response is nonlinear with receptor occupancy [15], the observed EC50s for agonist are often shifted to lower concentrations because only a small fraction of the channels have to be activated to elicit sufficient current to change the membrane potential. For antagonist assays, this property works against detection sensitivity and generally causes shifts in IC50s to higher compound concentrations. Only ~30 million ions
8.2 Membrane Potential Probes
need to pass through channels to change the membrane potential 60 mV in a 50 mm diameter cell [16]. Except for Ca2+, this small number of ions will not appreciably change the intracellular or extracellular ion concentrations under normal physiological conditions. In the case of a neuronal voltage-activated Na+ channel, only a small number of channels (10s to 100s) need to be opened to cause enough ions to enter into the cell and rapidly depolarize a cell with a high resistance. The approach is generic because changes in membrane potential can be generated by any electrogenic ionic flux across the membrane. Voltage-sensitive fluorescent probes therefore provide a basis for a universal HTS-compatible assay platform for ion channels. Researchers applying voltage-sensitive fluorescent probes must also be aware of some limitations, potential sources of experimental artifacts and important optimization parameters. Selective functional assays for the target of interest require a host cell line with low background currents that are not activated by the stimulation protocol. Occasionally, small currents that seem insignificant in voltageclamp recording can cause large effects on membrane potential assays. Once sufficient functional expression and stimulation conditions are established, it is important to optimize the cellular dye staining. Generally, staining conditions are selected for maximum dynamic range, signal over background, and response stability. For HTS applications, where reproducibility and efficiency is critical, it is important to select conditions that are not overly sensitive. Another potential issue is drug–dye interactions. The susceptibility of various membrane potential probes to such artifacts has been evaluated and it has been reported that FRET probes and FMP probes have decreased compound–dye issues than DiBAC4(3)[17]. The relatively high optical interference of DiBAC4(3) was also reported in a comparison with FMP in hERG [18] and KATP [4] channel assays. Normally these effects are fast and occur immediately after compound addition and do not affect the probes ability to respond to voltage changes. However, if the fluorescence intensities are significantly changed then the apparent voltage changes can be proportionately offset, causing significant error in the measured compound activity. For FRET probes, the ratiometric readout reduces some of these artifacts relative to single intensity dyes, and these effects can be readily identified as a large change in the donor or acceptor intensities compared to control wells in the absence of test compound. These interactions are to a large extent not observed at test compound concentrations below 3 mM, so potent compounds that interfere at higher concentration can usually be cleanly observed at lower concentrations. Finally, cell issues that affect membrane potential as well as most cell-based assays are confluence, health and stability of the cells. Sick cells or those cultured under variable conditions often have different functional channel expression and membrane properties that cause increased false positives or negatives and variability.
193
194
8 Ion Channel Assays Based on Ion and Voltage-sensitive Fluorescent Probes
8.3 Ion-sensitive Fluorescent Probes 8.3.1 Calcium Dyes
Calcium dyes have been used extensively for probing Ca2+ signaling in cells and tissue. All currently used exogenous small molecules indicators trace their origin to the seminal work from Roger Tsien and coworkers. These cellular probes are fluorescent Ca2+ chelators based on EGTA that have different excitation and emission wavelengths, ratiometric readouts, and Ca2+ affinities. Fluo3 and its progeny, such as Ca-Green and Fluo4 [19] have been the probes of choice for calcium channel assays using single wavelength excitation, which is typical for most plate readers. Fluo3, which is based on a fluorescein, is conveniently excited with the 488 nm argon ion laser line and its fluorescence can increase 100-fold upon binding Ca2+ (KD = ~ 400 nM) [20]. Fura2 has been favored for ratiometric measurements and requires instrumentation capable of dual excitation such as the plateimaging fluorimeter developed by the former SIBIA Neurosciences and SAIC group [21]. These intracellular indicators are typically loaded into cells as acetoxymethyl (AM) esters which are subsequently hydrolyzed via intracellular esterases to the tetraacid forms which are trapped within cells. The structures of commonly used fluorescent ion indicators are shown in Fig. 8.3.
Fig. 8.3 Structures of fluorescent ion probes.
8.3 Ion-sensitive Fluorescent Probes
8.3.2 Indicators of Other Ions
Cellular fluorescent indicators of other common ions that permeate through ion channels exist and have been successfully applied in a variety of cells and tissue, however they are not as commonly employed as Ca2+ indicators. This is particularly true for HTS applications that generally require a larger fluorescent change to ensure robustness, and compatibility with screening plate readers. Specific ion channel dependent fluorescence changes are modest because of limited ion selectivity and the difficulty of generating sufficiently large ion concentration changes due to target ion channels. All fluorescent indicators for Na+ and K+ are based on crown ethers, which confer ion selectivity, conjugated with two fluorescent probes. Upon binding to the macrocycle, the fluorescence intensity is enhanced. The sodium, SBFI, and potassium, PBFI, indicators use a benzofuran fluorphore [22]. These probes produce ratiometric changes in their excitation spectra that require similar detection instrumentation as Fura2. Sodium green was developed by Molecular Probes using fluorescein as the fluorophore which enables excitation in visible wavelengths similar to Fluo3 [23]. A key issue that limits the utility of these probes for sodium channels, as an example, is that the selectivity over K+ is not sufficiently high to eliminate competition with high intracellular K+. As a result large, >10 mM, intracellular Na+ concentrations are required to elicit significant fluorescence changes. This is not feasible for many channels that inactivate rapidly and/or express at low levels. For K+ channels the intracellular concentrations are essentially fixed, however, the use of thallium as a surrogate in combination with the low affinity Ca2+ indicator BTC has been shown to be capable of large K+ channel dependent signals[24] and is discussed later. Chloride indicators are primarily quinolinium or acridinium dyes that have decreased fluorescence when exposed to higher halide concentrations due to increasing collisional quenching. Different dyes, including SPQ and MQAE, have been applied to monitor CFTR activity [25]. An iodide-sensitive pteridine dye with improve optical properties has been reported [26] and applied to studying CFTR function [27]. An important property of these dyes is that their fluorescence is essentially insensitive to other physiological relevant anions and nitrate. This feature has been used to develop CFTR assays. CFTR expressing cells are loaded with Cl– or I– and a dye such as SPQ. The extracellular Cl– is replaced with nitrate and upon CFTR activation with forskolin intracellular Cl– leaves the cell via CFTR and is exchanged with nitrate, which causes the cellular fluorescence to increase. While routinely used for CFTR these approaches have not been generally used because they have limited brightness and signal changes, require the channel to stay open for a long time (like CFTR), and assay development complications that arise from other endogenous conductance pathways for halides and anion substitutes, can also be problematic.
195
196
8 Ion Channel Assays Based on Ion and Voltage-sensitive Fluorescent Probes
8.4 Fluorescence Assays for Ion Channels
In the remainder of the chapter, we will describe specific examples of how fluorescence probes have been applied to assay various ion channel sub-families. A large number of these applications are directed toward drug discovery because these assays can be automated and used cost effectively to profile large numbers of compounds. This is currently a necessity because of the inability to design modulators de novo. A summary of available and preferred options for the individual sub-families is listed in Table 8.2. Table 8.2 Fluorescent probes and assays for ion channel classes: the approach most often used is highlighted in bold. Target Membrane channel class potential
Ion indicators
Fluorescent proteins
Comments
Na+
FRET Redistribution
SBFI, Na-green
No
Fast FRET probes and E-VIPR use-dependence without gating modifiers
Ca2+
FRET Redistribution
Fluo-3&4, Fura2
Calcium-sensitive GFPs
Co-expression of inward rectifier used to assay voltage-dependence
K+
FRET Redistribution
Tl+ sensitive probes
No
Thallium as a substitute for K+ used for fluorescent flux assay
Cl–
FRET Redistribution
Quenching dyes (e. g. SPQ)
Halide-sensitive GFPs
Trafficking assays for CFTR
If Ca2+ permeable
yes
TRP nicotinic channels use both Ca2+ and membrane potential dyes
Ligand-gated FRET Redistribution
8.4.1 Calcium Channels
The ability to develop functional fluorescent calcium channel assays greatly benefited from the development of fluorescent intracellular calcium probes and screening instrumentation, such as the FLIPR [9, 28] and the voltage ion probe reader (VIPR) [12]. Typically these assays take advantage of the ~105-fold Ca2+ concentration gradient across the plasma membrane and high sensitivity and selectivity of probes such as Fura2 [29] and Fluo-3 [20]. The channels are typically activated with high K+ or ligand and the probes sensitively detect intracellular concentration changes from ~ 50–200 nM basal levels. Since the basal intracellular calcium con-
8.4 Fluorescence Assays for Ion Channels
centrations levels are so low very few activated channels are required to elicit a substantial fluorescence change. Voltage-gated calcium channels are the targets for established drugs such as dihydropyridines, phenylalkylamines, and benzothiazepines that block L or CaV1 channels and are used to treat cardiovascular disease, in particular hypertension and angina. The N-type or CaV2.2 channel is the target for the recently launched drug PrialtTM, which is used to treat chronic morphine resistant pain [30]. Functional calcium channel assays have been developed for most members of this target class. Velicelebi and coworkers have developed functional Ca2+ assays against CaV2 family channel complexes using stably expressing cell lines [21]. CaV-dependent calcium influx was stimulated with high K+ depolarization and the assay was able to reproduce the known molecular pharmacology differences of peptide toxins against these CaV subtypes. For example, they found that the m-conotoxin GVIA selectively blocked CaV2.2 over CaV2.1 and CaV2.3. This approach has also been applied to the screening of plant extracts for modulators of neuronal channels [31]. One of the limitations of assays designed to identify blockers for voltage-gated channels is that many blockers are state and voltage dependent. Xia and coworkers co-expressed the inward rectifier Kir2.3 with a CaV1 or L-type channel complex in order to probe the voltage dependence of block by incubating compound and cells at two extracellular K+ concentrations before applying a maximal high K+ stimulation [32]. They showed that blockers such as nimodopine, which are known to be voltage-dependent blockers with higher affinity at less negative membrane potentials, are more sensitively detected when the cells are K+-clamped at less negative potentials. In the case of nimodipine, they determined an IC50 of 3 nM at 30 mM external K+ versus 60 nM at 6 mM external K+. The ability to assay the relative statedependent activity of channel modulators in a HTS compatible format is a significant improvement over traditional high K+ induced assay protocols. 8.4.2 Non-voltage-gated Cation Permeable Channels
In addition to voltage-induced channel responses, fluorescent indicators have been widely used to measure activity of cation permeable channels that are activated by voltage-independent mechanisms. These channels can be classified as ligand-gated or ligand-activated, a characteristic property used to activate or induce channel activity at a defined time in the assay. For example exogenous application of a ligand to trigger external calcium entry has been essential to implement calcium indicator assays to aid the discovery of small molecules that pharmacologically modulate channel function. One important type of ligand-gated channel is the transient receptor potential (TRP) protein channel family. This class of proteins has gained considerable attention over the past several years as a novel and rapidly expanding channel superfamily. These primarily voltage-independent channels are most notable for their ability to respond to a variety of physical and chemical stimuli, such as temperature, mechanical force, phospholipids, oxidants and many others [33–35]. Because of these
197
198
8 Ion Channel Assays Based on Ion and Voltage-sensitive Fluorescent Probes
properties and where they are expressed, TRP channels have been implicated in a wide range of physiological processes including sensory physiology and immune response and related diseases. The calcium permeable nature of many TRP channels has been exploited to advance the field by assisting the identification and determination of the function of novel family members, for example TRPM3[36], as well as the search for new physiological stimuli and novel small molecules. TRPV1 (vanilloid receptor sub-family) has been defined by its activation by vanilloids such as capsaicin and noxious heat. Figure 8.4 demonstrates the use of the Fluo3 and Fura-Red calcium indicators to measure hTRPV1 response to capsaicin in a 3456-well nanoplate [37]. Capsazepine, a known TRPV1 antagonist dose-dependently inhibits the fluorescent response and is shown in the right panel. Using a similar assay, Rami and coworkers [38] used Fluo-3 to search for compounds that inhibited the capsaicin induced calcium-dependent fluorescent responses. In this study, they characterized a potent series of compounds that bind to an extracellular site to produce inhibition of temperature-induced activation. In a side-by-side comparison of the responses of the cold responsive channel TRPM8 and TRPV1 Behrendt and coworkers [39] found selective action from a library of odorants, providing new tools to help dissect the details of complex sensory signaling pathways that discriminate among different thermo-sensations. Fluorescent calcium indicators have also been used to identify compounds that modulate native channel complexes thought to be related to or comprised by members of the TRP family. Ishikawa et al. [40] examined the ability of small molecules to inhibit induced Fura-2 responses in human Jurkat T-cells to identify a potent inhibitor of
Fig. 8.4 hTRPV1 response in 3456 microtiter plate using fluorescent Ca2+ indicator Fluo-3/ Fura-red readout. (A) HEK cells stably expressing hTRPV1 were plated in a 3456-nanoplate. Cells were loaded with Fluo-3AM/Fura-red and capsaicin (200 nM) was added to evoke a calcium transient measured using a fluorescent plate reader. Each 6X6 square, one example is
enlarged, represents an 11 point concentration response analysis to a single compound in triplicate as well as capsaicin, positive (capsaicin plus antagonist) and negative (DMSO) controls with N = 1. (B) Concentration–response curve of capsazepine block of capscaicin-stimulated hTRPV1 is shown. (This figure also appears with the color plates.)
8.4 Fluorescence Assays for Ion Channels
Icrac, which was shown to alter immunosuppressant activity in a mouse contact hypersensitivity model. Appendino and colleagues have reported another example of extending studies from bench to preclinical stage. In this study, potent sub-nanomolar TRPV1 agonists were identified as well as sub-micromolar antagonists from channels exogenously expressed in HEK293 cells using Fluo-3 [41]. These agents were shown to pharmacologically modulate dorsal root ganglion neurons, bladder smooth muscle contraction and significantly altered micturition volume and frequency in an obstructed bladder function model. Thus the use of calcium indicators has greatly facilitated the identification of agents that modify in vivo physiology and are now being tested in clinical trials. In addition to elevating intracellular calcium levels, calcium ion entry can also produce membrane depolarization or hyperpolarization, via calcium-activated K+ channels. The nictoinic receptor, a calcium permeable cation selective channel is an example of a channel that has multiple functional roles. It is activated by acetylcholine and is important in excitation–contraction coupling as well as many central and peripheral neuronal functions. The cell signaling events that a channel participates in can vary and can lead to complex function. Recently, a number of different cell lines using both calcium and membrane potential probes to measure nicotinic receptor channel activity have been used to dissect and exploit these differences [42]. Data from both formats were complementary but demonstrated distinct differences in ligand-induced channel kinetics. Membrane potential measurements demonstrated greater sensitivity and potency to activation by agonists, while antagonists were slightly less potent as compared to calcium indicator measurements in the same cells. While some differences may be addressed by channel density or assay sensitivity importantly, this represents the ability to use fluorescent indicators to differentiate between channel functions. For voltage-gated calcium channels, participation of these channels in calcium entry versus membrane depolarization is also of interest as both functions have been shown to impact and regulate cellular physiology. Figure 8.5 compares the measured fluorescence membrane potential versus the fluorescence calcium esponse resulting from applying an electrical field to cells containing a CaV channel. Note the differences in waveform of the calcium and Vm signal. The membrane potential has an abrupt large transient change in signal that then reaches a plateau, however, the calcium indicator signal increases slowly building to a sustained level. The cellular property of calcium Ca2+ release (CICR) provides an additional complexity for the application of calcium dyes to the study of channel activity. CICR can greatly amplify the initial calcium entry through the plasma membrane-bound assay target. While providing high sensitivity, CICR can also limit the detection of slowly acting blockers and cause displacement of the dose response relationship relative to the actual compound activity on the target channel. The use of fast membrane potential probes, such as FRET probes, can be designed to provide complementary data that is not dependent on CICR. Using both formats as a measure of CaV activity, differences in pharmacology have been revealed and may be pursued to identify small molecules that differentiate between these CaV functions.
199
200
8 Ion Channel Assays Based on Ion and Voltage-sensitive Fluorescent Probes Fig. 8.5 Membrane potential versus calcium signal. Comparative measurements from CaV containing cells evoked using electric field stimulation using either an indicator of changes in membrane potential or intracellular calcium ions. HEK cells stably expressing a CaV channel were plated in a 96-well E-VIPR. The stimulation period is indicated. Cells were stained with membrane potential FRET dye pair (top) or loaded with Fluo3AM and Fura RedAM (bottom). All responses were blocked by the calcium channel blocker Mibefradil (10 mM, not shown).
8.4.3 Sodium Channels
Voltage-gated sodium channels (Navs) historically have been one of the most difficult ion channel classes for which to develop high-throughput assays. The difficulty has arisen from a combination of challenges including cellular expression, voltage dependence of activation and inactivation, millisecond activation and inactivation kinetics, and voltage and state dependence of many blockers. The use of fluorescence membrane potential probes is the primary approach for this target class because of its high sensitivity to small currents and high-speed readout. Because these channels open and inactivate in milliseconds most assays require the use of gating modifiers such as veratridine and batrachotoxin (BTX) to lock the channel in an open conformation to prolong Na+ influx and cause sufficient current to depolarize cells. The use of toxins in assays is generally not desired because test compounds must compete for these sites and can limit the sensitivity of the assays to the detection of competitive binders. Typically cells are incubated in the absence of extracellular Na+, substituting with organic cations such as tetramethylammonium, choline, or N-methyl-d-glucamine which do not permeate through open NaV channels. With the channel locked open, addition of Na+ results in an Nav-dependent inward current that rapidly depolarizes the cell [12]. Blockers are identified by their ability to inhibit this depolarization. The most commonly used fluorescent membrane potential probes are DiBAC4(3), FMP and FRET-based voltage sensor probes. Some recent examples of these approaches have been published. Vickery and coworkers have applied FMP dye and NaV activator evoked depolarizations to compare the molecular pharmacology of activators and blockers against rNaV1.8, rNaV1.2 and hNaV1.5 [43]. Using these assays they observed that different activators or gating-modifiers, including
8.4 Fluorescence Assays for Ion Channels
veratridine and the type II pyrethroids deltamethrin and venfalerate, had efficacies that were subtype-dependent. They reported that veratridine induced NaV-dependent depolarizations in rNaV1.2 and hNaV1.5 but was not effective against rNaV1.8. The type II pyrethroids were most effective in hNaV1.5 and rNaV1.8 and were potentiated with veratridine. Of the approximately15 known blockers examined, all blocked the three subtypes with approximately equal concentrations, except for the neurotoxin TTX. Felix and coworkers have used voltage-sensitive FRET probes to develop assays for NaV1.7 and NaV1.5. They have reported that pre-incubation of compounds prior to addition of activators enabled compounds to pre-bind to the native conformation and then subsequently added the activator prior to Na+ addition [44]. They observed that this protocol more accurately reported blocking activities for some classes of compounds. The use of electrical stimulation and high-speed FRET probes has been introduced recently and enables repetitive millisecond stimulation and detection of NaVs in standard 96 and 384 well plates. The electrical stimulation voltage ion probe reader (E-VIPR) eliminates the need for using pharmacological modifiers and enables repetitive stimulation. E-VIPR is very flexible and has been shown to robustly identify and characterize NaV blocker activity, including assessment of use-dependence (Huang et al. submitted). Figure 8.6 shows E-VIPR concentration–response traces of 5 drugs using an E-VIPR assay and stimulating at 5 Hz. The response includes contributions from both voltage and frequency. At intermediate concentrations use-dependence is detected as differential blockade of the first (tonic) and last (use-dependent) stimulated voltage-transients. TTX, which is not voltage-dependent, does not exhibit as much use-dependence as the local anesthetic drugs. 8.4.4 Potassium Channels
Potassium channels are deceptively challenging targets for which to develop useful fluorescence assays. The K+ channel family is relatively large and individual members are activated in a wide variety of voltage and nonvoltage mechanisms. While it is straightforward to add high K+ to cells and elicit a depolarization, it is much more challenging to develop an assay specific to a single target channel. The high and variable K+ channel background conductances make cell line and assay development difficult. The most common approach utilizes membrane potential dyes, although recently, the application of low affinity Ca2+ probes in combination with thallium as a K+ substitute has been used to develop HTS compatible fluorescence assays [24]. Membrane potential assays require the use of cell lines with minimal background conductances and relatively positive resting potentials. For blocker assays, expression of the target K+ channel will set the membrane potential at near its activation potential or near the K+ reversal potential (Krev), so that block will result in a depolarization (if there are no other significant K+ conductances). The block can be monitored continuously after compound addition or by probing for the re-
201
202
8 Ion Channel Assays Based on Ion and Voltage-sensitive Fluorescent Probes
Fig. 8.6 Activity of use-dependent NaV blockers in E-VIPR membrane potential assay. Concentration–response traces for TTX, tetracaine, amitriptyline, mexiletine, lidocaine and lamotrigine block of hNaV1.3-dependent depolarization transients using a 5 Hz stimulation protocol. The concentrations shown above the first ratio responses, at the left side
of the figure, are the highest concentrations tested. Each blocker was diluted 1 to 4 across the plates, as illustrated by the arrow. Use-dependent block is evident for all compounds except TTX, and is manifested as greater block at the end of the response train compared to the first response. Control responses without blockers are shown on the far right.
duction on high K+ induced depolarization. For activators or “openers”, the expressed channel is closed at less negative potentials and compound activation causes the cell to hyperpolarize toward the potassium equilibrium potential. This strategy has been employed for identifying and profiling KATP channels openers using redistribution dyes such as DiBAC4 in the bladder [8, 45, 46], myocytes [47] and in cells heterologously expressing SUR1 and the inward rectifier Kir6.2 [48]. DiBAC4 has also been used to identify blockers, for example fluoxetine inhibition of Ca-activated K+ channels [49]. Fluorescent probes that directly measure K+ are difficult to apply for monitoring + K channel-dependent [K+] changes because intracellular and extracellular [K+] do not change sufficiently under experimentally accessible conditions to elicit a large fluorescence change. Thus, K+ surrogates have been widely used for developing K+ channel assays. Rb86+ is routinely used as a radioactive substitute for K+ in flux assays (see Chapter 7). Recent developments in the application of atomic spectroscopy detection have led to nonradioactive Rb+ flux assays. Thallium (Tl+), another K+ surrogate, can readily enter cells via Na+/K+ ATPase and Na+/K+/Cl+ co-trans-
8.4 Fluorescence Assays for Ion Channels
port mechanisms [50]. Tl+ readily fluxes through K+ channels and is known to quench various water-soluble fluorophores. Its quenching properties have been exploited to probe the activity of acetylcholine receptors [51, 52]. Recently, Weaver and colleagues have identified a coumarin benzothiazole-based low affinity Ca2+ fluorescent probe BTC, structure shown in Fig. 8.3, that functions as an intracellular Tl+ sensor from a screen of fluorescent metal chelating probes [24]. Using this probe in combination with a Cl–-free assay buffer, which overcomes TlCl solubility limitations, they devised an HTS compatible fluorescent assays for measuring KCNQ2 and SK3 K+ channel modulator activities. Two-fold fluorescence changes were achieved with minimal interference from intracellular Ca2+ and fluorescent test compounds because of weak Ca2+ affinity and the visible excitation wavelengths. Researchers considering using Tl+ should be aware that it is readily absorbed and is known to be toxic [53]. 8.4.5 Chloride Channels
Extracellular and intracellular ligand-gated Cl– channels regulate membrane potential in excitable and nonexcitable cells and have been identified as drug targets for a number of different conditions and diseases, including neurological disorders and cystic fibrosis. For example, the GABAA receptor is a ligand-gated Cl– channel that is the primary target of many drugs for the treatment of anxiety, epilepsy, muscle spasms, sleep related conditions and is the target for benzodiazepine drugs such as diazepam. Despite GABAA being a well validated drug target, historically it has been difficult to develop fluorescence assays because of desensitization, limitations of Cl– sensitive probes, background conductances, and the desire to identify compounds that modulate the effects of GABA. Recently, FRET membrane potential probes have been successfully applied to develop assays for characterizing GABAA pharmacological agents, including potentiators [54, 55]. This approach has also helped to identify new selective allosteric modulators that can differentiate between beta subunits [56] and to illuminate the structural basis of the selectivity of neurosteriods on the receptor [57]. Cystic fibrosis is due to mutations in the gene encoding the epithelial PKAgated Cl– channel known as the cystic fibrosis transmembrane conductance regulator (CFTR) [58, 59]. The most common mutation is a deletion of phenylalanine at position 508 in the first nucleotide-binding domain of CFTR. This mutation leads to impaired trafficking and gating of CFTR to the apical membrane of respiratory epithelia, resulting in the defective anion transport that underlies the lung disease in patients [60]. Several fluorescent-based assays have been developed to identify and track the structure–activity relationship (SAR) of small molecules that directly target mutant CFTR to restore its defective gating or trafficking [61]. These include the use of fluorescent membrane potential and halide sensors, including genetically-targeted fluorescent proteins. Voltage-sensitive membrane potential probes offer a sensitive and convenient approach to the indirect monitoring of anion flux through chloride channels in a
203
204
8 Ion Channel Assays Based on Ion and Voltage-sensitive Fluorescent Probes
variety of different cells types, including recombinant cells and primary cell cultures. The excellent throughput, sensitivity, and reproducibility of such assays make them especially useful for tracking SAR to support medicinal chemistry optimization. However, because the membrane potential response is nonlinear, reaching equilibrium at the reversal potential for Cl–, the assay window and ability to monitor potency (Fig. 8.7) and efficacy (Fig. 8.8) can be compromised. Two approaches can be used to circumvent this problem. First, the rate of membrane potential change, rather than the absolute magnitude, can be used to monitor Cl– flux. To do this, the temporal response of the dye must be sufficiently fast to accurately track the membrane potential. Second, decreasing channel density or reducing agonist concentrations can reduce the assay sensitivity and expand the linear range. Although this is particularly useful for the identification of highly effica-
Fig. 8.7 CFTR membrane potential assay demonstrates efficacy limitations compared to flux assays: Response to CFTR activation between fluorescent-based and electrophysiological assay formats. To monitor the response to increasing amounts of CFTR activation cells expressing wild-type CFTR were mixed with parental cells in the indicated proportions (% wild-type CFTR). The response to CFTR activation using a maximal concentration of forskolin was monitored in both the fluorescence assay (black circles) and in an electrophysiological assay (red circles) under voltage-clamp control. The response in both assays was normalized to the response using
100 % wild-type CFTR-expressing cells. In the fluorescence-based assay, the half maximal response was observed at ~ 3% wild-type CFTR and was nonlinear as the concentration of wild-type CFTR expressing cells was increased. In contrast, the half-maximal response in the using chamber assay was reached at ~ 60 % wild-type CFTR and was linear. These results highlight the nonlinearity of the fluorescence-based assays, which can limit the SAR evaluation of agonist efficacy because the sensitive response saturates with low amounts of CFTR activation. (This figure also appears with the color plates.)
8.5 Assays for Monitoring Channel Trafficking
cious compounds, it can potentially reduce the range of potency, limiting SAR evaluation. Another assay approach is to monitor Cl– flux directly using genetically-encoded anion-sensitive fluorescent proteins, such as mutants of yellow fluorescent protein (YFP) [62, 63]. Anion binding shifts the YFP chromophore pKa values and results in quenched fluorescence. Cellular YFP sensor expression enables measurement of transmembrane halide fluxes. Similar to small molecule halide indicators, iodide and nitrate are more sensitively detected and consequently similar anion replacement strategies have been successfully applied to assaying CFTR activity, including high throughput screening [61, 64]. In another application, GABAA-dependent intracellular Cl– changes were measured in cultured hippocampal neurons using a Cl– sensitive YFP mutant fused to Cl– insensitive FRET acceptor protein [65]. The chimeric fluorescent probe provides emission ratio detection that reduces experimental artifacts compared to single intensity indicators. Despite successful applications of fluorescent protein probes for ion channel analysis, their broad use is limited by relatively small fluorescence changes and cellular expression.
8.5 Assays for Monitoring Channel Trafficking
In addition to using fluorescence-based assays to monitor changes in CFTR gating, they can be used to monitor increased channel density due to compound rescue of protein processing and/or trafficking. A few pharmacological agents have been identified that partially rescue the defective trafficking of mutant CFTR, leading to increased cell surface density. 4-Phenyl butyrate, for example, acts at millimolar concentrations to produce a modest increase in DF508-CFTR density in vitro [66], but had limited efficacy in vivo [67]. To enable the detection of compounds that alter protein trafficking, it is necessary to incubate the compounds for several hours prior to monitoring activity to allow for de novo synthesis and insertion into the membrane. This presents a potential issue in that incomplete wash-out of compounds that potentiate channel activity rather than increase its density may also be identified. To separate compounds that rescue the defective processing or trafficking from those that alter channel gating it is necessary to obtain independent confirmation using biochemical assays. For example, the correct processing and trafficking of mutant CFTR can be confirmed by monitoring its maturation via passage through the golgi network. Compounds may also act by increasing the gating activity, as well as trafficking to the cell’s surface, further complicating the assessments of channel density in the membrane. To directly monitor changes in the cell surface density and eliminate the impact of channel gating, fluorescence-based assays that monitor protein localization could be used. For example, GFP has been tagged to the cytoplasmic C terminal tail of CFTR and has been successfully used to monitor the cytoplasmic and cell surface protein localization [68]. Although the throughput of such assays is lim-
205
206
8 Ion Channel Assays Based on Ion and Voltage-sensitive Fluorescent Probes
8.6 Summary
ited, improvements in algorithms designed to separate cytoplasmic from nuclear staining could improve the utility of such assays. In addition to cystic fibrosis, protein trafficking and processing abnormalities of ion channels have been identified as the underlying cause of diseases, such as familial hyperinsulinism (Kir6.2), Sjögren’s syndrome (aquaporin-5), nephrogenic diabetes insipidus (aquaporin-2), Brugada Syndrome (NaV1.5), inherited long QT syndrome (KCNQ1, hERG), and Andersen-Tawil syndrome (Kir2.1). In some cases, channel ligands (i.e, agonists and antagonists) have been demonstrated to partially rescue the defective trafficking and membrane density of mutant channels, including NaV1.5 (mexiletine)[69], hERG (astemizole, terfenadine) [70], and Kir6.2 (sulfonylureas) [71]. Although a few agents have been identified, fluorescence-based assays similar to those used for mutant CFTR could be used to identify more drug-like, potent, and efficacious agents to correct the defective folding and/or trafficking of Na+ and K+ channels.
8.6 Summary
Fluorescence approaches to studying ion channel function and the interactions of modulating molecules offer significant advantages in sensitivity, ease of use, and high throughput analysis. Existing fluorescent cellular probes provide access to all classes of ion channels. These approaches primarily use membrane potential and ion-sensitive probes, which respond to the net ion flux through the channel. Each has it own assay development challenges and in some cases there are significant limitations on the types of information that can be obtained. Through a combination of improved probes and instrumentation, increasingly higher information content analysis is now possible, for example, fluorescence assays for use-dependent blockade and channel trafficking. Looking forward, additional detail should be attainable through fluorescent proteins and assays that can monitor different channel conformational states and their interactions with other proteins. 3 Fig. 8.8 CFTR membrane potential assay demonstrates the dependence of channel density on agonist sensitivity: Effects of CFTR density on agonist activity in fluorescent membrane potential assays. (A) Theoretical Michaelis– Menten type increase in open probability of CFTR by forskolin (Kd = 5 mM). (B) Simulation of the membrane potential response to CFTR activation by forskolin. The concentration dependent effects on membrane potential were calculated using the Goldman–Hodgkin–Katz equation incorporating the open probability at each agonist concentration and CFTR densities of 0.01 to 100 times the background permeability, which was assumed to be due to an endo-
genous K+ conductance. Only at very low CFTR densities did the EC50 approximate the Kd for forskolin. (C) To monitor the effects on CFTR density on agonist stimulation in a fluorescence-based membrane potential assay, CFTRexpressing cells were mixed with parental cells at the indicated proportions and expressed as % wild-type (wt) CFTR. As observed in the simulations, only at very low wt-CFTR proportions did the EC50 for forskolin approximate its Kd. These results illustrate that the potency of ion channel agonists in a fluorescence-based assay is highly sensitive to the channel density. (This figure also appears with the color plates.)
207
208
8 Ion Channel Assays Based on Ion and Voltage-sensitive Fluorescent Probes
References 1 J. E. Gonzalez, P. A. Negulescu, Intracellular detection assays for highthroughput screening, Curr. Opin. Biotechnol. 1998, 9(6), 624–631. 2 L. M. Loew, Potentiometric membrane dyes, in Fluorescent and Luminescent Probes for Biological Activity, W.T. Mason (Ed.), Academic Press, San Diego, 1993, pp. 150–160. 3 D. F. Baxter, et al., A novel membrane potential-sensitive fluorescent dye improves cell-based assays for ion channels, J. Biomol. Screen., 2002, 7(1), 79– 85. 4 K. L. Whiteaker, S. M. Gopalakrishnan, D. Groebe, C. C. Shieh, U. Warrior, D. J. Burns, M. J. Coghlan,V. E. Scott, M. Gopalakrishnan,Validation of FLIPR membrane potential dye for high throughput screening of potassium channel modulators, J. Biomol. Screen. 2001, (5), 305–312. 5 J. E. Gonzalez, R.Y. Tsien,Voltage sensing by fluorescence resonance energy transfer in single cells, Biophys. J. 1995, 69(4),1272–1280. 6 J. E. Gonzalez, R.Y. Tsien, Improved indicators of cell membrane potential that use fluorescence resonance energy transfer, Chem. Biol. 1997, 4(4), 269– 277. 7 R. F. Flewelling, W. L. Hubbell, The membrane dipole potential in a total membrane potential model. Applications to hydrophobic ion interactions with membranes, Biophys. J. 1986, 49(2), 541–552. 8 K. O. Holevinsky, Z. Fan, M. Frame, J. C. Makielski,V. Groppi, D. J. Nelson, ATP-sensitive K+ channel opener acts as a potent Cl- channel inhibitor in vascular smooth muscle cells, J. Membr. Biol. 1994, 137(1), 59–70. 9 K. S. Schroeder, B. D. Neagle, FLIPR: A new instrument for accurate, high throughput optical screening, J. Biomol. Screen. 1996, 1, 75–80. 10 J. Farinas, A. W. Chow, H. G. Wada, A microfluidic device for measuring cellular membrane potential, Anal. Biochem. 2001, 295(2), 138–142.
11 J. E. Gonzalez, M. P. Maher, Cellular fluorescent indicators and voltage/ion probe reader (VIPR) tools for ion channel and receptor drug discovery, Receptors Channels 2002, 8(5–6), 283–295. 12 J. E. Gonzalez, K. Oades,Y. Leychkis, A. Harootunian, P. A. Negulescu, Cell-based assays and instrumentation for screening ion-channel targets, Drug Discov. Today 1999, 4(9), 431–439. 13 M. Falconer, et al., High-throughput screening for ion channel modulators, J. Biomol. Screen. 2002, 7(5), 460–465. 14 D. E. Epps, M. L. Wolfe,V. Groppi, Characterization of the steady-state and dynamic fluorescence properties of the potential-sensitive dye bis-(1,3-dibutylbarbituric acid)trimethine oxonol (Dibac4(3)) in model systems and cells, Chem. Phys. Lipids. 1994, 69(2), 137– 150. 15 T. Kenakin, Stimulus-Response Mechanisms, in Pharmacologic Analysis of Drug-Receptor Interaction, Lippincott-Raven, Philadelphia, 1997, pp. 78–105. 16 J. Koester, Membrane Potential, in Principles of Neural Science, E. R. Kandel, J. H. Schwartz, T. M. Jessell (Eds.), Elsevier, New York, 1991, pp. 81–94. 17 C. Wolff, B. Fuks, P. Chatelain, Comparative study of membrane potentialsensitive fluorescent probes and their use in ion channel screening assays, J. Biomol. Screen. 2003, 8(5), 533–543. 18 W. Tang, J. Kang, X. Wu, D. Rampe, L. Wang, H. Shen, Z. Li, D. Dunnington, T. Garyantes, Development and evaluation of high throughput functional assay methods for HERG potassium channel, J. Biomol. Screen. 2001, 6(5), 325–331. 19 K. R. Gee, K. A. Brown,W. N. Chen, J. Bishop-Stewart, D. Gray, I. Johnson, Chemical and physiological characterization of fluo-4 Ca(2+)-indicator dyes, Cell Calcium 2000, 27(2), 97–106. 20 A. Minta, J. P. Kao, R.Y. Tsien, Fluorescent indicators for cytosolic calcium based on rhodamine and fluorescein chromophores, J. Biol. Chem. 1989, 264(14), 8171–8178.
References 21 G. Velicelebi, K. A.Stauderman, M. A. Varney, M. Akong, S. D. Hess, E. C. Johnson, Fluorescence techniques for measuring ion channel activity, Methods Enzymol. 1999, 294, 20–47. 22 A. Minta, R. Y. Tsien, Fluorescent indicators for cytosolic sodium, J. Biol. Chem. 1989, 264(32), 19449–19457. 23 G. P. Amorino, M. H. Fox, Intracellular Na+ measurements using sodium green tetraacetate with flow cytometry, Cytometry 1995, 21(3), 248–256. 24 C. D. Weaver, D. Harden, S. I. Dworetzky, B. Robertson, R. J. Knox, A thallium-sensitive, fluorescence-based assay for detecting and characterizing potassium channel modulators in mammalian cells, J. Biomol. Screen. 2004, 9(8), 671–677. 25 M. R. West, C. R Molloy, A microplate assay measuring chloride ion channel activity, Anal. Biochem. 1996, 241(1), 51–58. 26 S. Jayaraman, L. Teitler, B. Skalski, A. S. Verkman, Long-wavelength iodide-sensitive fluorescent indicators for measurement of functional CFTR expression in cells, Am. J. Physiol. 1999, 277(5 Pt. 1), C1008–1018. 27 F. Munkonge, et al., Measurement of halide efflux from cultured and primary airway epithelial cells using fluorescence indicators, J. Cystic Fibrosis 2004, 3 (Suppl. 2), 171–176. 28 E. Sullivan, E. M. Tucker, I. L. Dale, Measurement of [Ca2+] using the Fluorometric Imaging Plate Reader (FLIPR), Methods Mol. Biol. 1999, 114, 125–133. 29 G. Grynkiewicz, M. Poenie, R. Y. Tsien, A new generation of Ca2+ indicators with greatly improved fluorescence properties, J. Biol. Chem. 1985, 260(6), 3440–3450. 30 G. P. Miljanich, Ziconotide: neuronal calcium channel blocker for treating severe chronic pain, Curr. Med. Chem. 2004, 11(23), 3029–3040. 31 K. L. Rogers,W. F. Fong, J. Redburn, L. R. Griffiths, Fluorescence detection of plant extracts that affect neuronal voltage-gated Ca2+ channels, Eur. J. Pharm. Sci. 2002, 15(4), 321–330.
32 M. Xia, J. P. Imredy, K. S. Koblan, P. Bennett, T. M. Connolly, State-dependent inhibition of L-type calcium channels: cell-based assay in highthroughput format, Anal. Biochem. 2004, 327(1), 74–81. 33 D. E. Clapham, L. W. Runnels, C. Strubing, The TRP ion channel family, Nat. Rev. Neurosci. 2001, 2(6), 387– 396. 34 R. Inoue, TRP channels as a newly emerging non-voltage-gated CA2+ entry channel superfamily, Curr. Pharm. Des. 2005, 11(15), 1899–1914. 35 C. Montell, The TRP superfamily of cation channels, Sci. STKE 2005, (272): re3. 36 N. Lee, et al., Expression and characterization of human transient receptor potential melastatin 3 (hTRPM3), J. Biol. Chem. 2003, 278(23), 20890–20897. 37 L. Mere, T. Bennett, P. Coassin, P. England, B. Hamman, T. Rink, S. Zimmerman, P. Negulescu, Miniaturized FRET assays and microfluidics: key components for ultra-high-throughput screening, Drug Discov. Today 1999, 4(8), 363–369. 38 H. K. Rami, et al., Discovery of small molecule antagonists of TRPV1, Bioor. Med. Chem. Lett. 2004, 14(14), 3631– 3634. 39 H. J. Behrendt, T. Germann, C. Gillen, H. Hatt, R. Jostock, Characterization of the mouse cold-menthol receptor TRPM8 and vanilloid receptor type1 VR1 using a fluorometric imaging plate reader (FLIPR) assay, Br. J. Pharmacol. 2004, 141(4), 737–745. 40 J. Ishikawa, K. Ohga, T. Yoshino, R. Takezawa, A. Ichikawa, H. Kubota, T. Yamada, A pyrazole derivative,YM58483, potently inhibits store-operated sustained Ca2+ influx and IL-2 production in T lymphocytes, J. Immunol. 2003, 170(9), 4441–4449. 41 G. Appendino, et al., Development of the first ultra-potent “capsaicinoid” agonist at transient receptor potential vanilloid type 1 (TRPV1) channels and its therapeutic potential, J. Pharmacol. Exp. Ther. 2005, 312(2), 561–570.
209
210
8 Ion Channel Assays Based on Ion and Voltage-sensitive Fluorescent Probes 42 R. W. Fitch,Y. Xiao, K. J. Kellar, J. W. Daly, Membrane potential fluorescence: a rapid and highly sensitive assay for nicotinic receptor channel function, Proc. Natl. Acad. Sci. U S A 2003, 100(8), 4909–4914. 43 R. G. Vickery, et al., Comparison of the pharmacological properties of rat Na(V)1.8 with rat Na(V)1.2 a and human Na(V)1.5 voltage-gated sodium channel subtypes using a membrane potential sensitive dye and FLIPR, Receptors Channels 2004, 10(1), 11–23. 44 J. P. Felix, et al., Functional assay of voltage-gated sodium channels using membrane potential-sensitive dyes, Assay Drug Dev. Technol. 2004, 2(3), 260– 268. 45 W. A. Carroll, et al., Synthesis and structure-activity relationships of a novel series of tricyclic dihydropyridinebased KATP openers that potently inhibit bladder contractions in vitro, J. Med. Chem. 2004, 47(12), 3180–3192. 46 M. Gopalakrishnan, et al., Characterization of the ATP-sensitive potassium channels (KATP) expressed in guinea pig bladder smooth muscle cells, J. Pharmacol. Exp. Ther. 1999, 289(1), 551– 558. 47 K. L. Whiteaker, R. Davis-Taber,V. E. Scott, M. Gopalakrishnan, Fluorescence-based functional assay for sarcolemmal ATP-sensitive potassium channel activation in cultured neonatal rat ventricular myocytes, J. Pharmacol. Toxicol. Methods 2001, 46(1), 45–50. 48 M. Gopalakrishnan, et al., Pharmacology of human sulphonylurea receptor SUR1 and inward rectifier K(+) channel Kir6.2 combination expressed in HEK293 cells, Br. J. Pharmacol. 2000, 129(7), 1323–1332. 49 G. C. Terstappen, A. Pellacani, L. Aldegheri, F. Graziani, C. Carignani, G. Pula, C. Virginio, The antidepressant fluoxetine blocks the human small conductance calcium-activated potassium channels SK1, SK2 and SK3, Neurosci. Lett. 2003, 346(1–2), 85–88. 50 T. Brismar, S. Anderson,V. P. Collins, Mechanism of high K+ and Tl+ uptake in cultured human glioma cells,
51
52
53
54
55
56
57
58
59
60
61
Cell Mol. Neurobiol. 1995, 15(3), 351– 360. H. P. Moore, M.A Raftery, Direct spectroscopic studies of cation translocation by Torpedo acetylcholine receptor on a time scale of physiological relevance, Proc. Natl. Acad. Sci. U S A 1980, 77(8), 4509–4513. W. C. Wu, H. P. Moore, M. A. Raftery, Quantitation of cation transport by reconstituted membrane vesicles containing purified acetylcholine receptor, Proc. Natl. Acad. Sci. U S A 1981, 78(2), 775– 779. R. S. Hoffman, Thallium toxicity and the role of Prussian blue in therapy, Toxicol. Rev. 2003, 22(1), 29–40. C. E. Adkins, et al., GABA(A) receptors characterized by fluorescence resonance energy transfer-derived measurements of membrane potential, J. Biol. Chem. 2001, 276(42), 38934–38939. A. J. Smith, P. B. Simpson, Methodological approaches for the study of GABA(A) receptor pharmacology and functional responses, Anal. Bioanal. Chem. 2003, 377(5), 843–851. A. J. Smith, B. Oxley, S. Malpas, G. V. Pillai, P. B. Simpson, Compounds exhibiting selective efficacy for different beta subunits of human recombinant gamma-aminobutyric acid A receptors, J. Pharmacol. Exp. Ther. 2004, 311(2), 601–609. G. V. Pillai, A. J. Smith, P. A. Hunt, P. B. Simpson, Multiple structural features of steroids mediate subtype-selective effects on human alpha4beta3delta GABAA receptors, Biochem. Pharmacol. 2004, 68(5), 819–831. P. M. Quinton, Missing Cl conductance in cystic fibrosis, Am. J. Physiol. 1986, 251(4 Pt. 1), C649–652. J. R. Riordan, et al., Identification of the cystic fibrosis gene: cloning and characterization of complementary DNA, Science 1989, 245(4922), 1066– 1073. R. C. Boucher, New concepts of the pathogenesis of cystic fibrosis lung disease, Eur. Respir. J. 2004, 23(1), 146–58. H. Yang, et al., Nanomolar affinity small molecule correctors of defective
References
62
63
64
65
66
Delta F508-CFTR chloride channel gating, J. Biol. Chem. 2003, 278(37), 35079–35085. R. M. Wachter, S.J Remington, Sensitivity of the yellow variant of green fluorescent protein to halides and nitrate, Curr. Biol. 1999, 9(17), R628–629. R. M. Wachter, D. Yarbrough, K. Kallio, S. J. Remington, Crystallographic and energetic analysis of binding of selected anions to the yellow variants of green fluorescent protein, J. Mol. Biol. 2000, 301(1), 157–171. A. S. Verkman, S. Jayaraman, Fluorescent indicator methods to assay functional CFTR expression in cells, Methods Mol. Med. 2002, 70, 187–196. T. Kuner, G. J. Augustine, A genetically encoded ratiometric indicator for chloride: capturing chloride transients in cultured hippocampal neurons, Neuron 2000, 27(3), 447–459. R. C. Rubenstein, M. E. Egan, P. L. Zeitlin, In vitro pharmacologic restoration of CFTR-mediated chloride transport with sodium 4-phenylbutyrate in cystic fibrosis epithelial cells containing delta F508-CFTR, J. Clin. Invest. 1997, 100(10), 2457–65.
67 R. C. Rubenstein, P. L. Zeitlin, A pilot clinical trial of oral sodium 4-phenylbutyrate (Buphenyl) in deltaF508-homozygous cystic fibrosis patients: partial restoration of nasal epithelial CFTR function, Am. J. Respir. Crit. Care Med. 1998, 157(2), 484–490. 68 B. D. Moyer, B.A Stanton, Analysis of CFTR trafficking and polarization using green fluorescent protein and confocal microscopy, Methods Mol. Med. 2002, 70, 217–227. 69 C. R. Valdivia, et al., A trafficking defective, Brugada syndrome-causing SCN5A mutation rescued by drugs, Cardiovasc. Res. 2004, 62(1), 53–62. 70 E. Ficker, C. A. Obejero-Paz, S. Zhao, A. M. Brown, The binding site for channel blockers that rescue misprocessed human long QT syndrome type 2 ether-a-gogo-related gene (HERG) mutations, J. Biol. Chem. 2002, 277(7), 4989–4998. 71 F. Yan, C. W. Lin, E. Weisiger, E. A. Cartier, G. Taschenberger, S. L. Shyng, Sulfonylureas correct trafficking defects of ATP-sensitive potassium channels caused by mutations in the sulfonylurea receptor, J. Biol. Chem. 2004, 279(12), 11096–11105.
211
213
9 Approaches for Ion Channel Structural Studies Randal B. Bass and Robert H. Spencer
9.1 Introduction
Biological membranes are fascinating and essential components for all living cells. Whether they are actively transporting molecules into or out of the cell, transducing a signal, or allowing solutes to cascade down a concentration gradient, decades of work in numerous laboratories has illustrated the remarkable properties of these proteins that operate within what is essentially a thin layer of very complicated grease. Since the first atomic resolution structure of a membrane protein was reported over 20 years ago (the bacterial photosynthetic reaction center from Rhodopseudomonas viridis [1]), a wealth of information about how membrane proteins function has been gained from the determination of high-resolution structures from diverse classes of membrane proteins. The ability to visualize the structure of these proteins, as has been done for their soluble counterparts, has allowed keener understanding of their folding and functions. However, it has only been in the last decade that significant progress on the structure of ion channel proteins has been seen. Many advances in the field of membrane protein structural biology have contributed to the growing number of these proteins in the databank. For ion channels, breakthroughs on solving their structures have primarily come from the identification of bacterial homologs which could be expressed in sufficient quantities for labor-intensive crystallization trials. Although general structural information on mammalian ion channels has been obtained using electron microscopy, nearly all of the X-ray crystal structures of ion channels have come from bacterial sources – primarily due to the difficulties associated with their expression in heterologous systems and establishing biochemical conditions to stabilize them outside the lipid membrane. However, very recently the crystal structure of the first mammalian K+ channel has been reported and will hopefully be a harbinger of advances yet to come [2]. In this chapter, we hope to provide a broad overview of the issues and advances made in pursuing membrane proteins such as ion channels for structural studies, and touch on topics that are specifically relevant to their expression, biochemistry and structural analysis. In summarizing these advances, Expression and Analysis of Recombinant Ion Channels. Edited by Jeffrey J. Clare and Derek J. Trezise Copyright # 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim ISBN: 3-527-31209-9
214
9 Approaches for Ion Channel Structural Studies
we also aim to highlight those researchers who are at the cutting edge and changing the way in which the sometimes daunting problem of obtaining high-resolution structures is being approached. So just how many membrane proteins are out there, and is it really worth pursuing more? Most estimates that have come from sequencing the genomes from everything from humans to bacteria suggest that roughly 30 % of all proteins in the genomes are membrane proteins. For humans, that means around 10 000 of the ~30 000 protein-coding genes can be found in the lipid bilayer. Several authors have noted that the growth of structures for membrane proteins follows a similar trajectory to those of soluble proteins. In the 20 years following the first high-resolution structure of myoglobin (determined by Kendrew et al. in 1958 [3]), 132 stable protein structures had been solved whereas by 2003, in a similar period since the first membrane protein structure, 75 unique membrane protein structures had been solved [4]. Progress on the structures of membrane proteins is clearly slower than for soluble proteins, but it is possible that the end of 2005 will see the high-resolution determination of the 100th unique membrane protein. Many of these are structures of homologous proteins from other organisms, thus the number of unique folds is somewhat smaller. In stark contrast, the Protein Data Bank (www.rcsb.pdb.org) currently has nearly 30 000 deposited protein structures in total. One of the great hopes of membrane protein structural biology that drives the desire for more structures is that the knowledge gained can be applied to the treatment of disease and help refine the development of better, targeted drugs. Determining the structure of membrane proteins will not only continue to enlighten the molecular and structural basis for their functions, but is also likely to have a broader impact by facilitating lead optimization for the development of novel therapies. Due to the critical role that ion channels play in neuronal and cell signaling and ion homeostasis, they are, and will continue to be, valuable molecular targets for the treatment of human disease. Approximately one third of currently marketed drugs produce their pharmacological actions via ion channels; this also includes around 15 % of the top 100 best-selling medicines [5]. However, the development of novel, selective ion channel modulators has been difficult with only a single new ion channel drug being approved since 1997; the exception being ziconotide (Prialt), a recently approved Ca2+-channel blocking peptide for the treatment of severe chronic pain. Given current estimates of over 400 ion channel genes encoded within the human genome, there is an abundance of ion channel targets [6], and yet we have no structural information for any of these proteins. For that matter, there currently is not a single X-ray structure of a human transmembrane protein. Fortunately, we have the structure of a handful of bacterial and archaeal ion channels at atomic resolution which have been extremely enlightening and valuable for molecular modeling purposes (see Chapter 10), and now the first structure of the first mammalian K+ channel [2]. However, high-resolution structures of the many other mammalian ion channels, especially human, remain a significant need. A major breakthrough for structural biology, as for all branches of biology, was the molecular biology revolution that allowed the recombinant expression of vir-
9.1 Introduction
tually any protein. Reviewing the methods used to obtain protein samples used in crystallization attempts, one can quickly understand that the ability to clone, express and purify recombinant proteins is nearly universal in its application to obtaining structures. This technique has been widely applied in membrane proteins with great success. However, unlike the myriad of soluble bacterial and eukaryotic proteins successfully expressed in recombinant systems, there has been only limited success for the expression, purification and solution of a eukaryotic membrane protein crystal structure produced from a recombinant system. Certainly there are many cases of expression of membrane proteins in these systems, some of which have been useful for structural studies using electron microscopy (EM) [7–9]. The recent structure of the rat Kv1.2-b2 solved from protein expressed in the yeast Pichia pastoris is a tremendous advance and represents a significant breakthrough in the field that may open the door for many other mammalian membrane proteins [2, 10]. Why are the membrane proteins so recalcitrant to expression in these systems when the soluble proteins are not? One potential issue may be the vastly different lipids that make up eukaryotic bilayers, more specifically mammalian, compared to those that are found in prokaryotes. Whereas human cell membranes are composed largely of phosphatidylcholine, phosphatidylethanolamine, sphingomyelin and cholesterol, the E. coli inner membrane is mainly composed of phosphatidylethanolamine (75 %) [11]. The remainder of the lipid content in E. coli is largely cardiolipin and phosphatidylglycerol, neither of which are prevalent in human bilayers [12]. Even with a fantastic expression system, once a eukaryotic membrane protein enters the inner membrane of E. coli, it finds a lipid environment far different from the one in which it would normally reside. Thus, it is not surprising that many eukaryotic membrane proteins fail to express in E. coli, and if they do express they typically end up in inclusion bodies. This presents a new problem. Unlike soluble proteins, the successful refolding from inclusion bodies of a complex membrane protein containing multiple transmembrane helices has not yet been widely reported, although there is a proprietary technology reported by m·phasys GmbH (Tübingen, Germany) which suggests this approach may be feasible. Another promising advance was recently reported using a membrane-integrating protein called Mistic (membrane integrating sequence for translation of integral membrane protein constructs) identified from Bacillus subtilis that, when linked to the N-terminus of a eukaryotic membrane protein, produced significant quantities of several target proteins, including a voltage-gated K+ channel [13]. Although relatively hydrophilic, Mistic folds autonomously within the bacterial membrane and appears to facilitate the integration and folding of attached eukaryotic ‘cargo’ membrane proteins that have been extremely difficult to express in prokaryotes. This and other nascent technologies should be extremely valuable in moving forward the entire field of membrane protein structural biology. Certainly it is possible for mammalian membrane proteins to be expressed and correctly folded in bacterial cells, as several examples have been cited in the literature [8, 9, 14]. However, it is likely that many will not. This fact underscores the
215
216
9 Approaches for Ion Channel Structural Studies
need to broaden the search for suitable candidates for expression in these systems. One means to address this is using a multiple orthologue approach such as that applied in the Rees [15–17] and Chang laboratories [18, 19]. In all of these cases, orthologous bacterial proteins were expressed in E. coli to find combinations that could produce sufficient quantities of protein that were properly folded, and would produce crystals of sufficient diffraction quality. If these examples of bacterial membrane proteins are any indicator, the successful expression of eukaryotic membrane proteins is also likely to require this same approach.
9.2 Expression of Membrane Proteins for Structural Studies 9.2.1 Mammalian Expression
For mammalian membrane proteins, including ion channels, it seems intuitive to seek to isolate these from native tissues or to express them heterologously in cell culture. However, since ion channels are so efficient (with flux rates of ~107 ions s–1) mammalian cells natively express relatively low levels of these proteins, typically 10–1000 channels of any one type per cell, and thus are not good sources for structural studies. Nevertheless , there are a few examples (see Table 9.2, below) where sufficient quantities have been obtained for single-particle electron microscopy (EM) including the TRPC3 cation channel [20], L-type Ca2+ channel [21], type 1 IP3 receptor [22–24], and the ryanodine receptor [25–27]. There are also a few nonmammalian examples of abundant natural sources of ion channels which have been exploited for EM structures, including the nicotinic acetylcholine receptor (nAChR) from Torpedo ray [28] and the voltage-gated Na+ channel from electric eel [29]. Stably transfected recombinant cell lines are also relatively poor sources of ion channel protein as expression levels at the cell membrane rarely exceed a few thousand channels per cell. Additionally, there can often be a large pool of immature, incorrectly processed or folded protein remaining within the cell (see Chapter 4) resulting in a heterogeneous protein sample upon purification. One way to resolve this is by purifying only the mature, fully glycosylated channels. For example, in EM studies of both the L-type (dihydropyridine) and IP3 receptor-type Ca2+ channels, wheat germ lectin affinity chromatography was commonly used during their purification [30–32], and was also employed for the 20 Å resolution structure of human CFTR [33]. Another approach is to simply remove oligosaccharide altogether. In fact, given the heterogeneous nature of glycosylation the removal of oligosaccharide is likely to be beneficial, perhaps even essential, for high-resolution structural studies. This was found to be critical for obtaining well diffracting 2D crystals of human aquaporin-1 [34, 35] and, in the cases of Shaker, aquaporin-1, Kv1.3, and SkM1 the effects of deglycosylation on the function of these channels is minimal
9.2 Expression of Membrane Proteins for Structural Studies
[36–39]. Additionally, synchrotron radiation circular dichroism spectroscopy and thermal denaturation studies on the E. electricus Na+ channel show no net change in protein secondary structure resulting from the deglycosylation procedure [40]. In summary, given the current limitations on protein processing and expression level, the development of improved mammalian expression systems is likely to be required before this becomes a realistic option for the production of membrane proteins for structural studies. Viral expression systems, such as Semliki Forest virus (SFV) or BacMam (see Chapter 4), may provide a solution in the future. 9.2.2 Insect Expression
Employing insect cells for the expression of eukaryotic channels has shown some potential and has several advantages compared to mammalian systems since the cell culture is simpler and easier to scale up. The Drosophila Shaker K+ channel was one of the first ion channels successfully expressed heterologously in insect cells for the purpose of biochemical and structural studies [41]. The human Kv1.1 channel has been functionally expressed using baculovirus in Sf9 cells and proven useful for biochemical studies [42]. Based on whole-cell currents, the cell membrane contains no more than a few thousand channels per cell, but it is possible that a much larger intracellular pool of protein may be recoverable for structural studies. It has also been applied for biochemical and biophysical studies of CFTR, the glycine receptor, the KATP channel, and the heteromeric amiloride Na+ channel complex [43–46]. However, in spite of these advances, the general utility of insect cells for producing reasonable quantities of intact mammalian ion channel protein for structural studies is still in question. Although, the first images of the tetrameric structure of a voltage-gated K+ channel came from protein expressed in Sf9 cells, there has been little progress for other mammalian ion channels [47]. In the case of CFTR, no structural information has been gained for this channel expressed in insect cells. However, recently, 2D crystals of this channel were reported using heterologous expression in mammalian cells (BHK cells) that provided structural information at a resolution of 20 Å [33]. In spite of these issues, it should be recognized that the crystal structure of many soluble proteins has been obtained using protein derived from heterologous expression in insect cells and thus may yet prove similarly valuable for membrane proteins. 9.2.3 Yeast Expression
Although the possibility of using yeast for the production of mammalian membrane proteins has been pursued for many years, it was not until very recently that this was successfully applied for material suitable for structural studies. In 2003, Parcej and Eckhardt-Strelau reported the expression, purification and struc-
217
218
9 Approaches for Ion Channel Structural Studies
ture of the rat Kv1.2 channel in complex with the Kvb2 subunit at 21 Å resolution using single-particle electron microscopy [10]. The Kv1.2-b2 protein was expressed in the methylotrophic yeast Pichia pastoris and found to be in a relatively homogeneous form following the removal of a consensus N-glycosylation site and several putative phosphorylation sites. This is certainly one of the first breakthrough methods for the production of milligram quantities of eukaryotic membrane protein that appears to be both properly folded and functionally active based on ligand-binding analysis. This method was recently adapted for the determination of the crystal structure of this channel at 2.9 Å resolution [2] – a landmark achievement. It is certainly hoped that this system can be broadly used to explore the structure of many other mammalian membrane proteins in addition to ion channels. 9.2.4 Bacterial Expression
The heterologous expression of proteins in E. coli cells is certainly one of the most widely used systems applied to soluble proteins. However, in the case of membrane proteins, it is only in the past decade that significant progress has been seen on the successful application of this system for X-ray and EM structural studies. It was not until 1998 that the first successful X-ray crystal structures of ion channel proteins were determined (KcsA and MscL), and these paved the way for many other ion channel structures that were determined using similar methods [15, 48]. As clearly summarized in Table 9.1, with a single exception [2], all of the known ion channel structures determined using X-ray crystallography were derived from protein expressed and purified using E. coli as the host. The primary advantage of using a prokaryotic expression system is the ability to easily scale-up the process and the tremendously lower costs of production. In general, relatively traditional methods have been successfully employed for the expression of bacterial membrane proteins in E. coli. Plasmids such as the pET (Novagen, Madison, WI) or pQE (Qiagen, Valencia, CA) vectors are the most commonly utilized for expression in suitable E. coli strains [BL-21 (DE3), for pET vectors; XL-1 or SG13009 (pREP4) for pQE vectors]. Although the process appears relatively straightforward, the over expression of ion channels, or other membrane proteins, is often toxic to E. coli, requiring optimization of media, temperature, cell density and induction conditions for each protein of interest. Additionally, we have observed that over expression of these proteins will often result in cellysis at later stages of induction. Thus, it is necessary to manage this process carefully and consistently by harvesting the cells before this occurs. For bacterial membrane proteins, the use of E. coli will continue to be an important means for bringing forward new structures. However, the expression of eukaryotic membrane proteins in E. coli is still a significant challenge. There are scattered reports of limited success for a few mammalian membrane proteins, but none of these has yet been successful at providing any structural information using either EM or X-ray methods. The recent report on the expression of several
9.3 The Detergent Factor
eukaryotic membrane proteins in E. coli, including a GPCR and an ion channel, using the autonomously folding integral membrane protein, Mistic, is very encouraging [13]. It is hoped that this and similar advances will pave the way for the structure determination of the many eukaryotic membrane proteins that still evade our grasp.
9.3 The Detergent Factor
Successful crystallization is only possible if the membrane protein, once removed from the bilayer, is properly folded in a detergent. The list of detergents used in successful crystallizations is both lengthy and diverse. A table of successfully used detergents is shown in Fig. 9.1. The a-helical type membrane proteins have seen the greatest success n-octyl-b-d-glucopyranoside (OG), N,N-dimethyldodecylamine-N-oxide (LDAO) and n-dodecyl-b-d-maltopyranoside (DDM). Octylglucoside stands out as the detergent that has been used in the most successful crystallizations. However, it is critical to note that the success rates partly reflect the length of time that each of these have been available. Thus, it is not clear if there something universal about OG and LDAO that makes them so useful, or simply if they have they seen the greatest success because they have been utilized in membrane protein biochemistry for much longer. Similarly, chain length trends for the detergents, where multiple chains lengths are available, have been frequently speculated about. Again, it is important to note that the many variations within a given detergent family, such as the glucosides, maltosides and so forth, have come about only recently, precluding clear conclusions. While it is possible to observe some general correlations, for example a-helical-type proteins are more apt to crystallize in glucopyranosides then they are in polyoxyethylenes, few other trends beyond such cursory conclusions can be defined at present. It is no surprise that removal of membrane proteins from their native lipid environment poses one of the most serious challenges to maintaining a native fold conducive for crystallization trials. Certainly one of the most significant advances for membrane protein crystallography is the availability of a myriad of high quality, diverse detergents. In this respect, the work of the scientists at Anatrace (Maumee, OH) is of particular note. The development of several new detergent series, CYMAL, FOS-CHOLINE, FOS-MEA, CYFOS and C-HEGA was aided through a NIH Small Business Innovative Research grant. Recognition by the funding agencies of the need for better reagents for novel research is to be congratulated and encouraged. The effects of detergent on membrane proteins cannot be overstated. Proper choice of detergent will greatly impact on whether crystallization is possible. The choice of detergent can be broken into two critical components. The first step is to identify a detergent that can successfully extract the protein from the bilayer. Membrane proteins often contain multiple subunits so the detergent must be able to extract the protein while maintaining both its tertiary and its quaternary
219
220
9 Approaches for Ion Channel Structural Studies
Fig. 9.1 Graph of detergents successfully used for membrane protein structure determination. Data from the compilation of known membrane protein structures at http:// www.mpibp-frankfurt.mpg.de/michel/public/ memprotstruct.html. Beta-type proteins refer to outer membrane proteins of prokaryotes
with transmembrane spanning beta strands. Alpha type proteins, found in both prokaryotic and eukaryotic membrane proteins, span the bilayer with alpha helices. The x-axis denotes the number of successful structure determinations in each of the listed detergents.
structure. As illustrated in Fig. 9.2A, one can design screens to identify optimal expression constructs using multiple homologs, expression vectors, varying types or location of affinity tags, using a few key detergents. Once an optimized expression system has been identified, one can then broadly screen for detergent solubility, relative purity and integrity using a 96-well format as illustrated in Fig. 9.2B. Further analysis of the structural integrity of the protein must then be carried out. While spectroscopic techniques such as circular dichroism are helpful for determining the state of tertiary structure, assessing quaternary structure can be more challenging. Size-exclusion chromatography, although low resolution, works quite
9.3 The Detergent Factor
Fig. 9.2a Expression and detergent screening. (A) Schematic diagram for expression construct optimization and detergent screening. Multiple constructs, homologs, and detergents can be simultaneously screened to rapidly identify the optimal conditions for expression and solubilization. Ideally, each open reading frame (ORF) is cloned into several expression vectors that place an affinity tag at either the N- or C-terminus. These are transfected into a series of bacterial strains
developed for protein expression. Following induction of protein expression (typically using a range of conditions), culture samples are individually aliquoted, lysed and solubilized with a panel of unique detergents. Cleared lysates are analyzed by Western blot using an antibody to the affinity tag to determine relative expression for each construct expressed in each bacterial strain, as well as their relative detergent solubility.
221
222
9 Approaches for Ion Channel Structural Studies
Fig. 9.2b Expression and detergent screening. (B) Schematic diagram of a high throughput detergent screening procedure. To determine the relative solubility and yield for a membrane protein using a broad series of detergents, a 96-well extraction and purification procedure can be performed using a robotic liquid-handling system or manually. Briefly, a panel of unique detergents is added to each well of a deep 96-well block containing the cell lysate. After shaking gently, affinity resin is added across the wells to bind the target protein. The resin is then transferred to a 96well filter plate where it is washed, and subse-
quently eluted into a collection plate (available from several vendors including Novagen, San Diego, CA; Qiagen,Valencia, CA; Vivascience, Inc., Edgewood, NY). Protein recovery can be evaluated by UV spectrometry or other protein assay, and purity can be visualized by high-throughput SDS-PAGE (Invitrogen, Carlsbad, CA). Protein identity can be confirmed by Western blot analysis and mass spectrometry, and functional and/or structural integrity can be evaluated by size exclusion chromatography (SEC) or ligand-binding.
well for this, in spite of the presence of detergent micelles. Macromolecules and complexes of over 200 kDa can be routinely resolved with size-exclusion chromatography and perhaps more importantly, aggregated protein in the sample can be identified. The presence of aggregated protein is a significant obstacle to successful crystallization. Every attempt should be made to eliminate it, or significantly reduce it, either by reengineering constructs, altering growth and production of the protein or by chromatography, prior to setting up crystallization trials. To assess whether the protein of interest is maintained as a higher order, oligomeric structure in any detergent one can crudely, but quickly, address this using ultra filters of an appropriate molecular weight cutoff and simply look for the oligomeric protein in the retentate versus the filtrate (i. e. monomeric protein will
9.4 Purification Fig. 9.3 Crosslinking to determine proper oligomeric state. Polyacrylamide gel showing crosslinking of two homologs of MscL from M. tuberculosis (denoted Tb) and E. coli (Eco) with disuccinimidyl suberate (DSS). Shown on the left side of the gel are the positions of the molecular weight markers.. The two left hand lanes are monomers of the homologs without addition of the bifunctional cross-linking reagents DSS. The two right lanes are with the addition of DSS, showing a clear ladder of monomer, dimer, trimer, tetramer and finally pentamer that is the full oligomeric structure of MscL, as seen in the crystal structure. Reprinted with permission from Ref [15].
pass through the filter). A more visual approach uses bifunctional cross linking reagents to covalently link subunits together and subsequently visualize them on SDS-PAGE gels, as shown in Fig. 9.3. This technique has been used to determine the oligomeric state of the ion channel MscL [15]. An enhancement of this technique is cross linking in conjunction with capillary electrophoresis (CE) employing a sieving gel. The advantage of this latter technique is the ability to easily quantify peaks in an electropherogram and deduce the stoichiometry of a complex macromolecule. Although densitometry of Coomassie-stained gels can also accomplish this, CE offers the advantage of high resolution, ease of quantification, and excellent separation, even between subunits of similar molecular weight. On a practical note, although it may be advantageous to make membrane preparations for large-scale purification of membrane proteins, this technique may be unnecessary and misleading if used in conjunction with detergent screens. Care must be taken whenever sonication is used for detergent screening to avoid false positive results. Heavy sonication can produce very small (~5 mM diameter) vesicles that can remain suspended while attempting to spin down a detergent screen trial. If membrane preparation by sonication is employed, it is necessary to apply ultracentrifugation to pellet any small membrane vesicles, prior to gel and analysis.
9.4 Purification
The first membrane protein structures were all derived from naturally abundant proteins. For example Deisenhofer’s original membrane protein structure, the photosynthetic reaction center from Rhodopseudomonas virdis, is found in purple patches in the membrane in a highly concentrated “pre-purified” form [1]. Additionally, the first GPCR, bovine rhodposin, occurs naturally at high concentration in the outer rod segments and is relatively pure in that membrane. Although determining the structure of both these proteins certainly presented numerous challenges, considerable additional challenges are presented by proteins found in eu-
223
224
9 Approaches for Ion Channel Structural Studies
Table 9.1 Ion channel structures determined by X-ray crystallography. Examples of crystal structures determined for ion channels using X-ray crystallography. Name
Ref.
Year
Ion Selectivity
PDB ID
Resolution (A˚)
Subunits a)
TM a-helices Species (per subunit)
KcsA MscL KcsA b) EcCIC MscS Mthk StClC EcClC KirBac1.1
48 15 69 70 17 71 70 72 73
1998 1998 2001 2002 2002 2002 2002 2003 2003
K+ Non-selective K+ Cl – Non-selective K+ Cl – Cl – K+
1BL8 1MSL 1K4C 1KPK 1MXM 1LNQ 1KPL 1OTS 1P7B
3.2 3.5 2.0 3.5 3.9 3.3 3.0 2.5 3.6
4 5 4 2 6 4 2 2 4
2 2 2 17 3 2 17 17 2
KvAP Kv1.2-b-2
58 2
2003 2005
K+ K+
1ORQ 2A79
3.2 2.9
4 4
6 6
S. lividans M. tuberculosis S. lividans E. coli E. coli M. thermoautotrophicum S. typhimurium E. coli B. pseudomallei A. pemix R. norvegicus
Table 9.2 Ion channel structures determined by electron microscopy. Examples of ion channel structures determined using either single particle electron microscopy or electron crystallography from two-dimensional protein crystals. Year
Name
Ref.
Ion Selectivity
Resolution (A˚)
Subunits a)
TM a-helices (per subunit)
1994 1995 1997 1998 1999 2001 2001
47 74 38 75 76 77 29
K+ Cation K+ K+ Non-selective Cl – Na+
n/a n/a n/a 6 7.5 6.5 19
4 5 4 4 12 2 1
6 4 6 2 4 17 24
2001 2003 2003 2003 2004 2004 2004 2004 2005
Shaker 5-HT3 receptor Kv1.3 KcsA Cx43 EcCIC Voltage-sensitive Na+ channel Shaker L-type Ca2+channel nAChR Kv1.2-b2 CFTR IP3 receptor Kv4.2 / KChlP2 KvAP AMPA receptor
78 21 28 10 33 79 80 81 82
K+ Ca2+ Cation K+ Cl – Ca2+ K+ K+ Na+
25 23 4.0 21 20 15 21 10.5 42
4 1 5 4 1 4 4 4 5
6 24 4 6 12 6 6 6 3
2005 2005
Ryanodine receptor TRPC3
83 20
Ca2+ Cation
10 30
4 4
a) Stoichiomety for pore-forming domain.
b) Complex with Fab fragment.
6–8 6
9.4 Purification
225
Table 9.1 (continued)
Host
Affinity Tag
Tag Removal
Detergent (Extraction)
Detergent (Crystallization)
E. coli E. coli E. coli E. coli E. coli E. coli E. coli E. coli E. coli
His6, C-term His10, N-term His6, C-term His6, C-term His6, N-term His6, C-term His6, C-term His6, C-term His6, C-term
Yes No Yes Yes No Yes Yes Yes No
LDAO n-Dodecyl-b-D-maltoside n-Decyl-b-D-maltoside n-Octyl-b-D-maltoside Fos-Choline-14 LDAO n-Octyl-b-D-maltoside n-Decyl-b-D-maltoside Cymal-4, HEGA-10
E. coli P. pastoris
His6, C-term His6, N-term
Yes No
n-Decyl-b-D-maltoside n-Dodecyl-b-D-maltoside n-Decyl-b-D-maltoside n-Decyl-b-D-maltoside Fos-Choline-14 n-Decyl-b-D-maltoside n-Decyl-b-D-maltoside n-Decyl-b-D-maltoside n-Decyl-b-D-maltoside + n-Tridecyl-b-D-maltoside n-Decyl-b-D-maltoside n-Dodecyl-b-D-maltoside
n-Octyl-b-D-glucoside n-Decyl-b-D-maltoside
Table 9.2 (continued) Species
Host
Affinity Tag
Tag Removal
Detergent
D. melanogaster M. musculus H. sapiens S.lividans R. norvegicus E. coli E. electricus
Sf9 (Baculovirus) NG 108-15 CV-1 (Vaccinia) E. coli BHK E. coli Native
– – His6, N-term His6, N-term – His10, C-term –
– – N N – Y –
CHAPS n-Dodecyl-b-D-maltoside CHAPS n-Dodecyl-b-D-maltoside Tween-20 n-Dodecyl-b-D-maltoside Lubrol- PX
D. melanogaster O. cuniculus T. mamorata R. norvegicus H. sapiens M. musculus H. sapiens A. pernix R. norvegicus
COS Native Native P. pastoris BHK Native COS7 E. coli Native
1D4, C-term – – His9, N-term His10, C-term – 1D4, C-term His6, C-term –
N – – N N – N Y –
O. cuniculus M. musculus
Native HEK293
– FLAG, C-term
– N
CHAPS Digitonin – Tween-80 n-Dodecyl-b-D-maltoside CHAPS CHAPS n-Decyl-b-D-maltoside CHAPS or n-Decyl-b-D-maltoside CHAPS n-Decyl-b-D-maltoside
226
9 Approaches for Ion Channel Structural Studies
karyotic plasma membrane where many proteins of interest to both the academic and commercial structural biologist reside, such as transporters, channels and receptors. These proteins are found typically in low abundance and in a membrane that is host to hundreds of other proteins. It would be difficult, if not impossible, to obtain structural information for these proteins without the use of recombinant technology in conjunction with affinity purification tags. Many affinity tag systems are commercially available, such as FLAG, maltose binding protein, streptavidin, thioredoxin, and many more. A near universal tool for this approach is the use of polyhistidine tags and immobilized metal chelate chromatography (IMAC). So pervasive is its application, that it has been a significant technology for the high throughput world of structural genomics as well as a core technique for many membrane protein crystallography laboratories. The use of these tags is now so routine that they need not be thoroughly discussed here. Indeed, as shown in Tables 9.1 and 9.2, nearly all of the recombinant expressed ion channel proteins for which structures have been determined were prepared using some type of polyhistidine tag. However, a few specific points regarding the use of polyhistidine tags for membrane proteins should be made. Although hexahistidine (His6) tags were originally used, His8, His10 and even longer tags are now more common although, in our hands, the utility of tags longer than His10 is not clear. A critical advantage of polyhistidine tags is their ability to efficiently bind metal columns in the presence of detergents. Although other affinity tags can also be used in the presence of detergent in virtually all cases, our experience supports the use of polyhistidine tags in conjunction with the >60 detergents that are routinely used in our screens. In short, the presence of detergents above their critical micellar concentration (CMC) does not preclude the use of this type of tag, though it should be noted that the efficiency with which the tag binds the column is often diminished. It is this reduction in affinity that has led to the use of vectors encoding the elongated tags listed above. In the light of lowered affinity in the presence of detergent, very high levels of imidazole should be carefully tested when washing and eluting the column, in order to obtain maximum purity while balancing the need for high yield. These opposing forces can easily be monitored, even at very small scale, by employing Western blots with anti-hexahistidine antibodies which readily bind the longer polyhistidine tags, and which are commercially available and inexpensive. Regardless of the affinity tag used, the ability to manipulate the detergent in this capture step is often of great importance. The detergent used for extraction from the membrane may not be one in which the protein remains stable for long periods of time, or it may not prove successful in crystallization. At the affinity purification step, which is usually the first step after extraction, detergent concentration is often high (e. g. 0.1% or more), depending on the CMC of the detergent. This step is therefore an opportunity to lower the detergent concentration to a level at which, though still above the CMC, the protein remains stable and which allows for at least the possibility of crystal formation [19–23]. The organization of protein molecules into a three-dimensional crystal lattice requires the formation of intermolecular crystal contacts which stabilize its archi-
9.5 Crystallization
tecture. However, due to associated detergent, there is significantly less available surface area on purified membrane proteins for making these necessary contacts. The crystallization and X-ray structure of soluble proteins fused to large affinity tags, such as the maltose-binding protein (MBP), thioredoxin (TRX), or glutathione-S-transferase (GST), has recently been reported, and it has been suggested that these tags may aid membrane protein crystallization by increasing the available hydrophilic surface area [49]. This technique would be analogous to the approach taken by Kaback and coworkers using cytochrome b562 fused to lac permease [50], or Iwata and coworkers attaching protein Z to cytochrome bo3 to facilitate crystallization [51]. This approach has not yet been proven successful for obtaining high-resolution structures of any membrane protein, and moreover, in the two examples mentioned, their structures were finally solved as His-tag fusions [52, 53]. However, since MPB is targeted to the periplasm, some have speculated that an N-terminal fusion with MBP could potentially “drag” the first transmembrane spanning region through the bilayer, providing a critical foothold to proper folding and further insertions into the membrane. In addition another possible benefit is the “chaperone-like effect” provided by MBP, enhancing the likelihood of a proper fold. The successful expression of the human Na+/glucose co-transporter illustrates both the use of an alternative affinity tag, as well as the expression of functional human membrane protein in E. coli [9]. The authors used a FLAG-tagged protein expressed in E. coli to generate 0.3 mg of pure protein per liter of culture. The use of a lac promoter system, in simple LB medium, at reduced temperature (20 8C), and a bacterial strain defective in the outer membrane protein OmpT, illustrates the successful expression and purification of a human membrane protein in a bacterial system. Perhaps more importantly, the protein produced was functional when reconstituted into liposomes. This level of expression of a functional protein illustrates the utility of bacterial expression when one considers that a scale up to a fermentor would easily achieve the protein production required for crystallization attempts. Moreover, a functional assay, such as sugar transport in this example, adds to the likelihood that this method can produce material that has the ability to be crystallized.
9.5 Crystallization
In general, once successfully purified in a detergent-containing buffer system in which it is stable, the crystallization of a membrane protein, though challenging, is not that much different from that of soluble proteins. Crystals for membrane proteins are obtained either in batch mode or from vapor diffusion, with both hanging and sitting drop formats being successfully used. Sitting drops have the advantage of easier use with 96-well plates, enabling faster screening of more conditions, and smaller drop sizes, thus preserving valuable supplies of painstakingly purified protein as far as possible. An online list of crystal screening kits and the
227
228
9 Approaches for Ion Channel Structural Studies
composition of each tube or well is maintained at the Structural Biology Laboratories at the University of Uppsala (http://xray.bmc.uu.se/markh/php/xtalscreens.php). Membrane protein specific screens have been developed, most notably the MembFac screen originally developed by Michael Stowell and commercialized by Hampton Research, which was the first attempt at compiling the conditions used to successfully crystallize membrane proteins into a single screen [54]. As the number of successful structures continues to increase so does the breadth of conditions used for successful crystal formation. Making use of the commercially available screens is an excellent place to begin. When attempting to determine suitable conditions, if availability of material allows, it is worth trying them all, since they are relatively inexpensive and cover a wide range of precipitants, salts, buffers and pH. Commercial screens in 96-well format from Hampton Research, Nextal Biotechnologies, Emerald Biostructures and others are available, facilitating simple application of this higher throughput format. Microfluidics has the potential to vastly extend the amount of crystallization screening possible but, as of now, these systems are still in their infancy. The purification and crystallization of the light-harvesting complex of photosystem II (LHC-II) illustrates several techniques useful for the extraction and stabilization of membrane proteins [55]. In this case detergent (n-nonyl-b-d-glucoside), lipid [digalactosyldiacylglycerol (DGDG)], and chlorophyll were used to solubilize the complex. The use of a combination of detergent, a critical plant lipid present in the thylakoid membrane where the complex is found, as well as chlorophyll, is readily validated from the structure. Adjacent trimers of the LHC-II complex are largely mediated by two pairs of DGDG and two pairs of chlorophyll molecules. The crystallization solution contained a second detergent, N, N-bis-(3-d-gluconamidopropyl) deoxycholamide (BigCHAP). This illustrates a highly complicated extraction and stabilization of a membrane protein but perhaps most striking is the resulting crystalline lattice. The packing of the complexes resembles that of the highly symmetric (in this case icosahedral particles) packing of viruses. In a ‘Type-II’ membrane protein crystal the detergent micelle forms a torus around the membrane-spanning region and crystal contacts are mediated by polar regions of the molecule, as described above. In the case of LHC-II, the complex exists in a proteoliposome and contacts between adjacent molecules are made through hydrophobic contacts at the lipid. The resultant ‘Type III’ membrane protein crystal is clearly formed by the judicious addition of lipid, cofactor and detergents. This may prove especially important in maintaining the correct fold of eukaryotic membrane proteins expressed in organisms that do not have bilayers with a similar lipid composition. The recent Kv1.2-b2 structure also highlights the potential importance of exogenous lipid, including phosphatidylcholine, phosphatidylethanolamine and phosphatidylglycerol, which was reported to be essential during the purification and crystallization process [2]. Thus, the use of lipids in membrane protein crystallography has proved useful and should be explored further.
9.6 Use of Antibody Fragment
9.6 Use of Antibody Fragments
Crystal formation by membrane proteins, even assuming a stable homogenous protein is purified, can be hampered if limited polar surface area is present. The hydrophobic surface of membrane spanning regions is coated by the detergent micelle, allowing solubilization of the protein. The polar head groups of the detergent are of course identical and lack a regular structure that can promote crystal formation and growth. Polar protein surfaces are responsible for mediating crystal contacts which can be minimal for many integral membrane proteins. One way of enlarging these regions is to use antibody fragments, either Fab domains, or Fv fragments specific for the membrane protein. These antibody fragments bind specifically, and therefore regularly, to the membrane protein and provide additional polar surface area. The fragments are also rigid in structure, allowing a higher probability of producing a regular three-dimensional lattice. The application of antibody fragments, both Fab and Fv, has been successful with membrane proteins [56–58]. One route to produce these is to simply use a well established hybridoma protocol to generate suitable monoclonal antibodies and then create Fab fragments using proteolytic cleavage [58]. This technique works well, as long as the proteolytic enzymes cut uniformly and produce a homogenous Fab sample. Alternatively, variable region fragments (Fv), can be generated, either as independent light and heavy chain fragments or as a single chain with a linker sequence. Both forms of variable region fragments are readily expressed in E. coli. This latter approach can be initiated from the more well established hybridoma/Fab approach described above, by cloning of the variable region of the antibodies using RT-PCR amplification of the mRNA from the hybridoma cell line. Several well characterized examples are in the literature [59]. Regardless of the route used to create antibody fragments, one feature that likely contributes to crystal formation is the ability of the fragments to lock a single conformation of the target protein. Of course any perturbation of one protein by another, although sometimes required for a successful structure determination, leads to speculation as to the usefulness of the resulting structure. Transmembrane-spanning regions are particularly at risk of structural perturbation from antibody fragments since these regions are often more mobile, partly due to the lack of constraints that normally exist in the membrane. However, some membrane proteins, notably channels and transporters, are inherently flexible in these regions due to their function. The ability to “lock” the structure in a particular conformation using an antibody fragment is an extra benefit, in addition to increasing polar surface area, which can promote a successful crystallization. However, it should be noted that, for one of the better known structures determined in this manner (KvAP), significant skepticism remained about the relevance of the resulting model [60–62]. Another technique to generate antibodies is to use filamentous phage. Phage display offers several advantages in the creation of antibody fragments, most notably speed and recombinant expression of the resulting fragments. Fab or scFv
229
230
9 Approaches for Ion Channel Structural Studies
fragments can be generated in as little as one month. Recently Röthlisberger and coworkers created a phage display library that produces Fab fragments that are stable in detergent [63]. Screening of the library was carried out in the presence of 0.1% dodecylmaltoside, with subsequent binding analysis using Biacore performed in buffer containing the same concentration of detergent. The authors also provide guidance as to which affinity tags are most useful in the screening process. The resulting antibody fragments, Fab in this case, were shown to bind to their target in the low nanomolar range in a conformationally dependent manner.
9.7 Generation of First Diffraction Datasets
One striking thing that has been observed for a variety of membrane protein structures is the variability in the quality of diffraction from crystal to crystal, as well as the sensitivity of the crystals to cryoprotection. This latter complication may be due to the high solvent content of membrane protein crystals which can typically reach 80 % [15]. First diffraction images can produce less than inspiring data, when compared with a typical soluble protein. However, any diffraction is a significant step towards a structure and even the weakest of spots, often clustered just around the beamstop, are cause for celebration. Once any diffraction is obtained, a critical foothold in the process is reached, since this represents a milestone against which further modifications can be assessed. Now it is possible to assess any changes and ask, is this better or not? A long stream of blank images will likely precede this milestone, from which it is difficult to assess the impact of any changes made. As shown in Fig. 9.4, it can be seen that the first diffraction image may only produce a few spots around the beamstop, and cannot be successfully indexed with any confidence. However, the process of crystal screening and cryoprotection optimization is pursued to improve their diffraction qualities (Fig. 9.4B). Finally, in the case of MscL, heavy metal soaks of the crystals produced higher resolution reflections that produced the final structure (Fig. 9.4C and D). A similar approach has been successfully applied for the structures of the bacterial transporters MsbA and EmrE, bacterial homologs to the ABC transporter family [18, 19]. Advances in automation are having a major impact on several aspects of membrane protein crystallography, particularly for screening of crystals. 96 and 384well format crystallization trays now allow researchers to rapidly set up thousands of crystallization trials using a variety of robotic liquid-handling platforms such as the Mosquito (TTP Labtech, UK) or Hydra II (Matrix Tech. Corp., NH), but these need to be complemented with crystal imaging technology. The limit in the number of trials is often determined not by the chemical matrices that can be thought up for crystallization, but rather by restrictions on the amount of purified protein that can be produced. Since it is likely that many tens or hundreds of crystals will need to be produced and screened, obtaining sufficient protein preparations from the outset is critical.
9.7 Generation of First Diffraction Datasets
Fig. 9.4 Examples of X-ray diffraction images. (A) Initial diffraction pattern from a single crystal of the MscL ion channel protein from M. tuberculosis (Tb-MscL) prior to the optimization of crystallization and cryoprotection conditions. (B) Following optimization of the crystallization and cryoprotection conditions for a crystal of Tb-MscL that diffracted to a limiting resolution of 7 Å. (C) Diffraction pattern of a Tb-MscL crystal after soaking with the heavy atom compound, Na3Au(S2O3)2.
Note the significant improvement in the diffraction limit, as compared to (B), extending to 3.5 Å. This image also illustrates the significant anisotropy and intensity decay of the reflections often observed with membrane proteins. (D) Ribbon diagram of the resulting 3.5 Å structure of the M. tuberculosis MscL, a mechanosensitive channel, reproduced with permission from Ref. [15]. (This figure also appears with the color plates.) (This figure also appears with the color plates.)
231
232
9 Approaches for Ion Channel Structural Studies
Robots are also finding good use in the screening of crystals at the synchrotron. Anyone who has screened at the synchrotron will tell you that most of the time spent screening is taken up by mounting crystals and closing up the experimental hutch. With current state-of-the-art beam lines, the time taken to obtain the test diffraction image can be as short as 10 s. In contrast, getting the hutch open, mounting and centering the crystal and closing up the hutch takes many minutes. Robots such as the ACTOR system (Rigaku/MSC, TX) mounted at the beam line are equipped with cryo-pucks that hold dozens of crystals and are now in use at several synchrotrons. Due to the efficiency of not having to manipulate the crystals manually, one can now screen crystals at the beam line remotely. A Dewar flask loaded with pucks and dozens of crystals is shipped to the synchrotron facility and a technician can load up the robot. Automatic mounting and centering of the crystals at a preset exposure time will facilitate the screening of hundreds of crystals within a few hours. Once screened, each crystal is safely recovered and returned to the Dewar by the robot for future data collection. This is without doubt the way of the future since it massively increases the “open shutter” time at the beam lines, enhancing the efficiency of a seriously taxed resource.
9.8 Selenomethionine Phasing of Membrane Proteins
Congratulations! You’ve just successfully crystallized a membrane protein and got a dataset. Now all you have to do is obtain your phases and start model building. Membrane proteins involved in respiration and light harvesting have metal-containing cofactors that can be useful for phase determination. However, many membrane proteins, channels, receptors and transporters, typically do not. Although traditional multiple isomorphous replacement methods for obtaining phase information on membrane proteins and channels have been used, an increasingly useful technique widely used in soluble proteins, and just as valuable for membrane proteins, is selenomethionine multiple anomalous diffraction phasing (SeMet MAD). In spite of the often modest resolution of membrane proteins, this technique has seen spectacular successes and should not be discounted as a useful route for phase determination. A possible complication to using this technique is the large size of many membrane proteins. The larger the protein the higher the likelihood that a large number of methionine residues, and thus selenomethionines, will be present. Successful phasing using selenomethionine and MAD phasing have been reported for membrane proteins with up to 42 methionines per asymmetric unit, all at a resolution of 4 Å, underscoring the utility of this approach [17]. Incorporation of selenomethionine has been shown in E. coli (reviewed in Ref. [64]), in yeast [65], baculovirus [66], and mammalian tissue culture (BioXtal, Switzerland). In the Rees Lab at Caltech, bacterial expression of membrane proteins with incorporation of selenomethionine started with an M9 glucose minimal media with subsequent media augmentation. A modified M9 medium, containing
9.9 MAD Phasing and Edge Scanning
50 mg L–1 of selenomethionine and the other 19 amino acids at 40 mg ml–1 (or 80 mg, for d,l-amino acids) was sufficient for nearly complete incorporation of selenomethionine. As with any change in growth medium, optimization of induction reagents, induction time and growth temperature should be undertaken. The addition of naturally derived media components, such as tryptone and yeast extract, should be avoided since these reagents and others contain methionine, and will likely reduce the level of selenomethionine incorporation. As one would expect, the yield of recombinant protein in minimal media is considerably lower than that for rich media. Sufficient cell mass for purification and crystallization can require scaling up, in our case up to a 60 L fermentor. Similarly, for yeast expression, both extensive strain selection as well as media screening produced high incorporation rates [65]. In some cases, cells can be grown in methionine-containing media with subsequent transfer to selenomethionine-containing media, as was done for the first selenomethionine crystal structure that utilized insect cells and baculovirus infection. In this latter case, cells were grown and infected in methionine-rich media, then grown in methionine-depleted media and finally grown in selenomethionine-containing media. This approach is also readily applied to bacterial expression if cell growth in selenomethionine minimal media is insufficient. Current high throughput efforts in structural genomics have applied selenomethionine labeling right from the beginning. In fact, as with soluble proteins, some have suggested that it is advantageous to simply move immediately to expression of membrane proteins in selenomethionine incorporating systems. While this surely would speed the process to structure solution, it seems highly unlikely that the limitations imposed in media used for selenomethionine bioincorporation would lead to the successful membrane protein expression. A far more fruitful approach would seem to rely on a combination of multiple targets and expression systems, as mentioned throughout this chapter and a return to selenomethionine once diffraction quality crystals have been obtained.
9.9 MAD Phasing and Edge Scanning
A fluorescent scan of the crystal should be obtained before data are collected. Multiple wavelength anomalous diffraction (MAD) beam lines are equipped with -ray fluorescence detectors and obtaining the values of f ' and f '' for the anomalous scattering atom(s) aids in finding the precise wavelength you should use for collecting the peak and inflection datasets. Although it is possible to look up these values, the dataset will be better for choosing the wavelengths from the scan. The Blu-Ice program (developed at SSRL) can perform an edge scan and calculate these values for the user [67]. A minimum of two wavelengths are required to do MAD phasing (hence “multiple” wavelength anomalous diffraction); however, in practice, the peak wavelength, inflection, and a high-energy remote wavelength are usually collected. The peak wavelength is, as it sounds, the peak of the f '' plot.
233
234
9 Approaches for Ion Channel Structural Studies
A second wavelength, at the inflection point, is the wavelength at which the f ' value inflects at a minimum (very near the half maximal point on the f '' plot. The final wavelength is a high energy remote, usually collected at a lower wavelength distant from the edge of X-ray absorption of the anomalous scattering atom. Finally, a critical factor is to obtain highly redundant data. Anomalous scattering only produces relatively small differences between pairs of reflections (Bijvoet pairs). By obtaining data redundancy of 4-fold (each reflection measured 4 times), 5-fold or even higher, each of these pairs is measured multiple times, so the small differences in the intensity of each Bijvoet pair are statistically significant. Even high symmetry space groups, where a small angular wedge of data can produce a complete dataset, benefit, in terms of phasing, by collecting as much data as possible. Of course, the maximum would be to collect 3608 of data at each of the wavelengths chosen. If radiation damage is not a problem, as determined by integrating and scaling the data and obtaining the Rmerge of each dataset, then this approach should be used. Since the anomalous signal from selenium is small compared to heavy atoms used in multiple isomorphous replacement, such as gold, mercury and platinum, the data from a selenomethionine MAD experiment need special treatment. Searching for the anomalous scattering atoms, selenium in this case, which have relatively small differences in the intensities of Bijvoet pairs is enhanced by allowing the search programs (Solve, for example) to use local scaling. Local scaling of the data, scaling using a reciprocal space sphere of neighboring reflections, allows the best signal to noise ratio when obtaining these slight differences in intensities. In practice, what this means is stronger anomalous signals accurately measured to the highest resolution for that dataset, which will increase the chances of solving the crystal structure.
9.10 Negative B- factor Application (Structure Factor Sharpening)
Anisotropic decay in the intensity of high resolution reflections has been observed frequently for membrane proteins. Since many membrane protein crystals diffract to only modest resolution (3–4 Å) obtaining the most from these weak reflections in the highest resolution shell is crucial. A successful approach that has been used with great success is structure factor sharpening (the application of a negative B-factor to the observed structure factor, Fobs). The CCP4 program can be run to apply an overall B-factor and scale to the data that will increase the Fobs of the high-resolution reflection in a disproportional manner relative to the low-resolution reflections [68]. However, a disadvantage is that the application of negative B-factors increases the noise in electron density maps. Nevertheless, this technique is most valuable if noncrystallographic symmetry is present, since this excess noise is averaged out leaving maps that can be of excellent quality, thus producing a better model.
References
9.11 Conclusions
In spite of the somewhat daunting task of crystallizing a membrane protein such as an ion channel, there are emerging tools and techniques that will, without doubt, make structures of these intriguing proteins more common. Ion channels and membrane proteins in general have such tremendous therapeutic potential to be unlocked by the availability of high-resolution structures; one can expect the sheer number and speed of their solutions to only increase in the coming years.
References 1 J. Deisenhofer, O. Epp, K. Miki, R. Huber, H. Michel, X-ray structure analysis of a membrane protein complex. Electron density map at 3 A resolution and a model of the chromophores of the photosynthetic reaction center from Rhodopseudomonas viridis, J. Mol. Biol. 1984, 180, 385–398. 2 S. B. Long, E. B. Campbell, R. Mackinnon, Crystal Structure of a Mammalian Voltage-Dependent Shaker Family K+ Channel, Science 2005, 309, 897–903. 3 J. C. Kendrew, G. Bodo, H. M. Dintzis, R. G. Parrish, H. Wyckoff, D. C. Phillips. A three-dimensional model of the myoglobin molecule obtained by x-ray analysis, Nature 1958, 181, 662–666. 4 S. H. White, The progress of membrane protein structure determination, Protein Sci. 2004, 13, 1948–1949. 5 M. Wortman, The Re-Emergence of Ion Channel Drug Discovery, in StartUp: Windhover’s Review of Emerging Medical Ventures, 2003, p. 9. 6 J. C. Venter, et al., The sequence of the human genome, Science 2001, 291, 1304–1351. 7 R. Grisshammer, J. Tucker, Quantitative evaluation of neurotensin receptor purification by immobilized metal affinity chromatography, Protein Expr. Purif. 1997, 11, 53–60. 8 G. Fiermonte,V. Dolce, F. Palmieri, Expression in Escherichia coli, functional characterization, and tissue distribution of isoforms A and B of the phosphate carrier from bovine mitochondria, J. Biol. Chem. 1998, 273, 22782– 22787.
9 M. Quick, E. M. Wright, Employing Escherichia coli to functionally express, purify, and characterize a human transporter, Proc. Natl. Acad. Sci.U S A 2002, 99, 8597–8601. 10 D. N. Parcej, L. Eckhardt-Strelau, Structural characterisation of neuronal voltage-sensitive K+ channels heterologously expressed in Pichia pastoris, J. Mol. Biol. 2003, 333, 103–116. 11 M. A. Lesney, HTS: Seeking Signals Through the Noise, Modern Drug Discov. 2002, 5. 12 C. Tanford (ed.), The Hydrophobic Effect: Formation of Micelles and Biological Membranes, John Wiley & Sons, New York, 1980. 13 T. P. Roosild, J. Greenwald, M. Vega, S. Castronovo, R. Riek, S. Choe, NMR structure of Mistic, a membraneintegrating protein for membrane protein expression, Science 2005, 307, 1317–1321. 14 J. F.White, L. B. Trinh, J. Shiloach, R. Grisshammer, Automated largescale purification of a G protein-coupled receptor for neurotensin, FEBS Lett. 2004, 564, 289–293. 15 G. Chang, R. H. Spencer, A. T. Lee, M. T. Barclay, D. C. Rees, Structure of the MscL homolog from Mycobacterium tuberculosis: a gated mechanosensitive ion channel, Science 1998, 282, 2220–2226. 16 K. P. Locher, A. T. Lee, D. C. Rees, The E. coli BtuCD structure: a framework for ABC transporter architecture and mechanism, Science 2002, 296, 1091– 1098.
235
236
9 Approaches for Ion Channel Structural Studies 17 R. B. Bass, P. Strop, M. Barclay, D. C. Rees, Crystal structure of Escherichia coli MscS, a voltage-modulated and mechanosensitive channel, Science 2002, 298, 1582–1587. 18 G. Chang, C. B. Roth, Structure of MsbA from E. coli: a homolog of the multidrug resistance ATP binding cassette (ABC) transporters, Science 2001, 293, 1793–1800. 19 C. Ma, G. Chang, Crystallography of the integral membrane protein EmrE from Escherichia coli, Acta Crystallogr. D 2004, 60, 2399–2402. 20 K. Mio, T. Ogura,Y. Hara,Y. Mori, C. Sato, The non-selective cationpermeable channel TRPC3 is a tetrahedron with a cap on the large cytoplasmic end, Biochem Biophys Res Commun 2005, 333, 768–777. 21 M. Wolf, A. Eberhart, H. Glossmann, J. Striessnig, N. Grigorieff, Visualization of the domain structure of an L-type Ca2+ channel using electron cryo-microscopy, J. Mol. Biol. 2003, 332, 171–182. 22 P. C. da Fonseca, S. A. Morris, E. P. Nerou, C.W. Taylor, E. P. Morris, Domain organization of the type 1 inositol 1,4,5-trisphosphate receptor as revealed by single-particle analysis, Proc. Natl. Acad. Sci.U S A 2003, 100, 3936–3941. 23 Q. X. Jiang, E. C. Thrower, D. W. Chester, B. E. Ehrlich, F. J. Sigworth, Three-dimensional structure of the type 1 inositol 1,4,5-trisphosphate receptor at 24 A resolution, EMBO J. 2002, 21, 3575–3581. 24 Serysheva II, D. J. Bare, S. J. Ludtke, C. S. Kettlun, W. Chiu, G. A. Mignery, Structure of the type 1 inositol 1,4,5-trisphosphate receptor revealed by electron cryomicroscopy, J. Biol. Chem. 2003, 278, 21319–21322. 25 Serysheva II, S. L. Hamilton, W. Chiu, S. J. Ludtke, Structure of Ca2+ release channel at 14 A resolution, J. Mol. Biol. 2005, 345, 427–431. 26 E. V. Orlova, Serysheva II, M. van Heel, S. L. Hamilton,W. Chiu, Two structural configurations of the skeletal muscle calcium release channel. Nat. Struct. Biol. 1996, 3, 547–552.
27 Serysheva II, M. Schatz, M. van Heel,W. Chiu, S. L. Hamilton, Structure of the skeletal muscle calcium release channel activated with Ca2+ and AMP-PCP. Biophys. J. 1999, 77, 1936– 1944. 28 A. Miyazawa,Y. Fujiyoshi, N. Unwin, Structure and gating mechanism of the acetylcholine receptor pore, Nature 2003, 423, 949–955. 29 C. Sato,Y. Ueno, K. Asai, K. Takahashi, M. Sato, A. Engel,Y. Fujiyoshi, The voltage-sensitive sodium channel is a bell-shaped molecule with several cavities. Nature 2001, 409, 1047– 1051. 30 Serysheva II, S. J. Ludtke, M. R. Baker,W. Chiu, S. L. Hamilton, Structure of the voltage-gated L-type Ca2+ channel by electron cryomicroscopy, Proc. Natl. Acad. Sci.U S A 2002, 99, 10370– 10375. 31 M. C. Wang, G. Velarde, R. C. Ford, N. S. Berrow, A. C. Dolphin, A. Kitmitto, 3D structure of the skeletal muscle dihydropyridine receptor, J. Mol. Biol. 2002, 323, 85–98. 32 C. C. Chadwick, A. Saito, S. Fleischer, Isolation and characterization of the inositol trisphosphate receptor from smooth muscle, Proc. Natl. Acad. Sci.U S A 1990, 87, 2132–2136. 33 M. F. Rosenberg, A. B. Kamis, L. A. Aleksandrov, R. C. Ford, J. R. Riordan, Purification and crystallization of the cystic fibrosis transmembrane conductance regulator (CFTR), J. Biol. Chem. 2004, 279, 39051–39057. 34 A. K. Mitra, A. N. van Hoek, M. C. Wiener, A. S. Verkman, M. Yeager, The CHIP28 water channel visualized in ice by electron crystallography, Nat. Struct. Biol. 1995, 2, 726–729. 35 G. Ren,V. S. Reddy, A. Cheng, P. Melnyk, A. K. Mitra,Visualization of a water-selective pore by electron crystallography in vitreous ice, Proc. Natl. Acad. Sci.U S A 2001, 98, 1398–1403. 36 L. Santacruz-Toloza,Y. Huang, S. A. John, D. M. Papazian, Glycosylation of shaker potassium channel protein in insect cell culture and in Xenopus oocytes, Biochemistry 1994, 33, 5607–5613.
References 37 A. N. van Hoek, M. C. Wiener, J. M. Verbavatz, D. Brown, P. H. Lipniunas, R. R. Townsend, A. S. Verkman, Purification and structure-function analysis of native, PNGase F-treated, and endo-beta-galactosidase-treated CHIP28 water channels, Biochemistry 1995, 34, 2212–2219. 38 R. H. Spencer, et al., Purification, visualization, and biophysical characterization of Kv1.3 tetramers, J. Biol. Chem. 1997, 272, 2389–2395. 39 E. Bennett, M. S. Urcan, S. S. Tinkle, A. G. Koszowski, S. R. Levinson, Contribution of sialic acid to the voltage dependence of sodium channel gating. A possible electrostatic mechanism, J. Gen. Physiol. 1997, 109, 327–343. 40 N. B. Cronin, A. O’Reilly, H. Duclohier, B. A. Wallace, Effects of deglycosylation of sodium channels on their structure and function, Biochemistry 2005, 44, 441–449. 41 K. Klaiber, N. Williams, T. M. Roberts, D. M. Papazian, L. Y. Jan, C. Miller, Functional expression of Shaker K+ channels in a baculovirus-infected insect cell line, Neuron 1990, 5, 221–226. 42 R. A. Gubitosi-Klug, D. J. Mancuso, R. W. Gross, The human Kv1.1 channel is palmitoylated, modulating voltage sensing: Identification of a palmitoylation consensus sequence, Proc. Natl. Acad. Sci.U S A 2005, 102, 5964–5968. 43 C. E. Bear, C. H. Li, N. Kartner, R. J. Bridges, T. J. Jensen, M. Ramjeesingh, J. R. Riordan, Purification and functional reconstitution of the cystic fibrosis transmembrane conductance regulator (CFTR), Cell 1992, 68, 809– 818. 44 M. Cascio, N. E. Schoppa, R. L. Grodzicki, F. J. Sigworth, R.O. Fox, Functional expression and purification of a homomeric human alpha 1 glycine receptor in baculovirus-infected insect cells, J. Biol. Chem. 1993, 268, 22135– 22142. 45 M. V. Mikhailov, P. Proks, F. M. Ashcroft, S. J. Ashcroft, Expression of functionally active ATP-sensitive K-
46
47
48
49
50
51
52
53
54 55
56
channels in insect cells using baculovirus, FEBS Lett. 1998, 429, 390–394. U. S. Rao, A. Mehdi, R. E. Steimle, Expression of amiloride-sensitive sodium channel: a strategy for the coexpression of multimeric membrane protein in Sf9 insect cells, Anal. Biochem. 2000, 286, 206–213. M. Li, N. Unwin, K. A. Stauffer,Y. N. Jan, L. Y. Jan, Images of purified Shaker potassium channels, Curr. Biol. 1994, 4, 110–115. D. A. Doyle, et al., The structure of the potassium channel: molecular basis of K+ conduction and selectivity, Science 1998, 280, 69–77. D. R. Smyth, M. K. Mrozkiewicz, W. J. McGrath, P. Listwan, B. Kobe, Crystal structures of fusion proteins with largeaffinity tags, Protein Sci. 2003, 12, 1313– 1322. J. Zhuang, G. G. Prive, G. E. Werner, P. Ringler, H. R. Kaback, A. Engel, Two-dimensional crystallization of Escherichia coli lactose permease, J. Struct. Biol. 1999, 125, 63–75. B. Byrne, J. Abramson, M. Jansson, E. Holmgren, S. Iwata, Fusion protein approach to improve the crystal quality of cytochrome bo(3) ubiquinol oxidase from Escherichia coli, Biochim. Biophys. Acta 2000, 1459, 449–455. J. Abramson, I. Smirnova,V. Kasho, G. Verner, H. R. Kaback, S. Iwata, Structure and mechanism of the lactose permease of Escherichia coli, Science 2003, 301, 610–615. J. Abramson, et al., The structure of the ubiquinol oxidase from Escherichia coli and its ubiquinone binding site, Nat. Struct. Biol. 2000, 7, 910–917. M. H. Stowell, in UCLA Crystallization Workshop 1993. Z. Liu, et al., Crystal structure of spinach major light-harvesting complex at 2.72 A resolution, Nature 2004, 428, 287–292. S. Iwata, C. Ostermeier, B. Ludwig, H. Michel, Structure at 2.8 A resolution of cytochrome c oxidase from Paracoccus denitrificans, Nature 1995, 376, 660–669.
237
238
9 Approaches for Ion Channel Structural Studies 57 C. Hunte, J. Koepke, C. Lange, T. Rossmanith, H. Michel, Structure at 2.3 A resolution of the cytochrome bc(1) complex from the yeast Saccharomyces cerevisiae co-crystallized with an antibody Fv fragment, Struct. Fold. Des. 2000, 8, 669–684. 58 Y. Jiang, A. Lee, J. Chen,V. Ruta, M. Cadene, B. T. Chait, R. MacKinnon, X-ray structure of a voltage-dependent K+ channel, Nature 2003, 423, 33– 41. 59 G. Kleymann, C. Ostermeier, B. Ludwig, A. Skerra, H. Michel, Engineered Fv fragments as a tool for the one-step purification of integral multisubunit membrane protein complexes, Biotechnology (N Y) 1995, 13, 155–160. 60 L. G. Cuello, D. M. Cortes, E. Perozo, Molecular architecture of the KvAP voltage-dependent K+ channel in a lipid bilayer, Science 2004, 306, 491–495. 61 F. Bezanilla, The voltage-sensor structure in a voltage-gated channel, Trends Biochem. Sci. 2005, 30, 166–168. 62 M. Laine, D. M. Papazian, B. Roux, Critical assessment of a proposed model of Shaker, FEBS Lett. 2004, 564, 257– 263. 63 D. Rothlisberger, K. M. Pos, A. Pluckthun, An antibody library for stabilizing and crystallizing membrane proteins – selecting binders to the citrate carrier CitS, FEBS Lett. 2004, 564, 340–348. 64 W. A. Hendrickson, J. R. Horton, D. M. LeMaster, Selenomethionyl proteins produced for analysis by multiwavelength anomalous diffraction (MAD): a vehicle for direct determination of three-dimensional structure, EMBO J. 1990, 9, 1665–1672. 65 D. A. Bushnell, P. Cramer, R. D. Kornberg, Selenomethionine incorporation in Saccharomyces cerevisiae RNA polymerase II, Structure (Camb.) 2001, 9, R11–R14. 66 J. J. Bellizzi, J. Widom, C. W. Kemp, J. Clardy, Producing selenomethionine-labeled proteins with a baculovirus expression vector system, Struct. Fold. Des. 1999, 7, R263–R267.
67 T. M. McPhillips, et al., Blu-Ice and the Distributed Control System: software for data acquisition and instrument control at macromolecular crystallography beamlines, J. Synchrotron Radiat. 2002, 9, 401–406. 68 Collaborative Computational Project, N. The CCP4 suite: programs for protein crystallography, Acta Crystallogr. D 1994, 50, 760–763. 69 Y. Zhou, J. H. Morais-Cabral, A. Kaufman, R. MacKinnon, Chemistry of ion coordination and hydration revealed by a K+ channel-Fab complex at 2.0 A resolution, Nature 2001, 414, 43– 48. 70 R. Dutzler, E. B. Campbell, M. Cadene, B. T. Chait, R. MacKinnon, X-ray structure of a ClC chloride channel at 3.0 A reveals the molecular basis of anion selectivity, Nature 2002, 415, 287–294. 71 Y. Jiang, A. Lee, J. Chen, M. Cadene, B. T. Chait, R. MacKinnon, Crystal structure and mechanism of a calciumgated potassium channel, Nature 2002, 417, 515–522. 72 R. Dutzler, E. B. Campbell, R. MacKinnon, Gating the selectivity filter in ClC chloride channels, Science 2003, 300, 108–112. 73 A. Kuo, et al., Crystal structure of the potassium channel KirBac1.1 in the closed state, Science 2003, 300, 1922– 1926. 74 F. G. Boess, R. Beroukhim, I. L. Martin, Ultrastructure of the 5-hydroxytryptamine3 receptor, J Neurochem 1995, 64, 1401– 1405. 75 H. L. Li, et al., Two-dimensional crystallization and projection structure of KcsA potassium channel, J. Mol. Biol. 1998, 282, 211–216. 76 V. M. Unger, N. M. Kumar, N. B. Gilula, M. Yeager, Three-dimensional structure of a recombinant gap junction membrane channel, Science 1999, 283, 1176–1180. 77 J. A. Mindell, M. Maduke, C. Miller, N. Grigorieff, Projection structure of a ClC-type chloride channel at 6.5 A resolution, Nature 2001, 409, 219–223.
References 78 O. Sokolova, L. Kolmakova-Partensky, N. Grigorieff, Three-dimensional structure of a voltage-gated potassium channel at 2.5 nm resolution, Structure (Camb.) 2001, 9, 215–220. 79 C. Sato, et al. Inositol 1,4,5-trisphosphate receptor contains multiple cavities and L-shaped ligand-binding domains, J. Mol. Biol. 2004, 336, 155–164. 80 L. A. Kim, J. Furst, D. Gutierrez, M. H. Butler, S. Xu, S. A. Goldstein, N. Grigorieff, Three-dimensional structure of I(to); Kv4.2-KChIP2 ion channels by electron microscopy at 21 Angstrom resolution, Neuron 2004, 41, 513–519.
81 Q. X. Jiang, D. N. Wang, R. MacKinnon, Electron microscopic analysis of KvAP voltage-dependent K+ channels in an open conformation, Nature 2004, 430, 806–810. 82 T. Nakagawa,Y. Cheng, E. Ramm, M. Sheng, T. Walz, Structure and different conformational states of native AMPA receptor complexes, Nature 2005, 433, 545–549. 83 M. Samso, T. Wagenknecht, P. D. Allen. Internal structure and visualization of transmembrane domains of the RyR1 calcium release channel by cryoEM, Nat. Struct. Mol. Biol. 2005, 12, 539–544.
239
241
10 Molecular Modeling and Simulations of Ion Channels: Applications to Potassium Channels Daniele Bemporad, Alessandro Grottesi, Shozeb Haider, Zara A. Sands, and Mark S.P. Sansom
10.1 Introduction
The past decade has seen substantial advances in the structural biology of ion channels and of membrane proteins in general (see Refs. [1, 2] and Chapter 9). This has been primarily from X-ray diffraction methods, but electron microscopy (both from two-dimensional crystals [3] and from single particles [4]) is increasingly important, especially for defining the conformations of ion channels within a membrane environment. However, determining the structure of a channel protein is not an end in itself. Rather, one wishes to use structures to understand channel physiology and pharmacology. Understanding channel mechanisms at atomic resolution is both an intellectual challenge, and also an important step in the design of novel channel-perturbing ligands. The aim of this chapter is to describe how computational methods can be used to aid exploration of the relationship between ion channel structure and function. To achieve this we will focus on a particular class of ion channels, namely potassium channels [5]. K+ channels are of importance from a physiological and pharmacological perspective, for example in the central nervous system, and in the regulation of cardiac activity. They are also the best understood class of channels in terms of X-ray structures, and of experimental and computational studies of channel permeation and gating mechanisms. It is useful to review the structural biology of K+ channels. All of the structures that have been determined by X-ray diffraction are of bacterial homologues of mammalian K+ channels (Fig. 10.1). The structures include: (i) KcsA (PDB code 1K4C [6, 7]), a ‘minimalist’ K+ channel activated by low intracellular pH; (ii) KirBac1.1 and 3.1 (PDB codes 1P7B and 1XL4 [8, 9]), homologs of mammalian inward rectifier channels; (iii) KvAP (PDB code 1ORQ [10]), a voltage activated K+ channel; and (iv) MthK (PDB code 1LNQ [11]), a K+ channel activated by Ca2+ binding to an intracellular domain. Each of these structures shares a common architecture for the pore domain of K+ channels. K+ channels are composed of four subunits arranged symmetrically around a central pore. The pore-forming domain Expression and Analysis of Recombinant Ion Channels. Edited by Jeffrey J. Clare and Derek J. Trezise Copyright # 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim ISBN: 3-527-31209-9
242
10 Molecular Modeling and Simulations of Ion Channels: Applications to Potassium Channels Fig. 10.1 X-ray structures of K+ channels. In each case, for clarity, only two of the four subunits are present. Channels with the pore domain in a closed conformation (A) are represented by KcsA and KirBac3.1; channels with the pore domain in an open conformation (B) are represented by KvAP and MthK (for which only the transmembrane domain is shown). The dotted horizontal lines represent the approximate extent of the lipid bilayer.
is formed of a M1-P-F-M2 motif, where M1 and M2 are transmembrane (TM) a-helices, and the short P-helix and extended filter (F) region form a re-entrant loop between the two TM helices. (Note that in Kv channels the S5 and S6 helices are the equivalents of M1 and M2). The filter contains a sequence motif (TVGYG) that is characteristic of K+ channels and is the structural element responsible for the selective permeability to K+ ions. The four subunits of the pore domain thus form an eight TM helix bundle, with the M2 (or S6) helices lining the central pore in an inverted truncated conical geometry, whilst the P loop is inserted at the extracellular mouth of the M2 bundle where it forms the selectivity filter. The M2 (or S6) helices cross at the intracellular mouth of the pore, which corresponds to the activation gate of a K+ channel. In KcsA and KirBac the channel is crystallised in a closed state, and the M2 helices pack together to form a narrow hydrophobic region in the pore which corresponds to the (closed) gate. In MthK the protein was crystallised in the presence of Ca2+ and hence the channel was captured in an open state in which the M2 helices are kinked so as to open the intracellular gate. In KvAP the pore domain also appears to be in an open state with the S6 helices kinked.
10.2 Computational Methods
Given advances in the structural biology of K+ channels, what roles can computational methods play? The main uses of computational methods can be divided into two areas: (i) modeling; and (ii) simulation. Modeling is important because the available structures are all of bacterial K+ channels (as these are more readily
10.2 Computational Methods
expressed and crystallised), whereas from a physiological and pharmacological perspective we wish to understand structure/function relationships in eukaryotic (e. g. human) K+ channels. Simulations are important because an X-ray structure provides only a static snapshot of a protein, whereas a more dynamic description is needed in order to understand the functional behavior of channels. More explicitly, an X-ray structure is a spatial and temporal average structure of a channel protein in a crystal environment (i. e. with detergents and/or antibody fragments) at a temperature of ~100 K. We wish to characterise the dynamic behavior of a channel protein in a membrane (i. e. lipid bilayer) environment at a more physiologically relevant temperature of ~300 K. Modeling of mammalian ion channels on the basis of the structure of bacterial homologs is relatively well developed as a method [12]. Starting from a sequence alignment, homology modeling (Table 10.1) will yield a predicted structure for a mammalian channel based on a bacterial channel template. The quality of the resultant model depends to a large extent on that of the sequence alignment. Thus it is often necessary to adjust manually the initial sequence alignment to take account of: (i) experimental data identifying functionally important residues; (ii) experimental topology data; and (iii) preferential TM helix locations within the channel sequence. The latter may be predicted with ~80 % accuracy using a number of
Table 10.1 Computational methods. Method
Description
Code/Websites
homology modeling
generation of a mammalian channel model using a bacterial homolog structure as a template. atomistic simulations of protein motions (on a timescale of 10–50 ns).
Modeller [107, 108] http://salilab.org/modeller/ SwissModel [109] http://swissmodel.expasy.org/
molecular dynamics simulations network models Poisson– Boltzmann electrostatics ligand docking
coarse-grained descriptions of large scale protein motions calculation of the electrostatic field around and on the surface of a channel protein. prediction of ligand binding sites and interactions on the surface of a channel protein.
Gromacs [110] http://www.gromacs.org/ Charmm [111] http://www.charmm.org/ NAMD [112] http://www.ks.uiuc.edu/Research/namd/ GNM & ANM [51, 52] http://ribosome.bb.iastate.edu/software.html APBS [113] http://agave.wustl.edu/apbs/ UHBD [114] http://adrik.bchs.uh.edu/uhbd/ GRASP [115] http://honiglab.cpmc.columbia.edu/ Autodock [116] http://www.scripps.edu/mb/olson/doc/autodock/
This list of methods is representative of some readily available codes that have been used in channel modeling and simulation studies.
243
244
10 Molecular Modeling and Simulations of Ion Channels: Applications to Potassium Channels
TM helix prediction algorithms. However, rather than relying on a single algorithm, it is better to run multiple methods in order to generate a consensus prediction [13]. The major limitation of homology modeling and related approaches is that large (>15 residues) insertions (often corresponding to surface loops) in a mammalian channel sequence relative to that of the bacterial homolog are difficult to model with any certainty. In such cases it is preferable to omit the inserted region from the model. If the model with such omissions is to be used in simulations, a simple distance restraint can be used to mimic the effect of the missing region on the remaining structure. Molecular dynamics (MD) allows one to perform atomistic simulations of membrane protein motions on a timescale of 10 to 50 ns, through the application of an appropriate molecular mechanics forcefield (see Table 10.1). Such methods have been applied to a range of membrane proteins [14] including ion channels [15– 19], aquaporins [20–23], and bacterial outer membrane proteins [24–26]. The outcome of such a simulation is a trajectory, i. e. a description of the position and energetics of a membrane protein and its environment as a function of time, which can subsequently be analysed to yield insights into the relationship between the dynamic behavior and its biological function. A typical simulation system is illustrated in Fig. 10.2B. It consists of a K+ channel (here just the transmembrane domain) embedded in a phosphatidylcholine (PC) bilayer, with water molecules and ions on either side of the membrane. Typically, the size of such a simulation system is between 50 000 and 100 000 atoms. For such a system size, using current simulation codes (see Table 10.1) and relatively modest computational facilities (e. g. a small linux cluster) simulation times of the order of 10 ns can be achieved within a few days to weeks. This enables one to run multiple simulations to compare e. g. the dependence of the simulation behavior on the initial configuration of ions within the filter of a channel [27] or the behavior of various channel mutants [28]. More detailed calculations, such as the free energy landscape for ions within the filter of a channel [29], take a little longer but are still achievable with the same resources and simulation codes. Over the past few years, MD simulations have been widely exploited as a method of studying K+ channels [12, 17, 30–37]. It is important to note a major limitation of MD simulations, namely that of timescale. Even with substantial computational resources, for a channel simulation it is challenging to achieve a simulation time in excess of 100 ns (0.1 ms). To put this in context, the mean time for a single ion to pass through a channel is ~10 ns, and protein conformational changes underlying channel gating occur on a timescale of ~1 ms (106 ns). Thus, to use computational methods to obtain a complete description of single channel physiological function on the basis of molecular structure will require a hierarchy of simulation methods (see Fig. 10.2). As yet this hierarchy of methods is incompletely developed and integrated. Although the focus in the current article is on MD simulations, it is useful to consider other methods and their applications, real or potential, to ion channels. With respect to ion permeation a longer timescale can be addressed by the use of Brownian dynamics (BD) simulations. These are based on solution of the Pois-
10.2 Computational Methods Fig. 10.2 Molecular modeling and simulations. The overall aim (A) is to relate static X-ray structures of channels to the dynamic single channel physiological properties ((C) illustrated with single channel recordings from Kir6.2 channels, courtesy of F.M. Ashcroft [62]). Multilevel simulations are required to address a wide range of timescales (from ns to ms). In this chapter the focus is on atomistic modeling and simulations (B).
son–Boltzmann equation to describe the electrostatic field around a channel protein, followed by simulations in which the protein is treated as a static entity, the water as a continuum solvent, and ion diffusion in the electrostatic field created by the protein and membrane is simulated. This method has been used with some success in simulation of ion flow through high conductance, low ion selectivity channels such as bacterial outer membrane porins [38]. A detailed comparison of the results of MD and BD simulations has been performed for this system [39]. The application of these methods to lower conductance, high selectivity channels such as K+ channels is more challenging, and has been explored in detail by Chung and coworkers [40–43]. An even more challenging limitation of MD timescales occurs when trying to address the conformational changes underlying channel gating. One approach is to use ‘steered’ MD simulations in which a conventional MD simulation is modified by the inclusion of a biasing force that drives a conformational change such as channel gating [44]. Another MD-based approach is to identify the dominant low frequency protein motions from an MD simulation by way of principal components analysis (PCA; see Box 1). This method is also referred to as ‘essential dynamics’ [45–47], and has the advantage that it allows direct extrapolation from MD simulations to predict possible motions on longer timescales. However, it is well recognised that ~10 ns MD simulations incompletely capture the motions of membrane proteins [48], and thus biologically important long timescale motions may not be observed. There is consequently a considerable need to develop and apply coarsegrained (CG) models of ion channel motions. CG models of proteins have been explored in general for simulations of protein conformational changes and folding [49]. One promising class of CG methods is network models (Gaussian network models, GNM, and anisotopic network models, ANM) which represent each amino acid in a protein as a single ‘particle‘, coupled to other particles via a harmonic potential. Such methods have been shown to reproduce the temperature factors of proteins (a crystallographic measure of thermal fluctuations) [50] and have been used to explore the large scale dynamic behavior of various proteins [51, 52], includ-
245
246
10 Molecular Modeling and Simulations of Ion Channels: Applications to Potassium Channels
Box 1 Principal component analysis. Principal components analysis (PCA) of positional fluctuations is a valuable tool to extract the dominant low frequency protein motions from a molecular dynamics (MD) simulation. This enables one to extrapolate to the dynamics of the protein on a more extended timescale. The method is based on the definition of collective coordinates, C, that represent the major contribution to atomic fluctuations. These are obtained by diagonalizing the positional fluctuation covariance matrix A, whose elements are defined as: aij : {(xi – [xi]) (xj – [xj])} where […] denotes an average over time. A set of eigenvectors and eigenvalues are obtained solving the following equation:
AC = Cf where z = diag (zn) is a diagonal matrix whose nth element corresponds to the eigenvalues zn . Thus C is the eigenvector matrix. Generally 70–80 % of the total protein fluctuations are accounted for by the first 10 eigenvectors (that are accordingly referred to as ‘essential space’, [46]). Therefore, biological relevant motion can be captured by filtering the initial MD coordinate trajectory onto one of the essential degrees of freedom and then analyzing the resultant dynamics.
ing ion channel domains (see below, [53]). Further development and application studies are needed to explore fully the application of CG methods to channel gating, and their integration with MD simulations. Another computational approach that is widely used in studies of channels and receptors is computational docking of small molecules (i. e. ligands) to proteins. This is an important and active field of research in computational structural biology, and interested readers are referred to a recent review as a starting point [54]. An overview of a range of theoretical methods as applied to ion channels in general is provided in a special issue of IEEE Transactions on Nanobioscience [55]. In this chapter, we will focus on studies from the authors’ laboratory which are concerned with MD and related approaches to two classes of K+ channels, namely: inward rectifiers, and voltage-gated K+ channels.
10.3 Kir Channels 10.3.1 Structures
K+ channels of the inward rectifier (Kir) class have two main physiological roles: they regulate cell excitability by stabilizing the membrane potential close to the [K+] equilibrium potential, and they are involved in K+ transport across membranes [56, 57]. For example, Kir3.1/Kir3.4 channels modulate cardiac electrical
10.3 Kir Channels
activity, and Kir6.2 is involved in insulin release from pancreatic b-cells. Kir6.2 is the pore-forming component of the ATP-sensitive potassium (KATP) channel which couples cell metabolism to electrical activity by regulating K+ flux across the membrane. Channel closure is mediated by ATP, which binds to the intracellular domain of Kir6.2. Kir channels have two TM helices per subunit (similar to KcsA and MthK). Kir channels also have a large intracellular (IC) domain, composed of ~50 residues from the N-terminal tail of the protein plus a C-terminal domain of ~200 residues. This domain plays an important functional role via binding cytosolic regulators of Kir activity, such as ATP and PIP2. Three recent structures, of the isolated IC domain of a mammalian Kir (Kir3.1 = GIRK1 [58]) and of the two intact bacterial Kir homologues (KirBac1.1 [8] and KirBac3.1 [9]), offer a detailed understanding of structure/function relationships in this family of K+ channels (Fig. 10.3).
Fig. 10.3 The bacterial inward rectifier homolog KirBac. (A) Ca trace of KirBac1.1 showing the transmembrane (TM) and intracellular (IC) domains. The dotted horizontal lines represent the approximate extent of the lipid bilayer. (B), (C) TM pore-forming domains of KirBac1.1 and KirBac3.1, showing two of the four subunits as Ca traces, and the pore-lining surface (calculated using HOLE [117]).
10.3.2 Molecular Modeling
Molecular modeling studies have focussed on modeling mammalian Kir structures using bacterial K+ channel structures as templates. Earlier studies [28, 59, 60] focussed on the pore domain of Kir6.2. MD simulations were used to aid in evaluation of homology models of the Kir6.2 pore domain, derived using the KcsA pore domain structure as a template, and in particular to compare the relative con-
247
248
10 Molecular Modeling and Simulations of Ion Channels: Applications to Potassium Channels
formational stability of two alternative models of Kir6.2 which differed slightly in the sequence alignment of the M2 helices [59]. More recent studies have combined two templates in modeling the structure of Kir6.2 and related mammalian Kir channels. The IC domain of Kir6.2 has been modeled on the X-ray structure of the Kir3.1 C-terminal domain [58]. This has also enabled us to explore the conformational stability of the homology model of the Kir6.2 C-terminal domain on a 10 ns timescale. A homology model of the intact Kir6.2 molecule has been constructed [61] based on the crystal structures of KirBac1.1 and of the IC domain of Kir3.1. KirBac1.1 was used as a template for the TM domains of Kir6.2, whereas the IC domain was modeled on that of Kir3.1. This was because, over the region modeled, the IC domains of Kir3.1 and Kir6.2 exhibit a greater sequence identity (48 %) than those of Kir6.2 and KirBac1.1 (27 %). Further, the crystal structure of the IC domain of Kir3.1 was determined at a resolution higher (2 Å) than that of KirBac1.1 (3.6 Å). Each of the three segments of the model (TMs, N and C domains) were constructed separately, and then joined together. The spatial orientation of the IC domain, with respect to the TMs, was determined from the location of conserved residues in the IC domain of KirBac1.1. This model of Kir6.2 has been used as the basis of ligand docking and mutational studies (see below). 10.3.3 Simulations
Simulations of Kir channels have focussed on conformational changes and their relationship to channel gating. At a physiological level, Kir channel gating behavior may be (albeit somewhat crudely) divided into ligand gating, which occurs on a slow (multi-ms) timescale, and fast (‘filter’) gating which is seen as flickering transitions between open and closed within bursts of channel activity (see Fig. 10.2) [62]. However, even such fast gating is on a ms timescale. Thus, as discussed above, gating presents a considerable challenge to simulation studies, which cannot yet address ms timescales directly. However, by careful design of simulation ‘experiments’ and by suitable analyses, we may extrapolate from results on a multi-ns timescale and so formulate atomic resolution hypotheses relating to both aspects of channel gating. 10.3.4 Filter Flexibility
A number of simulations of KcsA have indicated that the original model of K+ channel function, which stressed a relatively rigid structure for the selectivity filter [6], requires modification to allow for filter flexibility [17, 27, 29, 63–65]. These simulation results correlate well with crystallographic studies of changes in the conformation of the selectivity filter of KcsA as a function of ion occupancy [7, 66] (Fig. 10.4), with crystallographic studies of conformational changes during block by tetraethyl ammonium [67], and with more indirect structural studies [68]. More
10.3 Kir Channels Fig. 10.4 Distortion of the K+ channel selectivity filter. The X-ray structure of the KcsA filter in the presence of (A) high and (B) low concentrations of K+ ions. Note the distortion to the filter in the presence of low [K+] due to mutual repulsion of the carbonyl oxygen atoms [7]. Distortions of the selectivity filter seen in simulations of inward rectifier channels. (C) The KirBac filter from a simulation in the absence of K+ ions [69]. (D) A Kir6.2 model filter for a mutation close to the filter (V127T) that changes open channel permeation and kinetic properties [28].
recently, simulations have revealed the filter of Kir channels to undergo distortions comparable to those seen in the low [K+] structure of KcsA. Thus in simulations of KirBac1.1 in which K+ ions were omitted from the filter (i. e. were replaced with water molecules) [69] the filter distorted via a ‘flip’ of one or more of the constituent polypeptide backbones resulting in carbonyl oxygen atoms being directed away from the pore. The close resemblance between the low [K+] X-ray structure and the zero-[K+] MD structure (Fig. 10.4) provides strong evidence for a model of K+ channel permeation in which the occupancy of the filter, by multiple (2 or 3) K+ ions simultaneously, is responsible for induced fit. In such a model there is a coupling of ion binding to a protein conformational change which underlies the high conduction rates of the channel [66]. Interestingly, a similar conformational change has been observed in simulations of models of mutants of Kir6.2 [28]. It had been noted that the single-channel conductance of Kir channels varies significantly between different members of the family. On this basis a mutation (V127T) was made close to the filter of Kir6.2 which was been shown to produce channels with reduced (40 % of wildtype) single-channel conductance [62]. Homology modeling (based on a KcsA template) combined with MD simulations was used to explore whether changes in structural dynamics of the filter were induced by the V127T mutations. Relative to the wild-type simulation, the V127T mutant showed significant distortion of the filter, such that ~50 % of the simulation time was spent in a distorted ‘filter-closed’ conformation (Fig. 10.4D). While in this conformation, translocation of K+ ions between adjacent binding sites within the filter was blocked. Significantly, the distorted filter conformation resembled that of KcsA crystallized in the presence of a low [K+]. Taken together, these studies suggest that local distortion of the selectivity filter may be a general model for determining the conductance of K+ channels and/or may be related to ‘fast’ gating of Kir channels.
249
250
10 Molecular Modeling and Simulations of Ion Channels: Applications to Potassium Channels
10.3.5 M2 Helices and Hinge Motion
KirBac1.1 and 3.1 have similar, but nonidentical, pore domain structures (Fig. 10.3B), differing in the extent to which the intracellular gate is closed. Multiple extended MD simulations (each of >20 ns duration) of the isolated TM domain of both KirBac channels in two membrane environments (a PC bilayer and a membrane-mimetic octane slab) have been performed in order to explore possible M2 helix motions underlying Kir channel gating [70]. In these simulations just the TM domain was used, in an attempt to uncouple the M2 helix gate from the IC ‘gate-keeper’ domain, thus enhancing the likelihood of observing conformational changes in the gate on a 20 ns timescale. Analysis of these simulations focussed on the conformational dynamics of the pore-lining M2 helices. Principal components analysis (see Box 1) was used to analyse bending of the M2 helix. Such bending occurs at a molecular hinge formed by a conserved glycine residue (Gly134 in KirBac1.1, Gly120 in KirBac3.1). This glycine has also been suggested to form a channel gating hinge on the basis of comparison of the structures of KcsA (i. e. a closed K+ channel) and MthK (an open K+ channel). A more detailed analysis of the M2 helix bundle dynamics was suggestive of a dimer-of-dimers motion in which opposing pairs of helices moved together. This may be related to a similar pattern of motions observed for the isolated IC domain (see below). The first two eigenvectors describing the motions of M2 in these simulations correspond to helix kink and helix swivel motions. The conformational flexibility of M2 seen in these simulations correlates well with differences in M2 conformation captured in the X-ray structures, as can be seen if one compares closed channels (KcsA and KirBac) in which the helix is undistorted, with open channels (e. g. MthK) in which the M2 helix is kinked (Fig. 10.5). Thus the simulations, albeit on
Fig. 10.5 M2 helices, comparing structure from molecular dynamics (MD) simulations with those from X-ray structures. (A) M2 helices from three crystal structures (KcsA – closed; KirBac1.1 – closed; and MthK – open). (B) M2 helices from an MD simulation of KirBac1.1. (C) M2 helices from an MD simulation of KirBac3.1. In each case the M2 helices are superimposed on the Ca atoms N-terminal to the conserved glycine hinge residue.
10.3 Kir Channels
a timescale substantially shorter than that of channel gating, support a gating model in which the intrinsic flexibility of M2 about a molecular hinge is used to modify the pore dimension at the intracellular mouth, enabling switching between a closed conformation (undistorted M2 helix, narrow hydrophobic pore mouth) and an open conformation (kinked M2 helix, wide pore mouth). Simulations of the closed structure of KcsA in comparison with simulations of a model of the KcsA open state (based on MthK) also support this model [71], as do steered MD simulations [72], and normal mode analysis [73] calculations based on the closed KcsA structure. 10.3.6 Intracellular Domain Dynamics
As noted above, the intracellular C-terminal (IC) domain of Kir channels regulates channel gating in response to changes in concentration of various ligands. MD simulations (~10 ns) have been used to probe the dynamics of two Kir C-terminal domain tetramers, namely Kir3.1 (a crystal structure) and Kir6.2 (a homology model). The Kir6.2 simulations were performed with and without bound ATP (introduced by docking). The results of these simulations revealed comparable conformational stability and dynamics for the crystal structure and for the homology model. However, principal components analysis (PCA) of the simulations did reveal a conserved pattern of motion of relevance to channel gating. Thus, in both the Kir3.1 and Kir6.2 tetramers PCA revealed loss of symmetry, consistent with a dimer-of-dimers motion of subunits in the IC domains of the corresponding Kir channels. Of course, the timescale on which this was observed is considerably shorter than that associated with channel gating. To test this hypothesis further, coarse-grained (anisotropic network model [52]) calculations were performed on the IC domain. These also revealed a dimer-of-dimers motion of the IC domain tetramer, essentially identical to that seen in the MD simulations (Fig. 10.6). This suggests that extrapolation from the MD simulation timescale to a channel gating timescale may be valid. Of course, to fully understand gating of Kir channels we need not only to understand the intrinsic flexibility of the TM and IC domains, but also the nature of the interactions of regulatory ligands with the IC domain. Unfortunately, on a 10 ns timescale, the flexibility of the Kir6.2 tetramer was not changed greatly by the presence of docked ATP, other than in two loop regions. 10.3.7 Interactions with Ligands
Ligand docking studies with a homology model of Kir6.2 have been used to help identify the ATP-binding site [61, 74]. The resultant intact model is consistent with a substantial body of functional data and has been tested by mutagenesis. Ligand binding occurs at the interface between two subunits. The phosphate tail of ATP interacts with two basic sidechains (R201 and K185) in the C-terminus of one subunit, and with a further basic sidechain (R50) in the N-terminus of another;
251
252
10 Molecular Modeling and Simulations of Ion Channels: Applications to Potassium Channels
Fig. 10.6 Kir intracellular C-terminal (IC) domain dynamics, with the protein shown as a Ca trace, and with arrows attached to each Ca atom to indicate the direction of an eigenvector and the magnitude of the corresponding eigenvalue. (A) The motion corresponding to the first eigenvector from PCA of an MD si-
mulation of a model of the IC domain tetramer from Kir6.2. (B) The first eigenvector (i. e. the lowest frequency mode) from an anisotropic network model [52] analysis of the IC domain tetramer of Kir3.1. (C) A schematic diagram summarising the motions of the four subunits as revealed by the eigenvectors.
the N6 atom of the adenine ring interacts with E179 and R301 in the same subunit. Significantly, mutation of residues lining the binding pocket reduced ATPdependent channel inhibition, lending support to the model. This provides a clear example of how a homology model can be used to rationalise existing mutation data, and to aid design of novel mutational experiments. Kir6.2 channels are modulated by ligands other than ATP, including PIP2. Simulations of PIP2 may be used to explore inositol headgroup conformations relative to the PC bilayer environment of the PIP2 molecule (Haider and Sansom, manuscript in preparation). In parallel, continuum electrostatics (Poisson–Boltzmann) calculations may be used to identify regions on the surface of the Kir6.2 channel model which have a highly positive potential and thus are likely to interact with the polyanionic headgroup of PIP2. By combining such calculations with docking studies of an IP3-fragment from the PIP2 simulations (Fig. 10.7) a model of the interactions of Kir6.2 with PIP2 may be generated and subsequently used in simulations to further explore how such ligands modulate the behavior of Kir channels (Haider and Sansom, unpublished work).
10.3 Kir Channels
Fig. 10.7 Interactions of PIP2 with Kir6.2. (A) A molecular surface representation of a model of the Kir6.2 channel calculated using GRASP [115], color on electrostatic potential (from –6.6 to 5.4 kT, red to blue). The region of positive electrostatic potential (blue) near the intracellular membrane/water interface (indicated by the circle) corresponds to a pos-
sible binding site for PIP2. (B) Snapshot from a simulation of three PIP2 molecules within a POPC bilayer. The PIP2 molecules are shown in space-filling format whilst the phosphorus atoms of the POPC headgroups are shown as green spheres. (This figure also appears with the color plates.)
10.3.8 Towards an Integrated Gating Model
It can be seen how simulation and modeling studies may provide insights into the intrinsic flexibility of the two major domains (TM and IC) of the Kir channel, and into the nature of the interactions of the IC domain with various ligands. One may attempt to integrate this information to formulate a gating model for Kir channels. In such a model (Fig. 10.8) a transition between exact tetrameric symmetry and dimer-of-dimers symmetry of the IC domains is associated with a change in TM helix packing coupled to gating of the channel. In this model, the open state of the channel has four kinked M2 helices which interact with a tetrameric IC domain, thus holding the helices in their open conformation. In contrast, in the closed channel the helices are no longer kinked, and pack together in a dimer-of-dimers conformation along with the IC domain. This model receives some support from the X-ray structures in that both KirBac1.1 and KirBac3.1 (which are in closed conformations) exhibit a degree of dimer-of-dimers symmetry, i. e. the crystallographic asymmetric unit is a dimer rather than a monomer. Of course, this model is speculative. However, it does provide a framework for further exploration, both computational (via more coarse-grained protein simula-
253
254
10 Molecular Modeling and Simulations of Ion Channels: Applications to Potassium Channels
Fig. 10.8 Proposed gating mechanism for Kir channels. The M2 helices of the TM domain of two opposite subunits are shown in ribbons format with the glycine hinge in dark gray. In the open state these helices are kinked. Asymmetric movements of the C-
terminal domains (pale and dark gray circles), as indicated by the arrows, result in closure of the channel via adoption of a dimer-of-dimers packing by the M2 helices, which also lose their kink upon channel closure.
tions) and experimental, of possible changes in symmetry associated with Kir channel gating. An even more challenging task will be to understand the way in which interactions with specific ligands stabilise either the open or closed form of the channel.
10.4 Kv Channels 10.4.1 Structures
The other major class of K+ channel for which structural data are available is the voltage-gated (Kv) channels. Kv channels open and close in response to changes in transmembrane voltage, and play a key role in electrical signalling by excitable cells. Kv channel subunits contain two distinct but functionally coupled TM domains (Fig. 10.9). The pore domain has a similar fold to that of other K+ channels, and shares significant sequence homology in the extracellular pore region with KcsA. The voltage sensor (VS) domain is responsible for triggering a change in conformation of the pore domain in response to changes in TM voltage so as to open the channel. Each Kv channel subunit consists of six a-helices (S1-S6). The first four TM helices form the VS domain (Fig. 10.9B), whereas the last two TM helices (S5-S6), along with an intervening re-entrant P loop, form the pore domain (Fig. 10.9C). Crystallographic studies of a bacterial Kv homologue, KvAP [10], have confirmed that the pore domain of KvAP has an architecture similar to
10.4 Kv Channels Figure 10.9 (A) Schematic diagram of the transmembrane topology of a Kv channel subunit, showing the voltage sensor (S1 to S4) and pore (S5 to S6) domains. The intact channel is made up of four such subunits. The intracellular (IC) and extracellular (EC) faces of the membrane are labelled. (B) Structure of the (isolated) voltage sensor domain of the KvAP, with the S4 helix in dark gray. (C) Structure of the pore domain taken from the X-ray structure of the intact KvAP channel.
that of other K+ channels. The main activation gate therefore lies at the intracellular mouth of the channel, at the crossing point of the S6 helices. The revelation of the structure of KvAP has resulted in some controversy concerning the VS domain. This domain is made of helices S1 to S4, with the positively charged S4 helix (which has every third residue positively charged in its Nterminal half) responding to a change in TM voltage in order to initiate the conformational change that results in channel activation gating. On the basis of the Xray structure of the intact KvAP channel it has been suggested that the S3 and S4 helices form a ‘paddle’ that lies close to the intracellular surface of the membrane in the resting state of the channel, and which is induced to cross the bilayer upon membrane depolarisation, thus triggering activation gating of the channel [75]. However, the exact conformation and orientation of the VS domain remains uncertain. In particular, the structure of the VS domain within the crystal of the full length channel construct is apparently at odds with various physiological and biophysical data [76, 77]. Interestingly, the orientation of the VS domain in the X-ray structure of the intact channel, and in a single particle EM structure [78] seem to be somewhat different, suggestive of a conformational transition associated with channel gating. Significantly, in addition to the crystal structure of the full length KvAP channel, the structure of an isolated VS domain fragment has also been solved (PDB code 1ORS) [10] (Fig. 10.9B). The isolated VS domain structure is perhaps more consistent with a range of biophysical and physiological data, and thus may be more representative of the conformation of the VS domain under physiological conditions. Combined with the relatively high resolution (1.9 Å) of the VS structure, this domain thus provides an attractive candidate for simulation studies of its conformational dynamics in the context of possible channel gating mechanisms. Furthermore, the structure of the isolated KvAP VS domain closely resembles that of the
255
256
10 Molecular Modeling and Simulations of Ion Channels: Applications to Potassium Channels
equivalent domain in a recent X-ray structure of an intact mammalian Kv1.2 channel [79]. However, let us first reconsider the pore domain and the gate per se. 10.4.2 S6 Helices, Hinges and Gating
Comparison of crystal structures of potassium channels and simulations of e. g. KirBac (see above) suggest that the gating mechanism of Kv channels may involve a key role for the pore-lining S6 helix. Within the S6 helix sequence there is a conserved PVP sequence motif. Mutations of this motif have been shown to alter the gating of Kv channels in a manner consistent with the formation of a molecular gating hinge in this region [80, 81]. Molecular dynamics simulations of isolated S6 helices in membrane-like environments [82] and of models of the pore-forming domain of Kv channels [83] have been used to explore the conformational dynamics of the S6 helix hinge. The results of these simulations indicate that the PVP motif may form a molecular hinge. In the isolated helices there is considerable (albeit anisotropic) motion about the PVP hinge (Fig. 10.10A). Substantial hinge-bending motion remains, even when the S6 helix forms part of a more complete pore domain model. Thus the intrinsic conformational dynamics of S6 are modulated by the remainder of protein but it remains flexible. These simulation results are suggestive of a channel gating model in which S6 bends in the vicinity of the PVP motif (Fig. 10.10B) in addition to the region around the conserved glycine that is N-terminal to the PVP motif. Evidence in favor of a helix distortion of S6 in the region of the PVP motif has also been obtained by cysteine modification studies [84, 85], and a PVP-hinge is also supported by the recent X-ray structure of a Kv1.2 channel [79, 86]. Thus, K+ channel gating may depend on a complex switch in conformation with three rigid helical sections linked by two molecular hinges, one at the conserved glycine and one at the PVP motif.
Fig. 10.10 (A) S6 helix structures taken from a simulation of the isolated helix in a membrane mimetic octane slab [82]. The Nterminus of the helix is at the top of the diagram, and the location of the PVP hinge motif is indicated. (B) Schematic model of the inner helix bundle of Kv channel pore as formed by four kinked S6 helices.
10.4 Kv Channels
10.4.3 The Barrier at the Gate
Homology modeling has been used to generate models of two states of the pore domain of the Shaker Kv channel: one, based on the X-ray structure of KcsA, represents a closed state of the channel, whilst the other, based on KvAP, represents an open state pore domain (Fig. 10.11A and B) [87]. It is evident that in the Kvclosed model the intracellular mouth of the channel (i. e. the activation gated) is much narrower (radius ~1.4 Å) than in the Kv-open model (radius >10 Å). A simple calculation of the energetic barrier presented by the hydrophobic gate region in the Kv-closed model may be obtained via a Poisson–Boltzmann calculation [16, 88], which provides a first approximation to the barrier height presented by a hydrophobic gate. The pore radius profile of the Kv-closed model has an average hydrophobic gate radius of 1.4 Å, extending over a length of 2 Å. This yields a broad
Fig. 10.11 Comparison of Kv-closed and Kvopen models. Homology models of the Shaker Kv pore domain, based on (A) KcsA (for the Kv-closed model) and (B) KvAP (for the Kv-open model). In both cases the model is restricted to the pore-forming domain and only two of the four subunits are shown. The approximate location of the lipid bilayer is
shown by the horizontal broken lines. The surface of the pore is shown as calculated using Hole [117]. (C) Poisson–Boltzmann energy as a function of position along the pore axis for a singly charged cation for the two models shown in (A). The black line corresponds to Kv-closed, and the gray line to Kv-open.
257
258
10 Molecular Modeling and Simulations of Ion Channels: Applications to Potassium Channels
barrier of maximum height ~80 kT in the centre of the gate (Fig. 10.11C). A comparison of Poisson–Boltzmann energies and atomistic simulation-based free energies for simple nanopore models [88] suggests that Poisson–Boltzmann calculations may overestimate the barrier height by a factor of about two. Correcting for this still yields an estimated barrier height of ~40 kT for the Kv-closed channel. Thus the model clearly corresponds to a fully closed conformation of the channel. In contrast, there is no barrier at all in the Kv-open model. Thus, S6 helix bending motions are sufficient to switch the pore fully from a functionally closed to a functionally open state. 10.4.4 The Nature of the Voltage Sensor
As discussed above, there is some uncertainty as to the conformation and/or localisation of the VS domain within Kv channels. Comparison of the X-ray structures of the VS in the intact KvAP channel (PDB code 1ORQ), and of the isolated VS domain (PDB code 1ORS) reveals a number of differences in their conformation. In particular, the isolated VS more closely resembles a conventional membrane protein. It consists of a bundle of four TM a-helices which are capable of spanning a lipid bilayer. The isolated VS domain forms crystals capable of diffracting to a high resolution [10]. It also forms a stable folded domain in detergent micelles [10, 75] and in lipid bilayers [89]. Given all this, the isolated VS domain structure thus may represent a conformation of the VS domain within the intact channel under physiological conditions. It is therefore of interest to explore the localisation of the VS domain in a membrane (and in the KvAP channel) and to examine its conformational dynamics. One approach to help localise the VS in a membrane is via a number of toxins that interact with the voltage sensor of Kv channels [76, 90, 91]. VSTX1 and related toxins from spider venoms bind to the voltage sensor of Kv channels. These toxins are thought to access the VS via the lipid bilayer phase. Consequently,VSTX1 provides a probe to help localise the VS of Kv channels relative to the centre of the bilayer. However, this requires knowledge of the location of the toxin within the bilayer. This may be approached via MD simulations. In a recent study [92] a series of simulations was used to explore the net drift of VSTX1 relative to the centre of a PC bilayer, starting from different locations of the toxin. The preferred location of the toxin appears to be at the membrane/water interface, ~15 Å from the centre of the bilayer. This interfacial location allows the toxin to maximise its H-bonding to lipid headgroups and to water molecules, whilst retaining extensive hydrophobic interactions with the hydrophobic core of the bilayer. Thus, the toxin partitions to the water/bilayer interface (in itself an extensive region, about 10 Å thick [93]), with its hydrophobic residues pointing down towards the hydrophobic core of the bilayer, and its hydrophilic sidechains interacting with the lipid headgroups and with interfacial water molecules. This localisation in turn allows one to develop a model of VSTX1 interactions with the voltage sensor of Kv channels.
10.4 Kv Channels
Mutational studies on voltage sensor-binding toxins combined with mutational studies of Kv channels point to an interaction between polar/charged residues on the toxin surface with residues towards the C-terminus of the S3 b helix of the voltage sensor [91, 94, 95]. Based on this information we can model the positions of VSTX1 and the KvAP voltage sensor relative to a POPC lipid bilayer. As seen in Fig. 10.12,VSTX1 is located such that a band of positively charged residues on its surface (which are likely to form the interactions between the toxin and the voltage sensor) is located exactly at the membrane interface. The VS crystal structure may be inserted into a POPC bilayer such that (i) it spans the bilayer symmetrically; and (ii) a surface-exposed tyrosine (Y46 of S1) is located at the interface. Note that tyrosine and tryptophan residues tend to be located in bands on the surfaces of membrane proteins corresponding to the lipid/water interface [96]. In this orientation, residues in the S3 b helix (which forms part of the crucial gating paddle [75, 76, 97] of the voltage sensor) are aligned at the correct depth to interact with the basic residues of VSTX1. Thus, it would seem that VSTX1 binds to the tip of the gating paddle (as defined by S3 b and the N-terminal region of S4), which is in turn located at the extracellular interfacial region of the membrane. A number of recent studies have focussed on the manner in which the VS and pore domains of Kv channels are packed together [98], and on exploring the changes in conformation and/or orientation of the VS in response to membrane depolarisation [76]. For example, site-directed spin label data [99] has been used to develop an alternative model of how the VS and pore domains of KvAP may pack together within a lipid bilayer environment. A key feature of this model is that the basic sidechains of S4 are exposed to the surrounding lipid environment. Several models of the conformational change underlying Kv channel gating have been
Fig. 10.12 A model for VSTX inhibition of Kv channels. VSTX1 is shown partitioned to the water/membrane interface, at which location it binds to the voltage sensor (VS). The toxin is shown in space filling format. The approximate location of the interface is shown by the phosphorus atoms of a POPC bilayer (small gray spheres), the other atoms of the lipid molecules being omitted for clarity. The crys-
tal structure of the isolated KvAP voltage sensor domain is shown such that a cluster of residues in the S3 b helix that may interact with VSTX1 is shown in space filling format. The tyrosine residue of S1 which defines the extracellular interfacial region (Y46) is also shown in space fill. The S3 b and S4 helices together form the ‘paddle’ of the voltage sensor domain (circled).
259
260
10 Molecular Modeling and Simulations of Ion Channels: Applications to Potassium Channels
proposed, namely: the S4 helical screw [100], transporter [101], paddle [75], and twisted S4 models [99]. Thus, it is of some interest to explore the intrinsic flexibility of the VS domain in order to explore further the plausibility of these models. MD simulations have been performed on the isolated VS from KvAP located in a detergent micelle environment (Sands et al., manuscript submitted). Simulations at two different temperatures (300 K and 368 K) were used to probe the intrinsic flexibility of this domain on a ~10 ns timescale (Fig. 10.13A). The VS contains a positively charged (S4) helix which is packed against a more hydrophobic S3 helix. The simulations at elevated temperature reveal an intrinsic flexibility of the S3 a region (i. e. the N-terminus of the S3 helix; Fig. 10.13B). It is also evident that the S4 helix undergoes hinge bending and swivelling about its central I130 residue (Fig. 10.13C). The conformational instability of the S3 a region may facilitate the motion of the N-terminal segment of S4 (i. e. S4 a). These simulations thus support a modified model of an S3b-S4 a paddle that can swivel relative to the rest of the VS domain. This movement may form the underlying basis of voltage sensing by Kv channels.
Fig. 10.13 The KvAP voltage sensor. (A) The voltage sensor embedded in a detergent (DMA) micelle shown at the end (t = 40 ns) of an MD simulation. (B) The structure of the S2–S3 region of the VS at the end of a 40 ns MD simulation at 368 K. The residues are colored according to the magnitude of root mean square fluctuations experienced by the
Cas during the course of each simulation (on a scale from blue = 0.0 Å to red = 4.8 Å). The loss of helicity in S3 a is evident. (C) KvAP S4 helix hinge-swivelling about residue I130, as revealed by eigenvector 1 of an MD simulation at 368 K. The colors indicate the range of motions represented by this eigenvector. (This figure also appears with the color plates.)
10.4.5 A Possible Gating Model
On the basis of these results it is possible to formulate a possible gating model for Kv channels (Fig. 10.14). In such a model the VS domain is envisaged as a ‘spring and swivel‘. The positively charged S4 helix responds to a change in transbilayer electrostatic field (i. e. membrane depolarisation) such that its S4 a region swivels relative to the C-terminal half of the S4 helix and the remainder of the voltage sensor. The conformational flexibility of S3 a is suggested to provide a restoring
10.5 Summary and Future Directions Fig. 10.14 A possible gating model for Kv channels. The VS domain is shown, with the S4 helix in black and the S3 a segment in dark gray. The positively charged S4 helix is suggested to respond to membrane depolarisation (DV) by the S4 a region swivelling relative to the S4b region which remains fixed relative to the remainder of the VS. The conformational flexibility of S3 a may provide a restoring ‘spring’ able to return the voltage-sensing paddle (formed by S3bS4 a) to its initial conformation once the resting membrane potential is restored.
‘spring’ connected to the voltage-sensing paddle (formed by S3b-S4 a) which resets the conformation upon return to the resting membrane potential. This model is therefore related to both the paddle model of MacKinnon and colleagues [75] and to models (reviewed in e. g. Ref. [97]) in which S4 is proposed to undergo a rotational motion, as suggested by e. g. fluorescence energy transfer experiments [102, 103]. It remains rather difficult to formulate detailed models of how a proposed motion of the VS upon depolarisation may be linked to the conformational change, involving bending of the S6 helices, that opens the Kv channel. The construction of such a model requires a more definitive structure and/or model of the way in which the VS and pore domains of Kv channels are packed together in the intact channel protein under physiological conditions. This remains elusive, although a number of methods can reveal important clues [99].
10.5 Summary and Future Directions
In summary, we have seen how homology modeling and simulation may be combined in order to extrapolate from static structures of bacterial ion channels to a dynamic description of the structural basis of function of their mammalian homologs. However, our discussion has been limited to K+ channels, for which substantial structural data are available. In contrast, for neurotransmitter-gated ion channels (e. g. the nicotinic acetylcholine receptor), more limited structural data are available [3, 104] which in turn restricts modeling [105] and simulation [106] studies. In brief, as structures for more channels are determined, computational studies relating structure to function will expand. Current simulation methodologies are able to address mechanisms of ion permeation through channels although further refinements are needed, especially with respect to understanding ion selectivity. However, modeling and simulation of gating remains challenging, as described above. We need improved methods that can address longer timescales and larger scale conformational transitions.
261
262
10 Molecular Modeling and Simulations of Ion Channels: Applications to Potassium Channels
Network models are promising, but are likely to be too approximate to describe fully, for example, voltage- or ligand-induced conformational changes leading to gating. It is therefore important to explore the application to ion channels of a variety of coarse-grained models of proteins [49]. It will also be important to understand the role of protein dynamics in the interactions of channels with their ligands. To date, computational studies have largely focussed on single channel proteins and their component domains. There will be a need in the future to address more complex assemblies, i. e. channel proteins interacting with other protein within and adjacent to the cell membrane. From a computational perspective, this will require the development of multi-scale simulation methods, that are able to model both large-scale events (e. g. protein–protein interactions) at the same time as more fine-grained processes (e. g. ion permeation). Thus, ion channels will continue to provide a focus for the development and exploitation of new computational tools to bridge the gap between structure and physiology.
Acknowledgements
Research in MSPS’s laboratory is supported by grants from The Wellcome Trust, the EPSRC, and the BBSRC. Our thanks to our many colleagues for their ongoing interest in this work.
References 1 J. M. Gulbis, D. A. Doyle, Potassium channel structures: do they conform? Curr. Opin. Struct. Biol. 2004, 14, 440– 446. 2 S. H. White, The progress of membrane protein structure determination, Prot. Sci. 2004, 13, 1948–1949. 3 N. Unwin, Refined structure of the nicotinic acetylcholine receptor at 4 Å resolution, J. Mol. Biol. 2005, 346, 967– 989. 4 O. Sokolova, L. Kolmakova-Partensky, N. Grigorieff, Three-dimensional structure of a voltage-gated potassium channel at 2.5 nm resolution, Structure 2001, 9, 215–220. 5 G. Yellen, The voltage-gated potassium channels and their relatives, Nature 2002, 419, 35–42. 6 D. A. Doyle, J. M. Cabral, R. A. Pfuetzner, A. Kuo, J. M. Gulbis, S. L. Cohen, B. T. Cahit, R. MacKinnon, The structure of the potassium chan-
7
8
9
10
nel: molecular basis of K+ conduction and selectivity, Science 1998, 280, 69–77. Y. Zhou, J. H. Morais-Cabral, A. Kaufman, R. MacKinnon, Chemistry of ion coordination and hydration revealed by a K+ channel-Fab complex at 2.0 Å resolution, Nature 2001, 414, 43– 48. A. Kuo, J. M. Gulbis, J. F. Antcliff, T. Rahman, E. D. Lowe, J. Zimmer, J. Cuthbertson, F. M. Ashcroft, T. Ezaki, D. A. Doyle, Crystal structure of the potassium channel KirBac1.1 in the closed state, Science 2003, 300, 1922–1926. J. M. Gulbis, A. Kuo, B. Smith, D. A. Doyle, A. Edwards, C. Arrowsmith, M. Sundstrom, Two intermediate gating state crystal structures of the KirBac3.1 K+ channel, to be published. Y. Jiang, A. Lee, J. Chen,V. Ruta, M. Cadene, B. T. Chait, R. Mackinnon, X-ray structure of a voltage-depen-
References
11
12
13
14
15
16
17
18
19
20
21
dent K+ channel, Nature 2003, 423, 33– 41. Y. Jiang, A. Lee, J. Chen, M. Cadene, B. T. Chait, R. MacKinnon, Crystal structure and mechanism of a calciumgated potassium channel, Nature 2002, 417, 515–522. A. Giorgetti, P. Carloni, Molecular modeling of ion channels: structural predictions, Curr. Opin. Chem. Biol. 2003, 7, 150–156. J. M. Cuthbertson, D. A. Doyle, M. S. P. Sansom, Transmembrane helix prediction: a comparative evaluation and analysis, Proteins Eng. Des. Sel. 2005, 18, 295–308. W. L. Ash, M. R. Zlomislic, E. O. Oloo, D. P. Tieleman, Computer simulations of membrane proteins, Biochim. Biophys. Acta 2004, 1666, 158–189. M. S. P. Sansom, I. H. Shrivastava, K. M. Ranatunga, G. R. Smith, Simulations of ion channels – watching ions and water move, Trends Biochem. Sci. 2000, 25, 368–374. D. P. Tieleman, P. C. Biggin, G. R. Smith, M. S. P. Sansom, Simulation approaches to ion channel structurefunction relationships, Quart. Rev. Biophys. 2001, 34, 473–561. M. S. P. Sansom, I. H. Shrivastava, J. N. Bright, J. Tate, C. E. Capener, P. C. Biggin, Potassium channels: structures, models, simulations, Biochim. Biophys. Acta 2002, 1565, 294– 307. B. Roux, T. Allen, S. Berneche,W. Im, Theoretical and computational models of biological ion channels, Quart. Rev. Biophys. 2004, 37, 15–103. B. Roux, K. Schulten, Computational studies of membrane channels, Structure 2004, 12, 1343–1351. M. O. Jensen, S. Park, E. Tajkhorshid, K. Schulten, Energetics of glycerol conduction through aquaglyceroporin GlpF, Proc. Nat. Acad. Sci. USA 2002, 99, 6731–6736. E. Tajkhorshid, P. Nollert, M. O. Jensen, L. J. W. Miercke, J. O’Connell, R. M. Stroud, K. Schulten, Control of the selectivity of the aquaporin water channel family by global or-
22
23
24
25
26
27
28
29
30
31
32
ientational tuning, Science 2002, 296, 525–530. B. Ilan, E. Tajkhorshid, K. Schulten, G. A. Voth, The mechanism of proton exclusion in aquaporin channels, Proteins: Struct. Funct. Bioinf. 2004, 55, 223–228. B. L. de Groot, H. Grubmuller,Water permeation across biological membranes: Mechanism and dynamics of aquaporin-1 and GlpF, Science 2001, 294, 2353–2357. D. P. Tieleman, H. J. C. Berendsen, A molecular dynamics study of the pores formed by Escherichia coli OmpF porin in a fully hydrated palmitoyloleoylphosphatidylcholine bilayer, Biophys. J. 1998, 74, 2786–2801. W. Im, B. Roux, Ions and counterions in a biological channel: A molecular dynamics simulation of OmpF porin from Escherichia coli in an explicit membrane with 1 M KCl aqueous salt solution, J. Mol. Biol. 2002, 319, 1177–1197. P. J. Bond, M. S. P. Sansom, The simulation approach to bacterial outer membrane proteins, Mol. Membr. Biol. 2004, 21, 151–162. I. H. Shrivastava, M. S. P. Sansom, Simulations of ion permeation through a potassium channel: molecular dynamics of KcsA in a phospholipid bilayer, Biophys. J. 2000, 78, 557–570. C. E. Capener, P. Proks, F. M. Ashcroft, M. S. P. Sansom, Filter flexibility in a mammalian K channel: models and simulations of Kir6.2 mutants, Biophys. J. 2003, 84, 2345–2356. S. Bernèche, B. Roux, Energetics of ion conduction through the K+ channel, Nature 2001, 414, 73–77. B. Roux, S. Bernèche,W. Im, Ion channels, permeation and electrostatics: Insight into the function of KcsA, Biochemistry 2000, 39, 13295–13306. B. Roux, Theoretical and computational models of ion channels, Curr. Opin. Struct. Biol. 2002, 12, 182–189. L. Guidoni,V. Torre, P. Carloni, Potassium and sodium binding in the outer mouth of the K+ channel, Biochemistry 1999, 38, 8599–8604.
263
264
10 Molecular Modeling and Simulations of Ion Channels: Applications to Potassium Channels 33 L. Guidoni,V. Torre, P. Carloni, Water and potassium dynamics in the KcsA K+ channel, FEBS Lett. 2000, 477, 37–42. 34 M. Compoint, P. Carloni, C. Ramseyer, C. Girardet, Molecular dynamics study of the KcsA channel at 2.0-Å resolution: stability and concerted motions within the pore, Biochim. Biophys. Acta 2004, 1661, 26–39. 35 J. Åqvist,V. Luzhkov, Ion permeation mechanism of the potassium channel, Nature 2000, 404, 881–884. 36 V. B. Luzhkov, J. Åqvist, A computational study of ion binding and protonation states in the KcsA potassium channel, Biochim. Biophys. Acta 2000, 1481, 360–370. 37 V. B. Luzhkov, J. Åqvist, Mechanisms of tetraethylammonium ion block in the KcsA potassium channel, FEBS Lett. 2001, 495, 191–196. 38 T. Schirmer, P. S. Phale, Brownian dynamics simulation of ion flow through porin channels, J. Mol. Biol. 1999, 294, 1159–1167. 39 W. Im, B. Roux, Ion permeation and selectivity of OmpF porin: A theoretical study based on molecular dynamics, Brownian dynamics, and continuum electrodiffusion theory, J. Mol. Biol. 2002, 322, 851–869. 40 S. H. Chung, T. W. Allen, M. Hoyles, S. Kuyucak, Permeation of ions across the potassium channel: Brownian dynamics studies, Biophys. J. 1999, 77, 2517–2533. 41 S. H. Chung, T. W. Allen, S. Kuyucak, Conducting-state properties of the KcsA potassium channel from molecular and Brownian dynamics simulations, Biophys. J. 2002, 82, 628–645. 42 S. H. Chung, T. W. Allen, S. Kuyucak, Modeling diverse range of potassium channels with Brownian dynamics, Biophys. J. 2002, 83, 263–277. 43 S. H. Chung, S. Kuyucak, Ion channels: recent progress and prospects, Eur. Biophys. J. 2002, 31, 283–293. 44 J. Gullingsrud, K. Schulten, Gating of MscL studied by steered molecular dynamics, Biophys. J. 2003, 85, 2087– 2099.
45 A. E. Garcia, Large-amplitude nonlinear motions in proteins, Phys. Rev. Lett. 1992, 68, 2696–2699. 46 A. Amadei, A. B. M. Linssen, H. J. C. Berendsen, Essential dynamics of proteins, Proteins: Struct. Funct. Genet. 1993, 17, 412–425. 47 A. Amadei, M. A. Ceruso, A. Di Nola, On the convergence of the conformational coordinates basis set obtained by the essential dynamics analysis of proteins’ molecular dynamics simulations, Proteins Struct. Funct. Genet. 1999, 36, 419–424. 48 J. D. Faraldo-Gómez, L. R. Forrest, M. Baaden, P. J. Bond, C. Domene, G. Patargias, J. Cuthbertson, M. S. P. Sansom, Conformational sampling and dynamics of membrane proteins from 10-nanosecond computer simulations, Proteins: Struct. Funct. Bioinf. 2004, 57, 783–791. 49 V. Tozzini, Coarse-grained models for proteins, Curr. Opin. Struct. Biol. 2005, 15, 144–150. 50 I. Bahar, A. R. Atilgan, B. Erman, Direct evaluation of thermal fluctuations in proteins using a single-parameter harmonic potential, Fold. Des. 1997, 2, 173–181. 51 O. Keskin, R. L. Jernigan, I. Bahar, Proteins with similar architecture exhibit similar large-scale dynamic behavior, Biophys. J. 2000, 78, 2093–2106. 52 A. R. Atilgan, S. R. Durell, R. L. Jernigan, M. C. Demirel, O. Keskin, I. Bahar, Anisotropy of fluctuation dynamics of proteins with an elastic network model, Biophys. J. 2001, 80, 505– 515. 53 S. Haider, A. Grottesi, F. M. Ashcroft, M. S. P. Sansom, Conformational dynamics of the ligand-binding domain of inward rectifier K channels as revealed by MD simulations: towards an understanding of Kir channel gating, Biophys. J. 2005, 88, 3310–3320. 54 S. J. Campbell, N. D. Gold, R. M. Jackson, D. R. Westhead, Ligand binding: functional site location, similarity and docking, Curr. Opin. Struct. Biol. 2003, 13, 389–395.
References 55 V. Krishnamurthy, S. H. Chung, G. Dumont, Special issue on ion channels – bionanotubes, IEEE Trans. Nanobiosci. 2005, 4, 1–2. 56 C. G. Nichols, A. N. Lopatin, Inward rectifier potassium channels, Annu. Rev. Physiol. 1997, 59, 171–191. 57 F. Reimann, F. M. Ashcroft, Inwardly rectifying potassium channels, Curr. Opin. Cell Biol. 1999, 11, 503–508. 58 M. Nishida, R. MacKinnon, Structural basis of inward rectification: cytoplasmic pore of the G protein-gated inward rectifier GIRK1 at 1.8 Å resolution, Cell 2002, 111, 957–65. 59 C. E. Capener, I. H. Shrivastava, K. M. Ranatunga, L. R. Forrest, G. R. Smith, M. S. P. Sansom, Homology modeling and molecular dynamics simulation studies of an inward rectifier potassium channel, Biophys. J. 2000, 78, 2929–2942. 60 C. E. Capener, M. S. P. Sansom, MD Simulations of a K channel model – sensitivity to changes in ions, waters and membrane environment, J. Phys. Chem. B 2002, 106, 4543–4551. 61 J. F. Antcliffe, S. Haider, P. Proks, M. S. P. Sansom, F. M. Ashcroft, Functional analysis of a structural model of the ATP-binding site of the KATP channel Kir6.2 subunit, EMBO J. 2005, 24, 229–239. 62 P. Proks, C. E. Capener, P. Jones, F. Ashcroft, Mutations within the P-loop of Kir6.2 modulate the intraburst kinetics of the ATP-sensitive potassium channel, J. Gen. Physiol. 2001, 118, 341– 353. 63 I. H. Shrivastava, D. P. Tieleman, P. C. Biggin, M. S. P. Sansom, K+ vs. Na+ ions in a K channel selectivity filter: a simulation study, Biophys. J. 2002, 83, 633–645. 64 C. Domene, M. S. P. Sansom, A potassium channel, ions and water: simulation studies based on the high resolution X-ray structure of KcsA, Biophys. J. 2003, 85, 2787–2800. 65 S. Y. Noskov, S. Bernèche, B. Roux, Control of ion selectivity in potassium channels by electrostatic and dynamic
66
67
68
69
70
71
72
73
74
75
76
properties of carbonyl ligands, Nature 2004, 431, 830 – 834. Y. Zhou, R. MacKinnon, The occupancy of ions in the K+ selectivity filter: charge balance and coupling of ion binding to a protein conformational change underlie high conduction rates, J. Mol. Biol. 2003, 333, 965–975. M. J. Lenaeus, M. Vamvouka, P. J. Focia, A. Gross, Structural basis of TEA blockade in a model potassium channel, Nature Struct. Mol. Biol. 2005, 12, 454– 459. L. G. Cuello, E. Perozo, Fast gating transitions determined by dynamic rearrangements in the selectivity filter of KcsA, Biophys. J. 2002, 82, 853. C. Domene, A. Grottesi, M. S. P. Sansom, Filter flexibility and distortion in a bacterial inward rectifier K+ channel: simulation studies of KirBac1.1, Biophys. J. 2004, 87, 256–267. A. Grottesi, C. Domene, M. S. P. Sansom, Conformational dynamics of M2 Helices in KirBac channels: helix flexibility in relation to gating via molecular dynamics simulations, Biochemistry 2005, submitted. J. Holyoake, C. Domene, J. N. Bright, M. S. P. Sansom, KcsA closed and open: modeling and simulation studies, Eur. Biophys. J. 2003, 33, 238–246. P. C. Biggin, M. S. P. Sansom, Openstate models of a potassium channel, Biophys. J. 2002, 83, 1867–1876. Y. F. Shen,Y. F. Kong, J. P. Ma, Intrinsic flexibility and gating mechanism of the potassium channel KcsA, Proc. Nat. Acad. Sci. USA 2002, 99, 1949–1953. S. Trapp, S. Haider, M. S. P. Sansom, F. M. Ashcroft, P. Jones, Identification of residues contributing to the ATP binding site of Kir6.2, EMBO J. 2003, 22, 2903–2912. Y. Jiang,V. Ruta, J. Chen, A. G. Lee, R. Mackinnon, The principle of gating charge movement in a voltage-dependent K+ channel, Nature 2003, 423, 42– 48. K. J. Swartz, Towards a structural view of gating in potassium channels, Nature Rev. Neurosci. 2004, 5, 905–916.
265
266
10 Molecular Modeling and Simulations of Ion Channels: Applications to Potassium Channels 77 D. J. S. Elliott, E. J. Neale, Q. Aziz, J. P. Dunham, T. S. Munsey, M. Hunter, A. Sivaprasadarao, Molecular mechanism of voltage sensor movements in a potassium channel, EMBO J. 2004, 23, 4717–4726. 78 Q. X. Jiang, D. N. Wang, R. MacKinnon, Electron microscopic analysis of KvAP voltage-dependent K+ channels in an open conformation, Nature 2004, 430, 806–810. 79 S. B. Long, E. B. Campbell, R. MacKinnon, Crystal structure of a mammalian voltage-dependent Shaker family K+ channel, Science 2005, in press. 80 D. H. Hackos, K. J. Swartz, Mutations of a conserved proline in the inner helix of the pore domain of the Shaker K+ channel with altered gating properties, Biophys. J. 2000, 78, 398A. 81 A. J. Labro, A. L. Raes, I. Bellens, N. Ottschytsch, D. J. Snyders, Gating of Shaker-type channels requires the flexibility of S6 caused by prolines, J. Biol. Chem. 2003, 278, 50724–50731. 82 J. N. Bright, I. H. Shrivastava, F. S. Cordes, M. S. P. Sansom, Conformational dynamics of helix S6 from Shaker potassium channel: simulation studies, Biopolymers 2002, 64, 303–313. 83 J. N. Bright, M. S. P. Sansom, The Kv channel S6 helix as a molecular switch: simulation studies, IEE Proc. -Nanobiotechnol. 2004, 151, 17–27. 84 D. D. Camino, M. Holmgren,Y. Liu, G. Yellen, Blocker protection in the pore of a voltage-gated K+ channel and its structural implications, Nature 2000, 403, 321–325. 85 S. M. Webster, D. del Camino, J. P. Dekker, G. Yellen, Intracellular gate opening in Shaker K+ channels defined by high-affinity metal bridges, Nature 2004, 428, 864–868. 86 S. B. Long, E. B. Campbell, R. MacKinnon,Voltage sensor of Kv1.2: structural basis of electromechanical coupling, Science 2005, 309, 903–908 87 A. Grottesi, C. Domene, S. Haider, M. S. P. Sansom, Molecular dynamics simulation approaches to K channels: conformational flexibility and physiolo-
88
89
90
91
92
93
94
95
96
97
98
gical function, IEEE Trans. Nanobiosci. 2004, 4, 112–120. O. Beckstein, K. Tai, M. S. P. Sansom, Not ions alone: barriers to ion permeation in nanopores and channels, J. Am. Chem. Soc. 2004, 126, 14694–14695. L. Cuello, D. M. Cortes, E. Perozo, Structure and dynamics of the KvAP voltage sensor domain reconstituted in lipid bilayers, Biophys. J. 2005, 86, 458A. H. Takahashi, J. I. Kim, H. J. Min, K. Sato, K. J. Swartz, I. Shimada, Solution structure of hanatoxin1, a gating modifier of voltage- dependent K+ channels: Common surface features of gating modifier toxins, J. Mol. Biol. 2000, 297, 771–780. V. Ruta, R. MacKinnon, Localization of the voltage-sensor toxin receptor on KvAP, Biochemistry 2004, 43, 10071– 10079. D. Bemporad, C. L. Wee, Z. Sands, F. Blaney, A. Grottesi, M. S. P. Sansom, Localising a Kv channel toxin in a membrane with atomistic simulations, Biophys. J. 2005, submitted. M. C. Wiener, S. H. White, Structure of a fluid dioleoylphosphatidylcholine bilayer determined by joint refinement of X-ray and neutron diffraction data. III. Complete structure, Biophys. J. 1992, 61, 434–447. S. Y. Lee, R. MacKinnon, A membrane-access mechanism of ion channel inhibition by voltage sensor toxins from spider venom, Nature 2004, 430, 232– 235. H. C. Lee, J. M. Wang, K. J. Swartz, Interaction between extracellular hanatoxin and the resting conformation of the voltage-sensor paddle in Kv channels, Neuron 2004, 40, 527–536. J. A. Killian, G. von Heijne, How proteins adapt to a membrane-water interface, Trends Biochem. Sci. 2000, 25, 429– 434. Z. Sands, A. Grottesi, M. S. P. Sansom,Voltage-gated ion channels, Curr. Biol. 2005, 15, R44–R47. M. Laine, D. M. Papazian, B. Roux, Critical assessment of a proposed model of Shaker, FEBS Lett. 2004, 564, 257– 263.
References 99 L. G. Cuello, D. M. Cortes, E. Perozo, Molecular architecture of the KvAP voltage-dependent K+ channel in a lipid bilayer, Science 2004, 306, 491–495. 100 C. C. Gandhi, E. Y. Isacoff, Molecular models of voltage sensing, J. Gen. Physiol. 2002, 120, 455–463. 101 D. M. Starace, F. Bezanilla, A proton pore in a potassium channel voltage sensor reveals a focused electric field, Nature 2004, 427, 548–553. 102 A. Cha, G. E. Snyder, P. R. Selvin, F. Bezanilla, Atomic scale movement of the voltage-sensing region in a potassium channel measured via spectroscopy, Nature 1999, 402, 809–813. 103 K. S. Glauner, L. M. Mannuzzau, C. S. Gandhi, E. Y. Isacoff, Spectroscopic mapping of voltage sensor movement in the Shaker potassium channel, Nature 1999, 402, 813–817. 104 K. Brejc,W. J. van Dijk, R. V. Klaassen, M. Schuurmans, J. van der Oost, A. B. Smit, T. K. Sixma, Crystal structure of an ACh-binding protein reveals the ligand-binding domain of nicotinic receptors, Nature 2001, 411, 269–276. 105 S. Amiri, K. Tai, O. Beckstein, P. C. Biggin, M. S. P. Sansom, The a7 nicotinic acetylcholine receptor: molecular modeling, electrostatics, and energetics, Mol. Membr. Biol. 2005, 22, 151–162. 106 R. J. Law, R. H. Henchman, J. A. McCammon, A gating mechanism proposed from a simulation of a human a7 nicotinic acetylcholine receptor, Proc. Natl. Acad. Sci. USA 2005, 102, 6813– 6818. 107 A. Sali, T. L. Blundell, Comparative protein modeling by satisfaction of spatial restraints, J. Mol. Biol. 1993, 234, 779–815. 108 A. Fiser, R. Kinh Gian Do, A. Sali, Modeling of loops in protein structures, Prot. Sci. 2000, 9, 1753–1773. 109 T. Schwede, J. Kopp, N. Guex, M. C. Peitsch, SWISS-MODEL: an auto-
110
111
112
113
114
115
116
117
mated protein homology-modeling server, Nucleic Acids Res. 2003, 31, 3381– 3385. E. Lindahl, B. Hess, D. van der Spoel, GROMACS 3.0: a package for molecular simulation and trajectory analysis, J. Mol. Model. 2001, 7, 306–317. B. R. Brooks, R. E. Bruccoleri, B. D. Olafson, D. J. States, S. Swaminathan, M. Karplus, CHARMM: A program for macromolecular energy, minimisation, and dynamics calculations, J. Comput. Chem. 1983, 4, 187– 217. L. Kalé, R. Skeel, M. Bhandarkar, R. Brunner, A. Gursoy, N. Krawetz, J. Phillips, A. Shinozaki, K. Varadarajan, K. Schulten, NAMD2: Greater scalability for parallel molecular dynamics, J. Comput. Phys. 1999, 151, 283–312. N. A. Baker, D. Sept, S. Joseph, M. J. Holst, J. A. McCammon, Electrostatics of nanosystems: application to microtubules and the ribosome, Proc. Nat. Acad. Sci. USA 2001, 98, 10037–10041. M. E. Davis, J. D. Madura, B. A. Luty, J. A. McCammon, Electrostatics and diffusion of molecules in solution: simulations with the University of Houston Brownian dynamics program, Comput. Phys. Commun. 1991, 62, 187–197. A. Nicholls, R. Bharadwaj, B. Honig, GRASP: graphical representation and analysis of surface properties, Biophys. J. 1993, 64, 166–170. D. S. Goodsell, G. M. Morris, A. J. Olson, Automated docking of flexible ligands: applications of AutoDock, J. Mol. Recog. 1996, 9, 1–5. O. S. Smart, J. G. Neduvelil, X. Wang, B. A. Wallace, M. S. P. Sansom, Hole: A program for the analysis of the pore dimensions of ion channel structural models, J. Mol. Graph. 1996, 14, 354– 360.
267
269
Subject Index a atomic absorption spectrometry (AAS) 166–175, 202 – BK channel assay 170–171 – detection 169 – SK channel assay 171–172 – analysis 169 – cell loading 169 – Na+ channel assay 174 – ligand-gated channel assay 174 – voltage-gated K+ channel assay 172–173 – Cl– assay 174 access resistance 116 accessory (auxiliary) factors 81, 88 affinity tags – see expression tags alanine scanning – see mutagenesis scanning allosteric modulators 203 anion indicators 195 antibody fragments for crystallisation 229 apamin 172, 180 automated electrophysiology 13–17, 82, 97–99, 145–162 b BacMam 87, 93–95 batrachatoxin 200 benzodiazepine drugs 203 bicistronic – see IRES biophysics simulation 142 BK channel – see K+ channels Boltzmann analysis 124–125, 127 Brownian dynamics 244 Brugada syndrome 20 BSC1 channel 17–18 BTC 195, 203 c calcium (Ca2+) channels – toxicity 90
– fluorescence assays 199 – high throughput screening 196–199 calcium channel, voltage-gated (CaV) – assays 85–86, 196–197 – blockers 197 – cDNA instability 80 – mutagenesis 47–48 caged amino acids 72 capacitance compensation 7, 117, 147, 157 capsaicin 198 capsazepine 198 cDNA instability 80 cDNA sub-cloning 28–36 cell attached recording 9 Cerenkov counting 167 channel activation analysis 127 channel block 132 channel inactivation analysis 127 channel run-down 134, 151 chaperones 87–88, 95 Cheng-Prussof equation 175 chimeric channels 36–43, 50 chloride channels 203 – assay formats 174, 203–205 – Ca2+-activated, oocytes 18, 114 – CFTR 195, 203 – halide sensors 195, 203–205 concatamers 52–53 coumarin dyes 190–192 crystallisation, automation 230–232 crystallisation screens 227–228, 230 current clamp 112 current-voltage relationship 119, 122–127 cysteine scanning – see mutagenesis, scanning cystic fibrosis transmembrane regulator (CFTR) 80, 88, 195, 203–205 cytotoxicity 85–87, 90, 95, 101, 218
Expression and Analysis of Recombinant Ion Channels. Edited by Jeffrey J. Clare and Derek J. Trezise Copyright # 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim ISBN: 3-527-31209-9
270
Subject Index d defective trafficking 205 Del-Castillo-Katz model 131–132 deltamethrin 201 desensitisation 129 detergent solubilisation 219–223 dihydropyridines 197 dissociation constant 128 domain swaps – see mutagenesis, chimeras dwell time analysis 140–141 dynaflow 154 e electrochromic probes 188 electrophysiology, automated 145–162 electrophysiology, analysis 111–142 electrophysiology, oocytes 5–12 electropositioning 150 endogenous channels – mammalian cells 81 endogenous channels – oocytes 3, 79, 114 envelope protocols 130 episomal vectors 91–92 E-VIPR 201–202 excised patch 9–12 expression systems – for structural studies 216–219 – inducible 101–103 – oocyte 1–3, 113–115 – stable 96–103, 115, 157, 216 – transient 91–92, 115, 148 – viral 87, 92–95, 217 expression tags 29–30, 50–52, 226 f flame photometry 168 FLIPR 189, 196 FLIPR membrane potential kit 190 fluorescence assays 187–200 – Ca2+ dyes 194 – FLIPR 189–190, 196 – FRET 188–192 – ion indicators 194–205 – TRP channels 197–198 – Ca2+ channels 196–197 – Na+ channels 200 – polarisation 180–181 fluorescence microscopy 18–19 fluorescent ion indicators 194 fluorescent polarisation 180–81 fluorimetric imaging plate reader (FLIPR) 189 fluoro-ligands 180
FMP kit 190, 200–201 FRET probes 188–192 g GABAA receptor 203 gating 18–19, 122–135 – activation 18–19 – charge 135 – currents 135–136 – deactivation 120, 130 – inactivation 18–19 – Kir channels 250–251, 253–254 – Kv channels 130, 256–258, 260–261 – modifiers 132 – valence 124 gene fusions 50–52, 227 Giga Ohm seals 150 Goldman-Hodgkin-Katz equation 119, 123–125 h halide sensors 195, 203 hERG – see K+ channels high throughput screening – K+ channel assays 175–180, 201–203 – membrane potential probes 190–193 – scintillation proximity assay 178–180 – membrane potential probes 193 – state dependence 197 – automated electrophysiology 159–161 – Na+ channel assays 155, 200–201 – radioligand binding assays 175–180 Hill equation 128, 132 Hodgkin and Huxley 112, 117 homology modelling 243 host cell lines 81–85 – accessory factors 81, 83–85 – adherence 83 – endogenous channels 81 – neuroblastoma 83 – resting potential 85 i iberiotoxin 170 ICRAC 199 inclusion bodies 215 inducible expression – see expression systems intracellular solution 7, 114 ion channel block 121 ion conductance 118 ion flux assays 166–175 ion selectivity 118–121 ion-sensitive fluorescent probes 194–199
Subject Index IonWorks HT 82, 147, 150 IonWorks Quattro 97, 159–161 IRES (internal ribosome entry site) expression vectors 82, 96–101 isochronous tail protocol 125–126 k kinetic analysis 129–132, 156 l Lambert-Beer-Bouger Law 168 leak correction 116–117, 151 ligand-gated channels 127–129, 154, 174, 197–199 – calcium assays 198–199 – radioligands 177 – Cl-channel 203 – desensitisation 152 – electrophysiology analysis 126–129, 131 liquid junction potentials 119 m macropatch, 9–11 macroscopic kinetics 129–136 macroscopic recordings 117–128 Markov model 139–141 Markov process 112, 129, 139 membrane capacitance 7, 10, 116, 147, 157 membrane potential probes 188–193 – advantages & limitations 192, 204 – DIBAC 188–192 – FRET 188 – high throughput screening 190–193 – oxonol dyes 188–192 microfluidics 153 microspheres 179 minK 157 molecular dynamic simulation – Kir channels 249-254 molecular dynamic simulation – Kv channels 256, 258–260 molecular dynamics 244 MQAE 195 multiplexing 161 multi-subunit channels 52–53, 91, 93, 98, 100–101 mutagenesis – chimeras 36–43, 50 – gene fusions 50–52 – scanning 18, 45–49 – site directed 20, 43–49 – unnatural amino acid 60–65
n neurotoxins 175 nicotinic acetylchloine receptor 65–66 nimodipine 197 noise analysis 133–134 nonsense suppression 59-60 o oocytes 21 – dominant genotype 115 – voltage-clamp 5–11 – biochemical analysis 12–13 – nuclear injection 1 – recording configurations 114–115 – surface expression 115 – vitelline membrane 10, 12, 79, 115 – endogenous currents 3, 79, 114 – intracellular chloride 114 – isolation & injection 2–4, 6, 11, 114 – for mutagenesis studies 46–48, 63, 64, 68–69 – single channel analysis 11 open probability 117, 122 optical recording 192 Opus Xpress 16–17 oxonol dyes 188–192 p patch clamp electrophysiology 112–115, 145 PCR 31–33, 39–43 perforated patch clamp 150 permeability analysis 118–121 phase determination 232–234 planar array electrophysiology 145–161 – capacitance compensation 151 – cell preparation 148–149 – cell sealing & recording 149–150 – drug application 152–154 – quality control 156 – series resistance compensation 151 – equipment 147 – experimental methods & design 147–148 – population patch clamp 159–161 – theory 146–147 population patch clamp – see planar array electrophysiology pore analysis 118–121 post-translational processing 85–90 – folding 85, 87–88, 215–216 – proteolytic cleavage 85 potassium (K+) channels – AAS assays 169–173
271
272
Subject Index – automated electrophysiology 148–150, 154, 157 – BK channel 170–171 – hEAG2 channel 34 – hERG channel 67–72, 82, 89, 103, 153, 173, 175, 208 – high throughput screening 201–203 – KCNK2 (TREK-1) channel 85 – Kir channels 246–254 – KATP channels 202 – radioligand binding – SK channel 171–172 – structure 215, 218, 241–242, 247 – Kv (shaker) channels – – dynamic simulation 256, 258–260 – – gating 130, 256–258, 260–261 – – structure 215, 218, 254–256 – – mutagenesis 18–19, 38, 39–42, 52 – – voltage sensor 258–260 – – Kv7.1 (KCNQ1) 98–99, 157 potassium (K+) depolarisation 202 potassium (K+) indicators 195 power spectrum 133 principle component analysis (PCA) 246 purification 223–227 purification, affinity 226, 227 purification tags – see expression tags r radioactive ion flux assays 167–168 radioligand binding 175–181 – assay interference 179 – filtration assays 177–178 – oocytes 13 – scintillation proximity assay 178–180 – SK channel assay 178–179 receptor desensitisation 152 recovery from inactivation 130 rectification 123 redistribution probes 188–190 reporter genes 85, 95 reversal potential 118–120, 123 RNA injection 2–4, 113 Robocyte 14–15 s SBFI, 195 scanning mutagenesis – see mutagenesis scintillation proximity assay (SPA) 167, 178–180 selection marker 90, 100 selenomethionine labeling 232–233 Semliki forest virus (SFV) 92
series resistance correction 113–114, 147, 157 shaker – see K+ channels signal filtering 137-138 single channel – analysis 136–137 – oocytes 11–12 – recording 136 – conductance 123 – kinetics 138 – missed events 141 site-directed mutagenesis (SDM) – see mutagenesis SK channel – see K+ channels sodium channels, voltage gated (NaV) – activators & gating modifiers 200 – automated electrophysiology 160 – cDNA instability 80 – expression in mamalian cells 83–85 – expression in oocytes 8 – high throughput screening 174, 200–201 – indicators 195 – mutations 19–21 sodium green 195 spin labelling 72 SPQ 195 stable cell lines – see expression systems state transitions 112, 125–130 state-dependence 197, 132 structure factor sharpening 234 SUR1 202 t tags – see expression tags tail-current analysis 119–121, 125 taqman 81 TEA 136 thallium 201–203 trafficking 88 trafficking, assays 205–207 trafficking abnormalities 207 transient receptor channels 197 transient transfection – see expression systems TRP channels 197–198 two-electrode-voltage-clamp 5, 113 u use-dependence 158, 201-202 v variance-mean analysis 134–135 venfalerate 201 veratridine 200
Subject Index viral expression – see expression systems, viral vitelline membrane 10–12, 79, 115 voltage ion probe reader (VIPR) 196, 201– 202 voltage-clamp 112, 115–116 – oocytes 5–11 – control 114 – errors 113, 116 voltage-dependence 158, 197 voltage-dependent block 121–126, 132, 197 voltage-independent channels 197 voltage-sensor 18–19, 136, 258–260
w whole-cell recording 115, 146, 150 Woodhull model 121–123, 132 x Xenopus laevis – see oocytes y yellow fluorescent protein (YFP) z Z' value 156
95, 205
273