Ion Channels: Methods and Protocols
Ion Channels: Methods and Protocols
Ion Channels: Methods and Protocols
Ion Channels
Methods and Protocols
Second Edition
METHODS IN MOLECULAR BIOLOGY™
Series Editor
John M. Walker
School of Life Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Second Edition
Edited by
Nikita Gamper
School of Biomedical Sciences, University of Leeds, Leeds, UK
Editor
Nikita Gamper
School of Biomedical Sciences
University of Leeds
Leeds, UK
Every cell in our body contains a great variety and number of permeability pathways for
various organic and inorganic ions, water, metabolites, nutrients, and signaling molecules.
Maintenance and precise control of gating within these pathways are fundamental principles
of life as these underlie basic cellular functions such as communication, contractility, and
metabolism. This book focuses on the strategies, approaches, methods, and protocols for
studying a large family of proteins that form ionic channels in the plasma membrane and
intracellular membranes of cells. Like other permeability pathways of biological membranes,
ion channels are essential for life as they generate action potentials and regulate synaptic
transmission in neurons and muscle cells, underlie intracellular Ca2+ signalling, and
contribute to the charge separation across plasma membranes. Not surprisingly, genetic
deficiencies or acute deregulations of ion channel activity, trafficking, or degradation often
cause or contribute to severe human disorders (often called “channelopathies”) and
pathologies, e.g., arrhythmias, epilepsies, chronic pains, deafness, diabetes, and many
others. Conversely, ion channels are increasingly recognized as therapeutic targets.
Slightly over half a century ago, ion-selective channels in the plasma membrane were
postulated by Alan Hodgkin and Andrew Huxley as a purely theoretical concept. Now, at
the beginning of the twenty-first century, hundreds of ion channel genes are cloned, and
the currents conducted by many of them are exhaustively characterized. Some ion channels
are assigned with clear physiological functions while some are linked to human diseases,
and, for a handful of them, functional structures are proposed. This is tremendous prog-
ress, yet there is even more that we do not know. The aim of the present book is twofold:
firstly, using practical examples from the cutting-edge current research, we will take a look
back at the major methods and approaches that allowed us to progress to our current
understanding of ion channel function, structural design, and biological roles; and sec-
ondly, we will try to look forward and identify approaches that will lead us to future
discoveries.
This book will be of interest to specialists in academia and industry looking for specific
methodology in studying ion channels. It will be helpful for lecturers and advanced stu-
dents in the university classroom as well as for anyone interested in the state-of-the art
biomedical toolkit.
v
Contents
Preface. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xi
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 437
Contributors
xi
xii Contributors
Abstract
Molecular pain research is a relatively new and rapidly expanding field that represents advancement in
conventional pain research. One of the fundamentals of molecular pain involves the cloning of genes and
especially the ion channels specifically involved in nociceptive processing at the periphery and centrally.
A variety of approaches were used to isolate these critically important genes. Cloning of these genes
involved innovative strategies based on existing molecular approaches. This review will discuss well-utilized
cloning approaches and their exploitation in molecular pain research.
Key words Molecular pain research, Subtractive cloning, Differential cloning, Functional cloning,
Expression cloning, Homology cloning
1 Introduction
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_1, © Springer Science+Business Media, LLC 2013
3
4 Armen N. Akopian
3 Functional Cloning
3.1 Expression The expression cloning strategy to isolate genes of interest was
Cloning widely utilized in pain research. Notable discoveries of ATP-gated
P2X2 (40), capsaicin-/heat-gated TRPV1 (7), and menthol/cold-
gated TRPM8 channels (9) were made using expression cloning
strategies. To successfully accomplish expression cloning, three
parameters should be defined: (a) decision on the use of a particu-
lar expression system needs to be made; (b) agonists activating
proteins of interests have to be known; and (c) methods for detec-
tion of protein activation should be selected. A majority of expres-
sion cloning experiments were performed either by injection of
mRNA in Xenopus oocytes (40) or transfection of cell lines (such
as HEK, COS-7, or CHO) with expression cDNA libraries (7).
However, occasionally these direct approaches could not be
Cloning Pain Related Genes 9
Fig. 2 Schematic representation of expression cloning employed to isolate TRPV1 (aka VR1). Positive clones
are marked as dark circles on dishes
pool was again divided into ten cDNA pools, and expression
screening procedure was repeated (Fig. 2; Step 4). Step-by-step,
positive cDNA pool diminished (in increment of ten) and only one
capsaicin or menthol-sensitive clone remained. The capsaicin-sen-
sitive clone became the founding member of a novel gene family,
the TRPV-family (7), while the menthol-sensitive clone expressed
a novel member (TRPM8) of the TRPM-family (9).
3.2 Isolation of There are two separate groups of genetic diseases, which are char-
Mutated Genes Using acterized by alterations in pain perceptions in patients. One group
Genetic Mapping is congenital insensitivity to pain (CIP), which is a rare syndrome
with various clinical expressions, characterized by a dramatic
reduction (or ablation) in pain perception since birth. According
to some classifications, there are five major types of CIP (42).
CIP types I–IV are manifested by hereditary sensory and auto-
nomic degeneration (i.e., neuropathy; HSAN) involving the Aδ
and C-fiber nociceptors (43). However, some CIP patients show
normal morphology for nerve biopsies (44). Another group
Cloning Pain Related Genes 11
3.3 Cloning of Genes Channels are often presented in cells as multi-subunit proteins.
Encoding Interacting Some channel-associated proteins promote functional channel
Proteins expressions (50). Thus, NaV1.8 channel transfection into superior
cervical ganglion (SCG) neurons shows exactly the same biophysi-
cal properties as those observed in DRG neurons (23). In contrast,
the NaV1.8 channel cannot be effectively and properly expressed in
popular mammalian cell lines such as HEK and CHO, even in the
presence of auxiliary β-subunits (51). Therefore, it could be sug-
gested that NaV1.8 channel requires an adapter protein or auxiliary
subunit for correct functioning.
12 Armen N. Akopian
3.4 Biochemical Although fairly outdated now, in the early days of molecular cloning,
Approach to many classical channels were cloned using biochemical approaches.
Functional Cloning The biochemical approach is based on cloning genes encoding
Cloning Pain Related Genes 13
4 Homology Cloning
5 Conclusion
Acknowledgements
References
1. Akopian AN, Abson NC, Wood JN (1996) binding protein regulates neurite outgrowth.
Molecular genetic approaches to nociceptor Eur J Neurosci 15:281–290
development and function. Trends Neurosci 14. Woolf CJ, Costigan M (1999) Transcriptional
19:240–246 and posttranslational plasticity and the genera-
2. Muir WW 3rd, Woolf CJ (2001) Mechanisms tion of inflammatory pain. Proc Natl Acad Sci
of pain and their therapeutic implications. U S A 96:7723–7730
J Am Vet Med Assoc 219:1346–1356 15. Woolf CJ, Salter MW (2000) Neuronal
3. Berg P (2008) Meetings that changed the plasticity: increasing the gain in pain. Science
world: Asilomar 1975: DNA modification 288: 1765–1769
secured. Nature 455:290–291 16. Befort K, Costigan M, Woolf CJ (2001)
4. Berg P, Baltimore D, Brenner S, Roblin RO Differential gene expression—how to find new
3rd, Singer MF (1975) Asilomar conference analgesic targets. Curr Opin Investig Drugs
on recombinant DNA molecules. Science 2:396–398
188:991–994 17. Indo Y, Tsuruta M, Hayashida Y, Karim MA,
5. Akopian AN, Sivilotti L, Wood JN (1996) A Ohta K, Kawano T, Mitsubuchi H, Tonoki H,
tetrodotoxin-resistant voltage-gated sodium Awaya Y, Matsuda I (1996) Mutations in the
channel expressed by sensory neurons. Nature TRKA/NGF receptor gene in patients with
379:257–262 congenital insensitivity to pain with anhidrosis.
6. Chen CC, Akopian AN, Sivilotti L, Colquhoun Nat Genet 13:485–488
D, Burnstock G, Wood JN (2005) A P2X puri- 18. Drenth JP, Waxman SG (2007) Mutations in
noceptor expressed by a subset of sensory neu- sodium-channel gene SCN9A cause a spec-
rons. Nature 377:428–431 trum of human genetic pain disorders. J Clin
7. Caterina MJ, Schumacher MA, Tominaga M, Invest 117:3603–3609
Rosen TA, Levine JD, Julius D (1997) The 19. Fertleman CR, Baker MD, Parker KA, Moffatt
capsaicin receptor: a heat-activated ion channel S, Elmslie FV, Abrahamsen B, Ostman J,
in the pain pathway. Nature 389:816–824 Klugbauer N, Wood JN, Gardiner RM et al
8. Story GM, Peier AM, Reeve AJ, Eid SR, (2006) SCN9A Mutations in paroxysmal
Mosbacher J, Hricik TR, Earley TJ, Hergarden extreme pain disorder: allelic variants underlie
AC, Andersson DA, Hwang SW et al (2003) distinct channel defects and phenotypes.
ANKTM1, a TRP-like channel expressed in Neuron 52:767–774
nociceptive neurons, is activated by cold tem- 20. Weiss J, Pyrski M, Jacobi E, Bufe B, Willnecker
peratures. Cell 112:819–829 V, Schick B, Zizzari P, Gossage SJ, Greer CA,
9. McKemy DD, Neuhausser WM, Julius D Leinders-Zufall T et al (2011) Loss-of-function
(2002) Identification of a cold receptor reveals mutations in sodium channel Nav1.7 cause
a general role for TRP channels in thermosen- anosmia. Nature 472:186–190
sation. Nature 416:52–58 21. Mogil JS, Yu L, Basbaum AI (2000) Pain
10. Waldmann R, Champigny G, Bassilana F, genes?: natural variation and transgenic
Heurteaux C, Lazdunski M (1997) A proton- mutants. Annu Rev Neurosci 23:777–811
gated cation channel involved in acid-sensing. 22. Diatchenko L, Slade GD, Nackley AG, Bhalang
Nature 386:173–177 K, Sigurdsson A, Belfer I, Goldman D, Xu K,
11. Chen CL, Broom DC, Liu Y, de Nooij JC, Li Shabalina SA, Shagin D et al (2005) Genetic
Z, Cen C, Samad OA, Jessell TM, Woolf CJ, basis for individual variations in pain percep-
Ma Q (2006) Runx1 determines nociceptive tion and the development of a chronic pain
sensory neuron phenotype and is required for condition. Hum Mol Genet 14:135–143
thermal and neuropathic pain. Neuron 49: 23. Akopian AN, Souslova V, England S, Okuse K,
365–377 Ogata N, Ure J, Smith A, Kerr BJ, McMahon
12. Kim AY, Tang Z, Liu Q, Patel KN, Maag D, SB, Boyce S et al (1999) The tetrodotoxin-
Geng Y, Dong X (2008) Pirt, a phosphoinosit- resistant sodium channel SNS has a specialized
ide-binding protein, functions as a regulatory function in pain pathways. Nat Neurosci
subunit of TRPV1. Cell 133:475–485 2:541–548
13. Ravenall SJ, Gavazzi I, Wood JN, Akopian AN 24. Caterina MJ, Leffler A, Malmberg AB, Martin
(2002) A peripheral nervous system actin- WJ, Trafton J, Petersen-Zeitz KR, Koltzenburg
Cloning Pain Related Genes 17
M, Basbaum AI, Julius D (2000) Impaired cloning of a mouse submaxillary gland renin
nociception and pain sensation in mice lacking cDNA fragment. Proc Natl Acad Sci USA
the capsaicin receptor. Science 288:306–313 78:6367–6371
25. Kwan KY, Allchorne AJ, Vollrath MA, 38. Fodor SP, Rava RP, Huang XC, Pease AC,
Christensen AP, Zhang DS, Woolf CJ, Corey Holmes CP, Adams CL (1993) Multiplexed
DP (2006) TRPA1 contributes to cold, biochemical assays with biological chips.
mechanical, and chemical nociception but is Nature 364:555–556
not essential for hair-cell transduction. Neuron 39. Diogenes A, Patwardhan AM, Jeske NA,
50:277–289 Ruparel NB, Goffin V, Akopian AN, Hargreaves
26. Souslova V, Cesare P, Ding Y, Akopian AN, KM (2006) Prolactin modulates TRPV1 in
Stanfa L, Suzuki R, Carpenter K, Dickenson A, female rat trigeminal sensory neurons.
Boyce S, Hill R et al (2000) Warm-coding J Neurosci 26:8126–8136
deficits and aberrant inflammatory pain in mice 40. Brake AJ, Wagenbach MJ, Julius D (1994)
lacking P2X3 receptors. Nature New structural motif for ligand-gated ion
407:1015–1017 channels defined by an ionotropic ATP recep-
27. Zurborg S, Piszczek A, Martinez C, Hublitz P, tor. Nature 371:519–523
Al Banchaabouchi M, Moreira P, Perlas E, 41. Drew LJ, Wood JN, Cesare P (2002) Distinct
Heppenstall PA (2011) Generation and char- mechanosensitive properties of capsaicin-
acterization of an Advillin-Cre driver mouse sensitive and -insensitive sensory neurons.
line. Mol Pain 7:66–69 J Neurosci 22:RC228
28. Stirling LC, Forlani G, Baker MD, Wood JN, 42. Axelrod FB, Hilz MJ (2003) Inherited auto-
Matthews EA, Dickenson AH, Nassar MA nomic neuropathies. Semin Neurol 23:
(2005) Nociceptor-specific gene deletion using 381–390
heterozygous Nav1.8-Cre recombinase mice. 43. Axelrod FB, Gold-von Simson G (2007)
Pain 113:27–36 Hereditary sensory and autonomic neuropa-
29. Meunier A, Pohl M (2009) Lentiviral vectors thies: types II, III, and IV. Orphanet J Rare
for gene transfer into the spinal cord glial cells. Dis 2:39
Gene Ther 16:476–482 44. Cox JJ, Reimann F, Nicholas AK, Thornton G,
30. Ro LS, Chang KH (2005) Neuropathic pain: Roberts E, Springell K, Karbani G, Jafri H,
mechanisms and treatments. Chang Gung Mannan J, Raashid Y et al (2006) An SCN9A
Med J 28:597–605 channelopathy causes congenital inability to
31. Hedrick SM, Cohen DI, Nielsen EA, Davis experience pain. Nature 444:894–898
MM (1984) Isolation of cDNA clones encod- 45. Fischer TZ, Waxman SG (2010) Familial pain
ing T cell-specific membrane-associated pro- syndromes from mutations of the Nav1.7
teins. Nature 308:149–153 sodium channel. Ann N Y Acad Sci 1184:
32. Hedrick SM, Nielsen EA, Kavaler J, Cohen 196–207
DI, Davis MM (1984) Sequence relationships 46. Yang Y, Wang Y, Li S, Xu Z, Li H, Ma L, Fan
between putative T-cell receptor polypeptides J, Bu D, Liu B, Fan Z et al (2004) Mutations
and immunoglobulins. Nature 308:153–158 in SCN9A, encoding a sodium channel alpha
33. Chen CC, England S, Akopian AN, Wood JN subunit, in patients with primary erythermal-
(1998) A sensory neuron-specific, proton- gia. J Med Genet 41:171–174
gated ion channel. Proc Natl Acad Sci U S A 47. Bejaoui K, Wu C, Scheffler MD, Haan G,
95:10240–10245 Ashby P, Wu L, de Jong P, Brown RH Jr
34. Akopian AN, Wood JN (1995) Peripheral (2001) SPTLC1 is mutated in hereditary sen-
nervous system-specific genes identified by sory neuropathy, type 1. Nat Genet 27:
subtractive cDNA cloning. J Biol Chem 270: 261–262
21264–21270 48. Bejaoui K, Uchida Y, Yasuda S, Ho M,
35. Coste B, Mathur J, Schmidt M, Earley TJ, Nishijima M, Brown RH Jr, Holleran WM,
Ranade S, Petrus MJ, Dubin AE, Patapoutian Hanada K (2002) Hereditary sensory neurop-
A (2010) Piezo1 and Piezo2 are essential com- athy type 1 mutations confer dominant nega-
ponents of distinct mechanically activated cat- tive effects on serine palmitoyltransferase,
ion channels. Science 330:55–60 critical for sphingolipid synthesis. J Clin Invest
36. Waldmann R, Bassilana F, de Weille J, 110:1301–1308
Champigny G, Heurteaux C, Lazdunski M 49. Slaugenhaupt SA, Blumenfeld A, Gill SP,
(1997) Molecular cloning of a non-inactivating Leyne M, Mull J, Cuajungco MP, Liebert CB,
proton-gated Na + channel specific for sensory Chadwick B, Idelson M, Reznik L et al (2001)
neurons. J Biol Chem 272:20975–20978 Tissue-specific expression of a splicing muta-
37. Rougeon F, Chambraud B, Foote S, Panthier tion in the IKBKAP gene causes familial dysau-
JJ, Nageotte R, Corvol P (1981) Molecular tonomia. Am J Hum Genet 68:598–605
18 Armen N. Akopian
50. Isom LL (2001) Sodium channel beta subunits: 62. Xu H, Ramsey IS, Kotecha SA, Moran MM,
anything but auxiliary. Neuroscientist 7: Chong JA, Lawson D, Ge P, Lilly J, Silos-
42–54 Santiago I, Xie Y et al (2002) TRPV3 is a cal-
51. Okuse K, Malik-Hall M, Baker MD, Poon WY, cium-permeable temperature-sensitive cation
Kong H, Chao MV, Wood JN (2002) Annexin channel. Nature 418:181–186
II light chain regulates sensory neuron-specific 63. Kayano T, Noda M, Flockerzi V, Takahashi H,
sodium channel expression. Nature 417: Numa S (1988) Primary structure of rat brain
653–656 sodium channel III deduced from the cDNA
52. Vaghy PL, Striessnig J, Miwa K, Knaus HG, sequence. FEBS Lett 228:187–194
Itagaki K, McKenna E, Glossmann H, Schwartz 64. Clapham DE (2003) TRP channels as cellular
A (1987) Identification of a novel 1,4-dihy- sensors. Nature 426:517–524
dropyridine- and phenylalkylamine-binding 65. MacKenzie AB, Surprenant A, North RA
polypeptide in calcium channel preparations. (1999) Functional and molecular diversity of
J Biol Chem 262:14337–14342 purinergic ion channel receptors. Ann N Y
53. Hartshorne RP, Catterall WA (1984) The Acad Sci 868:716–729
sodium channel from rat brain. Purification 66. North RA, Barnard EA (1997) Nucleotide
and subunit composition. J Biol Chem receptors. Curr Opin Neurobiol 7:346–357
259:1667–1675 67. Toledo-Aral JJ, Moss BL, He ZJ, Koszowski
54. Fields S, Song O (1989) A novel genetic sys- AG, Whisenand T, Levinson SR, Wolf JJ, Silos-
tem to detect protein–protein interactions. Santiago I, Halegoua S, Mandel G (1997)
Nature 340:245–246 Identification of PN1, a predominant voltage-
55. Noda M, Takahashi H, Tanabe T, Toyosato dependent sodium channel expressed princi-
M, Furutani Y, Hirose T, Asai M, Inayama S, pally in peripheral neurons. Proc Natl Acad Sci
Miyata T, Numa S (1982) Primary structure of U S A 94:1527–1532
alpha-subunit precursor of Torpedo californica 68. Guler AD, Lee H, Iida T, Shimizu I, Tominaga
acetylcholine receptor deduced from cDNA M, Caterina M (2002) Heat-evoked activation
sequence. Nature 299:793–797 of the ion channel, TRPV4. J Neurosci
56. Noda M, Takahashi H, Tanabe T, Toyosato 22:6408–6414
M, Kikyotani S, Furutani Y, Hirose T, 69. Xu XZ, Moebius F, Gill DL, Montell C (2001)
Takashima H, Inayama S, Miyata T et al (1983) Regulation of melastatin, a TRP-related pro-
Structural homology of Torpedo californica tein, through interaction with a cytoplasmic
acetylcholine receptor subunits. Nature 302: isoform. Proc Natl Acad Sci USA
528–532 98:10692–10697
57. Noda M, Takahashi H, Tanabe T, Toyosato 70. Waldmann R, Champigny G, Lingueglia E, De
M, Kikyotani S, Hirose T, Asai M, Takashima Weille JR, Heurteaux C, Lazdunski M (1999)
H, Inayama S, Miyata T et al (1983) Primary H(+)-gated cation channels. Ann N Y Acad Sci
structures of beta- and delta-subunit precur- 868:67–76
sors of Torpedo californica acetylcholine recep- 71. Miljanich GP, Ramachandran J (1995)
tor deduced from cDNA sequences. Nature Antagonists of neuronal calcium channels: struc-
301:251–255 ture, function, and therapeutic implications.
58. Noda M, Ikeda T, Kayano T, Suzuki H, Annu Rev Pharmacol Toxicol 35:707–734
Takeshima H, Kurasaki M, Takahashi H, Numa 72. Waxman SG, Cummins TR, Dib-Hajj SD,
S (1986) Existence of distinct sodium channel Black JA (2000) Voltage-gated sodium chan-
messenger RNAs in rat brain. Nature nels and the molecular pathogenesis of pain: a
320:188–192 review. J Rehabil Res Dev 37:517–528
59. Noda M, Ikeda T, Suzuki H, Takeshima H, 73. Kubo Y (1994) Towards the elucidation of the
Takahashi T, Kuno M, Numa S (1986) structural-functional relationship of inward
Expression of functional sodium channels from rectifying K+ channel family. Neurosci Res
cloned cDNA. Nature 322:826–828 21(2):109–117
60. Tanabe T, Takeshima H, Mikami A, Flockerzi 74. Noda M, Shimizu S, Tanabe T, Takai T,
V, Takahashi H, Kangawa K, Kojima M, Kayano T, Ikeda T, Takahashi H, Nakayama
Matsuo H, Hirose T, Numa S (1987) Primary H, Kanaoka Y, Minamino N et al (1984)
structure of the receptor for calcium channel Primary structure of Electrophorus electricus
blockers from skeletal muscle. Nature 328: sodium channel deduced from cDNA sequence.
313–318 Nature 312:121–127
61. Tanabe T, Beam KG, Powell JA, Numa S 75. Papazian DM, Schwarz TL, Tempel BL, Jan
(2002) Restoration of excitation-contraction YN, Jan LY (1987) Cloning of genomic and
coupling and slow calcium current in dysgenic complementary DNA from shaker, a putative
muscle by dihydropyridine receptor comple- potassium channel gene from drosophila.
mentary DNA. Nature 336:134–139 Science 237:749–753
Cloning Pain Related Genes 19
76. Boulter J, Evans K, Goldman D, Martin G, 78. Smith GD, Gunthorpe MJ, Kelsell RE, Hayes
Treco D, Heinemann S, Patrick J (1986) PD, Reilly P, Facer P, Wright JE, Jerman JC,
Isolation of a cDNA clone coding for a Walhin JP, Ooi L et al (2002) TRPV3 is a tem-
possible neural nicotinic acetylcholine perature-sensitive vanilloid receptor-like pro-
receptor alpha-subunit. Nature 319: tein. Nature 418:186–190
368–374 79. Dib-Hajj SD, Tyrrell L, Black JA, Waxman SG
77. Caterina MJ, Rosen TA, Tominaga M, Brake (1998) NaN, a novel voltage-gated Na channel,
AJ, Julius D (1999) A capsaicin-receptor is expressed preferentially in peripheral sensory
homologue with a high threshold for noxious neurons and down-regulated after axotomy.
heat. Nature 398:436–441 Proc Natl Acad Sci USA 95:8963–8968
Chapter 2
Abstract
To delineate the function of a single ion channel subtype amongst the multitude that normally constitutes
a signalling pathway, it is often insightful to study the function or signalling of that one ion channel in
the absence of the others. Mammalian cell lines that do not normally express the gene of interest can be
manipulated to do so via plasmid DNA expression vectors. However, large and highly charged molecules
like DNA cannot passively diffuse through cell membranes. Therefore introducing nucleic acid into mam-
malian cells may involve introducing pores transiently into the cell membrane to allow the passage of
circular plasmid DNA. This is relatively easily carried out using cationic lipids that form liposomes around
the DNA and fuse with the cell membrane to introduce the DNA inside the cell. Alternatively, a highly
successful mechanism for introduction of DNA involves utilizing viral vectors. These take advantage of
the molecular mechanisms that viruses have evolved to efficiently transport their genome inside cells.
Lipid-based transfection techniques and adenoviral delivery of plasmids encoding large genes (such as ion
channel genes) for expression in mammalian cells are the focus of this chapter.
Key words Transient transfection, Stable transfection, Adenovirus, Transfection efficiency, Reporter
gene, Bicistronic vector
1 Introduction
Since many ion channels are regulated by diverse posttranslational
modifications (such as phosphorylation, glycosylation, disulfide
bond formation) that can differ between species, in order to study
mammalian ion channel function in a heterologous system, it is
beneficial to use a mammalian expression system. Mammalian
cells can be transfected with plasmid DNA encoding ion channel
genes. Plasmid DNA may either be introduced into the cells in a
transient fashion or may integrate into the cell genome. Cells that
have incorporated the plasmid DNA into their genome can be
selected via the use of antibiotic resistance genes to generate stable
clones that express a protein of interest. This chapter focuses on
the methods of transient and stable transfection by lipid-based
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_2, © Springer Science+Business Media, LLC 2013
21
22 Daunia Laurenti and Lezanne Ooi
1.1 Using Reporters The efficiency of transfection is a major concern for many applica-
to Analyze the tions and should be monitored using a reporter, such as a
Efficiency of fluorescent protein, e.g., enhanced green fluorescent protein
Transfection (eGFP), to confirm the transfer of nucleic acid to the cell. This can
be achieved by (1) transfecting two separate plasmids, one of
which encodes eGFP. This is the weakest of the three options pre-
sented here; although it is likely that if one plasmid was able to
transit through a particular cell membrane then both plasmids
were able to do so, it cannot be guaranteed (see Note 1).
(2) Generation of a fusion protein, using a plasmid such as pEGFP-
N1 or pEGFP-C1 (Clontech), in which your protein of interest is
tagged with eGFP (as one continuous protein). Careful consider-
ation should be given as to the position of the eGFP relative to the
protein of interest (i.e., N-terminal or C-terminal) since the
27 kDa eGFP may affect folding, localization, or function of your
target protein. (3) Another option would be to use a bicistronic
vector (generates a single mRNA that encodes two proteins), such
as pIRES2-eGFP (Clontech). This vector contains an internal
ribosomal entry site (IRES) sequence, a nucleotide sequence that
promotes translation initiation in the middle of an mRNA and
thus allows the simultaneous expression of the protein of interest
and eGFP (as a separate protein) from the same mRNA transcript.
With this method eGFP expression can be used to determine
efficiency of transfection and identify those cells that are positively
transfected (since all cells expressing the eGFP were successfully
transfected) without the eGFP affecting the folding of the protein
of interest. One other important consideration is that the expres-
sion of any reporter may affect the expression of other proteins or
the function of the cell, thus control transfections should be car-
ried out in which the eGFP (in the absence of your protein of
interest) is expressed (see Note 2).
1.2 Transient Versus The type of transfection method used, the size of the DNA plas-
Stable Transfection mid to be transfected, the amount of DNA transfected relative to
the amount of reagent, and the number of cells plated will all
affect the efficiency of transfection. Many ion channel genes are
relatively large, often leading to low-transfection efficiencies in
mammalian cells. Particularly in cases where the efficiency of trans-
fection is an issue, or to standardize experiments, you may want to
consider generating a stable cell line, in which the plasmid has
integrated into the genome of the cell. This means that the cell
line stably expresses your protein(s) of interest through successive
rounds of division. With transient transfections, expression of the
proteins will reduce as the plasmid is lost through cell division.
Expression Systems and Transfection 23
For this reason, cells are often used for experiments 24–48 h after
transfection (division approximately once every 24 h in many
mammalian cell lines). Although more time consuming initially,
generating stable cell lines can save a large amount of time and
money throughout the project and may also lead to more consis-
tent results. Plasmids such as pEGFP-N1 and pIRES2-GFP con-
tain a neomycin resistance gene for selection of stably transfected
cells. Selection of stable clones is achieved by incubating the cells
with G418 (also known as geneticin) over a period of weeks. Cells
that have not integrated the plasmid into the genome die follow-
ing extended exposure to G418. Individual cells expressing the
reporter gene can then be picked and diluted to single cells that
then undergo clonal expansion.
1.3 Adenoviral Viral vectors permit high levels of transgene expression in many
Delivery cell types, often without affecting cell viability. Since the adeno-
viral DNA does not integrate into the genome and is not repli-
cated during cell division, adenoviral delivery mediates transient
expression of transgenes. The adenoviral DNA vector contains a
portion of the viral genome plus a gene of interest cloned into a
multiple cloning site inserted into the early region 1A (E1A) of
the genome. Recombinant adenoviral vectors are generated using
this viral DNA vector and a packaging cell line (e.g., HEK293,
which have been stably transfected with the E1A region of the
adenoviral genome). Via a method adapted from He and col-
leagues (1), we have previously successfully prepared adenoviral
vectors to transiently express genes and reporters in cell lines
(2, 3) and primary cells, including neurons (4) and cardiac myo-
cytes (5). The gene of interest is cloned into the pAdTrack-CMV
vector, linearized, and transformed into an Escherichia coli strain
BJ5183 that expresses pAdEasy adenoviral vector (pAdEasy-1).
The recombinant adenoviral construct is then transfected into a
packaging cell line (e.g., HEK293) for virus production. During
the generation of the HEK293 cell line, normal human embryonic
kidney cells were transformed with adenovirus five DNA and this
cell line can be utilized for the propagation of adenoviral vectors.
Since adenoviruses are pathogenic it is important to use adenoviral
vectors in which certain genes are deleted so that the viruses are
unable to replicate after entering a cell. HEK293 can be safely used
in combination with the adenoviral vector pAdEasy, in which the
E1 and E3 genes have been deleted (1). Using this system viral
production and transgene expression can be monitored with the
reporter eGFP.
In this chapter, we outline the lipid-based methods used to
transfect mammalian cells transiently and following on from this
how to generate stable cell lines. We then outline how to prepare
adenovirus particles, which can be used to infect mammalian cells
to express genes of interest.
24 Daunia Laurenti and Lezanne Ooi
2 Materials
1. Cell culture medium: Dulbecco’s Modified Eagle Medium
(DMEM), 10% fetal bovine serum, 2 mM glutamine, strepto-
mycin (10 g/L), and penicillin (10 g/L); pre-warmed to
37°C.
2. Serum-free media (e.g., Opti-MEM).
3. Lipid transfection reagent [e.g., Lipofectamine 2000,
Lipofectamine LTX (Invitrogen)].
4. Cell culture plates.
5. Phosphate-buffered saline (PBS).
6. Sterile, round bottomed 14 mL centrifuge tubes.
7. Purified plasmid DNA (e.g., pIRES2-GFP, pAdTrack-CMV).
8. Cell line for transfection, e.g., HEK293, CHO, Neuro2A
For stable transfection:
9. G418 (Sigma).
10. 96-well cell culture plates.
For adenovirus preparation:
11. Restriction enzymes PmeI, PacI.
12. Bacterial strain BJ5183 containing pAdEasy-1.
13. Bacterial strain XL1 Blue.
14. Electroporation cuvettes, electroporator (option to use chemi-
cally competent cells and heat shock for electroporation).
15. Luria-Bertani Media (LB Broth: Tryptone 1.0%, Yeast Extract
0.5%, NaCl 1.0%) + kanamycin (10 μg/mL) or ampicillin
(25 μg/mL). For plates, 10% agar.
16. Tris–HCl pH 8.0 0.1 M.
17. Sodium deoxycholate 5%.
18. Magnesium chloride 2 M.
19. DNaseI (100 mg DNaseI in 10 mL of 20 mM Tris–HCl pH
7.4, 50 mM NaCl, 0.1 mM DTT, 0.1 mg/mL BSA, and 50%
glycerol).
20. Cesium chloride 1.5 g/mL (90.8 g CsCl and 109.2 g 10 mM
Tris pH 8).
21. Cesium chloride 1.35 g/mL (70.4 g CsCl and 129.6 g 10 mM
Tris pH 8).
22. Cesium chloride 1.25 g/mL (54 g CsCl and 146 g 10 mM
Tris pH 8).
23. Polyallomer tubes.
24. Dialysis buffer (10 mM Tris–HCl pH 8, 10% glycerol).
25. Slide-a-lyzer dialysis cassette.
Expression Systems and Transfection 25
3 Methods
3.1 Transient 1. Seed cells at a density of 1.5 × 106 cells per well of a 6-well plate
Transfection 24 h prior to transfection.
2. Dilute 10 μg of plasmid in 250 μL of Opti-MEM (mix A).
3. Dilute 30 μL of Lipofectamine 2000 in 250 μL Opti-MEM
(mix B).
4. Incubate at room temperature for 20 min.
5. Add mix A to mix B.
6. Incubate for 25 min at room temperature.
7. Wash the cells with PBS.
8. Add the mix A + B to cells.
9. Incubate for 1 min with gentle agitation.
10. Add 500 μL of Opti-MEM medium.
11. Incubate for 1 h at 37°C.
12. Add 1.5 mL of DMEM: 10% fetal bovine serum and glutamine
added—NO ANTIBIOTICS.
13. Incubate the cells for 24 h at 37°C 5% CO2.
14. Change medium or add 1 mL of DMEM: 10% fetal bovine
serum and glutamine WITH antibiotics (if required).
15. After 24 h change medium and check fluorescence under
microscope.
3.1.1 Optimization Cell lines that transfect easily and show high levels of transfection
of Transfection Efficiency efficiency (>80%) using this method include human embryonic
kidney 293 (HEK293) and Chinese hamster ovary (CHO). Other
cell lines that show reduced transfection efficiencies may be required
for use, e.g., the mouse neuroblastoma cell line Neuro2A. In this
case, it is extremely important to optimize transfection; some hints
for this are included below. We tested Lipofectamine LTX (which
is suggested by the manufacturer for high-efficiency transfection of
Neuro2A cells) and Lipofectamine 2000 (which is not specifically
recommended by the manufacturer for transfection of Neuro2A
cells). Based on the original protocol from both products two dif-
ferent transfections were tested. For each, a control transfection
was carried out with the “empty plasmid” pEGFP-N1 (4.7 kb)
(control plasmid) and the test plasmid containing the cloned gene
of interest in pEGFP-GOI (11 kb).
A. Lipofectamine 2000, following the manufacturer’s protocol
for general transfection of 1 × 106 murine cells in a 6-well plate.
The protocol was as above except for:
1. Transfection with 4 μg DNA and 10 μL Lipofectamine
2000.
26 Daunia Laurenti and Lezanne Ooi
Table 1
Transfection efficiencies vary with plasmid size and reagent used
Table 2
Transfection efficiencies vary with ratio of DNA:lipophilic reagent and incubation time
Plasmid size (kb) Incubation time DNA (mg) Lipofectamine (mL) Efficiency of transfection (%)
4.7 10 10 30 10–15
4.7 20 10 30 15–20
4.7 25 10 30 20–30
4.7 30 10 30 20–30
4.7 35 10 30 10–15
11 10 10 30 5–10
11 20 10 30 15–20
11 25 10 30 20–30
11 30 10 30 30–35
11 35 10 30 30–35
3.3.3 Amplification 1. Take the lysate from step 9 of the previous section and use to
and Purification of Virus infect 2 × 10 cm tissue culture dishes of HEK293.
(See Note 6) 2. After 7–10 days the majority of cells will be floating and
expressing GFP. Harvest free-floating cells by centrifugation.
Resuspend cell pellet in 1 mL PBS per 10 mL media.
3. Freeze/thaw three times in −70°C/37°C, vortexing each
time.
4. Pellet cell debris at 18,000 × g 10 min at 4°C.
5. Transfer supernatant to fresh tube and store at −70°C.
6. Use the second lysate to infect 20 × 10 cm dishes of HEK293.
Lyse cells as above.
7. Use the third lysate to infect 40 × 15 cm dishes of HEK293 and
harvest and purify adenoviral particles as below.
30 Daunia Laurenti and Lezanne Ooi
4 Notes
1. Consider increasing the amount of the plasmid encoding your
protein of interest compared to the plasmid encoding eGFP
(e.g., 2:1 or 3:1) to increase the likelihood of expression of
your protein of interest in eGFP-positive cells. This may be
especially important if the plasmid encoding your protein
of interest is much larger than the plasmid encoding eGFP.
For efficient DNA transfection into cells, preparation of high-
quality plasmid DNA is essential. It is best to prepare DNA
using a commercial kit based on ion exchange chromatogra-
phy. Removal of endotoxins during the plasmid isolation pro-
cedure also enhances transfection efficiency and many
companies supply plasmid isolation kits that will remove endo-
toxin contamination.
2. Transfection can result in changes in gene expression and con-
sequently in altered cellular responses and/or morphology. It
is always advisable to compare responses to control transfected
cells. In the case of transfection with liposomes, the transfec-
tion reagent on its own can affect the cell membrane (and any
membrane proteins, such as ion channels); it may therefore
also be necessary to include a “transfection reagent” control in
experiments, in which transfection reagent is added as per the
above protocol but in the absence of plasmid DNA.
3. The incubation time of the single mixes affects transfection
efficiency (6). The “empty plasmid” pEGFP-N1 (4.7 kb) (con-
trol plasmid) was negatively affected by long incubation times
but the larger test plasmid containing the cloned gene of inter-
est pEGFP-GOI (11 kb) showed improved efficiency by
increasing the incubation time. Lipofectamine volume was
chosen based on the amount of cells transfected and the toxic-
ity to cells (confluence of cells compared with the initial
confluence). Finally, decreasing the amount of plasmid reduced
efficiency.
4. With the use of G418 for selection of resistant clones it may be
important to titrate the antibody concentration depending on the
32 Daunia Laurenti and Lezanne Ooi
References
1. He T-C, Zhou S, da Costa LT, Yu J, Kinzler KW, 4. Mucha M, Ooi L, Linley JE, Mordaka P, Dalle
Vogelstein B (1998) A simplified system for gen- C, Robertson B, Gamper N, Wood IC (2010)
erating recombinant adenoviruses. Proc Natl Transcriptional control of KCNQ channel genes
Acad Sci U S A 95:2509–2514 and the regulation of neuronal excitability.
2. Ooi L, Belyaev ND, Miyake K, Wood IC, Buckley J Neurosci 30:13235–13245
NJ (2006) BRG1 chromatin remodeling activity 5. Bingham AJ, Ooi L, Kozera L, White E, Wood
is required for efficient chromatin binding by IC (2007) The repressor element 1-silencing
repressor element 1-silencing transcription factor transcription factor regulates heart-specific
(REST) and facilitates REST-mediated repression. gene expression using multiple chromatin-
J Biol Chem 281:38974–38980 modifying complexes. Mol Cell Biol
3. Johnson R, Gamblin RJ, Ooi L, Bruce AW, 27:4082–4092
Donaldson IJ, Westhead DR, Wood IC, Jackson 6. Dalby B, Cates S, Harris A, Ohki EC, Tilkins
RM, Buckley NJ (2006) Identification of the ML, Price PJ, Ciccarone VC (2004) Advanced
REST regulon reveals extensive transposable transfection with Lipofectamine 2000 reagent:
element-mediated binding site duplication. primary neurons, siRNA, and high-throughput
Nucleic Acids Res 34:3862–3877 applications. Methods 33:95–103
Chapter 3
Abstract
Individual types of ion channels and other membrane proteins are typically expressed only at low levels
in their native membranes, rendering their isolation by conventional purification techniques difficult.
The heterologous over-expression of such proteins is therefore usually a prerequisite for their purification
in amounts suitable for structural and for many functional investigations. The most straightforward expres-
sion host, suitable for prokaryote membrane proteins and some proteins from eukaryotes, is the bacterium
Escherichia coli. Here we describe the use of this expression system for production of functionally active
polytopic membrane proteins and methods for their purification by affinity chromatography in amounts up
to tens of milligrams.
Key words Membrane protein, Protein expression, Nonionic detergent, Protein purification, Affinity
chromatography, Size exclusion chromatography
1 Introduction
Twenty to thirty percent of genes in most living organisms encode
membrane proteins (1), which play critical roles in many aspects
of biology, ranging from cell signalling to nutrition. Moreover,
such proteins represent the targets of more than 50% of currently
used therapeutic drugs (2). Isolation of membrane proteins for
structural and functional analysis is therefore of great biological
and medical importance. However, individual types of ion channels,
transporters, and other membrane proteins typically represent
less than 1% of the total protein content of natural biological mem-
branes and this low level of expression hinders their purification.
Moreover, the nonionic detergents required to solubilize the
proteins can interfere with purification by conventional approaches,
such as ion-exchange chromatography, by shielding some of the
charged groups on the protein surface within the hydrophilic head
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_3, © Springer Science+Business Media, LLC 2013
33
34 Vincent G.L. Postis et al.
2 Materials
2.1 Reagents and 1. E. coli host strains: BL21-gold(DE3) (Stratagene), BL21 Star™
Buffers for Bacterial (DE3) (Invitrogen), and C43(DE3) (Lucigen Corporation),
Cell Culture and all harboring the plasmid pRARE2 (Novagen) if required
Membrane Preparation (see Note 1).
2. Expression constructs: These should contain the open reading
frame (ORF) for the protein of interest, bearing appropriate
N- and/or C-terminal affinity tags, under the control of a lactose-
inducible tac promoter (i.e., in a derivative of the vector
pTTQ18 (6)) or another suitable promoter (see Note 2).
3. 1 M isopropyl-β-D-thiogalactoside (IPTG): Dissolve 2.38 g
IPTG in a final volume of 10 mL H2O, sterilize by passage
through a 0.22 μm filter, and store at −20°C.
4. 100 mg/mL carbenicillin: Dissolve 500 mg carbenicillin in a
final volume of 5 mL H2O, sterilize by filtration through a
0.22 μm filter, and store in the dark at −20°C.
5. 30 mg/mL chloramphenicol: Dissolve 150 mg chlorampheni-
col in 5 mL 100% ethanol and store at −20°C.
6. 50× 5052: 25% (w/v) glycerol, 2.5% (w/v) glucose, 10% (w/v)
α-lactose monohydrate. Weigh 25 g glycerol into a beaker and
then add 73 mL H2O, 2.5 g glucose, and 10 g α-lactose. Stir
until dissolved (see Note 3) then sterilize by filtration through
a 0.22 μm filter.
Membrane Protein Purification 35
3 Methods
Design of constructs for expression of membrane proteins in E. coli
needs to take into account both the origin of the gene to be
expressed and the topology of protein. For example, for proteins
from organisms in which codon usage differs from that of E. coli
use of host strains harboring a plasmid encoding tRNAs for rarely
used codons can sometimes improve expression levels (7) and is
the method described here. Alternatively, the corresponding gene
can be produced synthetically, a process that allows not only codon
optimization but also control of other features, such as mRNA
secondary structure and the beneficial presence of rare codon
clusters, which may influence membrane protein biogenesis (8).
Knowledge of the topology of the membrane protein of interest
is also useful in deciding how the sequence should be tagged; if
the topology has not yet been established experimentally it can be
predicted with reasonable success using a range of algorithms such
as TOPCONS (9). For the majority of membrane proteins, which
have cytoplasmic C-termini (10), addition of a C-terminal octahis-
tidine tag typically allows good expression and affinity purification.
An added advantage of C-terminal tagging is that, in combination
with N-terminal sequencing, the detection of the tag by Western
blotting indicates that the protein produced is full length.
Co-purification of endogenous histidine-rich E. coli proteins, such
as AcrB (11), can be avoided by incorporating a protease cleavage
site between the tag and the membrane protein, and rechromato-
graphing the protein after protease treatment (12). However,
addition of an oligohistidine tag to a protein terminus normally
located on the periplasmic/extracellular side of the membrane
typically results in misfolding and/or reduced levels of expression
40 Vincent G.L. Postis et al.
3.1 Initial The following procedure is designed for rapid screening of a range
Optimization of host strains and culture media, using autoinduction (15), to find
of Protein Expression those in which expression is optimal.
1. Freshly transform E. coli strains BL21-gold(DE3), BL21 Star™
(DE3), and C43(DE3) with a pTTQ18-derived expression
construct encoding an affinity-tagged ORF of the protein of
interest.
2. Inoculate 2 mL samples of LBglucose in a 24 deep-well plate
with single colonies from plates streaked with the above
transformants. Seal the plate with breathable seal then incu-
bate overnight at 37°C in a humidified plate shaker at
1,300 rpm.
3. Inoculate 3 mL samples of M9auto, LBauto, and SBauto in a 24
deep-well plate with 15 μL samples of the overnight cultures.
Seal the plate with breathable seal then incubate for 24 h at
37°C in a humidified plate shaker at 1,300 rpm.
4. Transfer 400 μL samples of the autoinduced expression cul-
tures to a 96-well plate and pellet the cells by centrifugation at
2,000 × gav for 15 min at 4°C.
5. Pour off the medium and tap on clean dry tissue to remove any
remaining liquid.
6. Freeze the cell pellets at −80°C for 1 h then add 100 μL cell
lysis reagent per well and incubate on a plate shaker at 1,000 rpm
for 30 min at room temperature.
7. Take duplicate 5 μL samples for assay of protein concentration
by the bicinchoninic acid (BCA) assay (17).
Membrane Protein Purification 41
3.2 Further The small-scale culture and dot blotting procedures described
Optimization of Protein above should provide an initial idea of the best expression host and
Expression growth medium required to achieve the desired expression level of
the protein of interest. However, before embarking on large-scale
expression it is wise to determine the optimum time of induction
and to compare yields obtained using autoinduction with those
obtained using IPTG induction, by performing cultures in flasks as
follows (see Note 8):
1. Inoculate 50 mL LBglucose in a 250 mL baffled flask with single
bacterial colony from a freshly streaked plate and incubate
overnight at 37°C with orbital shaking at 200 rpm.
2. Measure the D600nm of the overnight culture and inoculate a
series of 250 mL baffled flasks, each containing 50 mL of the
appropriate autoinduction medium identified as detailed in
Subheading 3.1 and supplemented with antibiotics as appropri-
ate, to give a theoretical D600nm value of 0.05. Incubate at 37°C
with orbital shaking at 200 rpm for 24, 36, 48, and 60 h then
prepare cytoplasmic membranes as detailed in Subheading 3.3
for analysis of expression levels by Western blotting.
42 Vincent G.L. Postis et al.
3.4 Large-Scale Once the optimal combination of host strain, medium, induction
Expression and method, temperature, and time of induction has been determined,
Membrane Preparation expression can be scaled up either in flask culture or, if available, in
a fermenter (14) to enable sufficient material to be produced for
Membrane Protein Purification 43
3.6 Purification In cases where protein topology precludes the use of an oligohisti-
of Strep-Tagged dine affinity tag, we have successfully used an attached Strep-tag II
Proteins by SAC sequence for membrane protein purification (3). This approach
circumvents the problem of co-purification of the protein with
endogenous His-rich proteins, sometimes seen in IMAC
purifications (11), although the relatively low affinity of the tag for
the Strep-Tactin affinity resin can result in low yields for some pro-
teins, especially if they are monomeric. The following method is
used in our laboratory for purification of MPSIL0347, a homo-
logue of the mammalian bestrophin family of chloride channels
(19), encoded by the gene alr2987 from the cyanobacterium Nostoc
sp. PCC 7120.
All purification steps should be done on ice or at 4°C as
appropriate.
1. Solubilize membranes as detailed in step 1 of Subheading 3.5,
except using 2× SAC solubilization buffer and incubating with
gentle mixing overnight.
2. Separate the detergent-soluble from the insoluble protein by
ultracentrifugation as detailed in step 2 of Subheading 3.5.
3. Pre-equilibrate 1 mL of a 50% slurry of Strep-Tactin® Superflow®
resin slurry (i.e., 0.5 mL packed resin [see Note 27]) in a
50 mL plastic tube by washing twice with 10 mL SAC wash
buffer. Do this by gently inverting the tube then centrifuging
at 700 × gav for 5 min each time.
4. Add the supernatant from the ultracentrifugation step to the
resin and allow protein binding to occur for 1 h with gentle
mixing.
5. Centrifuge the resin slurry again at 700 × gav for 5 min, remove
and discard the supernatant (but first keep a small sample, e.g.,
1 mL, for SDS-PAGE/Western blotting to assess the amount
of MPSIL0347 that has remained unbound), then gently
resuspend the resin in 5 mL of SAC wash buffer.
6. Next pack the resin into a column of appropriate dimensions,
then wash the resin under gravity with 20 mL (i.e., 40 CV) of
SAC wash buffer, collecting the eluate for further analysis if
required.
7. Elute the bound protein by successive addition of ten 0.5 mL
volumes of SAC elution buffer, collecting 0.5 mL fractions.
Membrane Protein Purification 47
4 Notes
1. E. coli BL21 strains are used for protein expression because
they lack the proteases OmpT and Lon, so minimizing protein
degradation. (DE3) strains are employed because they carry a
chromosomal copy of the T7 RNA polymerase gene under the
control of the lacUV5 promoter. They are thus intended for
expression of proteins under the control of a T7 promoter,
Membrane Protein Purification 49
22. This fraction should contain just the IMAC wash buffer 2
displaced by the elution buffer from the column, i.e., it should
not contain eluted protein, but it should be kept just in case!
23. Most batches of imidazole from commercial sources absorb
light at 280 nm. Thus, the first fraction will have an anoma-
lously low absorption, because it will contain a mixture of wash
and elution buffer, but the subsequent fractions will contain
only elution buffer and so the latter will represent an appropri-
ate blank. Imidazole with a low A280nm can be purchased,
although it is of course more expensive.
24. In our experience, slow removal of imidazole by dialysis is
preferable to its rapid removal by passage down a desalting
column, because the latter can result in precipitation of the
protein. A low concentration of DDM, at or above the CMC,
is included in the dialysis buffer to prevent loss of detergent
and thus precipitation of the membrane protein.
25. DDM forms roughly spherical micelles of reported size ranging
from about 40–75 kDa. To prevent concentration of free deter-
gent, a concentrator with a molecular weight cutoff (MWCO) of
100 kDa must therefore be used, and even so some increase in free
detergent concentration is to be expected. Because the protein-
detergent micelle is much larger than the protein alone, there is
usually little loss of proteins with molecular masses of ³ 50 kDa.
26. The methods used to assess whether the purified protein
remains in its native folded state and retains function will of
course depend on the identity of the particular protein under
investigation. In some cases, function can be assessed directly
using the detergent-solubilized protein, an example being the
use of scintillation proximity assays to measure substrate bind-
ing to transporters (23). Alternatively, function can be assessed
following reconstitution of the protein into a proteoliposome
and measurement, for example, of the influx of radiolabelled
substrate in the case of a transporter (24). The latter type of
assay confirmed that PepTSo purified by the method described
retains its expected peptide transport activity. In the case of ion
channels, electrophysiological assays are possible if the protein
is reconstituted into giant unilamellar vesicles (25) or planar
lipid bilayers (see Chapter 8 of this book).
27. The binding capacity of Strep-Tactin® Superflow® resin for
Strep-tagged proteins is 50–100 nmol per mL, so 0.5 mL resin
should bind between 1 and 2 mg Strep-tagged MPSIL0347
(Mr 38543).
28. In the case of MPSIL0347, the SAC dialysis buffer was deter-
mined to be the optimum for maintaining protein stability,
using the assay described by Postis et al. (26). Once such an
optimum buffer has been identified by this assay, it can also be
Membrane Protein Purification 53
Acknowledgements
References
1. Wallin E, von Heijne G (1998) Genome-wide 8. Norholm MH, Light S, Virkki MT, Elofsson
analysis of integral membrane proteins from A, von Heijne G, Daley DO (2012)
eubacterial, archaean, and eukaryotic organ- Manipulating the genetic code for membrane
isms. Protein Sci 7:1029–1038 protein production: what have we learnt so far?
2. Rask-Andersen M, Almen MS, Schioth HB Biochim Biophys Acta 1818:1091–1096
(2011) Trends in the exploitation of novel 9. Bernsel A, Viklund H, Hennerdal A, Elofsson
drug targets. Nat Rev Drug Discov 10: A (2009) TOPCONS: consensus prediction of
579–590 membrane protein topology. Nucleic Acids
3. Rahman M, Ismat F, McPherson MJ, Baldwin Res 37:W465–W468
SA (2007) Topology-informed strategies for 10. von Heijne G (2006) Membrane-protein
the overexpression and purification of topology. Nat Rev Mol Cell Biol 7:909–918
membrane proteins. Mol Membr Biol 24: 11. Glover CA, Postis VL, Charalambous K,
407–418 Tzokov SB, Booth WI, Deacon SE, Wallace
4. Low C, Jegerschold C, Kovermann M, Moberg BA, Baldwin SA, Bullough PA (2011) AcrB
P, Nordlund P (2012) Optimisation of over- contamination in 2-D crystallization of mem-
expression in E. coli and biophysical characteri- brane proteins: lessons from a sodium channel
sation of human membrane protein and a putative monovalent cation/proton anti-
synaptogyrin 1. PLoS One 7:e38244 porter. J Struct Biol 176:419–424
5. Bill RM, Henderson PJ, Iwata S, Kunji ER, 12. Huysmans GH, Chan N, Baldwin JM, Postis
Michel H, Neutze R, Newstead S, Poolman B, VL, Tzokov SB, Deacon SE, Yao SY, Young
Tate CG, Vogel H (2011) Overcoming barri- JD, McPherson MJ, Bullough PA, Baldwin SA
ers to membrane protein structure determina- (2012) A urea channel from Bacillus cereus
tion. Nat Biotechnol 29:335–340 reveals a novel hexameric structure. Biochem J
6. Stark MJR (1987) Multicopy expression vec- 445:157–166
tors carrying the lac repressor gene for regu- 13. Hilf RJ, Dutzler R (2008) X-ray structure of a
lated high-level expression of genes in prokaryotic pentameric ligand-gated ion chan-
Escherichia coli. Gene 51:255–267 nel. Nature 452:375–379
7. Burgess-Brown NA, Sharma S, Sobott F, 14. Deacon SE, Roach PCJ, Postis VLG, Wright GS,
Loenarz C, Oppermann U, Gileadi O (2008) Xia X, Phillips SE, Knox JP, Henderson PJ,
Codon optimization can improve expression of McPherson MJ, Baldwin SA (2008) Reliable
human genes in Escherichia coli: a multi-gene scale-up of membrane protein over-expression
study. Protein Expr Purif 59:94–102 by bacterial auto-induction: from microwell
54 Vincent G.L. Postis et al.
plates to pilot scale fermentations. Mol Membr rRNA processing in vivo. Mol Microbiol 33:
Biol 25:588–598 188–199
15. Studier FW (2005) Protein production by 21. Baldwin SA, Baldwin JM, Lienhard GE (1982)
auto-induction in high density shaking cul- Monosaccharide transporter of the human
tures. Protein Expr Purif 41:207–234 erythrocyte. Characterization of an improved
16. Wagner S, Klepsch MM, Schlegel S, Appel A, preparation. Biochemistry 21:3836–3842
Draheim R, Tarry M, Högbom M, van Wijk 22. Engel CK, Chen L, Prive GG (2002) Stability
KJ, Slotboom DJ, Persson JO, de Gier JW of the lactose permease in detergent solutions.
(2008) Tuning Escherichia coli for membrane Biochim Biophys Acta 1564:47–56
protein overexpression. Proc Natl Acad Sci 23. Harder D, Fotiadis D (2012) Measuring sub-
USA 105:14371–14376 strate binding and affinity of purified mem-
17. Smith PK, Krohn RI, Hermanson GT, Mallia brane transport proteins using the scintillation
AK, Gartner FH, Provenzano MD, Fujimoto proximity assay. Nat Protoc 7:1569–1578
EK, Goeke NM, Olson BJ, Klenk DC (1985) 24. Xie H, Patching SG, Gallagher MP, Litherland
Measurement of protein using bicinchoninic GJ, Brough AR, Venter H, Yao SY, Ng AM,
acid. Anal Biochem 150:76–85 Young JD, Herbert RB, Henderson PJ,
18. Newstead S, Drew D, Cameron AD, Postis Baldwin SA (2004) Purification and properties
VL, Xia X, Fowler PW, Ingram JC, Carpenter of the Escherichia coli nucleoside transporter
EP, Sansom MS, McPherson MJ, Baldwin SA, NupG, a paradigm for a major facilitator trans-
Iwata S (2011) Crystal structure of a prokary- porter sub-family. Mol Membr Biol 21:
otic homologue of the mammalian oligopep- 323–336
tide-proton symporters, PepT1 and PepT2. 25. Varnier A, Kermarrec F, Blesneac I, Moreau C,
EMBO J 30:417–426 Liguori L, Lenormand JL, Picollet-D’hahan N
19. Hagen AR, Barabote RD, Saier MH (2005) (2010) A simple method for the reconstitution
The bestrophin family of anion channels: of membrane proteins into giant unilamellar
identification of prokaryotic homologues. Mol vesicles. J Membr Biol 233:85–92
Membr Biol 22:291–302 26. Postis VL, Deacon SE, Roach PC, Wright GS,
20. Lopez PJ, Marchand I, Joyce SA, Dreyfus M Xia X, Ingram JC, Hadden JM, Henderson PJ,
(1999) The C-terminal half of RNase E, which Phillips SE, McPherson MJ, Baldwin SA (2008)
organizes the Escherichia coli degradosome, A high-throughput assay of membrane protein
participates in mRNA degradation but not stability. Mol Membr Biol 25:617–624
Chapter 4
Abstract
Nucleofection is a transfection method used to introduce substrates such as cDNA plasmids into primary
cells or other cell lines. The method can be successfully applied to cells that are considered difficult to
transfect or suffer from low transfection efficiency as seen with traditional transfection techniques. Neurons
in primary cultures retain many properties of their in vivo state and therefore, in many instances, are con-
sidered better experimental systems than immortalized cell lines, thus becoming increasingly desirable cell
types for biomedical research. However, being post-mitotic, primary neuronal cultures are particularly
difficult to transfect using routine transfection reagents. There is therefore a growing need for the efficient
delivery of expression vectors into such neuronal cultures. In this chapter we will discuss the application of
nucleofection for the heterologous expression of genes in primary neuronal cultures. We also discuss the
advantage of this technique relative to other conventional methods, and describe a reliable method for
transfection of cultured rat dorsal root ganglion (DRG) and trigeminal (TG) neurons.
1 Introduction
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_4, © Springer Science+Business Media, LLC 2013
55
56 Hannah M. Kirton et al.
2 Materials
3 Methods
All procedures are carried out under sterile conditions using a class
II inflow hood (unless indicated otherwise).
3.1 Dissociation of 1. Thoroughly spray the procedure table and all the instruments
Rat DRG (TG) Neurons with 70% alcohol.
2. Dissection solution is prepared by dissolving dispase (10 mg/mL)
and collagenase type 1A (1 mg/mL) in HBSS. The solution is
then sterilized by filtering through a 0.22 μM syringe filter and
stored at 37°C (tissue culture incubator) until required.
3. Rat is sacrificed using isoflurane overdose (or similar approved
method).
4. The spine is removed and cut in two halves in the transverse
plane. Each part is then cut in the sagittal plane using fine scis-
sors and placed into the small cell culture dish filled with Ca2+-
and Mg2+-free HBSS and kept on ice (see Note 5 for TG
dissociation).
5. Pin one of the four spine sections to the Styrofoam pad using
hypodermic needles with spinal canal facing up. Remove spinal
cord with fine (Dumont #55) forceps.
6. Pool DRG from all levels with fine forceps and place into a
small cell culture dish filled with the ice-cold Ca2+- and Mg2+-
free HBSS. Repeat the procedure for the remaining sections of
the spine. Refer to Chapter 25 of this book for further advice
on DRG extraction.
7. Under sterile conditions DRG (or TG) are transferred into the
U-bottomed tube containing the dissection solution, and incu-
bated for 10–30 min (see Note 6) in a humidified incubator
(37°C, air supplemented with 5% CO2).
8. After 10–30 min gently triturate the cell suspension with a
sterile 1 mL Gilson pipette to dissociate the ganglia (avoid
excessive trituration as this may damage the neurons due to
mechanical stress) (see Note 7).
Nucleofection of Primary Neurons 59
Fig. 2 Transfection of primary sensory neurons using nucleofection. (a, b) TG neurons transfected with GFP-
tagged delta-opioid receptors (DOR-GFP). Bright-field image (a) shows two neurons one of which overex-
presses DOR-GFP visible in epi-fluorescence illumination (b). (c, d) DRG neurons transfected with Venus-tagged
neuropeptide Y (Venus-NPY). Bright-field image (c) shows two neurons and several satellite glial cells; one
neuron displays vesicularly localized Venus-NPY fluorescence as imaged in total internal reflection fluorescence
(TIRF) mode. Imaging was performed using Nikon TE200E swept-field confocal/TIRF microscope
4 Notes
spinal column, (2) cutting of the column into two halves in the
transverse plane, and (3) cutting of the column in the sagittal
plane. Ganglia can be pulled out using the forceps or can be
cut out using the forceps and fine vannas scissors (the latter
becomes necessary if older animals are used). Dorsal roots can
be cut out if necessary (especially for older animals). We do not
remove dorsal roots for preparation from 7-day-old rats.
4. Coated sterile cell culture coverslips need to be prepared before
dissociation, preferably on the previous day. Sterile coverslips of
desired size are placed into each well of a 6- or 24-well plate and
subsequently coated with solution of laminin (50 μg/μL) and
poly-D-lysine (0.001%) in HBSS. Plates are incubated in a
humidified incubator for 6 h, and washed thereafter three times
with sterile culture-grade water or sterile HBSS and allowed to
thoroughly dry before storing at 4°C. On the day of the dissocia-
tion plates are pre-warmed to 37°C in a humidified incubator.
5. In the case of TG dissociation, the animal is decapitated (after
isoflurane overdose). The cranium is opened at the back of the
head and the brain is removed. Trigeminal ganglia are excised
using vannas scissors. The rest of the procedure is similar for both
TG and DRG. Each trigeminal ganglion hosts over 50,000 neu-
ronal cell bodies (9) while each DRG neuron contains ~6,000
neuronal cell bodies (10). Therefore, one pair of TG provides an
equivalent quantity of neurons comparable to that of 17 DRG.
6. This step is crucial for a good-quality culture. The timing of
incubation depends on (1) age of the animal (the older the
animal, the longer incubation time is required) and (2) the
strength of the collagenase (this will decline with time of stor-
age). For a 7-day-old Wistar rat and a fresh collagenase the
recommended time of incubation is 10–12 min. After incuba-
tion ganglia are gently triturated using a 1 mL Gilson pipette
(for even gentler trituration a disposable pipette tip can be cut
at the very end of the tip to enlarge the opening) 1–3 times. If
ganglia are still compact and there is little sign of dissociation, a
further 5 min incubation is recommended (dissociation is char-
acterized by a “cloudy” appearance to the media and few large
clumps of cells). Alternatively it is suggested to gently “flick”
the bottom of the tube to encourage the cells to de-aggregate.
When dissociation of the ganglia becomes apparent, ganglia are
triturated ten times in a similar manner as previously described.
Important: there will be visible threads of undigested tissue—
do not try to achieve a completely homogenized suspension as
this will lead to the overexposure of neurons to the digestion
enzymes and, as a result, lead to significant cell death. Presence
of undigested tissue will not impair quality of culture.
7. Many DRG and TG neurons are mechanosensitive (that is, they
express mechanosensitive ion channels that excite neurons upon
mechanical stimulation). Excessive trituration may therefore
Nucleofection of Primary Neurons 63
Acknowledgements
References
1. Abogadie FC, Vallis Y, Buckley NJ, Caulfield M current in sensory neurons by exogenous
MP (1997) Use of antisense-generating plas- proteases: a signaling pathway mediating
mids to probe the function of signal transduc- inflammatory nociception. J Neurosci 28:
tion proteins in primary neurons. Methods Mol 11240–11249
Biol 83:217–225 8. Liu B, Linley JE, Du X, Zhang X, Ooi L,
2. Gamper N, Shapiro MS (2006) Exogenous Zhang H, Gamper N (2010) The acute noci-
expression of proteins in neurons using the ceptive signals induced by bradykinin in rat
biolistic particle delivery system. Methods Mol sensory neurons are mediated by inhibition of
Biol 337:27–38 M-type K+ channels and activation of Ca2+-
3. Gresch O, Engel FB, Nesic D, Tran TT, activated Cl− channels. J Clin Invest 120:
England HM, Hickman ES, Körner I, Gan L, 1240–1252
Chen S, Castro-Obregon S, Hammermann R, 9. Forbes DJ, Welt C (1981) Neurogenesis in the
Wolf J, Müller-Hartmann H, Nix M, trigeminal ganglion of the albino rat: a quanti-
Siebenkotten G, Kraus G, Lun K (2004) New tative autoradiographic study. J Comp Neurol
non-viral method for gene transfer into primary 199:133–147
cells. Methods 33:151–163 10. Arvidsson J, Ygge J, Grant G (1986) Cell loss
4. Gartner A, Collin L, Lalli G (2006) in lumbar dorsal root ganglia and transgangli-
Nucleofection of primary neurons. Methods onic degeneration after sciatic nerve resection
Enzymol 406:374–388 in the rat. Brain Res 373:15–21
5. Linley JE et al (2012) Reactive oxygen species 11. Ernsberger U (2009) Role of neurotrophin
are second messengers of neurokinin signaling signalling in the differentiation of neurons from
in peripheral sensory neurons. Proc Natl Acad dorsal root ganglia and sympathetic ganglia.
Sci U S A 109(24):E1578–E1586 Cell Tissue Res 336:349–384
6. Linley JE, Pettinger L, Huang D, Gamper N 12. McMahon SB, Bennet DLH, Bevan S (2006)
(2012) M channel enhancers and physiological Inflammatory mediators and modulators. In:
M channel block. J Physiol 590:793–807 McMahon SB, Koltzenburg M (eds) Wall and
7. Linley JE, Rose K, Patil M, Robertson B, Melzack’s textbook of pain. Elsevier Churchill
Akopian AN, Gamper N (2008) Inhibition of Livingstone, Edinburgh, pp 49–72
Chapter 5
Abstract
HIV-derived lentiviral vectors (LVV) are among the most commonly used gene delivery vehicles. Their
production in high quantities, which enables concentration of viral particles to high titers, is important for
their successful application in both biomedical research and gene therapy. LVV are produced by co-trans-
fection of three or more plasmids into a packaging cell line followed by several purification and concentra-
tion steps. Protocols currently in circulation differ from each other but the direct comparison of their
efficacy based on the published information is extremely difficult because more than one variable may be
changed and essential information may be omitted. We systematically evaluated three protocols and found
that one single modification described here, using FuGene® 6 in the co-transfection step, increase LVV
output almost 20 times as compared to the most commonly used calcium phosphate (CaPO4) transfection
technique. Unexpectedly FuGene® 6 was also much more efficient than another widely used reagent,
Superfect. Dependent on requirements, this permits a dramatic downscaling of the packaging stage of viral
production, and/or super-concentration of LVV to achieve stronger expression. For example we were able
to prepare ~25 μL of high titer LVV suitable for injections into rodent brain using a single T75 cm2 cell
culture flask of packaging cells. The same output would require up to 20 times more packaging cells and
reagents following conventional protocols. We illustrate the potential of our approach using transfection
of primary neuronal cultures with LVV expressing an optogenetic actuator channelrhodopsin-2. Our
observations should help to achieve reproducible production of high titer LVV for experimental and
potential therapeutic applications.
Key words Lentivirus, High titer, FuGene, Transfection, Gene delivery, Transduction, Neuron,
Channelrhodopsin
1 Introduction
Lentiviral vectors (LVV) are widely used for gene delivery in vitro
and in vivo. They are non-immunogenic, fast to produce and offer a
significant packaging capacity for complex expression cassettes (1).
The most commonly used LVV are derived from HIV, and pseudo-
typed with the VSV-G protein from vesicular stomatitis virus. In the
brain LVV can help to achieve high levels of transgene expression
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_5, © Springer Science+Business Media, LLC 2013
65
66 James Hewinson et al.
2 Materials
2.1 Components 1. Lenti-X™293 T Cell Line (Clontech) (see Notes 1 and 2).
for Lentiviral Vector 2. Full Media: Remove 55 mL Dulbecco’s Modified Eagle Medium,
Preparation high glucose, with GlutaMAX™ (D-MEM; Invitrogen,) from a
500 mL bottle. Add 50 mL heat inactivated fetal bovine serum
(FBS; Invitrogen) and 5 mL penicillin–streptomycin (10,000
High Titer Lentiviral Production 67
3 Methods
Fig. 1 Comparison of LVV production using FuGene® 6 with commonly used trans-
fection reagents. Multiple preparations of LVV EF1α-PLAP were made and titered
directly using the PLAP enzymatic assay in TE671 cells. Using the protocol described
here, we found that transfection with FuGene® 6 resulted in an LVV titer approxi-
mately sevenfold higher compared to that using Superfect, and about a 20 times
higher compared with CaPO4. n = 7, 14, and 15 for LVV EF1α-PLAP preps using
CaPO4, Superfect, and FuGene® 6, respectively. Error bars are SEM. Unpaired t-tests:
CaPO4 versus Superfect, p = 0.06, Superfect vs FuGene® 6, p = 0.0002, CaPO4 ver-
sus FuGene® 6, p = 0.0025. Out of 15 preparations with FuGene® 6 protocol, the
lowest titer obtained was 1.2 × 109, while the highest titer was 1.5 × 1010
High Titer Lentiviral Production 69
3.1.1 Alternative For comparative purposes the optimized protocol described above
Transfection Protocols was run in parallel with established methods, i.e., using calcium
(Day 2) phosphate or Superfect-based transfection of the packaging cell
line (outlined below), producing LVV EF1α-PLAP or LVV
CamKII-hChR2:eYFP.
Calcium Phosphate: Procedures for days 1 and 3–5 are identical to the protocol for
Based Transfection FuGene® 6 described above (steps 1, 2, 8–18 of Subheading 3.1).
(Adapted from (8)) The volumes used in the calcium phosphate protocol described
in (8) were scaled down for use in a single T75 flask containing
packaging cells.
Transfection of Cells (Day 2):
1. Approximately 17 h postinoculation cells should be 60–70%
confluent.
72 James Hewinson et al.
Superfect-Based Procedures for days 1 and 3–5 are identical to the protocol for
Transfection (Adapted FuGene® 6 described above (steps 1, 2, 8–18 of Subheading 3.1).
from Coleman et al. (9))
Transfection of Cells (Day 2):
1. Approximately 17 h postinoculation cells should be 60–70%
confluent.
2. To prepare transfection mix, in a 1.5 mL centrifuge tube, add
lentiviral plasmids (see FuGene 6 procedure) to 400 μL serum-
and antibiotic-free D-MEM.
3. Add 30 μL Superfect (Qiagen) and vortex for three-1 s pulses
to mix. Allow to stand for 15 min at room temperature with no
agitation.
4. Transfer transfection mix into 5.5 mL full media and gently
mix.
5. Aspirate media from Lenti-X 293 T cells and add transfection
mix to the flask.
6. Incubate cells for 6 h.
7. Aspirate transfection mix and replace with 6 mL full media.
3.2 Lentiviral Vector LVV EF1α-PLAP can be prepared in parallel with your virus of
Titration: Placental interest and be used in the titration procedure describe below,
Alkaline Phosphatase therefore giving an indication of the titer of your virus of interest.
Staining Alternatively your virus of interest can be titered using commer-
cially available quantitative real-time PCR or viral capsid p24
ELISA kits (for example from Clontech).
Seed Cells (Day 1, pm)
1. Dissociate TE671 cells (see Note 3) using trypsin-EDTA and
seed into a 12-well cell culture plate at a density of 175,000
cells/well in a final well volume of 1 mL full media.
Infect Cells with Lentiviral Vector (Day 2, am)
2. Prepare a hexadimethrine bromide working solution by adding
100 μL hexadimethrine bromide stock to 9,900 μL full
media.
High Titer Lentiviral Production 73
4 Notes
Acknowledgments
References
Abstract
Two-electrode voltage clamp (TEVC) is a conventional electrophysiological technique used to artificially
control the membrane potential (Vm) of large cells to study the properties of electrogenic membrane proteins,
especially ion channels. It makes use of two intracellular electrodes—a voltage electrode as Vm sensor and a
current electrode for current injection to adjust the Vm, thus setting the membrane potential at desired values
and recording the membrane current to analyze ion channel activities. Here we describe the use of TEVC
in combination with exogenous mRNA expression in Xenopus oocytes for ion channel recording.
Key words Voltage clamp, Membrane potential, Voltage electrode, Current electrode, Xenopus
oocytes, Ion channel
1 Introduction
Ion channels of cell membranes may be gated by the membrane
potential (Vm) and/or by specific chemicals. Those belonging to
the superfamily of voltage-dependent ion channels are gated by the
Vm directly, whereas others, classified as ligand-gated ion channels,
are gated by their binding with specific chemicals called ligands.
On the other hand, voltage-dependent ion channels can be subject
to the modulation by chemical factors such as neurotransmitters,
hormones, intracellular messengers or exogenous drugs; and some
ligand-gated ion channels (such as NMDA receptors) are also
affected by the change in Vm. Thus, to study the voltage dependent
characteristics of ion channels readily, or to distinguish between
the effects of Vm versus chemicals, an experimental procedure is
needed to control the Vm (i.e., to change Vm in a desired pattern or
set it at a desired level). This procedure, known as voltage clamp,
was first designed by Cole and Marmont and improved by Hodgkin,
Huxley and Katz for application to giant axons of squids as two-
electrode voltage clamp (TEVC) in late 1940s (1). It utilizes two
intracellular electrodes, one to monitor Vm and the other to inject
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_6, © Springer Science+Business Media, LLC 2013
79
80 Bingcai Guan et al.
current from the cell. The output of A1, the potential at which is
approximately equal to Vm, is connected to one input terminal of a
clamping amplifier (A2), which is a high gain differential amplifier
and compares Vm with the voltage command signal (Vc) applied to
the other input terminal. The resulting output voltage of A2 forces
a current proportional to the difference (e) between Vm and Vc,
usually by means of a voltage-controlled current source mecha-
nism, to flow through the current electrode (electrode 2) into the
cell. The direction of the current is decided by the polarity of the
voltage at the output of A2, always reducing e. As long as the gain
(m) of A2 is sufficiently high (³103), Vm can approach Vc very closely
and thus achieves Vm clamp. Meanwhile, the current that passes
through electrode 2 is measured (either upstream of electrode 2 or
at the virtual grounding circuit) as the membrane current, since it
counterbalances the Vm deviation the membrane current would
have caused (3, 4).
Below we exemplify the TEVC procedures with the recording
of Kv7.2/7.3 voltage-gated potassium channel currents.
2 Materials
All solutions are prepared using analytical grade reagents and ultra-
pure water (resistivity 18 MW·cm). Stock solutions and working
solutions are stored at 4°C, and are used within 1 month. Longer
time of storage is not advised. All waste materials, especially the
discarded glass electrodes that may cause injury, should be securely
disposed of in accordance with local laboratory regulations.
fine scissors if needed (see Note 7), and store them in an electrode
container.
3. Prepare 100 mL of 3 M KCl in a beaker, add 1 g of agarose,
and microwave the mixture until the agarose is dissolved. Place
the beaker containing agarose–KCl solution into a 80°C water
bath to avoid agarose gelation. Warm up the syringe and its
needle, with which the agarose–KCl solution is then injected into
the glass electrode (see Note 8). Store the filled glass electrodes
in a container (see Note 9).
4. Install a filled glass electrode onto the electrode holder (see
Note 10), which, with Ag/AgCl wire pre-installed, has been
secured to the headstage, the latter having been installed on a
micromanipulator. Adjust the position of the tip of the glass
electrode using the micromanipulator.
5. Measure the resistance of the electrode after its tip has been
lowered into the bath solution with the micromanipulator.
The electrode resistance should be in the range of 0.5–1 MW
(see Note 11). If the electrode resistance is not within this
range, adjust the parameters of the puller or modify the length
of the tip for truncation.
3 Methods
(All the following operations are carried out at room temperature).
3.1 Checkup of the 1. Check the perfusion system making sure that there is no block-
Recording System age within it. Load solutions for the experiment, adjust the
perfusion rate to about 2 mL/min (or to the value required for
the specific experiment to be performed).
2. Check the aspiration system and drain tubing to make sure that
they work properly so that the surface of the bath solution is
stable.
3. Check the arrangement of the voltage headstage and current
headstage installed on the micromanipulators, and make
sure that they are positioned at a wide angle of over 90° (see
Note 12).
4. Check the shielding and grounding of the apparatus around
the input of the headstages to avoid introduction of external
interferences, especially the power line frequency pickup (see
Note 13).
5. Adjust the dissecting microscope so that the bath chamber to
accommodate the oocyte is well within the visual field and
clearly seen.
6. Turn on the system: turn the power of the digitizer on, and
then turn on the power of the voltage clamp amplifier.
7. Configure the acquisition software and edit the voltage com-
mand protocol. For users of Clampex: open the software, click
Two-Electrode Voltage Clamp 85
“File” → “Set Data File Names” to designate the file name for-
mat and the folder to store the data to be recorded. Click
“Configure” → “Digitizer” to change the configuration status
from “Demo” to the digitizer being used. Create/edit the
voltage command protocol for stimulation of an oocyte.
3.2 Load the Oocyte Transfer a Xenopus oocyte expressing exogenous ion channel(s)
and/or receptor(s) of interest gently to the bath chamber using a
sucking pipette.
3.3 Observation and Steps described below are specific to Geneclamp 500B amplifier
Recording of Currents and the Clampex software; however, other TEVC hardware and
software packages will have analogous steps/commands.
1. Lower the tips of the voltage electrode and current electrode
into the bath using the micromanipulators, and position the
electrode tips near (but not in contact with) the surface of
the oocyte.
2. Zero offset potentials: press the buttons “ZERO V1” and
“ZERO V2”on the front panel of the amplifier, and the “DC
METERS” will read 0.
3. Measure electrode resistances: Press buttons “R1” and “R2”
respectively, and the resistance of the electrodes will be dis-
played. The most appropriate values are within 0.5–1 MW
(see Note 11).
4. Impale the oocyte: Under the dissecting microscope, press the
tips of the electrodes against the surface of the oocyte gently
using the micromanipulator, and impale the oocyte by
further advancing the electrode tip slowly until it pops in (first
do cell impalement with the voltage electrode and then
with the current electrode) (see Note 14) (Fig. 2) (7).
Fig. 2 The visual field of the stereomicroscope showing the Xenopus oocyte in
the bath chamber and the two glass electrodes
86 Bingcai Guan et al.
Fig. 3 Exemplary Kv7.2/7.3 mediated currents and the voltage clamp protocol
used to induce the currents
Two-Electrode Voltage Clamp 87
3.4 Turning Off the First close the software, then turn off the power of the digitizer
Recording System and and the amplifier respectively. Empty the remaining liquid in the
Cleaning the Setup perfusion system, and wash the system thoroughly with water
in case its tubing(s) may be obstructed. Empty the waste liquid
collected from the outlet of the vacuum pump.
4 Notes
1. OR2 is used to dissolve collagenase II for digestion of the layer
of follicular cells surrounding the oocyte. It does not contain
Ca2+ so as to avoid calcium overload in the oocyte during the
digestion.
2. Only after all reagents are completely dissolved forming a
clear solution by stirring the mixture should the pH adjustment
be done.
3. The osmolality of normal external solutions for oocytes is
~189 mOsm/kg.
4. Silver wires can be well coated with AgCl layer after immersion
in newly purchased or well sealed chlorine bleach for 20–30 min.
Using outdated or poorly preserved bleach may take longer
time or even produce no visible AgCl layer.
5. For recording large currents, a virtual ground circuit is gener-
ally used. Its voltage sensing electrode should be placed in
the bath near the cell surface, so that the bath potential at the
outside of the cell membrane can be clamped at zero without
being affected by the large current flowing through the bath
solution.
6. The result of the first RAMP test should be discarded because
it often significantly deviates from those of the following tests
due to some destabilizing factors.
The RAMP value should be determined for three times to
get a mean value as reference for HEAT value setting.
7. This is because the opening of the electrode tips should not be
too thin. Otherwise the electrodes would have a large resistance
after being filled (see Note 11).
8. For filling the electrodes, the agarose–KCl solution should not
occupy more than half the total electrode length in case the
solution may spill out of the electrode and spoil the holder.
Care should be taken not to have air bubbles left in the agarose
between the electrode tip and the AgCl coated silver wire.
88 Bingcai Guan et al.
References
1. Hodgkin AL, Huxley AF, Katz B (1952) techniques, 3rd edn. Axon Instruments, Inc.,
Measurement of current–voltage relations in the Foster City, CA
membrane of the giant axon of Loligo. J Physiol 5. Du X, Zhang H, Lopes C, Mirshahi T, Rohacs T,
116:424–488 Logothetis DE (2004) Characteristic interac-
2. Neher E, Sakmann B, Steinbach JH (1978) The tions with phosphatidylinositol 4,5-bisphosphate
extracellular patch clamp: a method for resolving determine regulation of Kir channels by diverse
currents through individual open channels in modulators. J Biol Chem 279:37271–37281
biological membranes. Pflugers Arch 6. Operation manual P-97 Flaming/Brown
375:219–228 micropipette puller. (2009). Sutter Instrument
3. Halliwell JV, Plant TD, Robbins J, Standen NB Company, Novato
(1994) Voltage clamp techniques. In: Ogden D 7. Bierwirtz A, Schwarz W (2007) Two-electrode
(ed) Microelectrode techniques: the Plymouth voltage-clamp (TEVC). http://www.biophys.
workshop handbook, 2nd edn. Company of uni-frankfurt.de/~wille/prakt/anleitungen/03_
Biologists, Cambridge, pp 17–35 elektrophys.pdf. Accessed 20 May 2012
4. Sherman-Gold R (ed) (2008) The axon guide 8. GeneClamp 500B theory and operation (2002)
for electrophysiology and biophysics laboratory Axon Instruments, Inc, Union City
Chapter 7
Abstract
The patch clamp technique revolutionized the study of ion channels and is considered the gold standard
of measuring ion channel activity, from the academic laboratory to industrial-scale drug screening.
This technique enables the study of ion channels, from single molecules up to the whole-cell ion channel
population, and in their native environment. Whilst the study of single protein molecular behavior is the
ultimate goal of biophysicists from all fields, this is a routine ability for ion channel specialists. This chapter
is aimed at helping the beginner to design patch clamp experiments and to obtain the fundamental micropi-
pette configurations with mammalian cells: cell-attached patch, whole cell, inside-out patch, and outside-
out patch.
Key words Patch clamp, Electrophysiology, Ion channel, Whole-cell, Single channel
1 Introduction
The patch clamp technique was originally developed in order to
record ionic currents from a tiny patch of membrane (1).
A micrometer-aperture glass micropipette was pressed against a
cell membrane and currents were recorded using a high-quality
electronic amplifier. This revealed unitary currents in the picoam-
pere range that fluctuated between discrete and quantal levels that
were interpreted as the opening and closing of single ion-perme-
able pores in the membrane. We now know these pores or perme-
ation pathways as the integral membrane proteins that we call ion
channels. The two defining ion channel properties are permeation,
which describes the types of ion that pass through the open pore
and the speed at which this occurs, and gating, which describes the
stimuli that cause the ion channel to spend more or less time with
the pore open. Sometimes ion channels are further classified
according to the ligands or drugs that activate or inhibit them.
Patch clamp techniques enable us to determine all of these proper-
ties from cell membranes and much more. Even with automated
chip-based or planar patch clamp equipment now available with
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_7, © Springer Science+Business Media, LLC 2013
91
92 Jonathan D. Lippiat and David C. Wrighton
2 Materials
2.4 Patch Clamp The manufacturers of the following items are given as examples
Apparatus and are those presently in use in our laboratory. The whole sys-
tem should be assembled and tested thoroughly with the model
cell and then with micropipette and bath. Background noise and
50 Hz electrical noise should be eliminated to below 1 pA
(see Note 5).
1. Antivibration (air) table (e.g., Linos, Thorlabs) and compressed
air/gas supply.
2. Faraday cage. This could be supplied with the antivibration
table or custom-made in-house if workshop facilities are
available.
3. Inverted microscope (e.g., Nikon TE-2000) equipped with
4×, 10×, and 40× objective lenses (see Note 6).
4. Micromanipulators (e.g., Luigs-Neumann; microscope-specific
mount and fixings can usually be provided by the manufacturer
or fabricated in-house).
5. Patch clamp amplifier with headstage and micropipette holder
(e.g., HEKA EPC 10).
6. Model cell (provided with amplifier).
7. Reference electrode: Ag/AgCl pellet (e.g., World Precision
Instruments) soldered to thin wire and headstage pin
connector.
8. Benchtop or mini-tower computer, fitted with interface if sup-
plied with patch clamp amplifier, and installed with acquisi-
tion (e.g., Patchmaster, HEKA) and data analysis (e.g.,
Fitmaster, HEKA) software. If amplifier does not directly con-
nect to the computer then digital-analogue interface hardware
and software is required to be installed (e.g., Axon Digidata,
Cambridge Electronic Design, National Instruments, etc.) to
allow two-way communication between computer and
amplifier. The computer-based acquisition system also acts as
an oscilloscope.
9. Perfusion system: gravity-fed system fabricated in-house from
syringe reservoirs, silicone tubing, two- or three-way stopcocks,
and manifolds, or specialist system purchased from supplier
(e.g., Automate Scientific, Biologic, etc.).
10. Recording chamber or mount for 35 mm Petri dish (fabricated
in-house or supplied by World Precision Instruments, Warner
Instruments, ALA Scientific Instruments, etc.).
11. 1 mL syringe, three-way syringe stopcock, and silicone tubing
to attach to micropipette holder side-port to provide suction/
pressure to the micropipette.
Patch Clamp Techniques 95
3 Methods
3.1 Cell Culture All procedures should be carried out using aseptic technique and in
and Preparation a sterile vertical flow cabinet suitable for mammalian cell culture.
Procedures should also conform to local and national rules for
disposal of contaminated microbiological or genetically modified
material waste.
1. Aspirate culture medium from the culture flask.
2. Wash cells twice with PBS and harvest by adding trypsin-EDTA
dissociation solution. 200 μL of trypsin-EDTA solution in one
25 cm2 flask should suffice.
3. Rock the flask so that the dissociation solution covers the entire
surface area and observe cells on an inverted microscope.
4. Once cells appear rounded and become loosely adherent, add
5 mL of culture medium and transfer cell suspension into a
sterile plastic centrifuge tube (e.g., 15 mL).
5. Centrifuge cell suspension for 1 min at 200 × g.
6. Aspirate supernatant and resuspend in culture medium, diluting
if necessary.
7. Seed cells into a fresh 25 cm2 flask in a final volume of 6 mL for
further propagation of the cell line and so that the cells have
been diluted five- to tenfold.
8. Add 2 mL of cell suspension to each of the required number
of 35 mm Petri dishes and incubate overnight (see Note 1).
3.2 Micropipette 1. If required, cut capillary glass into shorter lengths (e.g.,
Fabrication and Filling 150 mm capillaries should be cut into two approximately equal
lengths). The capillary ends can be gently fire polished over a
Bunsen flame to prevent damage to the electrode wire.
2. Optimize pipette puller settings (heat, pulling distance, force)
for the specific device and for the type of glass capillary. For
micropipettes fabricated from thin-walled glass and intended
for whole-cell recording a final resistance as measured during
the experiment should typically be 2–3 MΩ, whilst patch (cell-
attached or excised) micropipettes are typically higher at
5–8 MΩ and are pulled from thick-walled glass. This resistance
is proportional to the cross-sectional area of the micropipette
tip and should be optimized to the cell type or size and channel
density (see Note 7).
3. Polish the tip of the micropipette using the microforge. This
enables fine control over the size of the aperture and makes a
clean, smooth, tip surface that enhances seal formation.
4. Fill the 5 mL syringe with the solution to be loaded into the
micropipette and attach a filter disc. For most experiments this
96 Jonathan D. Lippiat and David C. Wrighton
3.3 Cell-Attached This is the starting point for each of the different patch clamp
Configuration configurations described in this chapter. It can also be used to
monitor currents across the enclosed patch of membrane in the
intact cell. This is a particularly useful technique when determining
whether particular ion channels are regulated by cytosolic second
messengers, such as Ca2+ or cAMP, since the channels enclosed by
the patch pipette would be isolated from membrane-impermeant
receptor agonists applied by bath perfusion.
1. Check that the electrode wire in the micropipette holder has
an adequate coating of silver chloride otherwise the offset volt-
age will drift (see Note 8).
2. Replace tissue culture medium with extracellular solution and
mount Petri dish with cells onto microscope stage (or place
coverslip with adherent cells into recording chamber).
3. Position reference electrode directly into the dish or connect
by means of an agar bridge (see Note 9).
4. Fill a polished patch microelectrode with pipette solution
(see Subheading 3.2) and mount into the micropipette holder,
ensuring an airtight seal.
5. Position the micropipette above the bath solution and in the
field of view on the microscope.
6. Using the 1 mL syringe and tubing connected to the side-port
of the microelectrode holder, apply positive pressure. As an
example, depress the syringe plunger from 0.2 to 0.1 mL. This
prevents flow of debris from the bath into the micropipette
that might prevent seal formation.
7. Lower the micropipette into the bath solution and position the
tip in the proximity of the cells (see Note 10).
8. Zero the patch clamp amplifier using the control software
(“LJ,” in Patchmaster), or DC-offset if a manual amplifier (see
Note 11).
9. Apply a 1 mV pulse and observe or calculate the pipette resis-
tance (Fig. 1a).
10. Under high power magnification (40× objective) position the
micropipette so that the tip is touching the cell membrane.
11. Using the 1 mL syringe and/or stopcock, release the positive
pressure and apply negative pressure to the micropipette (e.g.,
retract syringe plunger by 0.1 mL). This should increase the
resistance of the seal between the micropipette and the cell
membrane, which will be reported by the control software
Patch Clamp Techniques 97
3.5 Outside-Out This configuration enables the recording of current from a small
Patch Configuration patch of membrane, isolated from the cell, with the extracellular
domains of the channel exposed to the bath. This permits the study of
Patch Clamp Techniques 99
3.6 Inside-Out Patch This configuration enables currents to be recorded from a small
Configuration patch of membrane, isolated from the cell, but with the intracellu-
lar-facing domains of the channel exposed to the bath. Here, uni-
tary or small numbers of channels can be studied whilst enabling
the application of intracellular reagents to the bath, e.g., nucle-
otides or Ca2+, when studying ATP- or Ca2+-sensitive potassium
channels, respectively.
1. Fabricate higher-resistance micropipettes from thick-walled
capillary glass and select the extracellular solution as the pipette
solution (see Subheading 3.2).
2. With the cells bathed in intracellular solution, obtain the cell-
attached configuration (see Subheading 3.3). If this is detri-
mental to the experiment it is possible to first bathe the cells in
extracellular solution, but start perfusing intracellular solution
once a gigaohm seal is formed and prior to patch excision.
3. Set the pipette potential to the intended holding potential
(e.g., −70 mV). The polarity may need to be reversed (i.e.,
+70 mV) if this has not been done when establishing the
100 Jonathan D. Lippiat and David C. Wrighton
3.7.1 Voltage-Gated 1. Select a holding potential that is below the activation threshold
Channels (Fig. 2) of the channel of interest, e.g., −80 mV.
2. Use square-wave voltage pulse protocols.
3. The duration of the voltage pulse should be sufficient for the
channel to reach steady state (e.g., 50 ms is sufficient for most
Kv channels).
4. If the channel undergoes inactivation, consider the use of
hyperpolarizing pre-pulses.
5. If the channel undergoes inactivation, ensure that the inter-pulse
interval is long enough to allow full recovery from inactivation,
unless you intend measuring time- and voltage-dependent
recovery from inactivation.
Patch Clamp Techniques 101
3.7.2 Voltage- 1. Select a holding potential close to the channel reversal poten-
Independent or Weakly tial (i.e., no net current ion flow).
Rectifying Channels 2. Do not use leak subtraction; voltage-independent currents are
considered by these protocols and amplifier functions as leak.
3. Use either voltage pulses or voltage ramp protocols and avoid
voltages that activate voltage-gated channels that might also be
present in the cell membrane.
4. There is little benefit in using a wide range (i.e., >50 mV) of
test voltages; measure channel activity as conductance from a
narrow voltage range through the reversal potential.
3.7.3 Ligand-Gated Ion 1. Holding potentials close to the native resting membrane
Channels potential (e.g., −60 mV) are often selected in order to deter-
mine whether the ligand-gated channel activation is excitatory or
inhibitory, whereby the ligand evokes either an inward or out-
ward current, respectively.
2. Research the ligand/agonist concentration range and select a
concentration that will stimulate the channel of interest, but not
affect other types of channel that may be present in the cell.
3. Take into consideration a possibility that the ligand or agonist
causes desensitization of the channel, this will determine for
how long the ligand is applied.
4. Low-bandwidth continuous recordings are often used to study
macroscopic currents.
5. Ligand-gated channels are usually voltage-independent so do
not use automated leak-subtraction methods.
6. Determine the reversal potential by applying voltage pulses or
a voltage ramp whilst the channels are exposed to ligand/
agonist.
4 Notes
1. 35 mm Petri dishes are convenient for culture of the cells and
can be placed directly onto an inverted microscope for electro-
physiological study. Alternatively, cells can be seeded and cul-
tured on glass coverslips, which can be transferred into the
electrophysiological recording chamber using forceps.
Depending on the cell type, the cell adherence can be improved,
if required, by purchasing pretreated coverslips or by preparing
substrates coated with poly-d-lysine, laminin, collagen, etc.,
depending on the cell type. To enable patches to be excised
successfully for inside-out and outside-out patch recording cell
adherence may need to be optimized. Alternately, cells could
be grown into clusters by seeding at higher density and cultur-
ing for >2 days so that adherence is improved via a greater
surface area contact between the substrate and cell monolayer.
This, however, is not desirable for whole-cell recording from
cells that form electrical synapses with neighboring cells via gap
junctions, and these include HEK293 cells.
2. Some ion channels require additional substances in order to be
significantly active, e.g., nucleotides or Ca2+. If the current of
interest is overwhelmed by other ionic conductances then the
conditions could be optimized to enhance the current of inter-
est or by reducing currents through other channels by including
impermeant ions or drugs.
3. The pH of the solutions should be adjusted using acid or base
that will add cations/anions that are already in high concentra-
tion, i.e., NaOH for Na+-rich extracellular solutions, and KOH
for K+-rich intracellular solution. Likewise, HCl is used if the
solution is required to be adjusted to more acidic pH. If the
final concentrations of certain ions are of particular concern,
e.g., [K+] in intracellular solution, then either the pH could be
104 Jonathan D. Lippiat and David C. Wrighton
15. The resistance of the cell membrane and the resistance between
the micropipette and the cell membrane are in series and form a
voltage divider. The current flow is the same throughout the
circuit, but the voltage (the pipette potential) across the two is
shared between each resistance according to its proportion of the
total resistance. The series resistance (Rs, a few MΩ) is usually
much lower than the membrane resistance when very few chan-
nels are active (Rm, a few 100 MΩ) and so the voltage across
the membrane (Vm) is very close to the pipette voltage (Vp):
Vm = Vp × (Rm/(Rm + Rs)).
If Rm decreases, i.e., through the opening of many ion
channels then the contribution of Rm to the total resistance
(Rm + Rs) decreases along with Vm. The application of Ohm’s
law, Vs = I × Rs, is a quick way of obtaining the voltage drop
across the series resistance, Vs, from the measured current
I and Rs; then Vm = Vp − Vs.
16. The formation of an excised vesicle is a common problem
during attempts to form an inside-out patch and occurs when
the excised membrane edges close up, in a manner akin to the
formation of outside-out patches, and results in a “cell-attached”
vesicle. A bathing solution that is very low in Ca2+ and Mg2+ ions
can help prevent this. The outward-facing part of the vesicle can
be ruptured to leave the inside-out patch by passing the tip of
the micropipette quickly out and then back into the bath solu-
tion, through the air-solution interface. This does however have
limited success and a complete loss of the patch occurs with at
least equal frequency. For more tips on dealing with such vesi-
cles refer to Chapter 9 of this volume.
References
1. Neher E, Sakmann B (1976) Single-channel system for studying ion channels with the
currents recorded from membrane of denervated patch-clamp technique. Methods Mol Biol 491:
frog muscle fibres. Nature 260:799–802 127–139
2. Hamill OP, Marty A, Neher E, Sakmann B, 5. de Wet H, Lippiat JD, Allen M (2008) Analysing
Sigworth FJ (1981) Improved patch-clamp tech- steroid modulation of BK(Ca) channels reconsti-
niques for high-resolution current recording tuted into planar lipid bilayers. Methods Mol
from cells and cell-free membrane patches. Biol 491:177–186
Pflugers Arch 391:85–100 6. Colquoun D, Sigworth FJ (1995) Fitting and
3. Lippiat JD (2008) Whole-cell recording using statistical analysis of single-channel records.
the perforated patch clamp technique. Methods In: Sakmann B, Neher E (eds) Single channel
Mol Biol 491:141–149 recording, 2nd edn. Plenum, New York, pp
4. Tammaro P, Shimomura K, Proks P (2008) 483–587
Xenopus oocytes as a heterologous expression
Chapter 8
Abstract
Planar lipid bilayer is an electrophysiological technique that enables study of functional activities of ion
channels, porins, and other pore-forming molecular complexes. The main purpose of this method is to
monitor ion channels’ behavior at the single molecule level in the artificial membranes. Here, I describe the
details of this technique that will underline formation of the lipid bilayers and incorporation and activation
of the ion channel protein.
Key words Planar lipid bilayer, Black lipid membranes, Ion channel, Ionic current, Conductance
1 Introduction
Electrophysiological approaches are designed to evaluate physical
properties and characteristics of ion channels. Planar lipid bilayer is
one of the unique electrophysiological techniques that is intended
to study specific channel properties of the purified complexes in a
well-controlled artificial environment (1). The effectiveness of this
method reveals the possibility to study and characterize ion channel
behavior at the single molecule level. Furthermore, this technique
is very useful for investigation of the direct effects of chemicals and
enzymes on the channel while excluding possible indirect effects,
which might be caused by the presence of regulatory proteins of
the native membranes.
Planar lipid bilayer experiments enable monitoring the transport
rates of ions across membranes through incorporated ion channels.
The experiments are performed in the chamber with cis- and trans-
compartments that are connected through a small aperture with a
diameter ranging from 50 to 250 mm. A lipid solution is applied
to the aperture with subsequent formation of a planar bilayer
membrane on the hole. This is followed by reconstitution of ion
channel proteins. Ion channels can be inserted into bilayer lipid
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_8, © Springer Science+Business Media, LLC 2013
109
110 Eleonora Zakharian
Fig. 1 Planar lipid bilayer chamber with inserted cuvette, lipid bilayers are painted
on the aperture in the middle of the cuvette. Silver-silver chloride electrodes are
connected to each compartment (cis and trans), those allow to obtain the ionic
current that is amplified using a voltage-clamp amplifier. Cups and chambers are
filled with equal volumes in the cis- and trans-sides, which results in a balanced
solution height, thus minimizing mechanical gradients across the bilayer membrane.
Below is a graphical representation for formation of planar lipid bilayers and
incorporated ion channel that conducts ions across the membrane
2 Materials
2.1 Solutions 1. Lipids include synthetic 1-palmitoyl-2-oleoyl-glycero-3-phos-
and Reagents phocoline (POPC) and 1-palmitoyl-2-oleoyl-glycero-3-phos-
phoethanolamine (POPE, Avanti Polar Lipids, Birmingham,
112 Eleonora Zakharian
AL). Lipids are stored in septum glass vials and screw caps with
Teflon discs inserts that are from Thermo Scientific Pierce
(Rockford, IL).
Short- and long-chain phosphoinositides are from Cayman
Chemicals (Ann Arbor, MI).
2. Organic solvent to dissolve the lipids—n-decane (Sigma-Aldrich,
St. Louis, MO).
3. Experimental bathing solution: 150 mM KCl, 0.2 mM MgCl2,
20 mM Hepes, pH 7.4. All salts are ultrapure (>99%) (Sigma-
Aldrich, St. Louis, MO). To increase the purity of the solutions
it is important to filter all the buffers with 0.22 mm filters.
4. Planar lipid bilayer chamber and cuvette are made of Delrin
(acetyl resin); we use cuvette with an aperture of ~150 mm in
diameter (Warner Instruments, Hamden, CT).
5. The TRPM8 protein is purified from the human embryonic
kidney cells (HEK-293), stably expressing the channel as pre-
viously described (7), for updates in the purification procedure
(see Note 1).
6. NCB buffer: 500 mM NaCl, 50 mM NaH2PO4, 20 mM Hepes,
2 mM Na-orthovanadate, 10% glycerol, pH 7.5.
7. Cell homogenization buffer: NCB buffer with addition of
1 mM of protease inhibitor PMSF, 5 mM b-mercaptoethanol.
8. Protein isolation buffer: NCB buffer with addition of one tab-
let of protease inhibitor cocktail, 1 mM PMSF, 20 mg/mL
DNase, 20 mg/mL RNase, 0.1% Nonidet P40 (Roche,
Indianapolis, IN), and 0.5% dodecyl-maltoside (DDM)
(CalBiochem, Darmstadt, Germany).
9. Phosphate buffer saline (PBS).
3 Methods
3.1 Preparing Preparation of the working equipment such as bilayer chambers
Working Equipment and cuvettes is the very first and important step and should be
for Planar Lipid carried before each experiment. To optimize the rate for a successful
Bilayer: Chambers, BLM experiment and eliminate false positive results due to possible
Cuvettes, and Glass contaminants, the bilayer chamber and cuvette have to be cleaned
Capillaries thoroughly.
1. Wash with dishwashing detergent in warm water; rinse well
several times with warm running water to remove detergent;
rinse three times with milli-Q water.
2. Wash with organic solvents—first with ethanol and then with
methanol; dry with nitrogen gas. It is useful to press on top of
cuvette when filled with solvent, the intense solvent flow
through the aperture helps to remove residual lipids and other
content from the aperture. When switching from one target-
protein to another, or in case of difficulties to remove contami-
nants, all washing steps can be done along with bath sonication
(see Note 2).
3. Delrin cuvettes can also be cleaned with chloroform, when
necessary to remove highly hydrophobic content from the
aperture. Note that not all materials are suitable for a chloro-
form use; be sure that your cuvette will not be damaged during
this cleaning procedure (see Note 3).
4. Prepare a set of air-bubble glass capillaries. Holding to the end
of capillary, flame the other end, simultaneously pooling the
edge with forceps until the thin prolonged glass tube is formed.
Remove the forceps and continue to flame the thin tube in
order to make the bubble-like structure.
3.2 Preparation 1. Open the ampoules with lipids in chloroform solution and
of the Lipids transfer the content into new and prerinsed with organic sol-
vent glass vials, add the molecular sieves (4 Å) and cover with
Teflon-side screw cap. For storage conditions see Note 4.
2. Prepare the mixture of POPC/POPE in 3:1 ratio and dry
the lipids by lyophilization in Speed-Vac at least for 2 h to
114 Eleonora Zakharian
3.3 Formation of 1. Fill the chamber in the following order: first add 1 mL of
Planar Lipid Bilayer bathing solution to the trans-compartment (cuvette); press
slightly on the top of cuvette until a tiny droplet will appear in the
area of aperture (see Note 5); add 1 mL of bathing solution to
the cis-compartment.
2. Immerse a pair of matched Ag-AgCl electrodes in the cis- and
trans-compartments. Adjust the background-leak current near
0 values, this can be done by changing the voltage in a range
of ±1 mV and adjusting manually the offset appropriately
so that at positive voltage baseline would go upwards and at
negative it would shift downwards (see Note 6).
3. Dip the air-bubble glass capillary in the lipid/decane solution
and carefully apply the lipids to the aperture in the cuvette by
“painting.” After several gentle movements of the air-bubble
in the area of aperture, the film will form. We prefer not to pre-
apply the lipidic solution beforehand; instead, we distribute
the lipids by painting with the air-bubble glass capillary when
the aqueous solvents are present (see Note 7).
4. Monitor the thinning of the lipid film by either observing it in
the microscope—planar lipid bilayers reflect no light and appear
black after formation, or alternatively estimate the capacitance
of the membrane—as lipid bilayers will form the capacitance
will gradually increase, indicating that the film is thinning.
The capacitance of the bilayer can reach 100–200 pF, depend-
ing on the size of aperture used (see Note 8).
3.4 Incorporation 1. After the bilayers are formed, add 0.2 mL of the TRPM8 micellar
and Activation of Ion solution (0.002 mg/mL) to the cis-compartment with gentle
Channel stirring. Normally it will take some time for the channel to
incorporate in the membrane. Fusion of the proteo-liposomes
or micelles can be observed by fusion spike appearance. After
the incorporation stop the stirring.
2. To activate TRPM8 in planar lipid bilayer, add its agonist
menthol (500 mM) to the cis- and/or trans-compartment.
TRPM8 inserts into lipid bilayer without preferences and may
be oriented either way. To induce TRPM8 channel openings,
apart from the ligand, it also requires the presence of its gating
factor phophoinositol 4,5-biphosphate (PIP2) (Fig. 2). The
short acyl-chain dioctanoyl diC8PIP2 (2.5 mM) can be added to
either side of the membrane, but it activates the channel from
the intracellular side (see Note 9). Channel current recordings
can be then obtained with different voltages. Orientation of
TRPM8 can be determined according to the outward rectification
Planar Lipid Bilayers 115
Fig. 2 Activation of TRPM8 channels in planar lipid bilayers by menthol and PIP2.
Representative single-channel current recordings of TRPM8 channels incorpo-
rated in planar lipid bilayers formed from POPC/POPE (3:1) in n-decane, between
symmetric bathing solutions of 150 mM KCl, 0.2 mM MgCl2 in 20 mM Hepes
buffer, pH 7.4 at 22°C. 0.2–0.5 mL of 0.2 mg/mL TRPM8 protein was incorporated
in POPC/POPE micelles, which were added to the cis-compartment. Clamping
potential was +60 mV. Upper and lower traces consist of three segments with
subsequent additions of components as indicated in the figure: 2 mM of diC8 PIP2
and 500 mM of menthol were added to both compartments (the figure is repro-
duced from ref. 8 with permission)
4 Notes
1. HEK-293 cells stably expressing Myc-tagged TRPM8 are
grown to ~80% confluence, washed and collected with PBS.
Cells are harvested and resuspended in cell homogenization
buffer. Then the cells are lysed by freeze-thawing method
and centrifuged at low speed to remove cell debris and DNA.
The supernatant is further centrifuged at 40,000 × g for 2.5 h.,
and the pellet is resuspended in protein isolation buffer.
The suspension is incubated overnight at 4°C on a shaker with
gentle agitation and then centrifuged for 1 h at 40,000 × g.
The supernatant is collected and incubated with the beads,
where the TRPM8 protein is purified by immunoprecipitation
with anti-Myc-IgG conjugated to A/G protein magnetic beads
(Pierce, Thermo Scientific), following the procedure provided
by the manufacturer. All steps of purification are performed at
4°C. For the planar lipid bilayer experiments the protein is
eluted with Myc-peptide (50 mg/mL).
2. Place chamber and cuvette into a beaker and perform all the
washing steps (warm water with detergent; rinse; organic
media) while using bath sonicator at low intensity, do not
exceed 5 minutes for each washing step. Longer and more
frequent sonication may affect the quality of the chamber.
3. Polystyrene cuvettes have poor resistance to organic solvents,
which can lead to degradation of the aperture; therefore these
cannot be cleaned with strong organic solvents. We also do not
recommend using chloroform-cleaning step for the bilayer
chamber, although it is made of Delrin, but the observation
window is made of a different plastic polymer that can be
entirely destroyed with chloroform.
4. Stock lipids/chloroform solutions are degassed with nitrogen
stream and stored in the presence of molecular sieves 4 Å
(Sigma) at −20°C. The molecular sieves are used for dehydration
Planar Lipid Bilayers 117
References
Abstract
The excised inside-out patch clamp technique gives rapid access to the intracellular surface of the plasma
membrane while measuring channel activity. This way the effects of intracellular regulators of ion channels
or transporters can be studied in isolation, in the absence of most of the cellular machinery. This chapter
summarizes our experience with this technique using large patches to study various ion channels expressed
in Xenopus oocytes.
Key words Xenopus oocytes, Excised patch, Macropatch, TRP channels, Patch clamp
1 Introduction
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_9, © Springer Science+Business Media, LLC 2013
119
120 Tibor Rohacs
Xenopus laevis oocytes have been used for quite a long time as
an expression system to study recombinant ion channels (9). There
are many articles that provide excellent description of oocyte prep-
aration and RNA injection (10); we will provide a brief description
here based on our experience.
2 Materials
2.1 Oocyte 1. OR2 solution for oocyte preparation and storage. The composi-
Preparation tion of OR2 is 82.5 mM NaCl, 2 mM KCl, 1 mM MgCl2,
and Injection 5 mM HEPES, pH = 7.4. We designate this Ca2+-free solution
as OR2(−). Since large volumes are used because of the repeti-
tive wash steps during oocyte isolation, it is practical to prepare
a 20× concentrate, and dilute several liters to working concen-
tration to keep in a large bottle. After dilution the pH needs to
be adjusted, and the solution can be kept at 4°C for several
weeks; it is not necessary to sterile filter the OR2(−) solution.
After digestion, the eggs are kept in OR2 solution with an
additional 1.8 mM Ca2+ and penicillin-streptomycin (desig-
nated as OR2(+) solution); we recommend that this solution is
sterile filtered.
2. Collagenase solution. We use 0.1–0.2 mg/mL of type IA col-
lagenase from Sigma. We dissolve it freshly in OR2(−) just
before starting the digestion process (see Note 1).
3. MS222 solution. 0.25% Ethyl 3-aminobenzoate methanesul-
fonate (tricaine, MS222) dissolved in H2O, pH adjusted to
~7.4. We prepare 1 L, which can be reused several times, when
kept in a cold room.
4. Scissors, forceps.
5. Sutures (Chromic gut).
6. Oocyte incubator (18°C) preferably with an electrical outlet
inside to power the rotating platform.
7. Dissecting microscope and light source.
8. Injector (Drummond Nanoject).
9. Nuclease-free water and mMessage Machine kit (Ambion).
2.2 Patch Clamp We recommend that all solutions are sterile filtered to avoid clog-
Measurements ging of the patch clamp pipette and the perfusion system.
1. Bath solution. The oocyte is placed in the bath solution during
the experiment, and the same solution is used to fill up the
perfusion system. The composition is 97 mM KCl, 5 mM
HEPES, and 5 mM EGTA, pH = 7.4. The purpose of includ-
ing EGTA in the bath solution is to avoid the activation of
the Ca2+-activated Cl− current endogenous in the oocytes
(see Notes 2 and 3).
Macropatch Recording 121
stone, also available from the same company. This is best done
under the dissecting microscope used for oocyte injection.
8. Patch clamp setup, equipped with a stereo microscope
(see Chapter 7 of this book for more details).
9. Micromanipulator. We use a three-axis manual manipulator
with one motorized axis (Siskiyou). The manual manipulators
can be used to approach the oocyte, and the fine final move-
ment is done with the motorized axis.
10. Perfusion system. Various gravity-based perfusion systems with
solenoid or pinch valves are available; we use models from ALA
Scientific and Automate Scientific. We use our custom-made
perfusion pencils, since we need very low perfusion rates, as we
often use expensive compounds that require keeping fluid flow
and dead space low. A perfusion pencil can be made by gluing
seven thin plastic tubes into a gel-loading tip using two-com-
ponent epoxy glue, available at any hardware store. Generally
it is possible to use less than 1 mL, even as low as 0.5 mL of
perfusion solution with a handmade system. As an alternative,
commercial perfusion pencils can be used for local perfusion of
the patch pipette, also available from ALA and Automate. To
achieve low volumes, thin tubes need to be used. It is impor-
tant to use as few connections as possible, since every connec-
tion is a source for air bubbles. We use 1 mL plastic syringes as
reservoirs, and instead of connecting them with a luer connec-
tor, we melt the bottom part, using a gas burner, and pull it to
form a thin tube, which we directly connect to the silicone tub-
ing used for the pinch valves.
3 Methods
usually remove far more oocytes than we need since many are
lost or damaged during the digestion, and in a poor quality
prep, sometimes a large number of oocytes need to be screened
to find enough good-quality ones (see Note 9).
3. Using dissolvable chromic gut sutures, the abdominal layer
and then the skin is sutured back with 3–4 stitches.
4. The frog is placed back to a recovery tank. They recover from
the anesthesia usually within 15–20 min. It is important not to
over-anesthetize them, since they can drown, if the recovery
time is excessive.
5. The oocyte sacs are either cut to smaller pieces with a pair of
scissors or torn to smaller pieces using two pairs of forceps.
After several washes with OR2(−), until the solution is not
turbid from debris, the oocytes are placed into a 0.1–0.2 mg/
mL collagenase solution, and the tube is rotated very gently at
18°C overnight. We use a 50 mL Falcon tube filled with
40 mL collagenase solution, and place it in a rotating device
inside an incubator, which is kept at 18°C. The temperature is
critical. The plane of rotation is parallel to the axis of the fal-
con tube, which creates a very gentle mechanical agitation
(see Note 10).
6. The next morning, the oocytes are washed several times with
OR2. This step is critical, since collagenase needs to be com-
pletely removed before the oocytes are placed into a Ca2+-
containing solution. Ca2+ greatly enhances the activity of
collagenase; thus even trace amounts of the enzyme can dam-
age the oocytes. Washing the oocytes is simple, they sediment
within seconds in a 50 mL tube, then the solution can be gently
poured off, and replaced with fresh, repeating several times.
7. Next the oocytes are shaken for an additional hour in OR2(−),
still Ca2+-free. This step removes the follicular layer by
mechanical agitation from most of those eggs that still have it
(see Note 11). We usually do this step with the rotation being
perpendicular to the axis of the 50 mL tube with lower solu-
tion levels (usually 35 mL) to create stronger mechanical
agitation.
8. Next the oocytes are placed in OR2(+) solution that contains
Ca2+ and antibiotics. Older protocols also included nutrients
such as pyruvate, this is unnecessary, since the yolk of the
oocytes provides enough nutrients, and additional nutrients
make infection of the solution more likely.
9. Select oocytes for injection, usually it is good to select at least
2× the required number to have non-injected controls, and
extra eggs for unforeseen purposes, they usually last a lot longer
than the ones that are not selected out from the debris and
damaged oocytes.
124 Tibor Rohacs
3.2 RNA Preparation 1. RNA is prepared from linearized DNA using the mMessage
and Injection Machine kit from Ambion (see Note 12 for oocyte vectors).
Gloves, RNAase-free solutions, pipette tips, and other tools are
necessary. Usually we store all tubes, pipette tips, and other
materials used for RNA work separately, and use them for this
purpose only.
2. A Drummond Nanoject injector is used; we usually inject
50 nL, even though the volume can be adjusted. After pulling,
the tip of the needle needs to be broken to a diameter of ~20–
25 μm. This can be achieved either by pushing the needle
through a Kimwipe paper or by carefully “cutting” the tip with
a scalpel. It is advisable to check the size of the tip after break-
ing it. Injecting pipettes with too small diameters get clogged
easily; too large ones damage the oocytes (see Notes 13–15).
3. The purpose of the digestion is to remove the oocytes from the
follicular sac, and to remove the follicular layer. Oocytes with-
out the follicular are easy to inject, since the sharp needle easily
penetrates the vitelline layer and the plasma membrane. Oocytes
with the follicular layer on are very difficult to inject and most
of the time injection causes significant damage to the oocytes,
reducing viability. The follicular layer can easily be noticed by
the thin blood vessels. If the follicular layer lacks visible blood
vessels it is a lot harder to notice; often it is only recognized
during injection by the difficulty of penetrating with the injec-
tion needle. If oocytes are under-digested, and a significant
number of them have the follicular layer, it is worth manually
defolliculating them with a fine pair of fine forceps before injec-
tion. This is a lot easier than devitellinization before patching,
see later, and generally pays back by better oocyte quality and
quantity.
3.3 Patch Clamp 1. The vitelline layer of the oocyte needs to be removed using
two pairs of sharp Dumont 55 forceps (see Note 16).
2. After the pipette is filled up with the pipette solution, the
pipette is lowered into the bath solution, and pipette potential
is zeroed. Gentle positive pressure is applied and the pipette is
moved close to the oocyte using the manual axes on the manip-
ulator (see Note 17).
3. Upon touching the oocyte, using the motorized axis, pipette
resistance increases slightly, then the positive pressure can
be relieved, which should further increase pipette resistance.
Macropatch Recording 125
4 Notes
a
1. Cell-attached 2. Vesicle
3. Inside-out
b PIP2
c
3 3
3
I (nA)
3
Excise
2 1 2
I (nA)
1 2 1
Air
0 -100 -50 2
50 100
0 60 120 180 240 mV
Time (s)
Fig. 2 (a) Cartoon depicting the patch pipette (thick grey lines), and the cell-attached configuration (1), forma-
tion of a vesicle after patch excision (2), and formation of an inside-out patch after a brief air exposure (3). Note
that most patches do not form a vesicle, but rather go straight into the inside-out configuration; see long arrow.
(b) Time course of a measurement with TRPM8 channels with 500 μM menthol in the patch pipette, current
traces are shown at −100 and +100 mV from a voltage ramp from −100 to +100 mV. After excision, the low
current levels correspond to a vesicle, which was broken by a brief exposure to an air bubble, generated with
a P20 pipette tip next to the patch pipette. The slow current decrease after the inside-out configuration is due
to the loss of phosphatidylinositol 4,5-bisphophate (PIP2) from the patch membrane, which is required for
TRPM8 activity. The application of 50 μM diC8-PIP2 is shown by the horizontal line
128 Tibor Rohacs
References
5. Rohacs T, Lopes CM, Jin T, Ramdya PP, 11. Csanady L, Torocsik B (2009) Four Ca2+ ions
Molnár Z, Logothetis DE (2003) Specificity of activate TRPM2 channels by binding in deep
activation by phosphoinositides determines crevices near the pore but intracellularly of the
lipid regulation of Kir channels. Proc Natl Acad gate. J Gen Physiol 133:189203
Sci U S A 100:745–750 12. Liman ER, Tytgat J, Hess P (1992) Subunit
6. Rohacs T, Lopes CM, Michailidis I, Logothetis stoichiometry of a mammalian K+ channel
DE (2005) PI(4,5)P2 regulates the activation determined by construction of multimeric
and desensitization of TRPM8 channels cDNAs. Neuron 9:861–871
through the TRP domain. Nat Neurosci 13. Baukrowitz T, Tucker SJ, Schulte U, Benndorf
8:626–634 K, Ruppersberg JP, Fakler B (1999) Inward
7. Thyagarajan B, Lukacs V, Rohacs T (2008) rectification in KATP channels: a pH switch in
Hydrolysis of phosphatidylinositol 4,5-bispho- the pore. EMBO J 18:847–853
sphate mediates calcium-induced inactivation 14. Fink M, Lesage F, Duprat F, Heurteaux C,
of TRPV6 channels. J Biol Chem Reyes R, Fosset M, Lazdunski M (1998) A
283:14980–14987 neuronal two P domain K+ channel stimulated
8. Zakharian E, Cao C, Rohacs T (2011) by arachidonic acid and polyunsaturated fatty
Intracellular ATP supports TRPV6 activity via acids. EMBO J 17:3297–3308
lipid kinases and the generation of PtdIns(4,5) 15. Venkatachalan SP, Bushman JD, Mercado JL,
P2. FASEB J 25:3915–3928 Sancar F, Christopherson KR, Boileau AJ
9. Barnard EA, Miledi R, Sumikawa K (1982) (2007) Optimized expression vector for ion
Translation of exogenous messenger RNA cod- channel studies in Xenopus oocytes and mam-
ing for nicotinic acetylcholine receptors pro- malian cells using alfalfa mosaic virus. Pflugers
duces functional receptors in Xenopus oocytes. Arch 454:155–163
Proc R Soc Lond B Biol Sci 215:241–246 16. Shih TM, Smith RD, Toro L, Goldin AL
10. Smith LD, Xu WL, Varnold RL (1991) (1998) High-level expression and detection of
Oogenesis and oocyte isolation. Methods Cell ion channels in Xenopus oocytes. Methods
Biol 36:45–60 Enzymol 293:529–556
Chapter 10
Abstract
Ion channels are integral membrane proteins that regulate membrane potentials and signaling of cells in
response to various stimuli. The patch-clamp technique enables the study of single channels or a population
of channels. The macroscopic recording approaches are powerful in revealing population-averaged behav-
iors of channels both under basal conditions and in response to various stimuli, modulators and drugs.
On their own, however, these approaches can be insufficient for determinations of channel gating mecha-
nisms as they do not accurately report channel open probabilities below 10−2 to 10−3. This obstacle can be
overcome with the use of single-channel recording techniques. Single-channel recording techniques can
be applied to one or a few channels to estimate Po over a larger range than macroscopic recordings.
The combination of heterologous overexpression of ion channels with macroscopic and single-channel
recordings can be applied to hundreds of channels to estimate Po between 1 and 10−8. Here, we describe
practical approaches of single-channel recordings that our laboratory utilizes. We also provide examples
where the combined macroscopic and single channel approach can be employed to study gating mecha-
nisms of the BK type, large conductance, Ca2+ and voltage activated potassium channel in a mammalian
expression system. The techniques presented should be generally applicable to the studies of ion channels
in heterologous expression systems.
Key words Patch clamp, Single channel recording, Heterologous expression, Ion channel, BK,
Mammalian expression system, Transient transfection
1 Introduction
Ion channels are integral membrane proteins that open or close
transmembrane ion conduction pathways in response to various
stimuli such as changes of temperature, pH, membrane potential, or
ligand binding. By increasing or reducing membrane permeabilities
to select ions, ion channels regulate membrane potentials and affect
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_10, © Springer Science+Business Media, LLC 2013
133
134 Nguyen V. Nguyen et al.
Fig. 1 Macroscopic and G–V relation alone do not distinguish unique gating
changes. (a) Simulated 0 Ca2+ G–V relations based on three sets of gating param-
eters a, b, and c. Relative to a (grey curve), b (dashed curve) represents a tenfold
increase of intrinsic gating and c (solid curve) represents a −34 mV shift of the
half-activation voltage of Qo (Vho (3). (b) Simulated 0 Ca2+ Log Po–V relations
based on the same sets of gating parameters. Log Po–V but not G–V relations
distinguish b and c
2 Materials
Fig. 2 Homemade sticky wax coating device and microforge. (a) A power transformer is connected to an
aluminum block with 30 W 25 W resistor (Dale) as a heating element. The aluminum block has been machined
to fit a small pyrex beaker for melting of wax. (b) A micromanipulator is mounted onto a 40× Nikon objective.
The micromanipulator places the heating filament within the focal plane of the objective. A glass electrode
holder is mounted to the stage in order to position the glass pipette tip in the proximity of the heating filament
and within the objective focal plane
3 Methods
Expression and analysis of BK channels in human embryonic kidney
(HEK293T) cells is a three-step process. First, cDNAs encoding
channel subunit(s) are transiently transfected into HEK293T cells
to express BK channels. Second, channel activities are assayed using
standard patch-clamp electrophysiology techniques that include
both macroscopic and single-channel recordings. Third, macro-
scopic and single-channel records are analyzed offline.
To obtain high quality single-channel recordings, it is essential
to carefully and consistently maintain the cultured cells. Healthy,
rapidly dividing cells have the highest transfection efficiency.
Moreover, high resistance and stable recordings at “extreme condi-
tions” (e.g., 0 Ca2+, very negative voltages) require healthy mem-
branes. Finally, analysis is facilitated by stable, low noise recordings.
The techniques for inside-out patch-clamp recording of BK
channel are standard. One aspect that is important for single-channel
recordings, particularly of channels with small single-channel
138 Nguyen V. Nguyen et al.
3.1 Expression of BK 1. HEK293T cells are thawed and passaged using sterile techniques
Channels in HEK-293T in a tissue culture hood with sterile solutions and supplies.
Cells (See Note 1) 2. HEK-293T cells are maintained in T25 tissue culture flasks in
a humidified tissue culture incubator at 37 °C and 5% CO2.
Each flask contains 5 ml of DMEM complete media (supple-
mented with 10% FBS, 2 mM L-glutamine and 5% streptomycin/
penicillin).
3. Cell cultures are passaged every 2–3 days to maintain a
confluence between 10 and 85% (see Note 2).
4. Passage of HEK cells: Remove media and wash cells twice with
PBS (without Ca2+ or Mg2+). Add 0.5 ml of trypsin/EDTA
(the minimum volume to cover the entire bottom of the flask)
to release the cells from the flask. Within a minute, tap the dish
against the palm of your hand to dislodge the cells for about
30 s. Inactivate trypsin with 6 ml of tissue culture media and
gently pipette the cell/media suspension to obtain a largely
single-cell suspension. Redistribute a fraction of cells into a
new T25 culture flask to maintain the stock (a 1:4 to 1:6 split
will generally achieve a 15–20% confluency). For transfection,
cells are placed into a 35 mm tissue culture dish at approxi-
mately 40% confluence (see Note 3). The goal is to obtain
~80–90% confluence the following day for transfection.
5. Prepare DNA mix for transfection: For BK channels composed
of only the pore-forming a subunit, 0.25 mg plasmid cDNA
encoding the a subunit and 0.25 mg of the pEGFP-C1 plasmid
encoding the EGFP marker are used. For BK channels com-
posed of pore-forming a subunit and the auxiliary b1 subunit,
0.25 mg plasmid cDNA encoding the a subunit and 1.5 mg of
Combined Single-Channel and Macroscopic Recording 139
Fig. 4 Analysis of NPo using all-point amplitude histogram or event detection. (a) Selected single channel
records of a patch at indicated voltages. There is one channel in the patch and currents between −60 and
−140 mV are not shown. (b1) Analysis of a −60 mV sweep using the all-point amplitude histogram. (b2)
Analysis of the same −60 mV sweep using event detection (only a fraction of the analysis is shown). Raw data
are shown in grey, idealized events are shown in black. (c) Estimated Po values (at −60 mV) using event detec-
tion (empty bars) or all-point amplitude histogram (solid bars) from five sweeps. Event numbers are indicated
in the empty bars. Averaged Po and SEM are shown on the right. (d) LogPo−V data obtained from the single-
channel recording
4 Notes
1. We choose HEK293T cells over Xenopus oocyte because of rela-
tive simplicity of cell culture and transfection. One potential
shortcoming of HEK293T cells is that the number of channels in
a patch can vary dramatically. However, the EGFP reporter allows
some selection of expression levels based on EGFP fluorescence
intensity. To better control channel expression having an induc-
ible promoter (such as tetracycline-regulated expression system)
in a stable cell line may be advantageous (29). Alternatively,
Xenopus oocyte can be used where expression levels are more
easily controlled by the amount of RNA injected (29).
For studying BK channel Ca2+ dependent gating, variations
in expression levels can be useful. For example, Po of BK chan-
nel a and ab (e.g., ab4) channels at high Ca2+ (>60 mM) and
very negative membrane potentials (where voltage sensors are
Combined Single-Channel and Macroscopic Recording 145
References
10. Sweet TB, Cox DH (2008) Measurements of 22. Orio P, Latorre R (2005) Differential effects of
the BKCa channel’s high-affinity Ca2+ binding beta 1 and beta 2 subunits on BK channel
constants: effects of membrane voltage. J Gen activity. J Gen Physiol 125:395–411
Physiol 132:491–505 23. Santarelli LC, Wassef R, Heinemann SH,
11. Sweet TB, Cox DH (2009) Measuring the Hoshi T (2006) Three methionine residues
influence of the BKCa (2)1 subunit on Ca2+ located within the regulator of conductance
binding to the BKCa channel. J Gen Physiol for K+ (RCK) domains confer oxidative sensi-
133:139–150 tivity to large-conductance Ca2+-activated K+
12. Wang B, Brenner R (2006) An S6 mutation in channels. J Physiol 571:329–348
BK channels reveals beta1 subunit effects on 24. Cui J, Aldrich RW (2000) Allosteric linkage
intrinsic and voltage-dependent gating. J Gen between voltage and Ca(2+)-dependent acti-
Physiol 128:731–744 vation of BK-type mslo1 K+ channels.
13. Wang B, Rothberg BS, Brenner R (2006) Biochemistry 39:15612–15619
Mechanism of beta4 subunit modulation of 25. Rothberg BS, Magleby KL (2000) Voltage and
BK channels. J Gen Physiol 127:449–465 Ca2+ activation of single large-conductance
14. Wang B, Rothberg BS, Brenner R (2009) Ca2+-activated K+ channels described by a two-
Mechanism of increased BK channel activation tiered allosteric gating mechanism. J Gen
from a channel mutation that causes epilepsy. Physiol 116:75–99
J Gen Physiol 133:283–294 26. Zagotta WN, Hoshi T, Dittman J, Aldrich RW
15. Gruslova A, Semenov I, Wang B (2012) An (1994) Shaker potassium channel gating. II:
extracellular domain of the accessory beta1 Transitions in the activation pathway. J Gen
subunit is required for modulating BK channel Physiol 103:279–319
voltage sensor and gate. J Gen Physiol 139: 27. Hirschberg B, Rovner A, Lieberman M, Patlak
57–67 J (1995) Transfer of twelve charges is needed
16. Yang H, Hu L, Shi J, Delaloye K, Horrigan to open skeletal muscle Na+ channels. J Gen
FT, Cui J (2007) Mg2+ mediates interaction Physiol 106:1053–1068
between the voltage sensor and cytosolic 28. Cox DH, Cui J, Aldrich RW (1997) Separation
domain to activate BK channels. Proc Natl of gating properties from permeation and
Acad Sci U S A 104:18270–18275 block in mslo large conductance Ca-activated
17. Yang J, Krishnamoorthy G, Saxena A, Zhang K+ channels. J Gen Physiol 109:633–646
G, Shi J, Yang H, Delaloye K, Sept D, Cui J 29. Trapani JG, Korn SJ (2003) Control of ion
(2010) An epilepsy/dyskinesia-associated channel expression for patch clamp recordings
mutation enhances BK channel activation by using an inducible expression system in mam-
potentiating Ca2+ sensing. Neuron 66: malian cell lines. BMC Neurosci 4:15
871–883 30. Hawley-Nelson P, Ciccarone V, Moore, ML
18. Koval OM, Fan Y, Rothberg BS (2007) A role (2008) Transfection of cultured eukaryotic
for the S0 transmembrane segment in voltage- cells using cationic lipid reagents. Curr Protoc
dependent gating of BK channels. J Gen Mol Biol Chapter 9: Unit 9.4
Physiol 129:209–220 31. Staruschenko A, Booth RE, Pochynyuk O,
19. Hou S, Horrigan FT, Xu R, Heinemann SH, Stockand JD, Tong Q (2006) Functional
Hoshi T (2009) Comparative effects of H+ and reconstitution of the human epithelial Na+
Ca2+ on large-conductance Ca2+- and voltage- channel in a mammalian expression system.
gated Slo1 K+ channels. Channels 3:249–258 Methods Mol Biol 337:3–13
20. Hou S, Xu R, Heinemann SH, Hoshi T (2008) 32. Magnus CJ, Lee PH, Atasoy D, Su HH,
The RCK1 high-affinity Ca2+ sensor confers car- Looger LL, Sternson SM (2011) Chemical
bon monoxide sensitivity to Slo1 BK channels. and genetic engineering of selective ion
Proc Natl Acad Sci U S A 105:4039–4043 channel-ligand interactions. Science 333:
21. Chen RS, Geng Y, Magleby KL (2011) Mg2+ 1292–1296
binding to open and closed states can acti- 33. Gamper N, Shapiro MS (2006) Exogenous
vate BK channels provided that the voltage expression of proteins in neurons using the
sensors are elevated. J Gen Physiol biolistic particle delivery system. Methods
138:593–607 Mol Biol 337:27–38
Chapter 11
Abstract
Perforated whole-cell patch-clamp is a variant of the patch-clamp technique used to measure the sum activ-
ity of ion channels in the plasma membrane of a single cell. Its defining feature is that electrical access to
the cell is obtained through inclusion of a pore-forming antibiotic in the patch pipette which perforates the
sealed patch of membrane in contact with the patch pipette. The antibiotic pores allow equilibration of
small monovalent ions between the patch pipette and the cytosol whilst maintaining endogenous levels of
divalent ions such as Ca2+ and signalling molecules such as cAMP. Therefore, the perforated patch-clamp
technique is ideal for studying ion channels whilst maintaining the integrity of second messenger signalling
cascades. Other benefits of using perforated patch-clamp over conventional patch-clamp include reduced
current rundown and stable whole-cell recording lasting >1 h. In this chapter, the application of the per-
forated patch-clamp technique for the study of heterologously expressed Kv7 potassium channels will be
discussed in detail including benefits and limitations of the technique.
1 Introduction
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_11, © Springer Science+Business Media, LLC 2013
149
150 John E. Linley
2 Materials
3 Methods
3.1 Cell Culture CHO cells are grown as an adherent culture in Ham’s F-12 supple-
mented with 10% fetal calf serum, penicillin, and streptomycin
(0.1%) in an incubator at 37°C, 5% CO2. Note that CHO cells
have an absolute requirement for proline in the media. To passage
cells, they are first washed in PBS then detached by trypsinization.
For electrophysiological recording from transfected CHO cells,
cells are plated onto 35 mm Nunclon culture dishes at a density of
~50% confluency and transfected 24 h later using a lipofection
reagent (e.g., Fugene HD) in accordance with the manufacturer’s
instructions. For transfection of Kv7.2/Kv7.3 channels we use
1 μg Kv7.2, 1 μg Kv7.3, 0.2 μg GFP plasmid cDNA and 3 μL
Fugene HD per transfection. 24 h post transfection cells are
trypsinized and replated onto sterile 10 mm glass coverslips pre-
coated with poly-l-lysine (0.001% poly-l-lysine solution for 20 min
followed by triple wash) in 24-well plates in a volume of 1 ml cul-
ture media. Cells are recorded from 48 to 72 h post transfection.
Plasmids encoding human Kv7.2 and human Kv7.3 (GenBank
Perforated Patch-Clamp Recording 153
3.3 Perforated 1. Pull patch pipettes using a micropipette puller and heat polish
Patch-Clamp: Setup to a resistance of 2–4 MΩ using a microforge.
2. Place patch pipettes in a covered holder or in a lidded petri dish
containing blu-tack in order to keep them clean. Note that
patch pipettes should be pulled fresh each day.
3. Remove glass coverslip from 24-well plate using fine forceps,
place in 35 mm dish containing extracellular solution, and rock
gently to wash cells (serum in media inhibits seal formation).
4. Place in perfusion chamber (break coverglass to obtain multi-
ple coverglass “chips” if required) on microscope stage.
5. Identify positively transfected cells by green fluorescence.
154 John E. Linley
3.4 Perforated 1. Apply a 5 mV voltage test pulse and zero off any offset
Patch-Clamp: potential.
Achieving Seal 2. Make a note of the open circuit resistance as this is indicative
and Whole-Cell of the patch pipette resistance.
3. Slowly micromanipulate the pipette in the z-axis towards the
cell whilst watching for a reduction in amplitude of the test
pulse (an increase in pipette resistance).
4. As soon as this occurs, apply light suction to the cell to obtain
a giga ohm seal. This can be trickier than in conventional
whole-cell as amphotericin B inhibits seal formation. If no seal
is obtained then increase the tip-dipping time (see Note 7).
5. Compensate for fast capacitance generated by the glass pipette
bath solution interface.
6. Change the membrane holding voltage to −70 mV. A slow
capacitance transient should begin to develop (see Fig. 1) over
a period of 5–15 min. This is caused by the gradual inclusion of
amphotericin pores in the patch of membrane forming the seal
allowing measurement of the cell capacitance. Initially this will
appear as a small (~20 pA) capacitance spike with a slow decay
kinetic (Fig. 1a). This will increase in magnitude and the decay
Fig. 2 Example recording from CHO cell expressing Kv7.2 and Kv7.3. (a) Current
trace in response to depolarizing voltage steps from a holding potential of
−70 mV in 20 mV increments from −100 to +60 mV. (b) Voltage dependence of
activation calculated from the magnitude of the deactivating tail current when
stepping from the test potential to −100 mV as indicated by the arrow in (a). Data
are normalized to the maximum tail current and presented as mean ± SEM from
ten cells
156 John E. Linley
4 Notes
References
1. Horn R, Marty A (1988) Muscarinic activation somes I. Specificity of the membrane permeability
of ionic currents measured by a new whole-cell changes induced by the polyene antibiotics.
recording method. J Gen Physiol 92:145–159 Biochim Biophys Acta 339:30–43
2. Hladky SB, Haydon DA (1970) Discreteness of 4. Rhee JS, Ebihara S, Akaike N (1994) Gram-
conductance change in bimolecular lipid mem- icidin perforated patch-clamp technique reveals
branes in the presence of certain antibiotics. glycine-gated outward chloride current in
Nature 225:451–453 dissociated nucleus solitarii neurons of the rat.
3. de Kruijff B, Gerritsen WJ, Oerlemans A, Demel J Neurophysiol 72:1103–1108
RA, van Deenen LL (1974) Polyene antibiotic- 5. Andreoli TE, Monahan M (1968) The interac-
sterol interactions in membranes of tion of polyene antibiotics with thin lipid mem-
Acholeplasma laidlawii cells and lecithin lipo- branes. J Gen Physiol 52:300–325
Chapter 12
Abstract
Mechanotransduction, the conversion of a mechanical stimulus into a biological response, constitutes the
basis of a variety of physiological functions such as the senses of touch, balance, proprioception, blood
pressure, and hearing. In vertebrates, mechanosensation is mediated by mechanosensory neurons, whose
cell bodies are located in trigeminal and dorsal root ganglia. Here, we describe an in vitro model of mecha-
notransduction that provides an opportunity to explore the properties of mechanosensitive channels in
mammalian sensory neurons. The mechano-clamp method allows applying local force on plasma mem-
brane of whole-cell patch-clamped sensory neurons. This technique uses a mechanical probe driven by a
computer-assisted piezoelectric microstage to repeatedly stimulate sensory neurons with accurate control
of stimulus strength, duration, and speed.
Key words Mechanosensation, Pain, Sensory neuron, Mechanoreceptor, Skin, DRG, Primary culture,
Mechanical probe, Piezoelectric microstage, Patch clamp, Ion channels
1 Introduction
Somatosensory neurons detect a wide variety of mechanical stimuli.
Some are specialized to detect external mechanical stimuli
(e.g., mechanoreceptors, mechanonociceptors), while others
inform the nervous system about self-generated stimuli (e.g.,
stretch receptors). The ability of these mechanoreceptors to detect
mechanical cues relies on the presence on the specialized sensory
endings of mechanotransducer channels that rapidly transform
mechanical forces into electrical signals and depolarize the sensory
ending (1–3). This local depolarization, called receptor potential,
eventually leads to the generation of action potentials that propa-
gate toward the central nervous system.
Progress has been made in establishing the functional proper-
ties, specificity, and perceptual functions of mechanoreceptors.
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_12, © Springer Science+Business Media, LLC 2013
159
160 Jizhe Hao et al.
2 Materials
2.1 Reagents for 1. Experimental animals: rats (male Wistar, 120–130 g). All animal
Recording DRG experiments have to be performed in accordance with the
Neurons in Primary guidelines on the use of animals by the relevant authorities.
Culture 2. Halothane (Belamont, Nicholas Piramal). Harmful by inhala-
tion, use adequate ventilation.
3. Collagenase IA (C9891, Sigma).
4. Bovine Serum Albumin (BSA, A9647, Sigma).
5. Cell culture media, DMEM (Invitrogen).
6. Hank’s balanced salt solution, HBSS (Invitrogen).
7. Penicillin/Streptomycin (Invitrogen).
8. Glutamine (Invitrogen).
9. Nerve growth factor, NGF (Millipore).
10. Glial cell derived neurotrophic factor, GDNF (Invitrogen).
11. Laminin (Sigma).
12. Tetrodotoxin (Ascent Scientific).
2.1.1 Solution Setup for Pipette (internal) solution for whole-cell recordings
Patch Clamp Recording
1. For voltage-clamp experiments, prepare pipette solution as
follows: 125 mM CsCl, 1 mM MgCl2, 4.8 mM CaCl2, 10 mM
HEPES, 10 mM EGTA, 4 mM Mg-ATP, and 0.4 mM Na-GTP.
Adjust pH 7.4 with CsOH. Adjust osmolarity to 300 mOsm
with CsCl.
Investigating Mechano-Gated Channels 161
Fig. 1 Mechano-clamp setup. (a) Photograph of the mechanical probe. (b) Schematic representation of the
mechano-clamp setup. Cultured sensory neurons are recorded using the patch clamp technique (1). A glass
pipette is filled with a pipette solution and an Ag/AgCl wire connects the cell to the patch clamp headstage,
which is a sensitive current-to-voltage converter. The patch clamp amplifier is connected to a computer
through an analog-digital interface, allowing data generation, acquisition and analysis (2). The mechanical
probe is connected to a piezoelectric ceramic actuator, which is ideal for applications requiring high-resolution
movements for micro- and nanopositioning (3). Piezoceramic actuators make use of the deformation of the
piezoelectric material when an electric field is applied. The piezoelectric actuator is connected to a linear
amplifier-driver and stimulus parameters, such as duration and speed of stimuli, are user configurable via
pCLAMP software
162 Jizhe Hao et al.
2.2.1 Setup for Patch There are many papers that illustrate the assembly of a functional
Clamp Recording (Fig. 1) patch-clamp setup and perfusion systems, i.e., (6, 7); see also
Chapter 7 of this book.
Investigating Mechano-Gated Channels 163
3 Methods
3.1 DRG Neuron 1. Preparation of coated Petri dishes. Nunclon dishes (35 mm ∅)
Preparation need to be prepared at least 1 h before seeding cells. With a
Pipetman loaded with 600 mL of laminin (10 mg/mL),
position the pipette at the center of the dish, then release the
solution until it forms a large droplet in the middle of the dish.
Avoid covering the entire surface of the dish to prevent cells
moving at the edge of the dish. Incubate the dishes for at least
1 h at 37°C. Before dissociation of DRGs, wash the dishes
twice with HBSS solution and once with culture medium.
Allow culture dishes to remain in a safe environment.
2. Dissection of DRGs. Young rats are deeply anesthetized with
halothane and humanly killed in accordance of local ethical
guidance. DRGs are removed from their connective tissue
sheaths and incubated in HBSS solution containing 2 mg/mL
collagenase IA for 45 min at 37°C. Fire-polish two glass Pasteur
pipettes so that relatively small tip diameters are achieved.
3. Remove the supernatant containing the enzymatic solution
and wash seven or eight times DRGs with HBSS solution.
Resuspend DRGs with 800 mL of HBSS.
4. Triturate DRGs with Pasteur pipettes having large then small
tip diameters (1 and 0.5 mm) until the suspension becomes
visually opaque. Trituration consists in filling and emptying the
barrel at a rate of about 2.0 mL/s. The trituration should be
gentle because mechanosensory neurons are damaged by shear
force produced by backward and forward motion (see Note 1).
5. The resulting suspension is plated at the center of Nunclon
dishes coated with laminin. Culture medium is DMEM sup-
plemented with 10% heat-inactivated fetal calf serum, 2 mM
L-glutamine, 100 U/mL penicillin–streptomycin, 25 ng/mL
NGF, and 2 ng/mL GDNF. Neurons are maintained in a
humidified atmosphere (5% CO2, 37°C) for 12–16 h before
recording.
3.2 Fabrication Fabricate mechanical probe from borosilicate glass tubing. Pull
of Mechanical Probe the pipette to have a final tip diameter of 1–2 mm on a Brown-
Flaming P-97 pipette puller. Fire-polish micropipette using a
164 Jizhe Hao et al.
3.3 Setting the 1. Prepare patch pipettes with resistances of 3–4 MW. The tip
Patch-Clamp Setup diameter is determined by the pulling parameters of the pipette
puller.
2. Make solutions as appropriate for your experiment (basic
solutions are given in Subheading 2.1.1).
3. Transfer the Nunclon dish containing DRG neurons into a
500 mL-recording chamber in the inverted microscope for visu-
alization and subsequent patch-clamping. Cells are superfused
using a gravity-fed bath perfusion system at a laminar flow rate
of ~4 mL/min under continuous suction. Position the inlet
tubing at one end of the recording chamber opposite from the
suction tubing. Keep the perfusion flowing at a constant rate in
order to avoid shear stress perturbation during recording.
4. Select a neuron based on cell health. Choose an isolated neu-
ron not bearing neurites (improved voltage clamp) if you aim
at characterizing biophysical properties of mechanosensitive
currents. Select a neuron-bearing neurites if you aim at investi-
gating mechanical responses of nerve processes. The success
rate of experiments is almost entirely based on cell health con-
ditions. Unhealthy cells display granular cytosol and poorly
refracting surface membrane (see Note 3).
5. Fill the patch pipette with the internal solution and place the
pipette on the electrode holder. Use a syringe to apply a small
positive pressure to the pipette solution.
3.4 Positioning 1. Bring both patch pipette and mechanical probe above the
Mechanical Probe and selected neuron. Place initially the mechanical probe at 1.4 mm
Getting Whole-Cell to the edge of the cell by estimating the distance between the
(See Note 4) probe and the cell surface by using cycling mechanical sweeps
of 0.7 or 1.4 mm.
2. Move patch pipette down into the bath solution. By focusing
back and forth between the target cell and the pipette tip, move
the pipette into position above the neuron. Readings of pipette
resistance and pipette offset are taken at this time. Move down-
ward the pipette until the pipette resistance increases by 0.1–
0.2 MW. Once contact is made, release the pipette positive
pressure. We usually observed a gradual rise in pipette resistance
from the values taken before contacting the cell, often accom-
panied by the appearance of small capacitive transients in the
recording. If seals do not form spontaneously apply a continu-
ous suction by mouth until seal resistance reaches 100 MW.
Apply negative potentials, ranging from −10 to −60 mV, via the
voltage clamp command to help seal formation (³1 GW).
Investigating Mechano-Gated Channels 165
Fig. 3 Investigating velocity dependence of mechanosensitive currents. (a, b) Effects of varying the rate of
onset mechanical stimulus on the amplitude of rapidly adapting (a) and ultraslowly adapting (b) mechanosen-
sitive currents. (c, d) Current-clamp responses evoked by mechanical stimuli of varying velocities (lower
traces) in DRG neurons expressing rapidly adapting (c) or ultraslowly adapting (d) mechanosensitive currents
RT ⎛ 4PX [X ]0 1 1 ⎞
E rev = ln ⎜ + − ⎟.
F ⎜⎝ PCs [Cs ]i 4 2 ⎟⎠
4 Notes
1. Adapt the trituration protocol to the type of neurons you are
interested in. Trituration serves to break up the DRGs following
incubation in the enzyme-containing solution. If done too
vigorously, low-threshold mechanosensory cells will be damaged
lowering viability; small nociceptive cells will be enriched. Too
weak trituration and tissue fragments will be left intact lower-
ing yield. You can best determine a suitable rate for your tissue
through trial and error.
2. The tip surface must to be smooth and relatively large in order
to avoid perforating cell membrane when stimulated. Probe
tips that are too small are harmful to the cells; those that are
too big can destabilize cell adhesion.
3. Select healthy DRG neurons, which appear as shiny, round
neurons in Petri dishes.
4. Fix the mechanical probe firmly on its holder to avoid unwanted
movements of the probe and oscillation during mechanical
indentation.
5. Design mechano-clamp protocol carefully. Avoid too rapid
onset speed for forward motion of the mechanical probe that
may dislodge the recording patch pipette and generate oscillation.
Avoid withdrawing the probe too rapidly to ensure stability of
patch clamp recording. Avoid high frequency stimulation that
causes cumulative desensitization of mechanosensitive currents
in successive sweeps. Avoid too large stimulus amplitude that
may alter cell integrity.
6. Record cells with small leak currents at negative holding poten-
tials. Discard cells that display mechanosensitive currents with
weird kinetics.
7. Make sure that voltage-clamp conditions are satisfactory, in
order to avoid contamination of mechanosensitive currents by
voltage-dependent sodium currents escaping voltage control.
8. Pay attention to several important parameters. Stimulus duration:
examining the entire cohort of relaxation kinetics of mechano-
sensitive currents in DRG neurons requires using mechanical
stimuli of various durations lasting from 100 ms to up to 4 s.
Interstimulus interval: this interval allows mechanosensitive
currents to fully recover between each stimulus; it depends on
stimulus duration too. Typically, a 10 s interval is required for
MS currents to fully recover. If the interstimulus interval is too
short, a use-dependent decrease in current amplitude will
manifest. Stimulus intensity: avoid using excessive stimulus
amplitude (>10 mm probe displacement) as it may cause irre-
versible damage to the cell. Stimulus velocity: rapid onset rate of
Investigating Mechano-Gated Channels 169
Acknowledgments
This work was supported by the CNRS and by grants from the
Agence Nationale de la Recherche, Fondation Schlumberger,
ARCInca-2006, UPSA, IRME, and Fondation pour la Recherche
Médicale.
References
1. Delmas P, Hao J, Rodat-Despoix L (2011) 6. Cummins TR, Rush AM, Estacion M, Dib-
Molecular mechanisms of mechanotransduc- Hajj SD, Waxman SG (2009) Voltage-clamp
tion in mammalian sensory neurons. Nat Rev and current-clamp recordings from mamma-
Neurosci 12:139–153 lian DRG neurons. Nat Protoc 4:1103–1112
2. Basbaum AI, Bautista DM, Scherrer G, Julius 7. Davie JT, Kole MH, Letzkus JJ, Rancz EA,
D (2009) Cellular and molecular mechanisms Spruston N, Stuart GJ, Hausser M (2006)
of pain. Cell 139:267–284 Dendritic patch-clamp recording. Nat Protoc
3. Lumpkin EA, Caterina MJ (2007) Mechanisms 1:1235–1247
of sensory transduction in the skin. Nature 8. Drew LJ, Wood JN, Cesare P (2002) Distinct
445:858–865 mechanosensitive properties of capsaicin-
4. McCarter GC, Reichling DB, Levine JD sensitive and -insensitive sensory neurons.
(1999) Mechanical transduction by rat dorsal J Neurosci 22:RC228
root ganglion neurons in vitro. Neurosci Lett 9. Hao J, Delmas P (2010) Multiple desensitiza-
273:179–182 tion mechanisms of mechanotransducer chan-
5. Hao J, Delmas P (2011) Recording of mecha- nels shape firing of mechanosensory neurons.
nosensitive currents using piezoelectrically J Neurosci 30:13384–13395
driven mechanostimulator. Nat Protoc 6: 10. Coste B, Crest M, Delmas P (2007) Pharma-
979–990 cological dissection and distribution of NaN/
170 Jizhe Hao et al.
Nav1.9, T-type Ca2+ currents, and mechani- Piezo proteins are pore-forming subunits of
cally activated cation currents in different mechanically activated channels. Nature 483:
populations of DRG neurons. J Gen Physiol 176–181
129:57–77 13. Rugiero F, Drew LJ, Wood JN (2010) Kinetic
11. Coste B, Mathur J, Schmidt M, Earley TJ, properties of mechanically activated currents in
Ranade S, Petrus MJ, Dubin AE, Patapoutian spinal sensory neurons. J Physiol 588:301–314
A (2010) Piezo1 and Piezo2 are essential com- 14. Hu J, Lewin GR (2006) Mechanosensitive
ponents of distinct mechanically activated cat- currents in the neurites of cultured mouse
ion channels. Science 330:55–60 sensory neurones. J Physiol 577:815–828
12. Coste B, Xiao B, Santos JS, Syeda R, Grandl J, 15. McCarter GC, Levine JD (2006) Ionic basis of
Spencer KS, Kim SE, Schmidt M, Mathur J, a mechanotransduction current in adult rat
Dubin AE, Montal M, Patapoutian A (2012) dorsal root ganglion neurons. Mol Pain 2:28
Chapter 13
Abstract
Ion channels are integral membrane proteins that regulate the flow of ions across the plasma membrane
and the membranes of intracellular organelles of both excitable and non-excitable cells. Ion channels are
vital to a wide variety of biological processes and are prominent components of the nervous system and
cardiovascular system, as well as controlling many metabolic functions. Furthermore, ion channels are
known to be involved in many disease states and as such have become popular therapeutic targets. For
many years now manual patch-clamping has been regarded as one of the best approaches for assaying ion
channel function, through direct measurement of ion flow across these membrane proteins. Over the last
decade there have been many remarkable breakthroughs in the development of technologies enabling the
study of ion channels. One of these breakthroughs is the development of automated planar patch-clamp
technology. Automated platforms have demonstrated the ability to generate high-quality data with high
throughput capabilities, at great efficiency and reliability. Additional features such as simultaneous intra-
cellular and extracellular perfusion of the cell membrane, current clamp operation, fast compound appli-
cation, an increasing rate of parallelization, and more recently temperature control have been introduced.
Furthermore, in addition to the well-established studies of over-expressed ion channel proteins in cell
lines, new generations of planar patch-clamp systems have enabled successful studies of native and pri-
mary mammalian cells. This technology is becoming increasingly popular and extensively used both
within areas of drug discovery as well as academic research. Many platforms have been developed includ-
ing NPC-16 Patchliner® and SyncroPatch® 96 (Nanion Technologies GmbH, Munich), CytoPatch™
(Cytocentrics AG, Rostock), PatchXpress® 7000A, IonWorks® Quattro and IonWorks Barracuda™,
(Molecular Devices, LLC); Dynaflow® HT (Cellectricon AB, Mölndal), QPatch HT (Sophion A/S,
Copenhagen), IonFlux HT (Fluxion Bioscience Inc, USA), which have demonstrated the capability to
generate recordings similar in quality to that of conventional patch clamping. Here we describe features
of Nanion’s NPC-16 Patchliner® and processes and protocols suited for this particularly flexible and suc-
cessful high-throughput automated platform, which is based on planar patch-clamp technology. However,
many of the protocols and notes given in this chapter can be applied to other automated patch-clamp
platforms, similarly.
Key words Automated electrophysiology, Planar patch-clamp, Planar chip, Micro-fluidics, Intracellular
perfusion, Voltage-gated ion channels, Ligand-gated ion channels, Voltage-clamp, Current-clamp,
Temperature control
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_13, © Springer Science+Business Media, LLC 2013
171
172 Carol J. Milligan and Clemens Möller
1 Introduction
The well established patch-clamp technique developed by Erwin
Neher and Bert Sakmann (1) involves the manual positioning of a
glass microelectrode onto the surface of a single cell. A seal is
formed, and with the application of a small amount of negative
pressure (or a voltage pulse), the cell membrane at the tip of the
pipette (the patch) ruptures allowing direct access to the interior of
the cell, enabling control of the membrane voltage. Here we
describe the automated planar patch-clamp approach using the
platform Patchliner as an example (Fig. 1a). This approach requires
the manual preparation of the cells in suspension, and because cap-
turing cells is a random process it is vital that they are of extremely
high quality with uniform homogeneity. The automated planar
approach involves the robotic delivery of solutions, cells, and
compounds onto the planar chip (Fig. 1b–d), which is a tiny boro-
silicate sheet of glass with a micron-sized aperture in the center.
Fig. 1 (a) The automated planar patch-clamp platform consists of the NPC-16 Patchliner, amplifiers, and a
computer. It has a much smaller foot print compared to a conventional rig. The Patchliner takes up to three
NPC-16 chips per session. (b) Image of part of the NPC-16 multi-well microfluidic cartridge with a solution
delivery pipette inserted into the extracellular inlet of one chamber. Waste chamber highlighted by ** and
intracellular chamber highlighted by *. (c) Image of the underside of the cartridge in which the intracellular
solution compartments are highlighted with dye. (d) Schematic of a single chamber showing the arrangement
of extracellular, intracellular, and waste chambers as well as the position of the planar chip
Planar Patch-Clamp 173
Fig. 2 (a) Schematic, illustrating the mechanism of double stacked solution application. Stacking solutions
allow brief compound applications with timed intervals. The pipette aspirates buffer first, followed by com-
pound. In this way, the cell is exposed to ligand first followed by rapid buffer washout. (b) Example responses
to 100 μM nicotine in the maintained presence of 10 μM PNU120596 at three different temperatures using a
triple stacked application protocol. 10 μM PNU120596 was applied to GH4C1 cells expressing the human
α7nAChR, followed by a brief co-application of 100 μM nicotine, followed by wash buffer. PNU120596 shows
strong temperature dependence. (c) Pooled data plotting the amplitude of response to 100 μM nicotine in the
presence of 10 μM PNU120596 at various temperatures. Data are sampled at 1 kHz and filtered at 333 Hz.
Data are normalized to the corresponding responses recorded at room temperature (n = 10 ± SEM)
2 Materials
2.1 Equipment 1. NPC®-16 Patchliner Probe Selector/Quattro/Octo,
and Reagents PatchControlHT software, NPC®-16 chips (single use, dispos-
able). NPC®-16 electrode set (see Note 2) (Nanion
Technologies GmbH).
2. Patch-clamp amplifiers (EPC-10 quadro, HEKA Instruments
Inc.).
3. Computer with 24-in. TFT monitor (Dell).
4. Software for multichannel data acquisition (Patchmaster,
HEKA Instruments Inc.) and analysis (GraphPad Prism
4.0; GraphPad Software Inc., La Jolla, USA or IGOR-Pro;
WaveMetrics Inc., Lake Oswego, OR).
5. Cell culture flasks (T25 & T75, BD Falcon).
176 Carol J. Milligan and Clemens Möller
2.2 Cell Culture 1. GH4C1 cells stably transfected with rat α7nAChRs (GH4C1-
rα7 cells) are cultured and passaged in standard T75 tissue
2.2.1 GH4C1-rα7 Cells culture flasks in DMEM supplemented with 10 % FBS and
100 μL/mL geneticin at 37 °C in a humidified atmosphere
composed of 95 % air and 5 % CO2.
2. The cells should be passaged every 2–3 days using an enzyme
free PBS-based cell dissociation buffer and kept to a confluence
of 60–80 % (see Note 3).
2.3 Planar Patch- Prepare solutions in deionized water, filter (see Note 6), and mea-
Clamp Recording sure the osmolarity (see Note 7) and the pH. Solutions can be
Solutions stored at 4 °C for up to 5 days. Solutions should be warmed to
room temperature (20–22 °C) before use.
1. Standard extracellular solution: 140 mM NaCl, 4 mM KCl,
1 mM MgCl2, 2 mM CaCl2, 5 mM d-Glucose monohydrate,
10 mM HEPES; adjust to pH 7.4 with NaOH. Osmolarity
298 mOsm.
2. Standard extracellular solution for enhancing seals and record-
ing with GH4C1-rα7 cells: 80 mM NaCl, 3 mM KCl, 10 mM
Planar Patch-Clamp 177
3 Methods
3.1 Harvesting Cells The use of healthy cells is absolutely critical to the success of planar
for Planar Patch- patch-clamp recordings because the capture of cells onto the pla-
Clamp nar chip is a random process. The health of the cell impacts on the
quality of the seal formed between the cell membrane and the pla-
nar chip, which ultimately influences the quality of the recording.
Unlike conventional patch-clamp recording where healthy cells
can be visually selected, once cells are prepared in suspension, there
is no opportunity to optically select the healthiest looking cells.
3.1.1 GH4C1-rα7 Cells 1. In sterile conditions, remove the medium from the cells and
gently wash with D-PBS with Ca2+ and Mg2+ (see Note 10).
2. Pipette 2 mL cold cell dissociation buffer on to the cells and
incubate at room temperature for 3 min.
3. Add 5 mL culture media and pipette up and down gently.
4. Transfer the cells in culture media from the flask to a 15 mL
conical centrifuge tubes and centrifuge at 100 × g for 2 min at
room temperature and discard the supernatant by decanting.
5. Resuspend the cell pellet in 2 mL of extracellular solution, cen-
trifuge at 100 × g for 2 min at room temperature and discard
the supernatant by decanting.
6. Resuspend the cell pellet in a mixture of extracellular solution
for capturing cells and culture media (50:50 ratio, see Note 11)
at a density of 1 × 106 to 5 × 107 cells/mL (see Note 12) (cells
can be counted using a hemocytometer).
7. Transfer the cells to the cell hotel on the Patchliner where they
are continuously pipetted up and down to maintain single cells
and viability.
3.1.2 mESC-Derived 1. Wash twice in 5 mL of D-PBS with Ca2+ and Mg2+ and incu-
Cardiomyocytes bate at 4 °C for 15 min.
2. Wash once with 5 mL of D-PBS without Ca2+ and Mg2+.
3. Dissociate with 2 mL pre-warmed trypsin/EDTA solution for
4–5 min at 37 °C and 5 % CO2 (see Note 13).
4. Transfer cell suspension to 6 mL Cor.At Complete Culture
Medium in a 50 mL tube.
178 Carol J. Milligan and Clemens Möller
3.2.1 General 1. Load a preprogrammed Tree and select the edit mode to make
Experimental Set-up modifications according to your cell type/characteristics. This
enables you to modify amplifier and pump parameters and also
allows access to the full range of commands, which can be
inserted into a protocol via a generic drag-and-drop function
(see Note 17).
2. Select the appropriate chip resistance for your cells and place
three chip cartridges on the chip wagon.
Planar Patch-Clamp 179
3.2.2 Stacked Solution As with most ligand gated ion channels, nAChRs exhibit receptor
Application with GH4C1- desensitization, which is a common phenomenon for ligand gated
rα7 Cells ion channels. The kinetics and level of desensitization of ligand
gated ion channels are determined by ligand concentration and
exposure time, or both. For rapidly desensitizing ion channels, it is
important that compound application is fast, so that the entire ion
channel population is exposed to maximum concentration before
entering the desensitized state. Therefore rapid solution exchange
combined with brief drug exposure times can minimize or correct
for the deleterious effect caused by receptor desensitization. This is
achieved by using a stacked solution application where two or three
zones of solution are aspirated into the pipette before administra-
tion onto the cells (see Note 21).
1. Load a preprogrammed ligand Tree for triple stacked solution
application. Volumes and speeds of applications can be adjusted
(see Note 22).
2. Select high resistant single hole chips (5–6 MW, ~10 pF) for
GH4C1-rα7 cell recordings and load three chips onto the chip
wagon.
3. Within the Tree, adjust the holding potential to −75 mV and
select a continuous recording protocol, with a 13 s sweep,
from the pulse generator file in the job list to record the fast
ligand activated currents.
4. Follow steps 3–6 in Subheading 3.2.1 (see Note 23).
In the example shown in Fig. 2b GH4C1-rα7 cells were
exposed to the modulator PNU120596 (10 μM), directly followed
by nicotine (100 μM) in the presence of PNU120596 and subse-
quently by wash buffer using a triple stacked solution application.
180 Carol J. Milligan and Clemens Möller
3.2.3 Temperature The measuring head, chip wagon, and solution inside the pipette
Controlled Compound can be heated simultaneously to maintain physiological tempera-
Application with GH4C1- tures during the course of an experiment. Alternatively, the solu-
rα7 Cells tion in the pipette can be heated independently, exposing the cells
to transient temperature increases, for the study of heat activated
channels.
1. Load a preprogrammed ligand Tree for triple stacked solution
application combined with temperature control. Temperature
settings can be adjusted (see Note 24).
2. Select high resistant single hole chips (5–6 MW, ~10 pF) for
GH4C1-rα7 cell recordings and load three chips onto the chip
wagon.
3. Within the Tree, adjust the holding potential to −75 mV and
select a continuous recording protocol, with a 13 s sweep,
from the pulse generator file in the job list to record the fast
ligand activated currents.
4. Follow steps 3–6 in Subheading 3.2.1.
In the examples shown in Fig. 2c, GH4C1-rα7 cells were
exposed to nicotine in the presence of the PNU120596 at differ-
ent temperatures (20, 25, 30, 35, 40, and 20 °C). Responses are
greatly attenuated when the temperature is raised to physiological
levels and higher. Current is partially recovered when the tempera-
ture is lowered back to 20 °C. Activation of α7nAChRs in the
absence of modulator is not significantly inhibited by elevated tem-
peratures (9). Therefore, these data demonstrate the strong tem-
perature dependent action of PNU120596 at α7nAChRs.
Fig. 3 (a–c) Parallel voltage clamp recordings of mESC-derived cardiomyocyte ion channel currents in the
automated patch-clamp system Patchliner. IK (a), INa (b), ICa,L (c) current traces and corresponding current–
voltage relationship curves (d). (e) APs in control solution are highly reproducible over a time period of 15 min.
Representative traces are shown at the beginning (t = 0–40 s) and after t = 14 min 20 s to 15 min, traces
represent the mean of four consecutively recorded APs. The error bars represent the mean at APD50 SEM.
Sweep interval was 10 s
4 Notes
1. For most automated patch-clamp systems on the market it is
challenging to use primary or transiently transfected cells
because of the blind approach of capturing cells. Transiently
transfected cells can have low expression rates so it is advisable
to co-transfect with GFP to determine good transfection
efficiency. Primary cells can also be problematic if the cell prep-
aration is impure containing more than one cell type.
2. Place electrodes in bleach filled chloridation chambers for
30 min and then rinse with deionized water.
3. Cells should be passaged every 2–3 days with dissociation buf-
fer to prevent the cells adhering too tightly to the support sub-
strate and to avoid the growth of clusters which are a
considerable problem for planar patch-clamp. Cells left to grow
to greater than 80 % confluency also leads to cell aggregates
and results in poor capture rates. Dissociation buffers encour-
age cell separation and isolation. Seal rates can sometimes be
improved by optimizing cell culture methods, e.g., by using
T75 flasks instead of smaller T25.
4. Cor.At culture medium is a complete medium that has been
optimized for use with Cor.At mESC-derived cardiomyocytes.
5. The continuous beating of mESC-derived cardiomyocytes up
to and after 4 days is a good indicator that the cells have main-
tained their quality and viability. However, after more than
4 days the cells tend to form aggregates, and it is more difficult
to obtain single cell suspensions without harming them, which
reduces the success rate.
6. All recording solutions should be filtered. The internal solu-
tion is particularly important and should be filtered with a
0.22 μm-pore diameter filter on the day of the experiment.
The internal solution should not be older than a few days, and
should be stored at 4 °C.
7. Recording solution osmolarity is measured directly using a
freezing-point osmometer and adjusted if necessary with man-
nitol or sucrose, non-permeating molecules, to 285 mOsm/L
for all internal solutions and between 290 and 310 mOsm/L
for all external solutions. External osmolarity should always be
higher than the internal osmolarity.
8. It is helpful to use seal enhancer solution to increase the chance
of giga-seal formation. The extracellular seal enhancer solution
is a high calcium containing solution, whereas the intracellular
solution for capturing cells contains fluoride. These seal
enhancing solutions are usually replaced once a giga-seal is
achieved and before establishing whole-cell access. To avoid
Planar Patch-Clamp 183
Acknowledgments
References
1. Neher E, Sakmann B (1976) Single channel modulator of the α7 neuronal nicotinic acetyl-
currents recorded from membrane of choline receptor: in vitro and in vivo character-
denervated frog muscle fibres. Nature 260: ization. J Neurosci 25:4396–4405
799–802 10. Sitzia F, Brown JT, Randall AD, Dunlop J
2. Farre C, Stoelzle S, Haarman C, George M, (2011) Voltage and temperature-dependent
Brueggemann A, Fertig N (2007) Automated allosteric modulation of α7 nicotinic receptors
ion channel screening: patch clamping made by PNU120596. Front Pharmacol 2:1–9
easy. Expert Opin Ther Targets 11:557–565 11. Witchel HJ, Hancox JC, Nutt DJ (2003)
3. Brüggemann A, Farre C, Haarmann C, Psychotropic drugs, cardiac arrhythmia and
Haythornthwaite A, Kreir M, Stoelzle S, sudden death. J Clin Psychopharmacol 23:
George M, Fertig N (2008) Planar patch 58–77
clamp: advances in electrophysiology. Methods 12. Fenichel RR, Malik M, Antzelevitch C,
Mol Biol 491:165–176 Sanguinetti M, Roden DM, Priori SG, Ruskin
4. Majeed Y, Bahnasi Y, Seymour VAL, Wilson JN, Lipicky RJ, Cantilena LR (2004) Drug-
LA, Milligan CJ, Agarwal AK, Sukumar P, induced torsades de pointes and implications
Naylor J, Beech DJ (2010) Rapid and for drug development. J Cardiovasc Electro-
contrasting effects of rosiglitazone on transient physiol 15:475–495
receptor potential TRPM3 and TRPC5 chan- 13. Möller C, Witchel H (2011) Automated elec-
nels. Mol Pharmacol 79:1023–1030 trophysiology makes the pace for cardiac ion
5. Xu SZ, Sukumar P, Zeng F, Li J, Jairaman A, channel safety screening. Front Pharmacol
English A, Naylor J, Ciurtin C, Majeed Y, 2:1–7
Milligan CJ, Bahnasi YM, Al-Shawaf E, Porter 14. Zhou Z, Gong Q, Ye B, Fan Z, Makielski JC,
KE, Jiang LH, Emery P, Sivaprasadarao A, Robertson GA, January CT (1998) Properties
Beech DJ (2008) TRPC channel activation by of HERG channels stably expressed in HEK
extracellular thioredoxin. Nature 451:69–72 293 cells studied at physiological temperature.
6. Li J, Sukumar P, Milligan CJ, Kumar B, Ma Biophys J 74:230–241
ZY, Munsch CM, Jiang LH, Porter KE, Beech 15. Polonchuk L (2012) Toward a new gold stan-
DJ (2008) Interactions, functions, and inde- dard for early safety: automated temperature-
pendence of plasma membrane STIM1 and controlled hERG test on the Patchliner®. Front
TRPC1 in vascular smooth muscle cells. Circ Pharmacol 3:1–7
Res 103:e97–104 16. Stoelzle S, Obergrussberger A, Brüggemann
7. Milligan CJ, Li J, Sukumar P, Majeed Y, Dallas A, Haarmann C, George M, Kettenhofen R,
ML, English A, Emery P, Porter KE, Smith Fertig N (2011) State-of-the art automated
AM, McFadzean I, Beccano-Kelly D, Bahnasi patch clamp devices: heat activation, action
Y, Cheong A, Naylor J, Zeng F, Liu X, Gamper potentials, and high throughput in ion channel
N, Jiang LH, Pearson HA, Peers C, Robertson screening. Front Pharmacol 2:1–11
B, Beech DJ (2009) Robotic multiwell planar 17. Stoelzle S, Haythornthwaite A, Kettenhofen
patch-clamp for native and primary mamma- R, Kolossov E, Bohlen H, George M,
lian cells. Nat Protoc 4:244–255 Brüggemann A, Fertig N (2011) Automated
8. Naylor J, Li J, Milligan CJ, Zeng F, Sukumar Patch Clamp on mESC-Derived Cardio myo-
P, Hou B, Sedo A, Yuldasheva N, Majeed Y, cytes for Cardiotoxicity Prediction. J Biomol
Beri D, Jiang S, Seymour VAL, McKeown L, Screen 16:910–6
Kumar B, Harteneck C, O’Regan D, 18. Jonsson MK, Vos MA, Mirams GR, Duker G,
Wheatcroft SB, Kearney MT, Jones C, Porter Sartipy P, de Boer TP, van Veen TA (2012)
KE, Beech DJ (2010) Pregnenolone Sulphate- Application of human stem cell-derived cardi-
and Cholesterol-Regulated TRPM3 Channels omyocytes in safety pharmacology requires
Coupled to Vascular Smooth Muscle Secretion caution beyond hERG. J Mol Cell Cardiol
and Contraction. Circ Res 106:1507–15 52:998–1008
9. Hurst RS, Hajos MM, Raggenbass M, Wall 19. Binah O, Dolnikov K, Sadan O, Shilkrut M,
TM, Higdon NR, Lawson JH, Rutherford- Zeevi-Levin N, Amit M, Danon A, Itskovitz-
Root KL, Mitchell B, Berkenpas MB, Eldor J (2007) Functional and developmental
Hoffmann WE, Piotrowski DW, Groppi VE, properties of human embryonic stem cells-
Allaman G, Ogier R, Bertrand S, Bertrand D, derived cardiomyocytes. J Electrocardiol 40:
Arneric SP (2005) A novel positive allosteric S192–S196
Planar Patch-Clamp 187
Abstract
Microdomains that form on the plasma membrane of cells are essential for signalling compartmentation
within cells. The localization of ion channels in these surface microdomains is important in defining what
signalling cascades will be generated. For example, in cardiomyocytes, similar to other excitable cells,
action potential propagation depends essentially on the properties of ion channels that are functionally and
spatially coupled. In this chapter we describe a novel advanced patch-clamp technique, “Smart patch-
clamp,” which enables the study of functional ion channels in the cell surface microdomains in a wide
variety of biological cells and tissues. Smart patch-clamp combines conventional patch-clamp and Scanning
Ion Conductance Microscopy (SICM). SICM uses a glass micropipette as scanning probe and generates a
high-resolution topography image of the cell surface. Next, same micropipette is used as a patch-clamp
pipette to record ion channel signals from specific spots on the cell surface as determined by SICM. In this
chapter we focus on recording single channel L-type calcium channel currents from T-tubules of adult rat
cardiomyocytes.
Key words Smart patch-clamp, Scanning ion conductance microscopy, Cardiomyocyte, L-type
calcium channel, Single-channel recording
1 Introduction
The spatial distribution of ion channels on the cell surface has been
investigated by microscopy based techniques such as immuno-
chemistry (1, 2). However, these techniques provide little informa-
tion on the functional characteristics of ion channels. By contrast,
the patch-clamp technique provides information about ion channel
currents and hence functionality of ion channels on the cell sur-
face. However, the conventional patch-clamp technique does not
allow the precise selection of a region of interest on the cell to be
investigated and thus provides limited spatial resolution (3, 4).
Smart patch-clamp technique combines Scanning Ion
Conductance Microscopy (SICM) and patch-clamp electrophysi-
ology using a glass micropipette as a dual probe: scanning probe
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_14, © Springer Science+Business Media, LLC 2013
189
190 Anamika Bhargava and Julia Gorelik
and patch-clamp pipette (5, 6). In this method, the glass micropi-
pette first scans the cell surface using current feedback and a high-
resolution topographic image of the cell surface is obtained. The
same glass micropipette is then positioned over the region of inter-
est on the cell surface as determined by SICM and single ion chan-
nel currents are recorded using patch-clamp technique. Therefore,
smart patch-clamp can record ion channel activity from precise
microdomains on the surface of live cells and thus can generate a
spatial functional map of surface ion channels. In contrast, in con-
ventional patch-clamp method the position of the pipette with
respect to the cell topography cannot be controlled with a nano-
scale precision. This technique, though recently developed, has
already led to the determination of the spatial distribution of ion
channels which may help to unravel the mechanisms of the local-
ized control of cell signalling and function (6). In this chapter we
describe the smart patch-clamp technique and its application to
various cell types.
probe was very close to the sample, it had limited ability to scan cell
surfaces with prominent tall structures such as microvilli. When the
structures were taller than the pipette-sample separation the probe
often broke itself or damaged the surface structures.
Therefore the technique was further developed and “hopping
mode” SICM was introduced (11). In hopping mode SICM, at each
imaging point, the pipette approaches the sample from a starting
position that is well above any of the surface features. At this starting
position the reference current is measured at an applied voltage. The
pipette then approaches the cell surface until the current is reduced
by a predefined set-point, usually 0.25–1 %. The position of
z-dimension actuator when the current achieves this reduction is
recorded as the height of the sample at this imaging point. The
z-dimension actuator then withdraws the pipette away from the
cell surface to the starting point and the sample is moved laterally
to the next imaging point. The reference current is continuously
updated while the pipette is away from the surface to account for
any slow drifts in the pipette current. This achieves unprecedented
accuracy in revealing the topography of various cell surfaces.
1.2 Applications Since its development in 2002 by Yuri Korchev and colleagues (5, 6),
of Smart Patch-Clamp smart patch-clamp technique has been applied in a wide variety of
experimental settings. The most popular application of smart patch-
clamp is recording ion currents from structures that are too small to
be resolved conventionally or that cannot be detected by light
microscopy. The best example of this kind is L-type calcium channel
192 Anamika Bhargava and Julia Gorelik
1.3 Future of Smart The full potential of smart patch-clamp is yet to be explored. For
Patch-Clamp instance, smart patch-clamp can potentially be used to identify
electrophysiological changes associated with morphological
changes that are associated with cell differentiation from precursor
cells. This approach may prove very useful in characterizing cells
when differentiating from either embryonic stem-cells or induced
pluripotent stem cells.
Another useful implementation would be to combine smart
patch-clamp with studying cell signalling. Ion channels are mem-
brane proteins involved in protein–protein interactions and are
also target proteins for many effector molecules such as second
messengers. Therefore smart patch-clamp can be applied to study
the role of ion channel in cell signalling in precise microdomains of
the cell. Modulation of ion channels by signalling molecules such
as cAMP or cGMP can be studied together with the use of biosen-
sors for these signalling molecules. Successful recent study of cAMP
signalling in different membrane domains of cardiomyocytes using
SICM and imaging techniques presented possible application of
smart patch-clamp to cell signalling studies (13).
2 Materials
2.1 Cells Smart patch-clamp can be applied to a wide variety of cells ranging
from large adult rat cardiomyocytes to small cells like sea urchin
spermatozoa and even to subcellular structures such as neuronal
synaptic boutons (5, 6). Isolated live cells can be evaluated for a
number of ion channels and receptors. In this chapter we describe
Smart Patch-Clamp 193
2.3 Micropipettes Pipettes used in smart patch-clamp are essentially no different from
those used in conventional patch-clamp. The best and most com-
monly used material is borosilicate glass. We use borosilicate glass
capillaries of 1.00 mm outer and 0.5 mm inner diameters (Intracel
LTD or Harvard apparatus). However, the size of pipette tip used
in smart patch-clamp is much smaller than in conventional patch-
clamp in order to obtain a high-resolution topography image.
Once pulled using a laser based puller, there is no need for addi-
tional fabrication such as fire polishing or Sylgard coating for smart
patch-clamp pipettes. We pull pipettes with inner tip diameter of
about 100 nm and when filled with pipette solution that gives an
average tip resistance of 100 MΩ.
2.4 Pipette Puller Although any puller can be used, from our experience laser based
puller (P-2000, Sutter Instrument Co., San Rafael, CA) is a reli-
able instrument for reproducible pulling of pipettes of same shape
and size.
2.5 Instrumentation The instrument uses an inverted microscope (e.g., Diaphot 200 or
for SICM TE200; Nikon Corporation, Tokyo, Japan). On the microscope
stage a computer controlled three axis piezotranslation system with
100 × 100 μm x–y piezo-stage for sample positioning and 25 μm
z-axis piezo-actuator for pipette movement is mounted (ICnano
sample scan system, Ionscope Ltd, UK). The glass micropipette is
mounted on the z-piezo stage and connected to the headstage of an
amplifier. A number of high quality patch-clamp amplifiers are now
available from a number of suppliers. The most common suppliers
are HEKA (Germany) and Molecular devices (Sunnyvale, CA,
USA). Smart patch-clamp can use any of the commercial amplifiers
with either a resistive or capacitive feedback headstage like Axopatch
200B or Multiclamp 700B (Molecular devices Sunnyvale, CA).
Ag/AgCl electrodes, in the bath and pipette, provide an electrical
connection in a conventional electrophysiological circuit.
194 Anamika Bhargava and Julia Gorelik
3 Methods
Fig. 2 Principle of operation of smart patch-clamp. (a) A micropipette approaches the cell surface and reaches
a defined separation distance (d ) whereupon the distance is kept constant by SICM feedback control. (b) The
SICM software scans this micropipette over the cell surface and positions it at a place of interest for patch-
clamp recording. (c) The micropipette is lowered to form a gigaohm seal for patch-clamp recording from the
selected structure. Reproduced with permission from ref. 5
4 Notes
a d
b e
Patch-clamping
0 mV
-80 mV
1 pA
50 ms
0.5pA
50ms
c f
Z-groove T tubule
S
L-type
T-tubule opening Ca2+ channels
(2.0 Ch/µm2)
Fig. 3 LTCC channel distribution in the cardiomyocyte membrane: mapping of ion channels by the smart patch-
clamp technique. (a) Adult rat cardiomyocyte and micropipette as seen optically by the microscope.
(b) Experimental topographic image of a representative rat cardiomyocyte membrane. Z-grooves, T-tubule
opening, and characteristic sarcomere units are marked. (c) Functional schematic of sarcomere units showing
position of the probed region (Z-groove, T-tubule opening and scallop crest). Probabilities of forming a GΩ seal
as a function of surface position are shown in parenthesis. (d) Cell-attached Ba2+ currents at voltages of +20,
0, −20 mV. (e) Several current traces elicited at 0 mV from one patch and ensemble average of 12 traces
showing typical LTCC current kinetics. (f) Statistical distribution of LTCCs with the highest density near the
T-tubule opening. Reproduced with permission from ref. 6
References
1. Alonso G, Widmer H (1997) Clustering of 8. Shevchuk AI, Gorelik J, Harding SE, Lab
KV4.2 potassium channels in postsynaptic MJ, Klenerman D, Korchev YE (2001)
membrane of rat supraoptic neurons: an ultra- Simultaneous measurement of Ca2+ and cellu-
structural study. Neuroscience 77:617–621 lar dynamics: combined scanning ion conduc-
2. Angelides KJ (1986) Fluorescently labelled tance and optical microscopy to study
Na + channels are localized and immobilized to contracting cardiac myocytes. Biophys J 81:
synapses of innervated muscle fibres. Nature 1759–1764
321:63–66 9. Hansma PK, Drake B, Marti O, Gould SA,
3. Alkondon M, Pereira EF, Albuquerque EX Prater CB (1989) The scanning ion-conduc-
(1996) Mapping the location of functional tance microscope. Science 243:641–643
nicotinic and gamma-aminobutyric acidA 10. Korchev YE, Milovanovic M, Bashford CL,
receptors on hippocampal neurons. J Pharmacol Bennett DC, Sviderskaya EV, Vodyanoy I, Lab
Exp Ther 279:1491–1506 MJ (1997) Specialized scanning ion-conduc-
4. Frosch MP, Dichter M (1992) Non-uniform tance microscope for imaging of living cells.
distribution of GABA activated chloride chan- J Microsc 188:17–23
nels in cultured cortical neurons. Neurosci 11. Novak P, Li C, Shevchuk AI, Stepanyan R,
Lett 138:59–62 Caldwell M, Hughes S, Smart TG, Gorelik J,
5. Gorelik J, Gu Y, Spohr HA, Shevchuk AI, Lab Ostanin VP, Lab MJ, Moss GW, Frolenkov GI,
MJ, Harding SE, Edwards CR, Whitaker M, Klenerman D, Korchev YE (2009) Nanoscale
Moss GW, Benton DC, Sanchez D, Darszon live-cell imaging using hopping probe ion
A, Vodyanoy I, Klenerman D, Korchev YE conductance microscopy. Nat Methods 6:
(2002) Ion channels in small cells and subcel- 279–281
lular structures can be studied with a smart 12. Vescovo G, Jones SM, Harding SE, Poole-
patch-clamp system. Biophys J 83:3296–3303 Wilson PA (1989) Isoproterenol sensitivity of
6. Gu Y, Gorelik J, Spohr HA, Shevchuk A, Lab isolated cardiac myocytes from rats with mono-
MJ, Harding SE, Vodyanoy I, Klenerman D, crotaline-induced right-sided hypertrophy and
Korchev YE (2002) High-resolution scanning heart failure. J Mol Cell Cardiol 21:1047–1061
patch-clamp: new insights into cell function. 13. Nikolaev VO, Moshkov A, Lyon AR, Miragoli
FASEB J 16:748–750 M, Novak P, Paur H, Lohse MJ, Korchev YE,
7. Korchev YE, Bashford CL, Milovanovic M, Harding SE, Gorelik J (2010) Beta2-
Vodyanoy I, Lab MJ (1997) Scanning ion con- adrenergic receptor redistribution in heart
ductance microscopy of living cells. Biophys J failure changes cAMP compartmentation.
73:653–658 Science 327:1653–1657
Part III
Abstract
Ion channels are integral membrane proteins that allow the flow of ions across membranes down their
electrochemical gradients and are a major determinant of cellular excitability. They play an important role
in a variety of biological processes as diverse as insulin release from beta cells in the pancreas through to
cardiac and smooth muscle contraction. We have used total internal reflection fluorescence (TIRF) micros-
copy to watch ion channels being transported in vesicles along microtubules within the cytoplasm of the
cell. Furthermore, we can directly observe the fusion of these vesicles with the plasma membrane and the
release and radial dispersion of single ion channels into the membrane. Finally, automated single-particle
tracking of these objects allowed us to determine their diffusional behavior.
Key words Total internal reflection fluorescence microscopy (TIRFM), Single molecule detection,
Potassium channel, Vesicle, Fluorescence, Plasma membrane
1 Introduction
Membranes consist of a lipid bilayer which makes them impermeable
for most molecules including ions. In biological membranes, ion
channels form pores which, with varying selectivity, allow ions to
pass at a high rate through the membrane down their electrochemical
gradient. Ion channel activity can be modulated directly by ligand
binding, membrane potential, second messengers (such as Ca2+ or
cyclic-AMP), protein–protein interactions, and phosphorylation
(1). Potassium channels are the largest family of ion channels and
they are important for maintenance of the membrane resting poten-
tial, hormone secretion, regulation of excitability, repolarization of
the action potential, and its shape and frequency (2–4). Potassium
channels are homo- or heteromeric proteins consisting of the pore-
forming alpha subunit and in some cases additional beta subunits,
which can be cytosolic or integral membrane proteins (5).
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_15, © Springer Science+Business Media, LLC 2013
201
202 Sarah Schwarzer et al.
2 Materials
2.1 Cloning 1. pEGFP-N1 vector from Clonetech.
2. mRFP vector (8).
3 Methods
3.1 Generation 1. Standard molecular cloning approaches can be used to fuse the
of Fluorescent relevant potassium channels or other genes of interest in frame
Constructs with a fluorescent protein.
2. In our studies a fusion between human KCNQ1 and the
enhanced variant of the green fluorescent protein (eGFP) was
created using PCR. The long isoform of KCNQ1 without the
termination codon was amplified by PCR using a high-fidelity
polymerase with EcoRI/XbaI restriction sites and subcloned
into pcDNA3. eGFP was amplified by PCR including 5¢ and 3¢
XbaI and ApaI restriction sites and cloned in frame to the
C-terminus of KCNQ1 in pcDNA3. An alternative strategy is
to use the eGFP vectors (see Note 2).
204 Sarah Schwarzer et al.
3.2 HL-1 Cell 1. Seed HL-1 cells at very low density (1:20) on 25 mm glass
Preparation and coverslips 48 h before transfection.
Transfection 2. Transfect the cells using FuGENE HD transfection reagent;
the transfection complex consists of 97 μL Claycomb medium,
5 μL FUGENE HD, and the 1–2 μg of DNA. Add this mix to
the cell culture and incubate for 24 h.
3. Perform experiments 24–48 h after transfection (see Note 3).
3.3 HEK293 Cell 1. Seed HEK293 cells at a higher density (1:5) in 35-mm dishes
Preparation and 24 h before transfection.
Transfection 2. Transfect the cells using Lipofectamine 2000 or TurboFECT
and split the day after onto 25 mm glass coverslips at a very low
density (1:15) (see Note 4).
3.4 Solution and 1. It is important to use Hank’s buffered salt solution for TIRF
Imaging Chamber microscopy to reduce background fluorescence; additionally it
buffers the pH in our sealed imaging chamber.
2. For lengthy experiments it is advisable to use medium with the
addition of 10% FCS. Our experience shows that the fully
sealed, custom-built, imaging chamber works best.
3. Cells cultured on 25 mm glass coverslips are placed in the
chamber topped by a silicone O-ring and covered by another
coverslip to seal the chamber. This custom-built chamber
allows one (via two ports on opposite sides of the chamber) to
insert hypodermic syringe needles connected to syringes to fill
the chamber with the medium.
4. The chamber is fixed securely to the stage of the microscope.
5. The temperature during the experiment is regulated by using a
microscope 37 °C incubator and a custom-made airflow heater
system.
Fig. 1 TIRF microscope images of HEK293 cells transfected with KCNQ1-GFP. (a) Image of a video showing
vesicles containing KCNQ1-GFP. (b) Tracking analysis of the video displaying the vesicle movements. (c) Single
molecules (KCNQ1-GFP) in the plasma membrane
the microscope light path. This beam was then directed onto a
position-sensitive photodiode to enable automatic focus
control.
2. To visualize eGFP and mRFP in the same specimen, appropri-
ate laser excitation (488 or 532 nm) and emission filter
(ET525/50M for eGFP; HQ590/50M for mRFP) were
selected.
3. Important criteria for the choice of camera are high sensitivity,
low readout noise and fast acquisition rate (20–50 frames s-1).
We used an electron multiplied CCD camera (EMCCD), which
is currently the best choice for high-speed single fluorophore
imaging within living cells because the signal generated at every
CCD element is multiplied before the readout giving increased
sensitivity and lower relative readout noise (see Note 5).
3.6 Image Analysis 1. Vesicles can be identified as bright punctate objects moving
just below the plasma membrane in following straight-line tra-
jectories (Fig. 1a, b). On occasion these vesicles would stall
and fuse with the membrane and single molecules (Fig. 1c)
would radially disperse from the site of fusion (see Note 6).
2. Video sequences are stored to computer hard disc and then
analyzed using custom-written image processing software,
which is available free of charge for academic users (http://
www.nimr.mrc.ac.uk/gmimpro/) (10). The computer pro-
gram automatically detects and tracks individual fluorescent
spots which are diffraction limited in size, have an average
intensity corresponding to that of a single fluorophore (mea-
sured under controlled conditions), and which show single-
step photobleaching. The software allows simultaneous
tracking of multiple objects (up to a thousand) within each
record and produces an output file that contains the inte-
206 Sarah Schwarzer et al.
4 Notes
1. Contact details: Professor William C. Claycomb, PhD,
Department of Biochemistry and Molecular Biology, 1901
Perdido St., Box P7-2, Medical Education Building Room
7238, New Orleans, LA 70112, [email protected].
2. Fluorescent proteins are convenient and widely used to track
protein localization and biogenesis. However the protein tag
can affect function and this has to be independently assessed.
For instance, here we were able to check the function of
KCNQ1-GFP channels by patch-clamping and comparing our
results with those obtained from wild-type KCNQ channels
(11). A drawback of fluorescent protein tags is that they are
not as bright and photobleach more rapidly than small organic
fluorophores (such as Cy3B). We have found that observation
of single eGFP fluorophores within live cells is readily achiev-
able but requires careful choice of equipment and optimization
of imaging conditions (12).
3. For single molecule experiments with TIRFM very low cDNA
concentrations are necessary. Systematic variations of the trans-
fection conditions are important to optimize a low level of pro-
tein expression. For example it may be necessary to vary the
amount of DNA, change the transfection reagent, the transfec-
tion time or cell density. For example, the transfected DNA
concentration for KCNQ1 was 40 ng in our studies (6). In
some circumstances we would transfect a range of concentra-
tions and use the one that worked best.
4. If several plasmids need to be transfected together it can be
helpful to sequentially transfect them one after the other to
ensure comparable expression at the site of interest. This arises
from differences in protein turnover rates of the various con-
structs. Similar titrations of transfection conditions might also
be necessary with HEK293 cells.
5. We found that image intensified CCD cameras (ICCDs) have
sufficient sensitivity and signal-to-noise ratio to image single
fluorophores, but the intensifier system creates spatial and tem-
poral noise which makes them inferior to EMCCD cameras.
TIRF Microscopy for Ion Channel Trafficking 207
Acknowledgments
References
1. Cooper EC, Jan LY (1999) Ion channel genes Ligand and voltage-gated ion channels. CRC,
and human neurological disease: recent prog- Boca Raton, FL, pp 1–71
ress, prospects, and challenges. Proc Natl Acad 4. Isomoto S, Kondo C, Kurachi Y (1997)
Sci U S A 96:4759–4766 Inwardly rectifying potassium channels: their
2. Jan LY, Jan YN (1997) Cloned potassium molecular heterogeneity and function. Jpn J
channels from eukaryotes and prokaryotes. Physiol 1997(47):11–39
Annu Rev Neurosci 20:91–123 5. Tinker A (2002) The assembly and targeting of
3. Chandy KG, Gutman GA (1995) Voltage- potassium channels. In: Henley J, Moss SJ (eds)
gated potassium channel genes. In: North RA The assembly and targeting of ion channels.
(ed) Handbook of receptors and channels. Oxford University Press, Oxford, pp 28–57
208 Sarah Schwarzer et al.
6. Mashanov GI, Nobles M, Harmer SC, Molloy use of cultured HL-1 cardiomyocytes for
JE, Tinker A (2010) Direct observation of studies of cardiac muscle cell structure and
individual KCNQ1 potassium channels reveals function. Am J Physiol Heart Circ Physiol
their distinctive diffusive behaviour. J Biol 286:H823–H829
Chem 285:3664–3675 10. Mashanov GI, Molloy JE (2007) Automatic
7. Claycomb WC, Lanson NA Jr, Stallworth BS, detection of single fluorophores in live cells.
Egeland DB, Delcarpio JB, Bahinski A, Izzo Biophys J 92:2199–2211
NJ Jr (1998) HL-1 cells: a cardiac muscle cell 11. Wilson AJ, Quinn KV, Graves FM, Bitner-
line that contracts and retains phenotypic char- Glindzicz M, Tinker A (2005) Abnormal
acteristics of the adult cardiomyocyte. Proc KCNQ1 trafficking influences disease patho-
Natl Acad Sci U S A 95:2979–2984 genesis in hereditary long QT syndromes
8. Campbell RE, Tour O, Palmer AE, Steinbach (LQT1). Cardiovasc Res 67: 476–486
PA, Baird GS, Zacharias DA, Tsien RY (2002) 12. Nenasheva TA, Mashanov GI, Peckham M,
A monomeric red fluorescent protein. Proc Molloy JE (2011) Imaging individual myosin
Natl Acad Sci U S A 99:7877–7882 molecules within living cells. Methods Mol
9. White SM, Constantin PE, Claycomb WC Biol 778:123–142
(2004) Cardiac physiology at the cellular level:
Chapter 16
Abstract
Understanding the molecular mechanisms of protein–protein interactions at the cell surface of living cells
is fundamental to identifying the nature of cellular processes. Here, we discuss how fluorescence-based
approaches have been successfully developed to visualize protein–protein interactions in living cells. Förster
resonance energy transfer (FRET) is unique in generating fluorescence signals between proteins that are
highly spatially sensitive. Furthermore, total internal reflectance fluorescence (TIRF) microscopy com-
bined with FRET is a robust technique used to assay protein/protein interactions and the functionality of
proteins assembled at the cell surface membrane.
Key words Protein–protein interactions, Cell surface, FRET, TIRF, Live-cell imaging
1 Introduction
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_16, © Springer Science+Business Media, LLC 2013
209
210 Sonya M. Bierbower and Mark S. Shapiro
2 Materials
2.2 Software 1. NIS-Elements for image acquisition and data analysis. The
software controls laser light delivery, microscope, and the
EMCCD camera (Nikon Instruments).
2. Microsoft Excel (used in conjunction with NIS-Elements for
formula-based data analysis).
2.3 Epifluorescence 1. Chinese Hamster Ovary (CHO) or other suitable cell line
Imaging expressing a membrane targeted tandem construct of eCFP
and eYFP (Rho-pYC) to act as a positive control for strong
FRET efficiency. It consists of the C-terminal prenylation site
of Rho (RQKKRRGCLLL) appended to the C-terminus of a
YFP-CFP fusion protein (10) (see Note 1).
2. CHO (or equivalent) cells expressing the CFP-tagged protein
to be tested and eYFP-M (membrane bound). These act as a
negative control for FRET efficiency, since they are known to
be too far apart in the membrane to exhibit FRET, and make a
good control for “spurious” or incidental FRET.
3. Tissue-culture cells expressing the eCFP-tagged and eYFP-
membrane bound proteins that you wish to assay for molecular
interactions.
4. Cell imaging solution: 160 mM NaCl, 5 mM KCl, 1 mM
MgCl2, 2 mM CaCl2, 10 mM HEPES, pH 7.4 with NaOH.
Other suitable saline solutions are equally suitable.
5. Glass-bottom culture dishes with No. 1.5 coverglass (0.16–
0.19 mm) on the bottom, 35 mm (Mat-tek) (see Note 2).
6. Objective immersion oil.
In this chapter we are not describing protocols (and therefore not
listing any materials required) for cell culture and transfection as these
can be found elsewhere (i.e., Chapters 2, 4, and 5 of this book).
3 Methods
3.1 TIRF/FRET It is most convenient to image eCFP and eYFP emission simulta-
Experiments on neously, which can be easily performed using the dual-view chip
Cultured Cells Using splitter, which is put between the side-camera port and the CCD
Epifluorescence camera. This device is equipped with a filter cube containing a
Microscopy dichroic mirror, and emission filters of your choice. For concur-
rent eCFP/eYFP imaging, we use an HQ470 nm/30 m and
Live TIRF-FRET Imaging 213
3.2 Imaging of Cells 1. Just prior to fluorescence imaging, replace the cell culture
medium with an appropriate cell imaging solution
(see Subheading 2.3, step 4).
2. Place the chamber on the stage on top of the oil immersion
60× objective (1.45 N.A.).
3. Using bright field transmitted light, focus on an isolated cell.
Note that there will not be contrast optics in the light path, so
focusing on the cells will be hard. We often “find” the cells
under epifluorescence, without using transmitted light
(see Note 3).
4. Using the 200 W metal-halide lamp (which gives similar spec-
tral output to more traditional mercury vapor lamps) and alter-
nating CFP and YFP filter cubes, pick a cell with robust
expression of both fluorophores (see Note 4).
5. Under TIRF illumination and either the TIRF CFP or YFP
filter cubes, the focal plane is adjusted immediately before each
image acquisition to obtain a “sharp” TIRF image. The mem-
brane proteins should appear punctate if the cell is focused
correctly (see Note 5).
6. Take the “before” photobleaching images of the cell using the
442 and 514 nm laser lines (see Note 6).
7. Photobleach the eYFP fluorophores by using the metal-halide
lamp and the wide-field YFP filter cube for a minimum of
7–10 min, which should be sufficient to achieve >80% photo-
bleaching of the eYFP. Photobleaching is done through the
wide-field metal-halide lamp since the molecules in live cells
are diffusible in the membrane and are likely to move. Thus,
wide-field illumination allows for the eYFP fluorophores in
214 Sonya M. Bierbower and Mark S. Shapiro
æ (eCFPpost - eCFPpre ) ö
%FRETefficiency = ç ÷ ´ 100,
è eCFPpre ø
where CFPpre and eCFPpost are the CFP emissions before and after
YFP photobleaching, respectively.
Using the protocol detailed in this chapter, Fig. 1 shows repre-
sentative images of cells transfected with CFP and YFP before and
after YFP photobleach. Specifically, Chinese hamster ovary cells
were transfected with CFP-tagged angiotensin II AT1 receptors
and YFP-tagged KCNQ3 K+ channels. Here, FRET was measured
under TIRF illumination by the donor dequenching method and
CFP emission was significantly stronger after YFP photobleach,
indicating robust FRET.
Positive controls are used as a measure of the highest per-
centage of FRET possible. Rho-pYC is a CFP and YFP tandem
Live TIRF-FRET Imaging 215
Fig. 1 Shown are images of Chinese hamster ovary cells transfected with CFP-tagged angiotensin II receptors
(AT1R) and YFP-tagged KCNQ3 K+ channels under TIRF illumination, using 442 nm or 514 nm laser lines,
respectively. Images of eCFP (left, in “rainbow pseudocolor” ) and eYFP (right, in yellow pseudocolor) emissions
are shown before or after YFP photobleach, as labeled. Note the significantly brighter eCFP emission (warmer
colors) after eYFP photobleach and the profoundly dimmer eYFP emission after 7 min photobleach under wide-
field illumination with the YFP filter cube
4 Notes
References
1. Centonze VE, Sun M, Masuda A, Gerritsen H, 7. Bal M, Zaika O, Shapiro MS (2008)
Herman B (2003) Fluorescence resonance Calmodulin binding to M-type K + channels
energy transfer imaging microscopy. Methods assayed by TIRF/FRET in living cells. J Physiol
Enzymol 360:542–560 586:2307–2320
2. Stryer L (1978) Fluorescence energy transfer 8. Bal M, Zhang J, Hernandez CC, Zaika O,
as a spectroscopic ruler. Annu Rev Biochem Shapiro MS (2010) Ca2+/calmodulin
47:819–846 disrupts AKAP79/150 interactions with
3. Erijman EA, Jovin T (2003) FRET imaging. KCNQ (M-Type) K + channels. J Neurosci
Nat Biotechnol 23:1387–1395 30: 2311–2323
4. Axelrod D, Thompson NL, Burghardt TP 9. Riven I, Iwanir S, Reuveny E (2006) GIRK
(1983) Total internal reflection fluorescent channel activation involves a local rearrange-
microscopy. J Microsc 129:19–28 ment of a preformed G protein channel com-
5. Funatsu TY, Harada M, Tokunaga K, Saito T, plex. Neuron 51:561–573
Yanagida T (1995) Imaging of single 10. Fowler CE, Aryal P, Suen KF, Slesinger PA
fluorescent molecules and individual ATP (2007) Evidence for association of GABA(B)
turnovers by single myosin molecules in aque- receptors with Kir3 channels and regulators of
ous solution. Nature 374:555–559 G protein signalling (RGS4) proteins. J Physiol
6. Boyer SB, Slesinger PA, Jones SV (2009) 580:51–65
Regulation of Kir2.1 channels by the Rho-
GTPase, Rac1. J Cell Physiol 218:385–393
Chapter 17
Abstract
Steady-state fluorescence spectroscopy is a biophysical technique widely employed to characterize
interactions between proteins in vitro. Only a few proteins naturally fluoresce in cells, but by covalently
attaching fluorophores virtually all proteins can be monitored. One of the first extrinsic fluorescent probes
to be developed, and that is still in use, is dansyl chloride. We have used this method to monitor the inter-
action of a variety of proteins, including ion channels, with the Ca2+-dependent regulatory protein calm-
odulin. Here we describe the preparation and use of dansyl-calmodulin (D-CaM).
Key words Fluorescence spectroscopy, Protein purification, In vitro binding, Protein labelling
1 Introduction
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_17, © Springer Science+Business Media, LLC 2013
217
218 Alessandro Alaimo et al.
a
N(CH3)2
NH NH O
C NH2 + C N S
H
HCl O
O C O C
O S O N(CH3)2
NH NH
Cl
Fluorescence Emission
(Arbitrary Units)
(Arbitrary Units)
Absorption
Excitation Emission
300 350 400 450 500 550 600
Wavelength (nm)
Fig. 1 Dansyl reactivity and its fluorescent properties. (a) Reaction scheme for the dansyl chloride labelling of
a lysine residue. (b) Fluorescence emission spectrum of D-CaM (solid line, excitation at 340 nm) and excitation
spectrum (dotted line, emission at 500 nm) in binding buffer
a 10 b 10
8 8
Emission (Arbitrary Units)
4 4
2 2
0 0
400 450 500 550 600 650 400 450 500 550 600 650
Wavelength (nm) Wavelength (nm)
Fig. 2 Changes in the D-CaM fluorescence spectrum induced by peptide and/or calcium. (a) Emission spec-
trum of D-CaM alone (12.5 nM, grey dotted spectrum) and after addition of 200 nM GST-Kv7.2 CBD (black solid
spectrum) in the absence of Ca2+ (10 mM EGTA). (b) Emission spectrum of D-CaM alone (12.5 nM, grey dotted
spectrum), and after the addition of 2 μM free Ca2+ (black solid spectrum) and the successive addition of
200 nM GST-Kv7.2 CBD (black bold spectrum). Note the blue shift in the spectra in the presence of Ca2+
2 Materials
2.1 Purification 1. LB medium (in grams per liter of distilled water: Tryptone 10,
of GST Fusion Proteins yeast extract 5, and NaCl 5) sterilized by autoclaving.
2. 100 mg/mL ampicillin stock solution sterilized by filtration
and stored at −20 °C.
3. 1 M isopropyl β-d-1-thiogalactopyranoside (IPTG) solution
sterilized by filtration and stored at −20 °C.
4. Dithiothreitol (DTT).
5. GST buffer: 20 mM Tris–HCl (pH 7.4), 100 mM NaCl, 0.5%
Triton X-100, 2 mM DTT, half a tablet of the protease inhibi-
tor cocktail “1× Complete” (Roche).
6. Solubilization buffer: 20 mM Tris–HCl (pH 7.4), 100 mM
NaCl, 2 mM DTT, and 6 M urea.
7. Refolding buffers: 20 mM Tris–HCl (pH 7.4), 100 mM NaCl,
and 2 mM DTT, containing 4, 2, 1, 0.5, or 0 M urea. Filter all
the buffers before use.
8. Glutathione-Sepharose 4B (GE Healthcare) and glutathione
reduced (Sigma-Aldrich).
9. Elution buffer: 50 mM Tris–HCl (pH 8.5) and 15 mM gluta-
thione reduced.
10. pGEX expression vectors (GE Healthcare).
11. BL21-DE3 (Novagen) and BL21-Codon plus (Agilent
Technologies) competent cells.
12. General laboratory equipment: Spectrophotometer, orbital
incubator, ultracentrifuge with fixed-angle rotor, microcentri-
fuge, ultrasonic probe sonicator, standard dialysis tubing,
filters, protein concentrators, magnetic stirrer, small columns
(1–5 mL bed volume), electrophoresis chamber and buffers,
SDS-PAGE gels, and standard material for protein detection
and staining.
3 Methods
3.1 Purification The method we use to produce recombinant proteins of the Kv7.2
of GST Fusion Proteins CBD (12) is detailed here. The constructs are cloned into pGEX
expression vectors and while the BL21-DE3 Escherichia coli is
strain generally employed to transform the plasmids, we also rec-
ommend the use of BL21-Codon plus.
1. Grow the cells at 37 °C in 1 L of LB medium containing
100 μg/mL ampicillin until A600 = 0.6–0.8. Induce protein
expression for 3 h at 30 °C with 0.3 mM IPTG.
2. Harvest the cells by centrifugation and resuspend the pellet in
20 mL of chilled GST buffer. After lysis by sonication, remove
the cell debris by centrifugation for 30 min at 80,000 × g and
4 °C. Transfer the supernatant (soluble fraction) to a fresh tube
while the insoluble cell lysate (inclusion bodies) is treated
separately.
3. Resuspend the inclusion bodies in 10 mL GST buffer and then
centrifuge again to remove any remaining soluble material. Repeat
this operation three times. Dissolve the precipitate in 5–10 mL
solubilization buffer for 30 min at 4 °C, mixing occasionally.
222 Alessandro Alaimo et al.
3.2 Purification The rat CaM gene cloned into the pET-14b expression vector is
and Dansylation transformed in BL21-DE3 E. coli (see Note 4). The protocol
of Calmodulin employed to purify CaM has been adapted from the literature (15)
and it yields large amounts of soluble protein (see below). Finally,
to some extent we follow the instructions described in the litera-
ture to covalently attach a dansyl group to CaM (8).
3.3 Fluorescence In this section, we describe the preparation of the samples and the
Experiments methods employed to perform fluorescence assays with D-CaM.
We present three experiments: two are spectrophotometric titra-
tions to measure fluorescence of a solution of D-CaM upon succes-
sive addition of ligand (Ca2+ and/or peptide), while the third is a
competition assay. Generally, the emission spectra are recorded
while adding increasing amounts of the target until no further
change is observed. Finally, the data are analyzed to obtain infor-
mation about the protein interactions, conformational changes in
224 Alessandro Alaimo et al.
Table 1
Calculated free Ca2+ concentrations as a function of total Ca2+ (EGTA
10 mM, pH 7.4)
(Ca2+) total (mM) (Ca2+) free (μM) (Ca2+) total (mM) (Ca2+) free (μM)
0 0 8.385 0.408
2.498 0.024 8.634 0.499
4.246 0.055 8.784 0.571
4.995 0.075 9.033 0.741
5.244 0.083 9.182 0.894
5.743 0.102 9.431 1.325
6.242 0.127 9.581 1.827
6.741 0.159 9.628 2.000
7.239 0.203 9.829 4.539
7.488 0.231 9.979 19.77
7.987 0.310 10.22 231.4
8.136 0.341 10.47 4,735
4 Notes
1. Rat CaM does not contain tryptophan residues but there are
five phenylalanines in the N-terminal domain and three in the
C-terminal domain; there are also two tyrosine residues in
Using Dansyl-Calmodulin 227
100
Relative fluorescence % 80
60
40
20
0
0 50 100 150 200
[Competing Peptide] (nM)
4
3 Competing
Ligand peptide
2
1
0
400 500 600
Fig. 3 Competition assay. Example of the competition of a peptide that does not cause an increase in D-CaM
fluorescence emission. First, the baseline is obtained by mixing D-CaM with a ligand at a concentration cor-
responding to its calculated EC50. The ligand is displaced from CaM by the increasing addition of a competing
peptide. Below, schematic representation of the experiment
Fig. 4 1H,15N-HSQC for dansylated (black) and wild type calmodulin (grey ) in the presence of 5 mM calcium.
The peak distribution is similar indicating that both proteins fold in a similar way. The changes in the chemical
shift observed throughout the molecule are an indication of multiple dansylation sites in the two protein
domains
Acknowledgments
References
1. Walker JM (1994) The dansyl method for activating) calmodulin derivatives. Biochemistry
identifying N-terminal amino acids. Methods 37:6188–6198
Mol Biol 32:321–328 9. Ehlers MD, Zhang S, Bernhadt JP, Huganir
2. Kincaid RL, Vaughan M (1986) Direct com- RL (1996) Inactivation of NMDA receptors
parison of Ca2+ requirements for calmodulin by direct interaction of calmodulin with the
interaction with and activation of protein NR1 subunit. Cell 84:745–755
phosphatase. Proc Natl Acad Sci U S A 83: 10. El Far O, Bofill-Cardona E, Airas JM,
1193–1197 O’Connor V, Boehm S, Freissmuth M, Nanoff
3. Johnson JD, Wittenauer LA (1983) A C, Betz H (2001) Mapping of calmodulin and
fluorescent calmodulin that reports the binding Gbg binding domains within the C-terminal
of hydrophobic inhibitory ligands. Biochem J region of the metabotropic glutamate receptor
211:473–479 7A. J Biol Chem 276:30662–30669
4. Kincaid RL, Vaughan M, Osborne JC Jr, 11. Schleiff E, Schmitz A, McIlhinney RA, Manenti
Tkachuk VA (1982) Ca2+-dependent interac- S, Vergères G (1996) Myristoylation does not
tion of 5-dimethylaminonaphthalene-1- modulate the properties of MARCKS-related
sulfonyl-calmodulin with cyclic nucleotide protein (MRP) in solution. J Biol Chem 271:
phosphodiesterase, calcineurin, and troponin 26794–26802
I. J Biol Chem 257:10638–10643 12. Alaimo A, Gomez-Posada JC, Aivar P,
5. Olwin BB, Storm DR (1983) Preparation of Etxeberría A, Rodriguez-Alfaro JA, Areso P,
fluorescent labeled calmodulins. Methods Villarroel A (2009) Calmodulin activation lim-
Enzymol 102:148–157 its the rate of KCNQ2 K + channel exit from
6. Malencik DA, Anderson SR (1983) Binding of the endoplasmic reticulum. J Biol Chem
hormones and neuropeptides by calmodulin. 284:20668–20675
Biochemistry 22:1995–2001 13. Dick IE, Tadross MR, Liang H, Tay LH, Yang
7. Mori M, Konno T, Ozawa T, Murata M, Imoto W, Yue DT (2008) A modular switch for spa-
K, Nagayama K (2000) Novel interaction of tial Ca2+ selectivity in the calmodulin regula-
the voltage-dependent sodium channel (VDSC) tion of CaV channels. Nature 451:830–834
with calmodulin: does VDSC acquire calmodu- 14. Bradford MM (1976) A rapid and sensitive
lin-mediated Ca2+-sensitivity? Biochemistry 39: method for the quantitation of microgram quan-
1316–1323 tities of protein utilizing the principle of protein-
8. Torok K, Cowley DJ, Brandmeier BD, Howell dye binding. Anal Biochem 72:248–254
S, Aitken A, Trentham DR (1998) Inhibition 15. Hayashi N, Matsubara M, Takasaki A, Titani
of calmodulin-activated smooth-muscle myo- K, Taniguchi H (1998) An expression system
sin light-chain kinase by calmodulin-binding of rat calmodulin using T7 phage promoter in
peptides and fluorescent (phosphodiesterase- Escherichia coli. Protein Expr Purif 12:25–28
Using Dansyl-Calmodulin 231
16. Wallace RW, Tallant EA, Cheung WY (1983) specific intrinsic phenylalanine and tyrosine
Assay of calmodulin by Ca2+-dependent fluorescence. Biophys J 83:2767–2780
phosphodiesterase. Methods Enzymol 102: 21. Gao J, Yao Y, Squier TC (2001) Oxidatively
39–47 modified calmodulin binds to the plasma mem-
17. Chen RF (1968) Dansyl labeled proteins: brane Ca-ATPase in a nonproductive and con-
determination of extinction coefficient and formationally disordered complex. Biophys J
number of bound residues with radioactive 80:1791–1801
dansyl chloride. Anal Biochem 25:412–416 22. Leclerc E, Corti C, Schmid H et al (1999)
18. Yus-Nájera E, Santana-Castro I, Villarroel A Serine/threonine phosphorylation of calmod-
(2002) The identification and characterization ulin modulates its interaction with the binding
of a noncontinuous calmodulin-binding site domains of target enzymes. Biochem J 344
in noninactivating voltage-dependent KCNQ (Pt 2):403–411
potassium channels. J Biol Chem 277: 23. Liu M, Chen TY, Ahamed B, Li J, Yau KW
28545–28553 (1994) Calcium-calmodulin modulation of the
19. Pitt GS, Zuhlke RD, Hudmon A, Schulman olfactory cyclic nucleotide-gated cation chan-
H, Reuter H, Tsien RW (2001) Molecular nel. Science 266:1348–1354
basis of calmodulin tethering and Ca2+- 24. Turner JH, Gelasco AK, Raymond JR (2004)
dependent inactivation of L-type Ca2+ chan- Calmodulin interacts with the third intracellu-
nels. J Biol Chem 276:30794–30802 lar loop of the serotonin 5-hydroxytryptam-
20. VanScyoc WS, Sorensen BR, Rusinova E, Laws ine1A receptor at two distinct sites: putative
WR, Ross JB, Shea MA (2002) Calcium binding role in receptor phosphorylation by protein
to calmodulin mutants monitored by domain- kinase C. J Biol Chem 279:17027–17037
Chapter 18
Abstract
This chapter describes immunochemistry-based methods to investigate recycling of membrane proteins at
the cell surface. Two methods are described, one qualitative and the other quantitative. Both methods
consist of two rounds of extracellular antibody capture. Firstly, a primary antibody is captured by an extra-
cellular epitope presented by the target membrane protein and is subsequently internalized. Secondly, the
primary antibody-labelled protein is recycled back to the membrane where it is captured by a probe-
conjugated secondary antibody. In the qualitative assay, the probe is a fluorophore, which can be imaged
by fluorescence microscopy. In the quantitative assay, the probe is horse-radish peroxidase (HRP) and
enzyme activity can be assayed by chemiluminescence.
1 Introduction
Recycling membrane proteins are internalized from the plasma
membrane into early and sorting endosomes from where they can
either return to the cell surface via a Rab4-dependent pathway (1)
or enter the endosomal recycling compartment (ERC) from where
their return to the plasma membrane occurs via Rab11-dependent
pathways (2). A limited number of membrane proteins use a some-
what convoluted pathway, involving trafficking from the ERC to
the cell surface via the trans-Golgi network (3). Trafficking mecha-
nisms for recycling are extensively reviewed by Maxfield and
McGraw (4) and Grant and Donaldson (5).
Recycling of internalized membrane proteins back to the cell
surface has been reported for a range of membrane proteins includ-
ing ion channels, receptors, and transporters. It represents an
efficient mechanism by which prompt delivery of proteins to the
plasma membrane occurs in response to changes in cell physiology.
For example, recycling endosomes supply APMA receptor for
long-term potentiation in neurons (6), b-adrenergic receptors for
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_18, © Springer Science+Business Media, LLC 2013
233
234 Christopher J. Cockcroft
2 Materials
2.1 Preparation of 1. No. 7 curved watchmaker forceps.
Coverslips and Cells 2. Borosilicate glass coverslips (13 mm).
3. HEK293 cell line (ECACC).
4. Dulbecco’s phosphate buffered saline (DPBS).
5. Poly-L-lysine coating solution: 0.01% poly-L-lysine
(70–150 kDa, Sigma), diluted to 0.001% in DPBS.
6. 70% ethanol.
7. Culture medium: DMEM + Glutamax-1, supplemented with
10% fetal bovine serum, penicillin (50 U), streptomycin
(50 mg).
8. Trypsin solution: 0.05% trypsin–EDTA.
9. Suitable transfection reagent available from a range of suppliers
(see Chapters 2, 4, and 5 of this book for more details on
transfection techniques and reagents).
Imaging of Ion Channel Recycling 235
Fig. 1 Visualization of KATP-HA channel recycling by confocal fluorescent microscopy. (a) Schematic diagram
showing the key stages of the antibody capture experiment. (b) Confocal images of cells expressing KATP-HA
following fluorescent-conjugated antibody capture by recycled channels at the cell surface. KATP-HA channel
recycling was detected after 30 min incubation at 37°C. Following fixation and permeabilization steps, non-
recycled channels were stained using anti-rat Cy3 secondary antibody (1:500 dilution, 1.25 mg/mL). The cell
nuclei were stained using DAPI (supplied in the mounting medium) (scale bar = 10 mm). Inset, grey value
profiles of line scans through cross sections of cell images (scale bar = 50 grey value)
a
INTERNALISATION COOLING RECYCLING
1° ANTIBODY
HRP CONJUGATED
2° ANTIBODY
b c 3.0
8x106 60 min
2.5
7x106
Normalised Recycling
6x106 2.0
Luminescence
50 min
5x106 40 min
30 min 1.5
4x106 20 min
1.0
3x106
10 min
2x106 0 min 0.5
6
1x10
Ctrl 0.0
0
0 10 20 30 40 50 60 70 80 0 10 20 30 40 50 60
Time (s) Time (min)
Fig. 2 Quantification of KATP-HA channel recycling channels by chemiluminescence. (a) Schematic diagram
showing the key stages of the antibody labelling experiment. (b) Representative real-time chemiluminescence
data from cell lysates following HRP-conjugated antibody capture by recycled channels at the cell surface after
0, 10, 20, 30, 40, 50, and 60 min at 37°C. Cells expressing SUR1alone were used as a control for nonspecific
antibody binding (Ctrl). (c) The time-course plot showing that KATP-HA is continuously recycled in a time-
dependent manner
3 Methods
3.1 Preparation HEK-293 cells are grown on poly-lysine-coated coverslips placed
of Cells in a 24-well dish. The cells are transfected with an expression vec-
tor construct containing the cDNA of interest engineered to
express an extracellular HA-epitope. If necessary, additional cells
should also be prepared for introducing controls for additional
variables, such as transfection efficiency and antibody specificity.
1. Gently mix 200–300 coverslips with 20 mL poly-L-lysine coat-
ing solution for 2 h at room temperature. Rinse ten times with
distilled water and store in 70% ethanol at 4°C until required.
2. In a tissue culture cabinet, using forceps, place a coverslip into
each well of a 24-well plate, using as many wells as appropriate
for the experiment. To speed up drying time, place each cover-
slip so that it rests against the wall of the well. Once dry, gently
tap the plate so that the coverslips fall to the bottom of the
well. Add 0.5 mL culture medium per well.
3. Remove the culture medium from HEK 293 cells grown in a
T-25 flask to ~80% confluence (grown in a humid CO2 incuba-
tor supplied with 5% CO2/95% air). Rinse cells with 5–10 mL of
DPBS, add 0.5 mL trypsin solution, and incubate until the cells
are detached. Then add 6 mL of culture medium and resuspend
gently.
4. Add 1–2 drops trypsinized cell suspension (~1 × 105 cells/mL)
per well and incubate overnight (see Note 5).
5. Transfect the cells with plasmid DNA constructs using an
appropriate transfection reagent, following the instructions of
the manufacturer (see Note 5). Incubate for 48–72 h.
238 Christopher J. Cockcroft
3.2 Imaging of This section describes a method for continuous labelling of recy-
Channel Recycling cled KATP-HA channels over a 30 min period using fluorophore-
conjugated secondary antibody for fluorescent microscopy imaging
of recycled channels (Fig. 1).
1. Pipette 30 mL droplets of primary antibody medium onto a
staining support tray.
2. Remove each coverslip from the 24-well plate (step 5,
Subheading 3.1) using forceps and rinse by gently dipping into
a beaker of ice cold PBS (200 mL) for 5–10 s (see Note 6).
Drain excess liquid from the coverslip by dabbing the edge
carefully onto tissue paper, taking care not to completely dry
out the cells on the coverslip.
3. Place each coverslip onto a 30 mL droplet of primary antibody,
with the cells facing toward the droplet (see Note 7). Incubate
at 37°C for 1 h to permit continuous labelling and internaliza-
tion of surface channels (see Note 8).
4. Cool the primary antibody staining support plate to £14°C for
15 min to prevent further trafficking (see Note 9). On ice,
remove each coverslip from the staining support tray and wash
with chilled PBS (as in step 2, using fresh PBS).
5. Place each coverslip onto a droplet of prechilled nonconju-
gated secondary antibody (prepared before use, as step 1).
Incubate at £14°C for 30 min to permit saturation of primary
antibody-labelled surface channels.
6. Remove each coverslip from the nonconjugated secondary anti-
body and wash with chilled PBS (as in step 2, using fresh PBS).
7. Place each coverslip onto a droplet of Fluorophore-conjugated
secondary antibody (prepared before use, as in step 1). Incubate
for 30 min at either 37°C to permit recycling or return to
£14°C conditions to prevent recycling.
8. Wash coverslips with chilled PBS to remove unbound secondary
antibody (as in step 2, using fresh PBS).
9. Fix and permeabilize the cells in Me:Ac (dispense sufficient ice
cold Me:Ac to fill a fixing tray). Submerge the coverslips in
Me:Ac and incubate for 10 min on ice (with the cells facing
upward). Gently perfuse the coverslips with PBS to remove
MeAc (see Note 10).
10. Remove each coverslip from the fixing tray and wash with
chilled PBS to remove any remaining fixative (as in step 2,
using fresh PBS).
Optional steps: the cells can now be stained for primary
antibody-labelled non-recycled channels or specific cellular
compartments, such as early endosomes (see Note 11).
11. Wash the coverslips by gently dipping into in a beaker of
Milli-Q water to remove buffer salts.
Imaging of Ion Channel Recycling 239
3.3 Quantification This section describes a method for quantifying KATP-HA channel
of Channel Recycling recycling in a time-course experiment (Fig. 2). For this experiment,
a full 24-well plate with coverslips is required. In the example given
below, the plate layout consisted of 21 coverslips with cells express-
ing KATP, three for each time period (0, 10, 20, 30, 40, 50, and
60 min). Three coverslips with cells expressing only SUR1 (no
Kir6.2-HA) were used as a negative control for nonspecific anti-
body binding.
Follow steps 1–3 from Subheading 3.2
4. Remove each coverslip from the primary antibody and wash
with chilled PBS (as in step 2, Subheading 3.2, using fresh
PBS).
5. Place each coverslip onto a 30 mL droplet of prechilled HRP-
conjugated secondary antibody (prepared before use as in step
1 of Subheading 3.2). Incubate at £14°C for 30 min to label
the remaining channels at the cell surface only.
6. Pipette 300 mL of lysis buffer into the wells of a 24-well plate
and store on ice or 4°C throughout the experiment.
7. Remove a set of three coverslips for measurement of channels
at the cell surface at the start of the time-course experiment
(0 min recycling). Wash with chilled PBS (as in step 2 of
Subheading 3.2, using fresh PBS), then place each coverslip
into a well of lysis buffer (from step 6), and gently pipette up
and down to dissociate the cells. Incubate overnight at 4°C
with gentle shaking or rocking to complete lysis.
8. Transfer the remaining coverslips on the staining plate to 37°C.
Remove sets of three coverslips at desired time intervals (e.g.,
10 min intervals). Wash with chilled PBS (as in step 2,
Subheading 3.2, using fresh PBS) and place each coverslip into
a well of lysis buffer (from step 7).
9. Assay the amount of protein in the cell lysate with the BCA
assay following the manufacturer’s instructions (see Note 12).
10. Pipette 60 mL of each lysate sample (in duplicate) into a white
microtiter 96-well plate. Assay chemiluminescence at 25°C
using a luminometric equipped plate reader. After recording a
baseline reading for 6 s, dispense 50 mL HRP substrate and
continue to record for a 70 s.
11. Data analysis: subtract the mean luminescence endpoint value
(final three measurements, after the addition of the HRP substrate)
240 Christopher J. Cockcroft
4 Notes
1. Epitope tags other than HA can be used for your protein of
interest (e.g. Myc tag).
2. The antibody concentrations used here is optimal for detection
of Kir6.2-HA in our hands and can be used as a guideline for
future experiments.
3. Alternatively, 1% BSA can also be used; ovalbumin is preferred
because of its relatively low hydrophobic binding properties
that can interfere with solubility of compounds used to disrupt
recycling (e.g., Phorbol-12-myristate-13-acetate).
4. Alternative plate readers can be used that are capable of dis-
pensing luminol into wells and measuring real-time or end-
point luminescence.
5. For imaging experiments, the cells should not exceed ~50%
confluency by the time of the experiment. Whereas for chemi-
luminescence experiments, cells should ideally reach 80%
confluency. The generation of stable cell lines can be useful,
especially for quantitative procedures where large numbers of
coverslips maybe advantageous for multiple experiments.
6. Coverslips can also be washed by perfusion with PBS. This is
useful for consistent high-throughput washes when using an
entire 24-well plate. An upturned lid of a 24-well plate (with
well rims) can be used as a tray for submersion of coverslips.
PBS can be gently perfused and aspirated using a 20 mL syringe
and a vacuum pump, respectively. Approximately 3 × 20 mL
wash steps are sufficient to rinse the coverslips in the tray.
7. Cells should be maintained in a humid environment during
incubation steps on the staining tray. Typically, a wet tissue in
a plastic box with a loose lid is sufficient to prevent the cover-
slips from drying out during incubation steps.
8. The antibody incubation time can be extended to 2 h to
increase the size of the labelled pool of internalized channels
for slowly internalizing channels.
9. The acid strip approach is an alternative to the nonconjugated sec-
ondary antibody block step. This approach involves stripping the
primary antibody from labelled channels at the cell surface. After
the chilled PBS wash step, cells should be washed twice with chilled
Imaging of Ion Channel Recycling 241
Acknowledgement
References
1. van der Sluijs P, Hull M, Webster P, Mâle P, 8. Shewan AM, van Dam EM, Martin S, Luen
Goud B, Mellman I (1992) The small GTP- TB, Hong W, Bryant NJ, James DE (2003)
binding protein rab4 controls an early sorting GLUT4 recycles via a trans-Golgi network
event on the endocytic pathway. Cell (TGN) subdomain enriched in Syntaxins 6 and
70:729–740 16 but not TGN38: involvement of an acidic
2. Wilcke M, Johannes L, Galli T, Mayau V, targeting motif. Mol Biol Cell 14:973–986
Goud B, Salamero J (2000) Rab11 regulates 9. Manna PT, Smith AJ, Taneja TK, Howell GJ,
the compartmentalization of early endosomes Lippiat JD, Sivaprasadarao A (2009) Constitutive
required for efficient transport from early endocytic recycling and protein kinase
endosomes to the trans-golgi network. J Cell C-mediated lysosomal degradation control
Biol 151:1207–1220 K(ATP) channel surface density. J Biol Chem
3. Gokool S, Tattersall D, Seaman MN (2007) 285:5963–5973
EHD1 interacts with retromer to stabilize 10. Smith AJ, Sivaprasadarao A (2008)
SNX1 tubules and facilitate endosome-to- Investigation of K(ATP) channel endocytosis
Golgi retrieval. Traffic 8:1873–1886 by immunofluorescence. In: Lippiat JD (ed)
4. Maxfield FR, McGraw TE (2004) Endocytic Potassium channels, methods in molecular
recycling. Nat Rev Mol Cell Biol 5: biology, vol 491. Humana Press, New York, pp
121–132 69–77
5. Grant BD, Donaldson JG (2009) Pathways 11. Smith AJ, Sivaprasadarao A (2008)
and mechanisms of endocytic recycling. Nat Chemiluminescence assays to investigate mem-
Rev Mol Cell Biol 10:597–608 brane expression and clathrin-mediated endo-
6. Park M, Penick EC, Edwards JG, Kauer JA, cytosis of K(ATP) channels. In: Lippiat JD
Ehlers MD (2004) Recycling endosomes supply (ed) Potassium channels, methods in molecu-
AMPA receptors for LTP. Science 305: lar biology, vol 491. Humana Press, New York,
1972–1975 pp 63–68
7. Yu SS, Lefkowitz RJ, Hausdorff WP (1993) 12. Zerangue N, Schwappach B, Jan YN, Jan LY
Beta-adrenergic receptor sequestration. A (1999) A new ER trafficking signal regulates
potential mechanism of receptor resensitization. the subunit stoichiometry of plasma membrane
J Biol Chem 268:337–341 K(ATP) channels. Neuron 22:537–548
Part IV
Abstract
There is demand for isoform-specific ion channel inhibitors as tools to investigate the biology of endogenous
ion channels and validate them as targets in drug discovery programs. There is also hope that such inhibi-
tors may be new therapeutic agents or provide the foundation for such agents. However, in practice, it is
commonly experienced that inhibitors lack sufficient specificity, fail to distinguish between members of a
class of ion channel, or have other (non-ion channel) off-target effects. Due to their extraordinary specificity,
antibodies offer a potentially attractive strategy for overcoming these problems. Inhibitory antibodies act-
ing at the extracellular face of ion channels are particularly attractive because there is enhanced possibility
for specificity and intracellular delivery methods are not required. Here we describe experience with such
an antibody approach and methodology for generating agents based on anti-peptide polyclonal
antibodies.
Key words Ion channel, Transient receptor potential, Functional antibodies, E3 targeting
1 Introduction
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_19, © Springer Science+Business Media, LLC 2013
245
246 Jacqueline Naylor and David J. Beech
Cav2.1/2
TRPM3
TRPM2
TRPC1
TRPC5
TRPV1
Nav1.7
Nav1.5
Kv3.1
Kv1.2
Eag1
E3
ions
2 Materials
3 Methods
3.1 Epitope Selection Custom-made antisera can be supplied by numerous companies
and Peptide Design with routine expertise in this field, which can be beneficial over
generating antibodies in-house. Regardless of the method chosen,
there are several key steps to follow (see Fig. 2).
250 Jacqueline Naylor and David J. Beech
Synthesise peptide
Add N or C terminal cysteine to allow conjugation to
support peptide (e.g. KLH). Retain excess peptide for
control.
Fig. 2 Methods’ schema. Summary of the methodology for generating anti-peptide polyclonal antibodies that
function as ion channel inhibitor agents
1. Obtain the amino acid sequence of the ion channel target. Use
Kyte-Doolittle hydrophobicity analysis (Protean Software,
Lasergene) (26) to map surface-exposed and membrane-
embedded regions of the protein in order to locate the third
extracellular loop.
2. Design a ~20-mer peptide based on the E3 sequence. Suppliers
of custom-made antisera will provide prediction tools to assist
with this step (see Note 1).
Antibody-Based Ion Channel Inhibitors 251
3.2 Generation 1. Establish which species you require for antibody generation
of Antisera (see Note 2).
2. Obtain an initial bleed prior to immunization in order to col-
lect preimmune sera, an important control for later
experiments.
3. Following an initial immunization with antigenic peptide,
standard immunization protocols are usually continued for at
least 1 month with weekly injections, but can vary as antibody
levels will vary animal to animal.
4. Intermittent test bleeds should be collected to monitor anti-
body titer using an enzyme-linked immunosorbent assay
(ELISA).
5. Final-bleed antisera are usually stored in the presence of sodium
azide as a preservative.
3.3 ELISA ELISA is an important analytical test for determining the titer of
antibodies present in a preparation of antiserum. If a titer is below
1:1,000 (i.e., dilutions of >1:1,000 fail to bind the antigenic pep-
tide) further immunizations are advisable. Ideally the titer will be
better than 1:10,000.
1. Coat wells of a 96-well MAXI-SORP plate with antigenic pep-
tide diluted to 4 mg/mL in a buffer suitable for the peptide.
Leave “no-peptide” control wells blank. Incubate overnight at
4 °C.
2. Wash wells three times with PBS/Tween to remove unbound
peptide.
252 Jacqueline Naylor and David J. Beech
3.4 Antibody Dialysis Antisera are usually preserved in the presence of sodium azide, which
and Purification is included to protect antibody stocks against contaminating infec-
tions. The sodium azide may, however, have unwanted effects on live
cells. Therefore dialysis of the antibody with buffer to remove the
sodium azide is advisable in advance of live cell or tissue experiments.
1. Remove membrane from the roll and cut to 10 cm (wear
gloves).
2. Boil for 10 min in 0.1 M sodium bicarbonate buffer.
3. Wash membrane in distilled water.
4. Boil again for 10 min in 0.1 M sodium bicarbonate buffer.
5. Wash membrane in distilled water. At this point membranes
may be stored in 25% ethanol at 4 °C.
6. For dialysis, soak membranes in PBS until flexible.
7. Fill membrane with 1 mL of antiserum and clamp both ends.
8. Place into a beaker filled with PBS at a volume 500 times that
of the antisera sample.
9. Leave for 48 h at 4 °C with gentle stirring to allow exchange.
10. During this time, change the PBS a total of three times.
11. Store dialyzed antibody in working aliquots −20 or −80 °C and
minimize freeze/thaw cycles.
The final-bleed serum provided will contain a mixture of immu-
noglobulin isotypes, typically 5–10 mg/mL of IgG, of which
100 mg/mL will be specific antibody. If required, antisera can be
subjected to further purification (see Note 3).
Antibody-Based Ion Channel Inhibitors 253
versatile method for the production of such agents, which have the
additional benefit of use in live cell and tissue experiments. However
the use of antibodies is not without limitations.
4 Notes
1. Algorithms are available that can predict regions of a protein
sequence that are likely to be highly antigenic and thus elicit the
best immune response (27). It is also useful to be aware of second-
ary structure and posttranslational modification for the peptide
region, as this may impede antibody access for the full protein.
2. You would usually choose a different species from the one you
will use in your biological studies. Rabbits will generate a large
volume of approximately 60 mL of antiserum and are com-
monly used. Mouse, goat, chicken, llama, etc. are alternatives.
3. Antisera can be purified by Protein A agarose columns. These
consist of bacterial proteins that recognize the non-antigen-
binding regions of antibodies, allowing the achievement of an
immunoglobin-only sample. A purer sample can be achieved
by affinity purification in which the antigenic peptide is cou-
pled to a column. If antiserum is passed through the column,
antibodies that bind to the antigenic peptide are preferentially
retained. They can subsequently be eluted. It is important to
note that problems may arise with such purification steps. For
example, high-affinity components may be lost or preferen-
tially selected depending on how you perform the assay.
Alternatively, a high-affinity fraction may be selected that does
not contain the active ion channel blocker. It is important to
reassess the antibody titer by ELISA following dialysis or
purification to determine whether there has been loss of
activity.
4. Blocking antibodies, like other antibodies, can expose the
investigator to the common technical difficulties experienced,
for example, in western blotting and immunocytochemistry
experiments. For this reason multiple experimental controls
are necessary to minimize off-target effects and increase the
likelihood that inhibition occurs because of an effect on the
ion channel of interest.
5. For the majority of E3 antibodies, 50% channel inhibition is
achieved in approximately 15 min. Such acute antibody effects
allow for a shorter period of exposure, reducing nonspecific or
off-target effects, but complete inhibition is rarely achieved
with over-expressed channels. The mechanism of inhibition
remains unknown, although at least for voltage-gated channels
it would not appear to be due to channel internalization or
down regulation of channels from the cell surface (6).
Antibody-Based Ion Channel Inhibitors 255
References
1. Nauli SM, Alenghat FJ, Luo Y, Williams E, 11. Xu SZ, Beech DJ (2001) TrpC1 is a
Vassilev P, Li X, Elia AE, Lu W, Brown EM, membrane-spanning subunit of store-operated
Quinn SJ, Ingber DE, Zhou J (2003) Ca2+ channels in native vascular smooth muscle
Polycystins 1 and 2 mediate mechanosensation cells. Circ Res 88:84–87
in the primary cilium of kidney cells. Nat Genet 12. Xu SZ, Muraki K, Zeng F, Li J, Sukumar P,
33:129–137 Shah S, Dedman AM, Flemming PK, McHugh
2. Li J, Sukumar P, Milligan CJ, Kumar B, Ma D, Naylor J, Cheong A, Bateson AN, Munsch
ZY, Munsch CM, Jiang LH, Porter KE, Beech CM, Porter KE, Beech DJ (2006) A sphin-
DJ (2008) Interactions, functions, and inde- gosine-1-phosphate-activated calcium channel
pendence of plasma membrane STIM1 and controlling vascular smooth muscle cell motil-
TRPC1 in vascular smooth muscle cells. Circ ity. Circ Res 98:1381–1389
Res 103:e97–e104 13. Xu SZ, Sukumar P, Zeng F, Li J, Jairaman A,
3. Raz-Prag D, Grimes WN, Fariss RN, English A, Naylor J, Ciurtin C, Majeed Y,
Vijayasarathy C, Campos MM, Bush RA, Milligan CJ, Bahnasi YM, Al-Shawaf E, Porter
Diamond JS, Sieving PA (2010) Probing KE, Jiang LH, Emery P, Sivaprasadarao A,
potassium channel function in vivo by intracel- Beech DJ (2008) TRPC channel activation by
lular delivery of antibodies in a rat model of extracellular thioredoxin. Nature 45(1):69–72
retinal neurodegeneration. Proc Natl Acad Sci 14. Naylor J, Li J, Milligan CJ, Zeng F, Sukumar
U S A 107:12710–12715 P, Hou B, Sedo A, Yuldasheva N, Majeed Y,
4. Chioni AM, Fraser SP, Pani F, Foran P, Wilkin Beri D, Jiang S, Seymour VA, McKeown L,
GP, Diss JK, Djamgoz MB (2005) A novel Kumar B, Harteneck C, O’Regan D,
polyclonal antibody specific for the Nav1.5 Wheatcroft SB, Kearney MT, Jones C, Porter
voltage-gated Na+ channel ‘neonatal’ splice KE, Beech DJ (2010) Pregnenolone sulphate-
form. J Neurosci Methods 147:88–98 and cholesterol-regulated TRPM3 channels
5. Fraser SP, Diss JK, Chioni AM, Mycielska ME, coupled to vascular smooth muscle secretion
Pan H, Yamaci RF, Pani F, Siwy Z, Krasowska and contraction. Circ Res 106:1507–1515
M, Grzywna Z, Brackenbury WJ, Theodorou 15. Ciurtin C, Majeed Y, Naylor J, Sukumar P,
D, Koyuturk M, Kaya H, Battaloglu E, De Bella English AA, Emery P, Beech DJ (2010)
MT, Slade MJ, Tolhurst R, Palmieri C, Jiang J, TRPM3 channel stimulated by pregnenolone
Latchman DS, Coombes RC, Djamgoz MB sulphate in synovial fibroblasts and negatively
(2005) Voltage-gated sodium channel expres- coupled to hyaluronan. BMC Musculoskelet
sion and potentiation of human breast cancer Disord 11:111
metastasis. Clin Cancer Res 11:5381–5389 16. Kumar B, Dreja K, Shah SS, Cheong A, Xu SZ,
6. Wyatt CN, Campbell V, Brodbeck J, Brice NL, Sukumar P, Naylor J, Forte A, Cipollaro M,
Page KM, Berrow NS, Brickley K, Terracciano McHugh D, Kingston PA, Heagerty AM,
CM, Naqvi RU, MacLeod KT, Dolphin AC Munsch CM, Bergdahl A, Hultgardh-Nilsson
(1997) Voltage-dependent binding and cal- A, Gomez MF, Porter KE, Hellstrand P, Beech
cium channel current inhibition by an anti- DJ (2006) Upregulated TRPC1 channel in
alpha 1D subunit antibody in rat dorsal root vascular injury in vivo and its role in human
ganglion neurones and guinea-pig myocytes. neointimal hyperplasia. Circ Res 98:557–563
J Physiol 502:307–319 17. Bergdahl A, Gomez MF, Dreja K, Xu SZ,
7. Xu SZ, Zeng F, Lei M, Li J, Gao B, Xiong C, Adner M, Beech DJ, Broman J, Hellstrand P,
Sivaprasadarao A, Beech DJ (2005) Generation Sward K (2003) Cholesterol depletion impairs
of functional ion-channel tools by E3 target- vascular reactivity to endothelin-1 by reducing
ing. Nat Biotechnol 23:1289–1293 store-operated Ca2+ entry dependent on
8. Zhou BY, Ma W, Huang XY (1998) Specific TRPC1. Circ Res 93:839–847
antibodies to the external vestibule of voltage- 18. Kwan HY, Shen B, Ma X, Kwok YC, Huang Y,
gated potassium channels block current. J Gen Man YB, Yu S, Yao X (2009) TRPC1 associ-
Physiol 111:555–563 ates with BK(Ca) channel to form a signal
9. Liao YJ, Safa P, Chen YR, Sobel RA, Boyden complex in vascular smooth muscle cells. Circ
ES, Tsien RW (2008) Anti-Ca2+ channel anti- Res 104:670–678
body attenuates Ca2+ currents and mimics cer- 19. Rosado JA, Brownlow SL, Sage SO (2002)
ebellar ataxia in vivo. Proc Natl Acad Sci U S A Endogenously expressed Trp1 is involved in
105:2705–2710 store-mediated Ca2+ entry by conformational
10. Naylor J, Milligan CJ, Zeng F, Jones C, Beech coupling in human platelets. J Biol Chem
DJ (2008) Production of a specific extracellu- 277:42157–42163
lar inhibitor of TRPM3 channels. Br J 20. Klionsky L, Tamir R, Holzinger B, Bi X,
Pharmacol 155:567–573 Talvenheimo J, Kim H, Martin F, Louis JC,
256 Jacqueline Naylor and David J. Beech
Treanor JJ, Gavva NR (2006) A polyclonal anti- antibody blocking M2 ion channel protein.
body to the prepore loop of transient receptor PLoS One 6:e28309
potential vanilloid type 1 blocks channel activa- 24. Lo AS, Zhu Q, Marasco WA (2008) Intracellular
tion. J Pharmacol Exp Ther 319:192–198 antibodies (intrabodies) and their therapeutic
21. Gomez-Varela D, Zwick-Wallasch E, potential. Handb Exp Pharmacol 181:343–373
Knotgen H, Sanchez A, Hettmann T, 25. Miller KM, De Ryck MR, Wolff CGJ, Lawson
Ossipov D, Weseloh R, Contreras-Jurado C, ADG, Finney HM, Baker TS (2011) Function
Rothe M, Stuhmer W, Pardo LA (2007) modifying Nav1.7 antibodies. Patent number
Monoclonal antibody blockade of the human WO/2011/051350
Eag1 potassium channel function exerts 26. Kyte J, Doolittle RF (1982) A simple method
antitumor activity. Cancer Res 67: for displaying the hydropathic character if a
7343–7349 protein. J Mol Biol 157:105–132
22. Tradtrantip L, Zhang H, Saadoun S, Phuan 27. Kolaskar AS, Tongaonkar PS (1990) A semi-
PW, Lam C, Papadopoulos MC, Bennett JL, empirical method for prediction of antigenic
Verkman AS (2012) Anti-aquaporin-4 mono- determinants on protein antigens. FEBS Lett
clonal antibody blocker therapy for neuromy- 276:172–174
elitis optica. Ann Neurol 71:314–322 28. Hecquet CM, Ahmmed GU, Vogel SM, Malik
23. Wei G, Meng W, Guo H, Pan W, Liu J, Peng T, AB (2008) Role of TRPM2 channel in medi-
Chen L, Chen CY (2011) Potent neutraliza- ating H2O2-induced Ca2+ entry and endothe-
tion of influenza A virus by a single-domain lial hyperpermeability. Circ Res 102:347–355
Chapter 20
Abstract
Ion channels mediate a wide variety of physiological processes by forming small pores across the membranes
that allow regulated flow of ions into or out of the cell. The primary linear sequences of ion channel pro-
teins, like any proteins, are composed by 20 different amino acids, each of which is determined by specific
triplet codon in their genes. Site-directed mutagenesis is a widely used molecular biology method to
change the triplet in the coding sequence and thereby the amino acid residue in the protein sequence.
Functional characterization of the ion channels carrying point mutations allows us to interrogate the
structure–function relationships of the ion channels. Here, we will describe the site-directed mutagenesis
procedures, in which the wide-type cDNA or plasmid is used as a template to synthesize the complemen-
tary mutation-containing DNAs from two mutagenic primers in the polymerase chain reaction.
1 Introduction
Ion channels are integral membrane proteins that form small pores
to allow ions to cross the cell membranes and thereby transduce
important signals for a variety of physiological functions. More
than three hundreds of genes encoding ion channel proteins have
been identified (1). Ion channels are known to undergo closed,
open, inactivated, or desensitized states, each of which often has
more than one step, but the gating mechanisms are still not well
understood. X-ray crystallography in general provides a snapshot
of the structure of proteins in one particular state or step, and it is
notoriously challenging to determine the crystal structure of mem-
brane proteins such as ion channels. Nonetheless, a handful of ion
channels have been solved at the atomic level, including voltage-
gated potassium channel (2–4), ionotropic glutamate receptor (5),
acid-sensing ion channels (6, 7), and P2X receptors (8, 9). Such
structural information is tremendously useful and become particu-
larly powerful, when combined with the results from functional
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_20, © Springer Science+Business Media, LLC 2013
257
258 Wei Yang and Lin-Hua Jiang
Table 1
PCR protocols used for site-directed mutagenesis
2 Materials
2.1 Reagents 1. High-fidelity DNA polymerase (e.g., Turbo or Ultra Pfu DNA
polymerase).
2. 10× Pfu reaction buffer: 100 mM KCl, 100 mM (NH4)2SO4,
200 mM Tris–HCl pH 8.8, 20 mM MgSO4, 1% Triton X-100,
and 1 mg/mL nuclease-free bovine serum albumin.
3. DpnI enzyme.
4. 2.5 mM dNTP mix.
5. Sense and antisense mutagenic primers.
6. Chemically competent E. coli cells (see Note 1).
7. Luria-Bertani (LB) media and LB agar plates.
8. Ampicillin: 100 mg/mL stock solution made in sterile water
and stored at −20°C, and 60 mg/mL used in LB media or LB
agar plates.
9. 50× Tris-acetate (TAE) electrophoresis solution: 242 g Tris
base, 57.1 mL glacial acetic acid, and 100 mL 0.5 M EDTA
pH 8. The working solution is prepared by diluting the stock
solution within water.
10. Agarose gel for electrophoresis.
11. Ethidium bromide: 5 mg/mL stock solution made in water.
Add 5 mL to 100 mL of agarose solution to visualize DNA in
agarose gel using a transilluminator.
3 Methods
3.1 Design 1. The mutagenic primers should be 25–40 bases long, with a
of the Primers melting temperature (Tm) of ³78°C. The following formula is
commonly used to estimate the Tm value of the primers:
Tm = 81.5 + 0.41 (%GC ) − 675 / N − % mismatch
Site-Directed Mutagenesis 261
3.2 PCR 1. Set up the following PCR sample in a sterile thin-wall Eppendorf
tube for each mutation: 5 mL of 10× reaction buffer, 1–2 mL
(50–100 ng) of cDNA template (see Note 2), 2 mL of 10 mM
sense mutagenic primers (see Note 3), 2 mL of 10 mM anti-
sense mutagenic primers, and 4 mL of 2.5 mM dNTP mix. Add
sterile DNAase-free H2O to a final volume of 49 mL. Finally,
add 1 mL of 2.5 U/mL Pfu DNA polymerase.
2. Overlay the PCR sample in each tube with ~30 mL of mineral
oil, if the thermal cycler does not have a hot-top assembly.
3. Use the PCR cycles as detailed in Table 1 (see Notes 4 and 5).
4. Add 1 mL of DpnI enzyme (10 U/mL) to each PCR sample,
and mix gently and thoroughly by pipetting the samples sev-
eral times before incubated at 37°C for 1 h to remove the
cDNA templates (see Notes 6 and 7).
3.3 Transformation 1. Thaw competent cells, which are normally stored at −80°C, on
ice just before use. Transfer 50–100 mL of competent cells into
a 1.5 mL Eppendorf tube for each sample.
2. Add 1–3 mL of PCR sample to the competent cells, mix by
gentle flicking (not pipetting), and incubate on ice for 30 min.
262 Wei Yang and Lin-Hua Jiang
4 Notes
Fig. 2 An agarose gel shows the PCR products after DpnI treatment. Lane 1, the DNA ladder; lanes 2 and 14, the
plasmids used as the template; lanes 3–13, PCR products using gradient PCRs with the annealing temperature from
56 to 67°C. The better PCR yield was observed with the annealing temperature higher than 60°C (lanes 7–13 )
Fig. 3 Nucleotide sequence alignment of the DNA sequencing results for the E960Q and D987E two pore mutations against the original human TRPM2 cDNA sequence
using the multiple sequence alignment ClustalW2 program. The numbers on the right indicate the position of the last nucleotide in each line in the human TRPM2
cDNA sequence, or the readable sequencing results for the E960Q or D987E mutation. All the nucleotide sequences are identical as denoted by the asterisks
underneath, with the exception of changes in the G2894C for the E960Q mutation and the C2977G for the D987E mutation
Site-Directed Mutagenesis 265
References
1. Gabashvili IS, Sokolowski BH, Morton CC, affect ATP-activated P2X7 receptor functions.
Giersch AB (2007) Ion channel gene expres- J Psychiatr Res 44:347–355
sion in the inner ear. J Assoc Res Otolaryngol 13. Sorge RE, Trang T, Dorfman R et al (2012)
8:305–328 Genetically determined P2X7 receptor pore
2. Jiang Y, Lee A, Chen J, Ruta V, Cadene M, formation regulates variability in chronic pain
Chait BT, MacKinnon R (2003) X-ray struc- sensitivity. Nat Med 18:595–599
ture of a voltage-dependent K+ channel. Nature 14. Kunkel TA (1985) Rapid and efficient site-
423:33–41 specific mutagenesis without phenotypic selec-
3. Jiang Y, Lee A, Chen J, Cadene M, Chait BT, tion. Proc Natl Acad Sci 82:488–492
MacKinnon R (2002) Crystal structure and 15. Taylor JW, Ott J, Eckstein F (1985) The
mechanism of a calcium-gated potassium chan- rapid generation of oligonucleotide-directed
nel. Nature 417:515–522 mutations at high frequency using phospho-
4. Long SB, Campbell EB, Mackinnon R (2005) rothioate modified DNA. Nucleic Acids Res
Crystal structure of a mammalian voltage- 13:8765–8785
dependent Shaker family K+ channel. Science 16. Weiner MP, Costa GL, Schoettlin W, Cline J,
309:897–903 Mathur E, Bauer JC (1994) Site-directed
5. Sobolevsky AI, Rosconi MP, Gouaux E (2009) mutagenesis of double-stranded DNA by the
X-ray structure, symmetry and mechanism of polymerase chain reaction. Gene 151:119–123
an AMPA-subtype glutamate receptor. Nature 17. Vandeyar MA, Weiner MP, Hutton CJ, Batt
462:745–756 CA (1988) A simple and rapid method for the
6. Jasti J, Furukawa H, Gonzales EB, Gouaux E selection of oligonucleotide-directed mutants.
(2007) Structure of acid-sensing ion channel 1 Gene 65:129–133
at 1.9 A resolution and low pH. Nature 18. Ishii TM, Zerr P, Xia XM, Bond CT, Maylie J,
449:316–323 Adelman JP (1998) Site-directed mutagenesis.
7. Gonzales EB, Kawate T, Gouaux E (2009) Methods Enzymol 293:53–71
Pore architecture and ion sites in acid-sensing 19. Nagy ZB, Felfoldi F, Tamas L, Puskás LG
ion channels and P2X receptors. Nature 460: (2004) A one-tube, two-step polymerase chain
599–604 reaction-based site-directed mutagenesis method
8. Kawate T, Michel JC, Birdsong WT, Gouaux E with simple identification of the mutated prod-
(2009) Crystal structure of the ATP-gated P2X uct. Anal Biochem 324:301–303
(4) ion channel in the closed state. Nature 20. Monks SA, Needleman DJ, Miller C (1999)
460:592–598 Helical structure and packing orientation of
9. Hattori M, Gouaux E (2012) Molecular mech- the S2 segment in the Skaker K+ channel. J Gen
anism of ATP binding and ion channel activa- Physiol 113:415–423
tion in P2X receptors. Nature 485:207–212 21. Yang W, Manna PT, Zou J, Luo J, Beech DJ,
10. Browne LE, Jiang LH, North RA (2010) New Sivaprasadarao A, Jiang LH (2011) Zinc inacti-
structure enlivens interest in P2X receptors. vates melastatin transient receptor potential 2
Trends Pharmacol Sci 31:229–237 channels via the outer pore. J Biol Chem
11. Hermosura MC, Cui AM, Go RC, Davenportb 286:23789–23798
B, Shetlera CM, Heizerb JW, Schmitzc C, 22. Yang W, Zou J, Xia R, Vaal ML, Seymour VA,
Mocza G, Garrutod RM, Perraudb A-L (2008) Luo J, Beech DJ, Jiang L-H (2010) State-
Altered functional properties of a TRPM2 vari- dependent inhibition of TRPM2 channel by
ant in Guamanian ALS and PD. Proc Natl Acad acidic pH. J Biol Chem 285:30411–30418
Sci U S A 105:18029–18034 23. Liu X, Surprenant A, Mao HJ, Roger S, Xia R,
12. Roger S, Mei ZZ, Baldwin JM, Dong L, Bradley H, Jiang L-H (2008) Identification of
Bradley H, Baldwin SA, Surprenant A, Jiang key residues coordinating functional inhibition
LH (2010) Single nucleotide polymorphisms of P2X7 receptors by zinc and copper. Mol
that were identified in affective mood disorders Pharmacol 73:252–259
266 Wei Yang and Lin-Hua Jiang
24. Liu X, Ma W, Surprenant A, Jiang L-H (2009) macaque monkey P2X7 receptor. Br J Pharmacol
Identification of the amino acid residues in the 164:743–754
extracellular domain of rat P2X(7) receptor 27. Hanahan D (1983) Studies on transformation
involved in functional inhibition by acidic pH. of Escherichia coli with plasmids. J Mol Biol
Br J Pharmacol 156:135–142 166:557–580
25. Zou J, Yang W, Beech DJ, Jiang L-H (2011) A 28. Xia R, Mei ZZ, Mao HJ, Yang W, Dong L,
residue in the TRPM2 channel outer pore is Bradley H, Beech DJ, Jiang LH (2008)
crucial in determining species-dependent sensi- Identification of pore residues engaged in
tivity to extracellular acidic pH. Pflugers Arch determining divalent cationic permeation in
462:293–302 transient receptor potential melastatin sub-
26. Bradley HJ, Browne LE, Yang W, Jiang L-H type channel 2. J Biol Chem 283:
(2011) Pharmacological properties of the rhesus 27426–27432
Chapter 21
Abstract
Cysteine contains a highly reactive thiol group and therefore under oxidizing conditions a disulfide bond
can form between a pair of cysteines that are juxtaposed in the close vicinity, which can be only reversed by
reducing agents. These attributes have been elegantly exploited to study the functional role of an interac-
tion or contact between two adjacent domains that are present in ion channels or virtually in any proteins,
by introducing double cysteine substitutions at the domain interface and measuring changes in the ion
channel functions arising from cross-linking the two substituted cysteines via formation of a disulfide
bond. Here I describe this cysteine-based cross-linking approach.
Key words Disulfide bond, Inter-domain interaction, Double cysteine substitution, Cross-linking,
Ion channel, Ligand binding, Gating
1 Introduction
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_21, © Springer Science+Business Media, LLC 2013
267
268 Lin-Hua Jiang
structures have been solved at the atomic level and such structures
reveal numerous interactions or contacts between domains located
within the same or neighboring subunits. Some of these interac-
tions have been demonstrated to be crucial in coordinating ligand
binding, e.g., ACh binding in the nAChR (6), and ATP and Zn2+
binding in the P2XR (3), or mediating ion channel gating, e.g., in
the GABAAR (7), KV channels (8), and P2XR (9). Thus, a fuller
understanding of inter-domain/subunit interactions in the closed,
open, and desensitized states can help to provide mechanistic
insights into the principles of how the ion channels function. Such
insights will facilitate elucidation of the molecular mechanisms of
diseases arising from mutational perturbance of ion channels, e.g.,
(10), and structure-guided design of therapeutic drugs.
Cysteine amino acid is one of the building blocks of virtually
every protein including ion channels. Uniquely, cysteine contains a
highly reactive thiol group. Thus, when a pair of cysteines is
exposed to each other in the close vicinity, a disulfide bond can
form between them, catalyzed by ambient oxygen or oxidizing
agents. The thiol group in cysteine can also form disulfide bonds
with many of the cysteine-modifying reagents (e.g., methanethio-
sulfonates). Such a unique chemical property of cysteine underpins
the substituted cysteine accessibility method, which was elegantly
developed by Karlin and his colleagues to study the ion permeation
and gating properties in the nAChR (11, 12), and has since been
used to study many other ion channels. In this method, single-
cysteine substitution is introduced into the region of interest. One
assumes that the residue replaced with cysteine occupies a func-
tionally crucial position in the channel, formation of a disulfide
bond between the introduced cysteine and cysteine-modifying
reagents confers discernible changes in the ion channel functional
properties.
This chapter describes cysteine-based cross-linking approach,
which follows the same principle of disulfide bonding and extends
the single substituted cysteine accessibility method. It is primarily
used to probe the role of an interaction between two neighboring
domains or regions, predicted on the basis of the atomic structures
or educated guess, in determining the functional properties of ion
channels. This approach thus involves introduction of double
cysteine substitutions into two positions located on the opposite
side of an interface between two domains, either from adjacent
subunits (Fig. 1a) or within the same subunit (Fig. 1b), and takes
the assumptions that if one or both of these positions or the sur-
rounding regions are engaged in, e.g., ligand binding, permeation,
or conformational changes accompanying the channel gating,
cross-linking of the substituted cysteines as a result of forming a
disulfide bond alters such ion channel functional properties. Such
functional alterations can be reversed using dithiothreitol (DTT)
or other reducing agents. It is easy to implement, and it is versatile,
Cysteine-Based Cross-Linking 269
a
Oxidisation
SH HS S S
Reduction
Oxidisation
SH HS S S
Reduction
i.e., applicable to virtually any ion channel types, and can yield
important mechanistic understanding of the role of inter-domain
interactions in coordinating ligand binding and mediating channel
gating and ion permeation, as have been demonstrated in a num-
ber of ion channels (7–9, 13–18).
2 Materials
3. Microforge.
4. Solution perfusion system (e.g., RSC-160 rapid solution
change system from BioLogic).
5. Glass capillary.
6. 1-mL plastic syringe.
7. Syringe filter disk with 0.2-mm pores.
8. Internal/pipette solution: 145 mM NaF, 10 mM EGTA, and
10 mM HEPES, pH 7.3.
9. External/bath solution: 147 mM NaCl, 2 mM CaCl2, 2 mM
KCl, 1 mM MgCl2, 13 mM glucose, and 10 mM HEPES,
pH 7.3.
10. 10 mM DTT freshly prepared in external solution.
3. Methods
3.1 General The starting point is obvious: which pair of residues is chosen to be
Considerations replaced with cysteine? The decision can be straightforward if
atomic structural information exists to indicate juxtaposition of
two residues in two neighboring domains, and some functional
evidence to suggest their importance in the ion channel functions.
For example, structural models indicate the close vicinity of Asp57
and Asp149 residues in the extracellular domain and Lys279 residue
in the extracellular transmembrane linker region in the GABAAR
(7) or Glu63 in one subunit and Arg274 in the adjacent subunit in
the extracellular domain of the P2X2R (17), and functional studies
of the single-point mutant channels implied their engagement in
the channel gating (7, 17). Cysteine-based cross-linking approach
yielded unambiguous evidence to support a crucial role of the elec-
trostatic interactions mediated by these residues in the channel gat-
ing (7, 17). On other occasions where structural information is
limited or not available, the choice of double cysteine substitution
is largely reduced to educated guess, even trial and error. For
example, conserved charged residues including Lys68, Lys70, Lys190,
Arg292, Arg305, and Lys309 in the extracellular domain of P2X1R and
equivalent residues in other P2XR are essential for P2XR activation
by ATP (3). Double cysteine substitution introduced into two sep-
arate subunits and cross-linking of the co-expressed single-cysteine
mutant subunits, in combination with Western blotting and func-
tional studies, showed that K69C and K309C formed an inter-
subunit disulfide (16). This provides evidence to indicate that Lys68
and Lys309 residues contribute to formation of a novel inter-subunit
ATP binding site, which has been substantiated by the atomic
structural determination (3). Another example is shown in Fig. 2;
Val48 and Ile328 residues, located at the extracellular end of the first
and the second transmembrane domains, respectively (Fig. 2),
Cysteine-Based Cross-Linking 271
Fig. 2 Cross-linking of V48C in the first transmembrane domain (TM1) and I328C
in the second transmembrane domain (TM2) impairs P2X2R activation. (a) A rep-
resentative whole-cell recording of ATP-evoked inward currents in a HEK293 cell
expressing [V48C, I328C] double-mutant rat P2X2R. The initial ATP-induced cur-
rent was small but drastically increased by exposure to 10 mM DTT. The current
gradually declined upon washing DTT and was reversed upon reapplication of
DTT (modified from (13). This research was originally published in Journal of
Biological Chemistry. Jiang LH et al. Amino acid residues involved in gating
identified in the first membrane-spanning domain of the rat P2X2 receptor. 2001;
276: 14902–14908. © the American Society for Biochemistry and Molecular
Biology). (b) A structural model of the transmembrane domains of the trimeric rat
P2X2R in the closed state viewed from extracellular side, showing close juxtapo-
sition of V48 residue in the TM1 on the periphery and I328 residue in the TM2
pore-forming domain (reproduced from (3) with permission)
3.2 Cell Preparation The following procedures should be conducted in a sterile tissue
for Transient culture hood except indicated otherwise (refer to Chapters 2, 4
Transfection and 5 of this book for alternative transfection methods and
optimization).
1. Grow cells in DMEM culture medium in a T25 flask media in
a humid 37 °C, 5% CO2 tissue culture incubator until 80%
confluent.
2. Rinse cells with PBS.
3. Detach cells using trypsin–EDTA solution, transfer cell sus-
pension in a 15-mL Falcon tube, and collect cells by centrifu-
gation at 200 × g for 5 min.
4. Aspirate supernatant and resuspend cell pellet in 2 mL of cul-
ture medium by gentle pipetting.
5. Seed 0.2–0.3 mL of cell suspension in each 35-mm Petri dish,
add 1.5 mL of culture medium, and leave in the incubator to
grow until >80% confluent (see Note 2). Prepare one Petri
dish of cells for each transfection.
6. For each transfection, dilute 0.1 μg of plasmid for GFP (see
Note 3) and 1 μg of plasmid for the ion channel of interest into
100 μL of transfection medium in one 1.5-mL Eppendorf tube
(see Note 4) and 3 μL Lipofectamine 2000 into 100 μL of
transfection medium in another Eppendorf tube. Leave them
in the tissue culture hood at room temperature for 5 min.
7. Combine plasmid and Lipofectamine 2000 containing trans-
fection medium, and mix thoroughly by gentle pipetting.
Leave it in the tissue culture hood at room temperature for
20 min.
8. Add 0.8 mL of culture medium into transfection medium.
9. Replace existing culture medium with transfection medium.
10. Return cells to the incubator and leave overnight (see Note 5).
3.4 Whole-Cell 1. Make recording pipettes from glass capillary using a pipette
Patch-Clamp puller, and if necessary, polish the tip using a microforge.
Recording 2. Backfill the recording pipette with the pipette solution (see
Subheading 2.2), and place in the electrode holder.
3. Apply positive air pressure to the recording pipette via the side-
port of the electrode holder (see Note 7).
4. Immerse the recording pipette into the external or bath solu-
tion, and offset the liquid junction potential (see Note 8).
5. Apply a 5 mV test pulse every 5 s from 0 mV holding potential
to examine the recording pipette resistance, which should be
in the range of 2–4 MΩ.
6. Select single GFP-positive cells under the fluorescent microscope.
7. Position the recording pipette closely to the cell and impale
gently onto it using a micromanipulator.
8. Apply negative air pressure to the recording pipette to obtain a
giga-ohm (GΩ) seal (see Note 9).
9. Set a negative holding potential (e.g., −60 mV), and cancel the
pipette capacitance using amplifier fast capacitance (see Note 8).
10. Apply further negative air pressure to the recording pipette to
break the membrane patch underneath the tip of the recording
pipette to achieve the whole-cell figuration.
11. Compensate the access resistance (Rs) using whole-cell (slow)
capacitance cancellation (see Note 8).
12. Once Rs is below an acceptable value (£10 MΩ), and if
required, carry out series resistance compensation (see Note 8;
see also Chapter 7 of this book for more detailed protocol of
the patch-clamp recording).
13. Begin recording. Figure 2a shows an example of whole-cell
current patch-clamp recording made from a HEK293 cell
expressing double cysteine mutant [V48C, I328C] of rat
P2X2R at a holding potential of −60 mV. During this record-
ing, 30 μM ATP was applied for 2 s every 2 or 4 min, using
RSC-200 fast solution changer, to the cell to activate the
mutant receptor channels. After two initial ATP applications,
10 mM dithiothreitol (DTT) was applied to the cell (denoted
by 0 min), using RSC-200. The initial ATP-induced current
was small but drastically increased and reached maximum after
application of DTT for 20 min. The current gradually declined
upon washing DTT, and was reversed upon reapplication of
274 Lin-Hua Jiang
4 Notes
References
1. Hille B (2001) Ion channels of excitable mem- 6. Corringer PJ, Le Novère N, Changeux JP
branes, 3rd edn. Sinauer associates, Sunderland, (2000) Nicotinic receptors at the amino acid
MA level. Annu Rev Pharmacol Toxicol 40:
2. Khakh BS, North RA (2006) P2X receptors as 431–458
cell-surface ATP sensors in health and disease. 7. Kash TL, Jenkins A, Kelley JC, Trudell JR,
Nature 442:527–532 Harrison NL (2003) Coupling of agonist
3. Browne LE, Jiang LH, North RA (2011) New binding to channel gating in the GABA(A)
structure enlivens interest in P2X receptors. receptor. Nature 421:272–275
Trends Pharmacol Sci 31:229–237 8. Elliott DJ, Neale EJ, Aziz Q, Dunham JP,
4. Yu FH, Yarov-Yarovoy V, Gutman GA, Munsey TS, Hunter M, Sivaprasadarao A
Catterall WA (2005) Overview of molecular (2004) Molecular mechanism of voltage sen-
relationships in the voltage-gated ion channel sor movements in a potassium channel. EMBO
superfamily. Pharmacol Rev 57:387–395 J 23:4717–4726
5. Kashlan OB, Kleyman TR (2011) ENaC struc- 9. Roberts JA, Allsopp RC, El Ajouz S, Vial C,
ture and function in the wake of a resolved Schmid R, Young MT, Evans RJ (2012)
structure of a family member. Am J Physiol Agonist binding evokes extensive conforma-
Renal Physiol 301:F684–F696 tional changes in the extracellular domain of the
276 Lin-Hua Jiang
ATP-gated human P2X1 receptor ion channel. 15. Spelta V, Jiang LH, Bailey RJ, Surprenant A,
Proc Natl Acad Sci USA 109:4663–4667 North RA (2003) Interaction between
10. Sorge RE, Trang T, Dorfman R, Smith SB, cysteines introduced into each transmembrane
Beggs S, Ritchie J, Austin JS, Zaykin DV, domain of the rat P2X2 receptor. Br J
Meulen HV, Costigan M, Herbert TA, Pharmacol 138:131–136
Yarkoni-Abitbul M, Tichauer D, Livneh J, 16. Marquez-Klaka B, Rettinger J, Bhargava Y,
Gershon E, Zheng M, Tan K, John SL, Slade Eisele T, Nicke A (2007) Identification of an
GD, Jordan J, Woolf CJ, Peltz G, Maixner W, intersubunit cross-link between substituted
Diatchenko L, Seltzer Z, Salter MW, Mogil JS cysteine residues located in the putative ATP
(2012) Genetically determined P2X7 receptor binding site of the P2X1 receptor. J Neurosci
pore formation regulates variability in chronic 27:1456–1466
pain sensitivity. Nat Med 18:595–599 17. Jiang R, Martz A, Gonin S, Taly A, de Carvalho
11. Akabas MH, Stauffer DA, Xu M, Karlin A LP, Grutter T (2010) A putative extracellular
(1992) Acetylcholine receptor channel struc- salt bridge at the subunit interface contributes
ture probed in cysteine-substitution mutants. to the ion channel function of the ATP-gated
Science 258:307–310 P2X2 receptor. J Biol Chem 285:
12. Karlin A, Akabas MH (1998) Substituted- 15805–15815
cysteine accessibility method. Methods Enzymol 18. Jiang R, Taly A, Lemoine D, Martz A,
293:123–145 Cunrath O, Grutter T (2012) Tightening of
13. Jiang LH, Rassendren F, Spelta V, Surprenant the ATP-binding sites induces the opening
A, North RA (2001) Amino acid residues of P2X receptor channels. EMBO J 31:
involved in gating identified in the first mem- 2134–2143
brane-spanning domain of the rat P2X(2) 19. Trouet D, Nilius B, Voets T, Droogmans G,
receptor. J Biol Chem 276:14902–14908 Eggermont J (1997) Use of a bicistronic GFP-
14. Jiang LH, Kim M, Spelta V, Bo X, Surprenant expression vector to characterise ion channels
A, North RA (2003) Subunit arrangement in after transfection in mammalian cells. Pflugers
P2X receptors. J Neurosci 23:8903–8910 Arch 434:632–638
Chapter 22
Abstract
Regulator of K+ conductance (RCK) domains form a conserved class of ligand-binding domains that con-
trol the activity of a variety of prokaryotic and eukaryotic K+ channels. Structural analysis of these domains
by X-ray crystallography has provided insight toward mechanisms underlying ligand binding and channel
gating, and thus the experimental strategies aimed at determining structures of liganded and unliganded
forms of the domains may be useful in analysis of other ligand-binding domains. Here, we describe a basic
strategy for crystallographic analysis of the RCK domain from the MthK channel, for determination of its
Ca2+-bound structure.
Key words Crystallization, Cytoplasmic domain, Calcium, Binding site, Potassium channel
1 Introduction
Large-conductance Ca2+-activated K+ channels (BK channels) are
found in a wide range of tissues, and play a critical role in linking K+
efflux to increases in cytoplasmic Ca2+ levels, thus tying Ca2+ signal-
ing to electrical hyperpolarization of the cell membrane (1, 2). In
electrically excitable nerve and muscle cells, this linkage provides an
important feedback mechanism to allow for rapid repolarization of
the membrane, promoting termination of action potentials and
smooth muscle relaxation (3–7). While structural analysis of the
mammalian BK channel can be technically complex and has so far
yielded low-resolution structural information (8–10), the prokary-
otic Ca2+-activated K+ channel, MthK, can be expressed and crystal-
lized prodigiously, and has served as a model system for understanding
gating mechanisms in these channels (11–16).
MthK and BK channels are regulated by Ca2+ binding to a con-
served cytoplasmic domain, known as the regulator of K+ conduc-
tance (RCK) domain (8, 9, 13, 14, 17–20). RCK domains are
ubiquitous among prokaryotic and eukaryotic K+ channels and
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_22, © Springer Science+Business Media, LLC 2013
277
278 Frank J. Smith and Brad S. Rothberg
2 Materials
All solutions are prepared using deionized, distilled water
(ddH2O).
2.1 Plasmids and 1. E. coli strain BL21(DE3), purchased from Stratagene, La Jolla,
E. coli Strains CA.
2. MthK RCK plasmid: MthK RCK domain cDNA (residues
M107-A336 in MthK protein sequence) in pET21a vector
with D184N mutation generated using QuickChange
(Stratagene, La Jolla, CA). The MthK RCK domain sequence
was followed by a thrombin recognition/cleavage sequence
(amino acids LVPRGS) and C-terminal hexahistidine tag, and
the coding region was codon-optimized for E. coli expression.
3 Methods
3.1 BL21(DE3) E. coli 1. Pre-warm one agar plate (with ampicillin) to room
Transformation temperature.
2. Thaw BL21(DE3) cells on ice for 5 min.
3. Add 0.1 μg/μL MthK RCK plasmid solution to 25 μL
BL21(DE3) cells and store on ice for 15 min.
4. Heat shock the cells for 45 s at 42°C.
5. Cool cells on ice immediately for 5 min.
6. Add 400 μL NZYM + Glu medium.
7. Incubate for 45 min at 37°C with vigorous shaking (on a
shaker table at 225–250 rpm).
8. Plate the cell suspension on agar plate.
9. Incubate the plate overnight at 37°C.
3.4 Affinity 1. Prepare HiTrap chelating HP (5 mL) column (see Note 1):
Chromatography (a) Perfuse with 20–30 mL ddH2O at 2.5 mL/min.
(b) Perfuse with 20–30 mL 0.1 M CoCl2 buffer at 2.5 mL/min.
(c) Perfuse column with 20–30 mL ddH2O at 2.5 mL/min.
(d) Perfuse column with 20–30 mL Buffer B + 400 mM imi-
dazole at 2.5 mL/min.
(e) Perfuse column with 20–30 mL Buffer B + 20 mM imida-
zole at 2.5 mL/min.
2. Load cleared lysate (from Subheading 3.3) onto column at
4.5 mL/min. Monitor protein elution by UV detection (see
Note 1).
3. Once lysate is loaded onto column, perfuse column with Buffer
B + 20 mM imidazole buffer at 4.5 mL/min until unbound
protein is eluted, as indicated by reduction in UV signal to
baseline level (approximately 20–30 mL).
4. Once a baseline is reached, perfuse column with Buffer
B + 400 mM imidazole buffer to elute bound protein. Monitor
UV signal, and collect protein upon increase of UV signal in a
sterile 50 mL conical tube. Stop collection once UV signal
decreases to baseline level (around 5 mL).
5. Immediately add 4 U thrombin (two aliquots of thrombin
solution) to eluate and incubate 2 h at room temperature with
gentle shaking.
6. Filter protein using 0.22 μm SPIN-X tubes for 1 min in refrig-
erated desktop centrifuge at 18,000 × g, at 4°C.
3.5 Gel Filtration 1. Equilibrate Superdex-200 10/300 column with two column
Chromatography, volumes of degassed Buffer B, at a flow rate of 1 mL/min
Concentration, and (see Note 2).
Protein Storage 2. Load protein onto column via 1 mL sample loop and elute with
Buffer B, at a flow rate of 0.5 mL/min. Collect fractions begin-
ning at approximately 11 mL elution volume, and continue col-
lection until the UV signal falls to baseline, typically at approximately
13 mL elution volume. If automated fraction collection is used,
pool fractions collected during the major elution peak, which is
centered at approximately 12 mL elution volume.
3. Measure protein concentration using Bradford or similar
assay.
282 Frank J. Smith and Brad S. Rothberg
4 Notes
1. Perfusion of the HiTrap metal-affinity chromatography column
and fraction collection is facilitated by the use of a peristaltic
pump coupled to a UV detector (i.e., Model EP-1 Econo-
Pump and Model EM-1 UV Monitor, Bio-Rad), with the UV
detector signal output connected to a chart recorder.
2. Buffers used in gel filtration chromatography steps should be
degassed by dispensing buffer into a sidearm flask, sealing the
top with a rubber stopper, and connecting the sidearm to a
vacuum (via a vacuum trap) for a minimum of 1 h. Alternatively,
this could be performed immediately subsequent to filtration
using a vacuum-filtration unit, by leaving the buffer in the col-
lection bottle with the sidearm connected to vacuum for at
least 1 h after filtration. Gel filtration chromatography is per-
formed using a preparative fast protein liquid chromatography
apparatus (i.e., ÄKTA FPLC system, GE Healthcare), with elu-
tion monitored by UV detection. This chromatography proce-
dure is facilitated through use of automated fraction
collection.
3. Crystallization screens can alternatively be set in hanging-drop
format.
4. For this crystallization experiment, we were particularly inter-
ested in discovering conditions in which crystal growth was
more successful in drops containing 50 mM CaCl2, whereas
no crystal growth was noted in the drop with no added CaCl2
in the same well. It should be further noted that calcium (and
other divalent cations) is particularly prone to formation of
insoluble complexes and salt crystals with anions that are com-
monly included as additives in commercially available screens.
These anions include phosphate, sulfate, and citrate.
Distinguishing between calcium salt crystals and protein crys-
tals can be facilitated by setting a “control” tray, in which the
screen is set by mixing the well solution with the calcium-
containing protein buffer (i.e., with no protein). In this con-
trol screen, protein crystals will not form—only salt crystals.
284
Table 1
Example of a pos sible grid screen for optimization of crystal growth of the MthK RCK domain with D184N mutation in 50 mM CaCl2, using PEG3350 as
precipitant, at pH ranging from 5.9 to 6.3
100 μl 1 M MES 100 μl 1 M MES 100 μl 1 M MES 100 μl 1 M MES 100 μl 1 M MES 100 μl 1 M MES
pH 6.1 pH 6.1 pH 6.1 pH 6.1 pH 6.1 pH 6.1
50 μl 1 M CaCl2 50 μl 1 M CaCl2 50 μl 1 M CaCl2 50 μl 1 M CaCl2 50 μl 1 M CaCl2 50 μl 1 M CaCl2
560 μl ddH2O 490 μl ddH2O 450 μl ddH2O 410 μl ddH2O 370 μl ddH2O 330 μl ddH2O
pH 6.3 320 μl PEG3350 360 μl PEG3350 400 μl PEG3350 440 μl PEG3350 480 μl PEG3350 520 μl PEG3350
(50% stock) (50% stock) (50% stock) (50% stock) (50% stock) (50% stock)
100 μl 1 M MES 100 μl 1 M MES 100 μl 1 M MES 100 μl 1 M MES 100 μl 1 M MES 100 μl 1 M MES
pH 6.3 pH 6.3 pH 6.3 pH 6.3 pH 6.3 pH 6.3
50 μl 1 M CaCl2 50 μl 1 M CaCl2 50 μl 1 M CaCl2 50 μl 1 M CaCl2 50 μl 1 M CaCl2 50 μl 1 M CaCl2
530 μl ddH2O 490 μl ddH2O 450 μl ddH2O 410 μl ddH2O 370 μl ddH2O 330 μl ddH2O
pH 6.5 320 μl PEG3350 360 μl PEG3350 400 μl PEG3350 440 μl PEG3350 480 μl PEG3350 520 μl PEG3350
(50% stock) (50% stock) (50% stock) (50% stock) (50% stock) (50% stock)
100 μl 1 M MES 100 μl 1 M MES 100 μl 1 M MES 100 μl 1 M MES 100 μl 1 M MES 100 μl 1 M MES
pH 6.5 pH 6.5 pH 6.5 pH 6.5 pH 6.5 pH 6.5
50 μl 1 M CaCl2 50 μl 1 M CaCl2 50 μl 1 M CaCl2 50 μl 1 M CaCl2 50 μl 1 M CaCl2 50 μl 1 M CaCl2
530 μl ddH2O 490 μl ddH2O 450 μl ddH2O 410 μl ddH2O 370 μl ddH2O 330 μl ddH2O
X-Ray Crystallography of MthK 285
Fig. 1 Examples of MthK D184N RCK domain crystals grown in the presence of
calcium. Crystals were grown in a hanging drop, over a reservoir solution contain-
ing 20% PEG3350, 20 mM CaCl2, and 100 mM MES pH 5.9. These crystals display
a hexagonal morphology, and formed in 2–3 weeks under these conditions
Fig. 2 MthK D184N RCK domain crystal, mounted in a nylon loop and frozen in a
nitrogen stream at 100 K. This crystal was grown in a hanging drop, over a res-
ervoir solution containing 20% PEG3350, 100 mM CaCl2, and 100 mM MES pH
5.9 ( The photograph was captured at the National Synchrotron Light Source,
beamline X25. The dimension (see planaons) of the black box is 100 × 150 μm)
286 Frank J. Smith and Brad S. Rothberg
Acknowledgements
References
1. Vergara C, Latorre R, Marrion NV, Adelman ring in the high-conductance Ca2+-activated K+
JP (1998) Calcium-activated potassium chan- channel. Nature 481:94–97
nels. Curr Opin Neurobiol 8:321–329 10. Wu Y, Yang Y, Ye S, Jiang Y (2010) Structure
2. Hille B (2001) Ion channels of excitable mem- of the gating ring from the human large-con-
branes, 3rd edn. Sinauer Associates, Inc., ductance Ca2+-gated K+ channel. Nature
Sunderland, MA 466:393–397
3. Jaffe DB, Wang B, Brenner R (2011) Shaping 11. Ye S, Li Y, Chen L, Jiang Y (2006) Crystal
of action potentials by type I and type II large- structures of a ligand-free MthK gating ring:
conductance Ca2+-activated K+ channels. insights into the ligand gating mechanism of
Neuroscience 192:205–218 K+ channels. Cell 126:1161–1173
4. Brayden JE, Nelson MT (1992) Regulation of 12. Dong J, Shi N, Berke I, Chen L, Jiang Y
arterial tone by activation of calcium-dependent (2005) Structures of the MthK RCK domain
potassium channels. Science 256:532–535 and the effect of Ca2+ on gating ring stability.
5. Herrera GM, Heppner TJ, Nelson MT (2000) J Biol Chem 280:41716–41724
Regulation of urinary bladder smooth muscle 13. Jiang Y, Lee A, Chen J, Cadene M, Chait BT,
contractions by ryanodine receptors and BK MacKinnon R (2002) Crystal structure and
and SK channels. Am J Physiol Regul Integr mechanism of a calcium-gated potassium chan-
Comp Physiol 279:R60–R68 nel. Nature 417:515–522
6. Storm JF (1987) Action potential repolariza- 14. Pau VP, Smith FJ, Taylor AB, Parfenova LV,
tion and a fast after-hyperpolarization in rat Samakai E, Callaghan MM, Abarca-Heidemann K,
hippocampal pyramidal cells. J Physiol 385: Hart PJ, Rothberg BS (2011) Structure and func-
733–759 tion of multiple Ca2+-binding sites in a K+ channel
7. Lancaster B, Nicoll RA (1987) Properties of regulator of K+ conductance (RCK) domain. Proc
two calcium-activated hyperpolarizations in Natl Acad Sci U S A 108:17684–17689
rat hippocampal neurones. J Physiol 389: 15. Niu X, Qian X, Magleby KL (2004) Linker-
187–203 gating ring complex as passive spring and
8. Yuan P, Leonetti MD, Pico AR, Hsiung Y, Ca(2+)-dependent machine for a voltage- and
MacKinnon R (2010) Structure of the human Ca2+-activated potassium channel. Neuron
BK channel Ca2+-activation apparatus at 3.0 A 42:745–756
resolution. Science 329:182–186 16. Hou S, Xu R, Heinemann SH, Hoshi T (2008)
9. Yuan P, Leonetti MD, Hsiung Y, MacKinnon Reciprocal regulation of the Ca2+ and H + sen-
R (2011) Open structure of the Ca2+ gating sitivity in the SLO1 BK channel conferred by
X-Ray Crystallography of MthK 287
the RCK1 domain. Nat Struct Mol Biol 23. Roosild TP, Castronovo S, Miller S, Li C,
15:403–410 Rasmussen T, Bartlett W, Gunasekera B, Choe
17. Schreiber M, Salkoff L (1997) A novel cal- S, Booth IR (2009) KTN (RCK) domains reg-
cium-sensing domain in the BK channel. ulate K+ channels and transporters by control-
Biophys J 73:1355–1363 ling the dimer-hinge conformation. Structure
18. Wei A, Solaro C, Lingle C, Salkoff L (1994) 17:893–903
Calcium sensitivity of BK-type KCa channels 24. Zhang X, Solaro CR, Lingle CJ (2001)
determined by a separable domain. Neuron Allosteric regulation of BK channel gating by
13:671–681 Ca2+ and Mg2+ through a nonselective, low
19. Bao L, Kaldany C, Holmstrand EC, Cox DH affinity divalent cation site. J Gen Physiol
(2004) Mapping the BKCa channel’s “Ca2+ 118:607–636
bowl”: side-chains essential for Ca2+ sensing. J 25. Xia XM, Zeng X, Lingle CJ (2002) Multiple
Gen Physiol 123:475–489 regulatory sites in large-conductance calcium-
20. Bao L, Rapin AM, Holmstrand EC, Cox DH activated potassium channels. Nature 418:
(2002) Elimination of the BK(Ca) channel’s 880–884
high-affinity Ca2+ sensitivity. J Gen Physiol 26. Zeng XH, Xia XM, Lingle CJ (2005) Divalent
120:173–189 cation sensitivity of BK channel activation sup-
21. Kuo MMC, Haynes WJ, Loukin SH, Kung C, ports the existence of three distinct binding
Saimi Y (2005) Prokaryotic K+ channels: From sites. J Gen Physiol 125:273–286
crystal structures to diversity. FEMS Microbiol 27. Zhang X, Zeng X, Lingle CJ (2006) Slo3 K+
Rev 29:961–985 channels: voltage and pH dependence of mac-
22. Kroning N, Willenborg M, Tholema N, Hanelt roscopic currents. J Gen Physiol 128:
I, Schmid R, Bakker EP (2007) ATP binding 317–336
to the KTN/RCK subunit KtrA from the K+- 28. Yuan A, Santi CM, Wei A, Wang ZW, Pollak K,
uptake system KtrAB of Vibrio alginolyticus: Nonet M, Kaczmarek L, Crowder CM, Salkoff
Its role in the formation of the KtrAB complex L (2003) The sodium-activated potassium
and its requirement in vivo. J Biol Chem channel is encoded by a member of the Slo
282:14018–14027 gene family. Neuron 37:765–773
Chapter 23
Abstract
As large, multimeric, integral membrane proteins, ion channels pose technical challenges to analysis by
NMR spectroscopy. Here we present a strategy to overcome some of these technical hurdles, using a rep-
resentative ion channel modulatory domain, the regulator of K+ conductance (RCK) domain from a K+
channel cloned from Thermoplasma volcanium. By introducing a mutation to limit the stoichiometry of
the octameric RCK domain “gating ring” complex to its dimeric building block, NMR spectral resolution
can be greatly improved. Here we present protocols for efficient production of highly deuterated, uni-
formly 15N-labeled protein, as well as protein containing 15N-labeling to specific amino acid types. These
labeling strategies can be applied to improve spectral resolution and facilitate sequential resonance
assignments.
1 Introduction
Analysis of ion channel structure has, over the last 10 years, become
an indispensible component of molecular physiology (1–3). Yet
while structural analysis of ion channels seems to be dominated by
the static (but remarkably detailed) pictures provided by X-ray
crystallography, a quantitative understanding of channel confor-
mational dynamics through nuclear magnetic resonance (NMR)
spectroscopy has remained relatively limited. This inequity may
arise in part from technical hurdles posed by ion channels and their
modulatory domains, which are frequently beyond the size limit
inherent to solution NMR, except (notably) in the cases of KcsA
and the glutamate receptor ligand-binding domain (4–8). Large
proteins yield broad spectral lines and consequently have an intrin-
sically low signal-to-noise ratio. In addition, the large number of
amino acids that makes up a large protein increases spectral over-
lap. Both of these phenomena contribute to difficulties in making
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_23, © Springer Science+Business Media, LLC 2013
289
290 Karin Abarca-Heidemann et al.
Fig. 1 Comparison of structural properties of the TvoK WT RCK domain and RCK(ΔNAI) mutant. (a) Gel-filtration
elution profiles (measured by absorbance at 280 nm) of the wild-type RCK domain and RCK(ΔNAI) mutant. The
wild-type domain complex elutes from the Superdex-200 column at ~11 mL, consistent with a ~200 kDa
octamer, whereas the RCK(ΔNAI) mutant elutes at ~15 mL, consistent with a ~50 kDa dimer. (b) Circular
dichroism spectra for the TvoK RCK WT and RCK(ΔNAI) are nearly identical to one another. Together these
results are consistent with the (ΔNAI) mutation disrupting assembly of dimers to form an octameric complex,
but leaving secondary and tertiary structure intact
Fig. 2 15N,1H-TROSY-HSQC spectra obtained from deuterated, uniformly 15N-labeled protein. (a) TvoK RCK WT:
150 mM KCl, 50 mM HEPES, 2 mM β-mercaptoethanol, pH 7.5, 10% D2O, 47°C; (b) TvoK RCK(ΔNAI): 50 mM
KCl, 50 mM MES, 2 mM β-mercaptoethanol, pH 6.5, 10% D2O, 37°C. Both spectra were acquired using a
Bruker Avance 900 MHz spectrometer equipped with a cryoprobe. The ΔNAI (dimer) spectrum exhibits sharper,
more clearly distinguished crosspeaks than the spectrum from the octameric complex
2 Materials
a 10 9 8 7
b 10 9 8 7
15
105 N uniform labeling 105 105 105
15
N Lys
15
125 125 125 125
10 9 8 7 10 9 8 7
1 1
H chemical shift (ppm) H chemical shift (ppm)
c 10 9 8 7
d 10 9 8 7
15
105 105 105 N uniform labeling 105
15
N Lys
15 15
110 N Val 110 110
N Val 110
N chemical shift (ppm)
15
10 9 8 7 10 9 8 7
1 1
H chemical shift (ppm) H chemical shift (ppm)
Fig. 3 15N,1H-TROSY-HSQC spectra obtained from deuterated, uniformly 15N-labeled and uniformly deuterated,
amino acid-specific 15N-labeled RCK(ΔNAI). (a) Spectrum from deuterated, uniformly 15N-labeled protein;
(b) deuterated, 15N-Lys labeled protein; and (c) deuterated, 15N-Val labeled protein. (d) Overlay of spectra in
(a–c), illustrating superimposition of crosspeaks arising from Lys and Val N-H groups with those observed with
uniform 15N-labeling. Experimental conditions are the same as those described for Fig. 2b
10. Kanamycin (Sigma, St. Louis, MO): 200× solution, 100 mg per
10 mL H2O, filtered through 0.22 μm filter, stored at −20°C.
11. Isopropyl-β-D-1-thiogalactoside (IPTG; Sigma, St. Louis, MO).
12. Vitamin mix, 500×: per 100 mL, 250 mg thiamin, 500 mg
biotin, 500 mg choline chloride, 500 mg folic acid, 500 mg
niacinamide, 500 mg D-panthothenic acid, 50 mg riboflavin.
Store aliquots at −20°C.
13. Trace elements, 2,000×: per 100 mL, 2 g FeCl2, 25.6 mg
H3BO3, 7.2 mg CoCl2, 1.6 mg CuCl2, 136 mg ZnCl2, 242 mg
NaMoO4, 16 mg MnCl2. Store aliquots at −20°C.
14. Low glucose M9 medium: 47.7 mM Na2HPO4, 22 mM KH2PO4,
8.5 mM NaCl, 18.6 mM NH4Cl. After autoclaving, add 2 mM
MgSO4, 5.5 mM glucose, 100 μM CaCl2, 0.1 mL vitamin mix
(500×), 2 mL trace elements (2,000×) 0.5 mL, pH 7.4.
15. 15N,2H-Celtone: Celtone Base Powder (D, 97%+; 15
N 98%+;
Cambridge Isotope Laboratories, Andover, MA).
16. 2H-only-Celtone: Celtone Base Powder (D, 97%+; Cambridge
Isotope Laboratories, Andover, MA).
17. Spectra 9 medium (D, 97%; Cambridge Isotope Laboratories,
Andover, MA).
18. α-15N-Lys: L-Lysine:2HCl (ALPHA-15N, 95–99%; Cambridge
Isotope Laboratories, Andover, MA).
19. 15N-Val: L-Valine (15N 98%; Cambridge Isotope Laboratories,
Andover, MA).
20. 15N-Leu: L-Leucine (15N 98%; Cambridge Isotope Laboratories,
Andover, MA).
21. 15N,2H-Lys: L-Lysine:HCl (D9, 98%; 15
N2 98%; Cambridge
Isotope Laboratories, Andover, MA).
3 Methods
3.1 Preparation 1. Inoculate 6 mL LB medium with CT19 E. coli cells and incu-
of Competent CT19 bate the culture overnight at 37°C.
E. coli (RbCl Method) 2. Add 0.8 mL of the overnight culture to 80 mL pre-warmed
LB containing 20 mM MgCl2 and grow culture at 30–37°C
until OD600 ~ 0.4–0.5.
3. Chill the culture on ice at least for 10 min.
4. Spin at 2,500 × g for 15 min at 4°C.
5. Resuspend cells by gentle swirling in 32 mL ice-cold TfB1.
6. Incubate the cell suspension on ice for 10 min.
Isotope Labeling for NMR Spectroscopy 295
4 Notes
Acknowledgements
References
Abstract
Dendrites emerging from the cell bodies of neurons receive the majority of synaptic inputs. They possess
a plethora of ion channels that are essential for the processing of these synaptic signals. To fully understand
how dendritic ion channels influence neuronal information processing, various patch-clamp techniques
that allow electrophysiological recordings to be made directly from dendrites have been developed. In this
chapter, I describe one such method that is suitable for making electrophysiological recordings from the
apical dendrites of hippocampal and cortical pyramidal neurons.
1 Introduction
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_24, © Springer Science+Business Media, LLC 2013
303
304 Mala M. Shah
2 Materials
3 Methods
3.1 Slicing 1. Make and freeze ~400 mL of cutting solution until ~45% of it
Procedure is ice. Depending on the freezer, this can take between 45 min
and 1 h.
2. In the meantime, lay out the tools for surgery, make the external
solution (usually 1 L), set up the slicer [including cleaning the
blade (e.g., Gillette blade) used for cutting slices with acetone
and/or ethanol], and prepare an incubation chamber for storing
slices. This usually consists of a beaker containing either an inter-
face chamber or a submerged chamber with cotton mesh on it in
which the slices rest. Add external solution to the chamber and
bubble it with 95% O2/5% CO2. The solution in this chamber is
pre-warmed to 37°C in a water bath prior to slices being added.
3. When the cutting solution is adequately frozen, take it out of
the freezer and use a handheld blender or equivalent to make
a slushy icy solution. This solution is bubbled with 95% O2/5%
CO2 and kept on ice for the remainder of the procedure.
4. Terminally anesthetize the animal in accordance with local eth-
ical guidelines (e.g., with an injectable anesthetic such as ket-
amine/xylazine solution).
5. When the animal is fully anesthetized (pinching the paw pro-
duces no reflexes), cut open the chest cavity and intracardially
perfuse the slushy cutting solution until the liver turns pale.
Decapitate the head and speedily remove the brain into the
ice-cold cutting solution. Appropriate regulations for animal
experimentation must be followed.
6. Place the brain on a suitable cold surface (block of cold metal
or petri dish with a sylgard bottom) and submerge it in ice-
cold cutting solution. For hippocampal slices from which
recordings from CA1 pyramidal dendrites can be made, first
hemi-sect the brain. Each half is then placed on the cut (medial
surface) and a cut at approximately 30°C to the base of the
half-brain is made on the dorsal surface. If the experiment
involves patching entorhinal cortex pyramidal cell dendrites,
then a few millimeters parallel to the ventral surface is cut from
the dorsal side (see Note 5).
7. This cut surface is then glued onto the slicing stage of a vibratome
using a thing film of cyanoacrylate glue or equivalent. The slic-
ing stage can be cooled prior, though this is not necessary.
8. Transfer the slicing stage into the slicing chamber of the
vibratome. Submerge the blocks of tissue with ice-cold cutting
solution. The solution in the chamber is also bubbled with 95%
O2/5% CO2.
9. Lower the blade for cutting slices to the appropriate position.
Cut thin tissue slices (200–400 μm) by advancing the blade at
Dendritic Ion Channel Recording 307
3.2 Dendritic 1. Pull patch pipettes using thick-walled borosilicate glass with a
Electrophysiological suitable patch pipette puller.
Recordings 2. Add the slice to the slice chamber being perfused at a constant
rate (1–2 mL/min) with external solution bubbled with
95%O2/5% CO2. The slice should be orientated such that the
dendrites to be patched are perpendicular to the patch pipette
(Fig. 2).
3. The slices are held down in the slice chamber using a “harp.”
This is a horseshoe-shaped flat metal (either silver or platinum)
onto which thin threads are glued.
4 Notes
References
1. Nusser Z (2009) Variability in the subcellular 4. Davie JT, Kole MH, Letzkus JJ, Rancz EA,
distribution of ion channels increases neuronal Spruston N, Stuart GJ, Häusser M (2006)
diversity. Trends Neurosci 32:267–274 Dendritic patch-clamp recording. Nat Protoc
2. Shah MM, Hammond RS, Hoffman DA (2010) 1:1235–1247
Dendritic ion channel trafficking and plasticity. 5. Nevian T, Larkum ME, Polsky A, Schiller J
Trends Neurosci 33:307–316 (2007) Properties of basal dendrites of layer 5
3. Sjostrom PJ, Rancz EA, Roth A, Hausser M pyramidal neurons: a direct patch-clamp record-
(2008) Dendritic excitability and synaptic plas- ing study. Nat Neurosci 10:206–214
ticity. Physiol Rev 88:769–840
Chapter 25
Abstract
Electrophysiological recordings from an acutely sliced preparation provide information on ionic currents
and excitability of native neurons under near physiological conditions. Although this technique is com-
monly used on central nervous system structures such as spinal cord and brain, structures within the
peripheral nervous system (including sensory ganglia and fibers) have proven to be much more difficult to
study in acute preparations. Here we describe a method for patch-clamp recordings from rat dorsal root
ganglion (DRG) slices.
Key words Dorsal root ganglion, Patch-clamp recording, Slice preparation, Ion channel, M-type K+
channel
1 Introduction
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_25, © Springer Science+Business Media, LLC 2013
311
312 Kirstin E. Rose et al.
2 Materials
3 Methods
3.1 Removal and 1. Wash all tools (Fig. 2a) and sterilize with 70% v/v ethanol/
Embedding of DRG deionized water prior to tissue removal.
2. Following humane sacrifice of rat to local ethical standards,
place the body on a dissection board with the dorsal surface
facing uppermost. Cut away the skin overlying the lumbar
region and proceed to carry out a laminectomy of the exposed
lumbar region. Specifically, remove the entire posterior back-
bone along with overlying ligaments and muscles, which will
normally include DRGs L3–L6.
314 Kirstin E. Rose et al.
Fig. 1 Patch-clamp setup for slice recording. (1) Vibration isolation table; (2) Faraday cage; (3) upright
microscope equipped with IR-DIC; (4) microscope stage; (5) water-immersion objectives; (6) video camera (in
our case, connected to a monitor located in a rack beside the patch-clamp setup); (7) headstage and pipette
holder attached to a micromanipulator; (8) micromanipulators remote control panels (one controlling the
microscope stage (8a) and another one controlling the pipette (8b)), mounted on a bench separated from the
vibration isolation table; (9) 2 peristaltic pumps [one controlling inflow (9a) and the second the outflow (9b)];
(10) erlenmeyer containing carbogen-bubbled ES
3. Pin the excised section onto a dissection board with the ante-
rior surface facing uppermost. Make cuts through the lamina
body of the spinal vertebrae, starting at the anterior end, until
the left and right sides of the column are separated. Using fine
forceps remove the spinal cord and meninges to expose the
DRGs under the binocular stereo dissecting microscope. They
should look like little pearls at regular intervals along the col-
umn (Fig. 2b).
4. Use fine forceps to carefully remove the L4 and L5 DRGs (see
Note 1). Place the DRG into HBSS buffer on ice (see Note 2).
While in buffer on ice, remove any attached nerves and roots
using fine scissors (see Note 3).
5. Prepare a 2% w/v agar solution by diluting for example 0.5 g
agar in 25 mL ES. Heat the solution using a microwave owen
and stop the heating just before the solution boils. Pour liquid
2% w/v agar into a small weighing boat and carefully place
DRG into agar using fine forceps (see Note 4). Place weighing
boat containing embedded DRGs on ice for 5–10 min to allow
agar to set.
Patch Clamp Recording from DRG Slices 315
Fig. 2 Major steps of the procedure to prepare DRG slices. (a) General view of the setup for removal of DRGs.
(1) Binocular stereo dissecting microscope; (2) light source; (3) dissecting tools (from left to right: 2 watch-
maker forceps with very fine tips, fine scissors, scissors, Pasteur pipette, flat tip tweezers); (4) 2 ES-containing
Petri dishes (one large to keep the spine and one small to keep the DRGs) placed on ice. (b) View of DRGs
(indicated by the red arrows) still within the cut open vertebral column (with spinal cord removed) observed
with binocular stereo dissecting microscope. (c) General view of the slicing setup. (1) Vibrating blade micro-
tome; (2) brain slice keeper; (3) tools (from left to right : fine scissors, watchmaker forceps with very fine tips,
flat tip tweezers, spatula to prepare the agar solution, Pasteur pipette to transfer the slices to the slice keeper,
specimen disc, scalpel). (d) DRGs are embedded in cooled agar in a weighing boat placed on cold ES. (e) Blocks
of agar (each containing one DRG) dried by placing them on filter paper. (f) Each block of agar is glued on the
specimen disc, placed in the buffer tray (containing cold carbogen-bubbled ES) of the vibratome and cut into
slices. (g) Lateral view of a slice keeper containing ES continuously “bubbled” with carbogen. The flow rate of
the gas is adjusted with the needle valve
3.2 Slicing and Slice 1. Using a scalpel cut cubes of agar containing one DRG and glue
Incubation onto the specimen disc (see Note 5).
2. Slice cube containing DRG on a vibroslicer (i.e., vibrating
blade microtome Leica VT100S) in ES solution on ice. Set the
316 Kirstin E. Rose et al.
3.3 Perforated 1. Place slice in recording chamber (see Note 9) and hold in place
Patch-Clamp of Acute using a slice anchor usually made from a high-density material
DRG Slice stable in water (i.e., platinum, gold, or tungsten). Our experi-
ments were performed at room temperature. Optionally, a
desired temperature can be maintained by a temperature con-
troller heating solution prior its entry into the recording
chamber.
2. Perfuse the chamber with ES continuously saturated with car-
bogen at a flow rate of 3–5 mL/min (see Note 10).
3. When selecting a cell to patch generally smaller cells that did
not have any covering/surrounding glia are more amenable to
patching.
4. For perforated patch-clamp technique pipettes are backfilled
with amphotericin solution containing Lucifer yellow (optional;
see Note 11) and then tip is dipped in amphotericin-free IS for
30 s to 1 min.
5. The pipette is mounted into the headstage amplifier and small
amount of positive pressure is applied before the pipette enters
into the bath solution to maintain a clean electrode tip; the posi-
tive pressure can be maintained with bulldog clip up until the
contact with the cell surface is made (see below and Note 12).
The pipette is maneuvered down through bath until it is
approximately level with the cell of interest.
6. Once in close approximation to the cell, the pipette is slowly
advanced onto cell membrane until pipette resistance reaches
~10 MΩ (see Note 13).
7. Positive pipette pressure is then released and negative pressure
is progressively applied to pipette by mouth until GΩ seal is
achieved. Negative pressure is then slowly released.
8. The pipette capacitance (Cfast) is then compensated using an
amplifier/software. The cell membrane capacitance (Cslow) will
start increasing with time as the amphotericin perforation
developed. This can be monitored on the oscilloscope online;
the perforation usually takes up to 10 min (see Chapter 11 of
this book for more details).
9. Once membrane capacitance has reached a plateau it is cancelled
with the use of the amplifier/software. The method normally
Patch Clamp Recording from DRG Slices 317
a b c
d e f Capsaicin
XE991
Current density at –30 mV, pA/pF
–60 pA 9 –20
20 pA
Control 8
200 ms –40
7
XE991 –60
...................................... 6
0 pA
0 200 400 600 800 1000 0 200 400 600
Time, s Time, s
Fig. 3 Patch-clamp recording from DRG slice. (a) Low-magnification (×10) micrograph of the DRG.
(b) Micrograph depicting DRG slice with a patch-clamp pipette making a GΩ seal with a DRG neuron.
(c) Fluorescence micrograph of the DRG slice taken after the perforated patch recording was completed and
the recorded cell was filled with Lucifer yellow through the patch pipette (see Note 11). (d) Perforated patch
recording of M-like current from a small-diameter neuron in an acute DRG slice in the absence (control) or
presence (XE991) of specific M channel blocker XE991 (3 μM); voltage protocol is depicted above. (e) Time
course of M-current inhibition by 3 μM XE991 (black bar). (f) Activation of TRPV1 channel with capsaicin
(1 μM); perforated patch recording, currents were elicited with the same voltage pulse shown in (d), plotted is
a steady-state current at −60 mV. Panels (d) and (e) are modified from (9) with permission
4 Notes
Acknowledgments
References
1. Safronov BV, Bischoff U, Vogel W (1996) signaling in peripheral sensory neurons. Proc
Single voltage-gated K+ channels and their Natl Acad Sci USA 109:E1578–E576
functions in small dorsal root ganglion neu- 4. Linley JE, Pettinger L, Huang D, Gamper N
rones of rat. J Physiol 493:393–408 (2012) M channel enhancers and physiological
2. Crozier RA, Ajit SK, Kaftan EJ, Pausch MH M channel block. J Physiol 590:793–807
(2007) MrgD activation inhibits KCNQ/M- 5. Linley JE, Rose K, Patil M, Robertson B,
currents and contributes to enhanced neuronal Akopian AN, Gamper N (2008) Inhibition of
excitability. J Neurosci 27:4492–4496 M current in sensory neurons by exogenous
3. Linley JE, Ooi L, Pettinger L, Kirton H, Boyle proteases: a signaling pathway mediating
JP, Peers C, Gamper N (2012) Reactive oxy- inflammatory nociception. J Neurosci
gen species are second messengers of neurokinin 28:11240–11249
320 Kirstin E. Rose et al.
6. Mucha M, Ooi L, Linley JE, Mordaka P, Dalle 10. King CH, Scherer SS (2012) Kv7.5 is the
C, Robertson B, Gamper N, Wood IC (2010) primary Kv7 subunit expressed in C-fibers.
Transcriptional control of KCNQ channel genes J Comp Neurol 520:1940–1950
and the regulation of neuronal excitability. 11. Devaux JJ, Kleopa KA, Cooper EC, Scherer SS
J Neurosci 30:13235–13245 (2004) KCNQ2 is a nodal K+ channel.
7. Passmore GM, Reilly JM, Thakur M, Keasberry J Neurosci 24:1236–1244
VN, Marsh SJ, Dickenson AH, Brown DA 12. Roza C, Lopez-Garcia JA (2008) Retigabine,
(2012) Functional significance of M-type the specific KCNQ channel opener, blocks
potassium channels in nociceptive cutaneous ectopic discharges in axotomized sensory
sensory endings. Front Mol Neurosci 5:63 fibres. Pain 138:537–545
8. Passmore GM, Selyanko AA, Mistry M, 13. Shen W, Hamilton SE, Nathanson NM,
Al-Qatari M, Marsh SJ, Matthews EA, Surmeier DJ (2005) Cholinergic suppression of
Dickenson AH, Brown TA, Burbidge SA, KCNQ channel currents enhances excitability
Main M, Brown DA (2003) KCNQ/M cur- of striatal medium spiny neurons. J Neurosci
rents in sensory neurons: significance for pain 25:7449–7458
therapy. J Neurosci 23:7227–7236 14. Scholz A, Vogel W (2000) Tetrodotoxin-
9. Rose K, Ooi L, Dalle C, Robertson B, Wood resistant action potentials in dorsal root gan-
IC, Gamper N (2011) Transcriptional repres- glion neurons are blocked by local anesthetics.
sion of the M channel subunit Kv7.2 in chronic Pain 89:47–52
nerve injury. Pain 152:742–754
Chapter 26
Abstract
Mature human or mouse erythrocytes functionally express an unexpected diversity of ion channels that
endows these small enucleated cells with a toolkit for electrosignaling. Being largely dormant under resting
conditions, these ion channels enable erythrocytes to quickly respond to internal or external stimuli. They
are integral modules of complex programs such as oxygen-regulated ATP release or stress-induced pro-
grammed erythrocyte death. This article summarizes electrophysiologial and non-electrophysiological
methods to analyze erythrocyte ion channels and provides protocols for channel activation.
Key words Gardos channels (IK, SK4, KCa3.1), ClC-2, Organic osmolyte and anion channels, Protein
kinase A-activated anion channels, Ca2+-permeable nonselective cation channels, Patch-clamp,
Isosmotic hemolysis, Flow-cytometry, Tracer flux, ATP release
1 Introduction
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_26, © Springer Science+Business Media, LLC 2013
321
322 Stephan M. Huber
2 Materials
2.4 Other Solutions 1. Poly-L-lysine (1:100 in phosphate buffered saline for 10 min).
and Reagents 2. Triarylmethane-34 (TRAM-34).
3. Tert-Butylhydroperoxide (tBHP).
4. 5-Nitro-2-(3-phenylpropylamino)-benzoic acid, NPPB.
5. Ionomycin.
6. Sodium (meta)arsenite (NaAsO2).
7. Ca2+-pump inhibitor Na3VO4.
8. Fluo-3/AM or Fluo-4/AM.
9. Amiloride.
10. Ethylisopropylamiloride, EIPA.
11. 1-Ethyl-2-benzimidazolinone, 1-EBIO.
12. Gramicidin D.
13. Valinomycin.
14. Luciferin–luciferase assay kit (Roche Diagnostics, Mannheim,
Germany).
15. ELISA kit for the detection of trace amounts of free hemoglo-
bin (i.e., E-90HM; Immunology Consultants Laboratory,
Newberg, OR, USA).
16. Inosine.
45
17. Radioactive tracer Ca2+ (handling requires radioisotope
laboratory).
18. Trichloroacetic acid (6%).
3 Methods
3.1 Patch-Clamp 1. Mount Petri dish in your patch-clamp setup, superfuse with
Recording in Human NaCl solution and preheat the system to 37°C.
and Mouse 2. Earth the bath solution via salt bridge filled with NaCl
Erythrocytes solution.
3. Stop superfusion and add few microliter of erythrocyte
suspension.
4. Let the cells sediment and attach to the dish bottom. Remove
nonattached cells by fast superfusion (see Note 1).
5. Apply a constant superfusion of about 200 μL/min at 37°C.
6. Use borosilicate pipettes with resistances of 8–14 MΩ.
7. If necessary, improve giga-ohm seal formation (to 10–100 GΩ)
by applying negative pipette potential (−30 mV) at constant
(low) negative pipette pressure for up to 5 min and/or tran-
siently increase the bath CaCl2 concentration to 5 mM.
8. If your system is mechanically vulnerable, lift the giga-ohm sealed
cell from the bottom (not possible with poly-L-lysineated dish).
9. In cell-attached mode, continuously control for spontaneous
entry into whole-cell mode by microscopy (see below and
Fig. 1).
10. For entry into the whole-cell mode, rupture the membrane by
additional suction and/or brief electrical pulses (700 mV dur-
ing 100–200 μs). The whole-cell recording configuration is
indicated by a minute increase in capacitance and a simultane-
ous bleaching of the erythrocyte due to dialysis of hemoglobin
by the pipette solution (see Fig. 1 and Note 2).
11. Since whole-cell currents are low, control for formation of an
excised outside-out patch by monitoring the presence of a
“ghost” at the pipette tip (see Fig. 1c).
12. For patch excision, increase erythrocyte attachment to the dish
bottom by pre-coating the dish with poly-L-lysine.
13. Elicit currents by applying standard voltage pulse protocols
from −100 mV to +100 mV. Adapt protocol if high positive
voltages (³ +80 mV) result in loss of seal resistance. Sample
and low-pass filter currents at 10 and 3 kHz, respectively.
14. Estimate the leak current fraction by changing the electro-
chemical equilibrium of the charge-carrying ion species and/
or by pharmacologically inhibition of the involved ion chan-
nels (see Note 3).
15. Compute liquid junction potentials between pipette and NaCl
bath solution and between NaCl salt bridge and further bath
solution as reported (29) and correct voltages.
326 Stephan M. Huber
3.2.3 Activation of 1. Wash twice and suspend freshly drawn human erythrocytes in
Organic Osmolyte and Anion glucose-free NaCl solution to a hematocrit of 5% and store
Channels (See Note 8) them at 8°C for 1–7 days in order to energy deplete the cells
and to lower the oxidative defense.
2. Wash aliquots of 500 μL cell suspension with glucose-free
NaCl solution, pellet the cells (500 × g for 5 min), and discard
the supernatants.
3. Resuspend every 20 s a cell pellet in 1 mL of glucose-free NaCl
solution containing tert-butylhydroperoxide (tBHP, 0 or
1 mM). Prepare additional oxidized samples for isosmotic
hemolysis control (see Subheading 3.3.4).
328 Stephan M. Huber
3.2.4 Activation of ClC-2 1. Infect human erythrocytes (blood group 0) with laboratory
Cl− Channels (See Note 12) strains of Plasmodium falciparum and culture the infected cells
asynchronously at a hematocrit of 5% and a parasitemia of
2–10% in RPMI 1640 medium supplemented with Albumax II
(0.5%) in an atmosphere with reduced oxygen tension.
2. Mount a Petri dish in your patch-clamp setup and start super-
fusion with NaCl solution at 37°C. Transfer a 10 μL aliquot of
the infected culture directly in the NaCl solution. Prior to that,
stop superfusion.
3. Restart superfusion when erythrocytes have been sedimented
and attached to the dish bottom.
4. Identify erythrocytes parasitized with the trophozoite stage of
Plasmodium falciparum by light microscopy (see Note 13).
Ion Channels in Human Erythrocytes 329
Fig. 3 Activation of the 18 pS Cl− channels by arsenite. (a) Macroscopic cell-attached currents of a control erythrocyte
(left ) and an erythrocyte pretreated for 3 h with arsenite (1 mM, right). Currents were elicited by the indicated pulse
protocol (inset ) and recorded with K-gluconate pipette and NaCl bath solution. (b) Single-channel tracings recorded
at different voltages as in (a). (c) Relationship between channel amplitude and voltage
3.3.5 ClC-2 Chloride 1. Wash (three times) and equilibrate erythrocytes (5% hemat-
Channels Measured ocrit) for 15 min with glucose-free KCl/NaCl solution
by Cell Volume Changes (100 μL) containing 4 μM gramicidin D, 2 μM valinomycin,
(See Note 19) and 50 μM NPPB in order to Na+-permeabilize and to K+-
permeabilize the erythrocyte membrane and to inhibit organic
osmolyte and anion channels, respectively.
332 Stephan M. Huber
3.3.6 Pannexin-1 1. Wash (three times) and resuspend erythrocytes (5% hemat-
and VDAC ATP Channels ocrit) in NaCl solution.
Measured by Extracellular 2. Place 200 μL aliquots in a 96-well plate on ice.
ATP Accumulation
3. Let the erythrocytes sediment.
(See Note 20)
4. To avoid ATP release induced by mechanical perturbation,
replace carefully 180 μL supernatant by the same volume of
ice-cold NaCl solution containing inducer (e.g., 0 or 200 μM
of tert-butylhydroperoxide) or/and inhibitors of ATP release
(e.g., 0 or 50 μM of NPPB).
5. Heat the cells to 37°C.
6. Harvest immediately after re-sedimentation (background val-
ues) and every 60 min thereafter the supernatants (100 μL).
7. Spin down supernatants (500 × g for 5 min) to remove residual
erythrocytes immediately after harvesting; again harvest super-
natant (90 μL) and freeze it at −25°C.
8. Quantify the extracellular ATP concentration of the aliquots
(50 μL) by a luciferin–luciferase assay kit and a luminometer
according to the supplied experimental protocol.
9. Subtract mean background values and calculate ATP release
per number of cells and time.
10. Run further ATP standard curves in the presence of the applied
inducers/inhibitors to control for impairment of the luciferin–
luciferase reaction by the drugs.
11. Determine the hemoglobin concentration of the remaining
supernatant volume by ELISA according to the instructions of
the supplier to control for hemolysis-mediated contamination
by intracellular ATP.
3.3.7 Tracer Flux 1. Wash erythrocytes in KCl/NaCl solution (see Note 22).
(See Note 21) Protocol 2. Pre-incubate the cells (10% hematocrit) for 10 min at 37°C in
for 45Ca2+ Uptake 400 μL of KCl/NaCl solution supplemented with 10 mM
inosine (to replenish the erythrocytic nucleoside pool which
diminishes by ATP release) and 1 mM Na3VO4 (to inhibit the
Ca2+-ATPase).
3. Add 400 μL of the same solution (37°C) additionally contain-
ing the radioactive tracer (~2 μCi/mL 45Ca2+) and 0.4 mM
CaCl2 and incubate at 37°C under slight agitation.
Ion Channels in Human Erythrocytes 333
4 Notes
10. Autocrine purinergic signaling via ATP release and P2Y recep-
tors facilitates the induction of active organic osmolyte and
anion channels (27).
11. Unusual features of organic osmolyte and anion channel-gen-
erated whole-cell currents are that a negative holding potential
(−30 mV) induces time-dependent inactivation of the inward
currents and that micromolar concentrations of serum albu-
min stimulate outward rectification (33, 34).
12. Human and mouse erythrocytes express volume-sensitive
ClC-2 Cl− channels which are not activated by cell volume
changes in unstimulated erythrocytes. Infection by the malaria
parasite Plasmodium or oxidative stress “resurrects” the dor-
mant channels which are then reversibly activated by cell
swelling and inactivated by cell shrinkage. ClC-2 channels
have very low single-channel conductance (3–4 pS) and are
best recorded by fast whole-cell mode where they exhibit
typical inward rectification and time-dependent activation at
negative voltages (5, 6). Here a protocol for Plasmodium-
infected human erythrocytes is given. This protocol can also
be used to activate the organic osmolyte and anion channels
(see Subheading 3.2.3).
13. Trophozoites detoxify hem as hemozoin which is clearly visible
as black dot in the food vacuole of the intraerythrocytic para-
site. These “dots” typically moves by Brownian motion.
14. The ClC-2 generated whole-cell current fraction is sensitive to
Zn2+ (1 mM).
15. The 18 pS anion channel of human erythrocytes is activated in
inside-out patches by protein kinase A (8). Sodium (meta)
arsenite confers oxidative stress to cells and perturbs intracellu-
lar phosphorylation/dephosphorylation pathways by targeting
several kinases (35). Here a simple protocol is provided to
activate 18 pS anion channels by pretreating the cells with
arsenite. Figure 3 gives an example of such activation.
16. Erythrocytes lack intracellular Ca2+ stores. Hence, steady-state
cytosolic free Ca2+ concentration [(Ca2+)i] depends solely on
Ca2+ entry and extrusion across the plasma membrane as
well as on cytosolic Ca2+ buffering by Ca2+-binding proteins.
The latter have high affinity but low capacity and low resting
Ca2+ concentrations are mainly maintained by the activity of
the powerful Ca2+ pump in the erythrocyte membrane. Thus,
perturbations of the pump leak equilibrium results in instanta-
neous changes in [Ca2+]i which can be measured by Ca2+-
specific dyes such as Fluo-3 or Fluo-4. Increase in fluorescence
(expressed here as a slope of the fluorescence increase with
time) in Ca2+-depleted erythrocytes after re-addition of extra-
336 Stephan M. Huber
Acknowledgments
References
1. Huber SM, Duranton C, Lang F (2005) Patch- 7. Skals M, Jensen UB, Ousingsawat J,
clamp analysis of the “new permeability path- Kunzelmann K, Leipziger J, Praetorius HA
ways” in malaria-infected erythrocytes. Int Rev (2010) Escherichia coli alpha-hemolysin trig-
Cytol 246:59–134 gers shrinkage of erythrocytes via K(Ca)3.1
2. Schillers H (2008) Imaging CFTR in its native and TMEM16A channels with subsequent
environment. Pflugers Arch 456:163–177 phosphatidylserine exposure. J Biol Chem
3. Verloo P, Kocken CH, Van der Wel A, Tilly 285:15557–15565
BC, Hogema BM, Sinaasappel M, Thomas 8. Egee S, Lapaix F, Decherf G, Staines HM,
AW, De Jonge HR (2004) Plasmodium falci- Ellory JC, Doerig C, Thomas SL (2002) A
parum-activated chloride channels are defec- stretch-activated anion channel is up-regulated
tive in erythrocytes from cystic fibrosis patients. by the malaria parasite Plasmodium falciparum.
J Biol Chem 279:10316–10322 J Physiol 542:795–801
4. Lange T, Jungmann P, Haberle J, Falk S, 9. Huber SM, Uhlemann AC, Gamper NL,
Duebbers A, Bruns R, Ebner A, Hinterdorfer Duranton C, Kremsner PG, Lang F (2002)
P, Oberleithner H, Schillers H (2006) Reduced Plasmodium falciparum activates endogenous
number of CFTR molecules in erythrocyte Cl− channels of human erythrocytes by mem-
plasma membrane of cystic fibrosis patients. brane oxidation. EMBO J 21:22–30
Mol Membr Biol 23:317–323 10. Huber SM, Lang C, Lang F, Duranton C
5. Huber SM, Duranton C, Henke G, Van De Sand (2008) Organic osmolyte channels in malaria-
C, Heussler V, Shumilina E, Sandu CD, Tanneur infected erythrocytes. Biochem Biophys Res
V, Brand V, Kasinathan RS, Lang KS, Kremsner Commun 376:514–518
PG, Hubner CA, Rust MB, Dedek K, Jentsch 11. Bouyer G, Cueff A, Egee S, Kmiecik J,
TJ, Lang F (2004) Plasmodium induces swell- Maksimova Y, Glogowska E, Gallagher PG,
ing-activated ClC-2 anion channels in the host Thomas SL (2011) Erythrocyte peripheral type
erythrocyte. J Biol Chem 279:41444–41452 benzodiazepine receptor/voltage-dependent
6. Shumilina E, Huber SM (2011) ClC-2 chan- anion channels are upregulated by Plasmodium
nels in erythrocytes. Open Biol J 4:18–26 falciparum. Blood 118:2305–2312
338 Stephan M. Huber
12. Sridharan M, Bowles EA, Richards JP, Krantic 24. Lang KS, Duranton C, Poehlmann H, Myssina
M, Davis KL, Dietrich KA, Stephenson AH, S, Bauer C, Lang F, Wieder T, Huber SM
Ellsworth ML, Sprague RS (2011) Prostacyclin (2003) Cation channels trigger apoptotic
Receptor-Mediated ATP Release from death of erythrocytes. Cell Death Differ
Erythrocytes Requires the Voltage-Dependent 10:249–256
Anion Channel (VDAC). Am J Physiol Heart 25. Barksmann TL, Kristensen BI, Christophersen
Circ Physiol 302:H553–H559 P, Bennekou P (2004) Pharmacology of the
13. Kucherenko YV, Morsdorf D, Lang F (2009) human red cell voltage-dependent cation chan-
Acid-sensitive outwardly rectifying anion chan- nel; Part I. Activation by clotrimazole and ana-
nels in human erythrocytes. J Membr Biol logues. Blood Cells Mol Dis 32:384–388
230:1–10 26. Lang PA, Kempe DS, Myssina S, Tanneur V,
14. Foller M, Bobbala D, Koka S, Boini KM, Birka C, Laufer S, Lang F, Wieder T, Huber
Mahmud H, Kasinathan RS, Shumilina E, SM (2005) PGE(2) in the regulation of pro-
Amann K, Beranek G, Sausbier U, Ruth P, grammed erythrocyte death. Cell Death Differ
Sausbier M, Lang F, Huber SM (2010) 12:415–428
Functional significance of the intermediate 27. Tanneur V, Duranton C, Brand VB, Sandu
conductance Ca2+-activated K+ channel for the CD, Akkaya C, Kasinathan RS, Gachet C,
short-term survival of injured erythrocytes. Sluyter R, Barden JA, Wiley JS, Lang F, Huber
Pflugers Arch 460:1029–1044 SM (2006) Purinoceptors are involved in the
15. Foller M, Kasinathan RS, Koka S, Lang C, induction of an osmolyte permeability in
Shumilina E, Birnbaumer L, Lang F, Huber malaria-infected and oxidized human erythro-
SM (2008) TRPC6 contributes to the Ca2+ cytes. FASEB J 20:133–135
leak of human erythrocytes. Cell Physiol 28. Binh VQ, Luty AJ, Kremsner PG (1997)
Biochem 21:183–192 Differential effects of human serum and cells
16. Hirschler-Laszkiewicz I, Tong Q, Conrad K, on the growth of Plasmodium falciparum
Zhang W, Flint WW, Barber AJ, Barber DL, adapted to serum-free in vitro culture condi-
Cheung JY, Miller BA (2009) TRPC3 activa- tions. Am J Trop Med Hyg 57:594–600
tion by erythropoietin is modulated by TRPC6. 29. Barry PH, Lynch JW (1991) Liquid junction
J Biol Chem 284:4567–4581 potentials and small cell effects in patch-clamp
17. Bennekou P (1993) The voltage-gated non- analysis. J Membr Biol 121:101–117
selective cation channel from human red cells 30. Duranton C, Huber SM, Tanneur V, Brand
is sensitive to acetylcholine. Biochim Biophys VB, Akkaya C, Shumilina EV, Sandu CD, Lang
Acta 1147:165–167 F (2004) Organic osmolyte permeabilities of
18. Locovei S, Bao L, Dahl G (2006) Pannexin 1 the malaria-induced anion conductances in
in erythrocytes: function without a gap. Proc human erythrocytes. J Gen Physiol
Natl Acad Sci U S A 103:7655–7659 123:417–426
19. Hoffman JF, Dodson A, Wickrema A, Dib- 31. Akkaya C, Shumilina E, Bobballa D, Brand VB,
Hajj SD (2004) Tetrodotoxin-sensitive Na+ Mahmud H, Lang F, Huber SM (2009) The
channels and muscarinic and purinergic recep- Plasmodium falciparum-induced anion channel
tors identified in human erythroid progenitor of human erythrocytes is an ATP-release path-
cells and red blood cell ghosts. Proc Natl Acad way. Pflugers Arch 457:1035–1047
Sci U S A 101:12370–12374 32. Staines HM, Alkhalil A, Allen RJ, De Jonge
20. Lang PA, Kaiser S, Myssina S, Birka C, HR, Derbyshire E, Egee S, Ginsburg H, Hill
Weinstock C, Northoff H, Wieder T, Lang F, DA, Huber SM, Kirk K, Lang F, Lisk G, Oteng
Huber SM (2004) Effect of Vibrio parahae- E, Pillai AD, Rayavara K, Rouhani S, Saliba KJ,
molyticus haemolysin on human erythrocytes. Shen C, Solomon T, Thomas SL, Verloo P,
Cell Microbiol 6:391–400 Desai SA (2007) Electrophysiological studies
21. Alper SL, Vandorpe DH, Peters LL, Brugnara of malaria parasite-infected erythrocytes: cur-
C (2008) Reduced DIDS-sensitive chloride rent status. Int J Parasitol 37:475–482
conductance in Ae1-/- mouse erythrocytes. 33. Staines HM, Powell T, Ellory JC, Egee S,
Blood Cells Mol Dis 41:22–34 Lapaix F, Decherf G, Thomas SL, Duranton
22. Duranton C, Huber SM, Lang F (2002) C, Lang F, Huber SM (2003) Modulation of
Oxidation induces a Cl− dependent cation con- whole-cell currents in Plasmodium falciparum-
ductance in human red blood cells. J Physiol infected human red blood cells by holding
539:847–855 potential and serum. J Physiol 552:177–183
23. Huber SM, Gamper N, Lang F (2001) 34. Duranton C, Tanneur V, Lang C, Brand VB,
Chloride conductance and volume-regulatory Koka S, Kasinathan RS, Dorsch M, Hedrich
nonselective cation conductance in human red HJ, Baumeister S, Lingelbach K, Lang F,
blood cell ghosts. Pflugers Arch 441:551–558 Huber SM (2008) A high specificity and affinity
Ion Channels in Human Erythrocytes 339
interaction with serum albumin stimulates an pathways in the mouse erythrocyte membrane.
anion conductance in malaria-infected erythro- FEBS Lett 585:3430–3435
cytes. Cell Physiol Biochem 22:395–404 38. Kirk K (2001) Membrane transport in the
35. Ventura-Lima J, Bogo MR, Monserrat JM malaria-infected erythrocyte. Physiol Rev
(2011) Arsenic toxicity in mammals and aquatic 81:495–537
animals: a comparative biochemical approach. 39. Lang PA, Kasinathan RS, Brand VB,
Ecotoxicol Environ Saf 74:211–218 Duranton C, Lang C, Koka S, Shumilina E,
36. Sridharan M, Adderley SP, Bowles EA, Egan Kempe DS, Tanneur V, Akel A, Lang KS,
TM, Stephenson AH, Ellsworth ML, Sprague Foller M, Kun JF, Kremsner PG, Wesselborg
RS (2010) Pannexin 1 is the conduit for low S, Laufer S, Clemen CS, Herr C, Noegel AA,
oxygen tension-induced ATP release from Wieder T, Gulbins E, Lang F, Huber SM
human erythrocytes. Am J Physiol Heart Circ (2009) Accelerated clearance of Plasmodium-
Physiol 299:H1146–H1152 infected erythrocytes in sickle cell trait and
37. Qiu F, Wang J, Spray DC, Scemes E, Dahl G annexin-A7 deficiency. Cell Physiol Biochem
(2011) Two non-vesicular ATP release 24:415–428
Chapter 27
Abstract
Ion channels play key roles in physiology. They function as protein transducers able to transform stimuli
and chemical gradients into electrical signals. They also are critical for cell signaling and play a particularly
important role in epithelial transport acting as gateways for the movement of electrolytes across epithelial
cell membranes. Experimental limitations, though, have hampered the recording of ion channel activity in
many types of tissue. This has slowed progress in understanding the cellular and physiological function of
these channels with most function inferred from in vitro systems and cell culture models. In many cases,
such inferences have clouded rather than clarified the picture. Here, we describe a contemporary method
for isolating and patch-clamping renal tubules for ex vivo analysis of ion channel function in native tissue.
Focus is placed on quantifying the activity of the epithelial Na+ channel (ENaC) in the aldosterone-sensitive
distal nephron (ASDN). This isolated, split-open tubule preparation enables recording of renal ion chan-
nels in the close to native environment under the control of native cell signaling pathways and receptors.
When combined with complementary measurements of organ and system function, and contemporary
molecular genetics and pharmacology used to manipulate function and regulation, patch-clamping renal
channels in the isolated, split-open tubule enables understanding to emerge about the physiological func-
tion of these key proteins from the molecule to the whole animal.
Key words Patch clamp, Isolated renal tubules, Ion channel recording, Collecting duct, ENaC
1 Introduction
Ion channels are key intrinsic membrane proteins. They are respon-
sible for the diversity of electrical signaling found in every living cell.
In addition, electrolytes and water, the latter of which follows the
movement of electrolytes via osmosis, are transported across epithe-
lial barriers of the integumentary system, gastrointestinal tract, the
kidney, secretory glands, and many other organs, in part, via ion
channels. Vectorial ion transport across epithelia is a consequence of
the selective expression of ion channels and transporters in apical
and basolateral membranes of epithelial cells. Understanding the
basics of ion channel biophysics and function as well as regulation is
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_27, © Springer Science+Business Media, LLC 2013
341
342 Elena Mironova et al.
2 Materials
2.1 Mouse Sacrifice 1. Six-week-old C57BL/6J male mice (this mouse strain is
and Kidney Isolation broadly commercially available, for instance from Jackson
Laboratory, USA; strain code 000664) (see Note 1).
2. CO2 gas tank and hermetic chamber for sacrifice.
3. Cold Hanks balanced salt solution (HBSS) (Sigma Aldrich,
USA).
4. 6-cm-diameter plastic Petri dishes (BD Falcon, USA).
5. Standard straight forceps (Fine Science Tools, USA).
6. Straight surgical scissors (Fine Science Tools, USA).
7. Nitrile gloves (Kimberly-Clark, USA).
2.3 Single-Channel 1. Patch clamp amplifier (we use Axopatch 200B Molecular
Analysis of ENaC Devices., Downingtown, PA, USA; see Note 3).
Activity in Isolated 2. Digitizer, i.e., Digidata 1322A or 1400 A/D board (Molecular
Tubules Using the Devices) interfaced with a PC running appropriate data acqui-
Patch-Clamp Method sition and analysis software (i.e., pClamp 9.2 or newer software
suite from Molecular Devices).
344 Elena Mironova et al.
3 Methods
slicing
isolation of kidney
Microscopic surgery:
Tubule Isolation
Fig. 1 Schematic illustration of the method used to isolate cortical collecting ducts for patch-clamp analysis.
The kidney is isolated from the mouse and then cut into thin slices (<1 mm). Segments of interest are then
mechanically isolated from these slices with microdissection typically using watchmaker forceps and a
stereomicroscope
Fig. 2 Shown here is a segment of the cortical collecting duct. The top of this segment has been split open to
allow patch-clamp access to the apical membranes of lining epithelial cells
a b 0 mV c c V (mV)
LiCl –60 –50 –40 –30 –20 –10
–20 mV c
c–a NaCl –0.2
–40 mV c –0.4
–0.6
–60 mV c
–0.8
I (pA)
1 pA –1.0
2 sec
Fig. 3 Recording ENaC at the apical membrane of a principal cell in an isolated, split-open collecting duct.
(a) Patch-clamp recording in cell-attached configuration. Major ions in bath and pipette solutions are shown.
(b) Representative single-channel current traces for ENaC from a principal cell. All recordings were performed
in the cell-attached configuration in the voltage-clamp mode. Current was recorded at test potentials that
ranged from 0 to −60 mV. Inward Li+ currents are depicted as downward deflection, and the dashed lines show
the 0 current level (closed state) at each voltage. (c) Single-channel current–voltage relationship for ENaC in
cell-attached patches that were made on the apical membrane of principal cells in isolated split-open mouse
collecting ducts
a b
1 AVP 2 1 ATP 2
1 pA 1 pA
60 sec 20 sec
1. before 1. before
c c
2. + AVP
c 2 sec
2. + ATP
c
2 sec
Fig. 4 Activation of ENaC in principal cells of isolated cortical collecting ducts by arginine vasopressin (AVP) (a)
and inhibition by ATP (b). Shown here are representative single-channel current traces for ENaC in a cell-
attached patch from a principal cell before and after application of 1 μM AVP (a) and 10 μM ATP (b). Arrows
indicate addition of AVP and ATP to the external bath solution. These patches were clamped to a holding potential
of −Vp = −60 mV. Closed state noted with c and areas below 1 (before) and 2 (after addition of AVP or ATP) shown
at an expanded time scale below. Data originally presented in (39, 46) and reproduced here with permission
3.1 Mouse Kidney 1. Make sure that your handling of animals adheres to the appro-
Isolation priate ethical guidance and legislation (see Note 5).
2. Animal work is performed using protective gloves under appro-
priate safety conditions.
348 Elena Mironova et al.
b
0.5
Po
0.4
0.3
a
0.2
0.01 % [Na+]
0.1
c n=29 n=20 n=18
0.0
<0.01% 0.32% 2.0% [Na+]
4
N
3
0.32 % [Na+] 2
c
1
0
<0.01% 0.32% 2.0% [Na+]
0.4
1 pA 0.2
2 sec 0.0
<0.01% 0.32% 2.0% [Na+]
Fig. 5 ENaC activity and density are controlled by dietary NaCl intake in mice. (a) Representative current traces
from cell-attached patches containing ENaC from mice kept on low (<0.01%)-Na+ (top), regular (0.32%)-Na+
(middle), and high (2%)-Na+ (bottom) diets. C denotes the closed state. (b) Summary of ENaC Po, fNPo, and N
for mice kept on low, regular, and high-salt diets. *P < 0.05 vs. regular (0.32%)-Na+ diet; data are expressed
as means ± SE. Numbers inside bars indicate number of experiments. Figure originally presented in (45) and
reproduced here with permission
a b
0.32 % [Na+] 0.5 ∗
c Po
0.4
0.3
1 pA
2 sec 0.01 % [Na+] 0.2
c 0.1
n=27 n=13
0.0
0.32% <0.01% [Na+]
Fig. 6 ENaC activity and density are controlled by dietary NaCl intake in rats. (a) Representative current traces
from cell-attached patches containing ENaC from Sprague–Dawley rats kept on low (<0.01%)-Na+ (bottom)
and regular (0.32%)-Na+ (top) diets. C denotes the closed state. (b) Summary of ENaC Po for rats kept on low
(<0.01%)-Na+ and regular (0.32%)-Na+ diets. *P < 0.05 vs. regular (0.32%)-Na+ diet; data are expressed as
means ± SE. Numbers inside bars indicate number of experiments. Figure originally presented in (32) and
reproduced here with permission
Channels in the Split-Open Tubule 349
3.2 Isolation of 1. The kidney is taken from the HBSS solution and decapsulated
Mouse Renal Tubules using forceps. Figure 1 illustrates the steps of this procedure
for reference.
2. The decapsulated kidney is cut into thin slices (<1 mm) with a
razor blade in a 10-cm-diameter plastic Petri dish.
3. One of the slices is placed into a new 6-cm-diameter plastic
Petri dish with approximately 3 mL of fresh ice-cold HBSS
solution. This Petri dish is put under the stereomicroscope.
4. Using two watchmaker forceps the slice is divided into sections
using blunt dissection. Each section should contain approxi-
mately 10–20 tubules. From the cortical part of each section,
the collecting tubules of individual nephrons are carefully iso-
lated (see Note 8).
5. Isolated tubules are transferred to poly-D-lysine pre-coated
5 × 5 mm glass chips (see Note 9).
3.3 Splitting 1. A chip containing tubules is placed into the recording chamber
Open Tubules affixed to the stage of an inverted microscope. The chip is per-
fused with bath (extracellular) solution for 2–3 min to remove
cell debris (see Note 10).
2. The tubule is then split open with two sharp micropipettes con-
trolled with separate micromanipulators to gain access to the
apical plasma membrane of epithelial cells within the tubule.
3. The tubule is oriented in the chamber so that its midline is
perpendicular to the front of the microscope.
4. The tubule is gently pinned with the left micropipette, taking
care not to press through to the bottom cells. With the left
pipette stationary, the top surface of the tubule is scraped away
with the right pipette to open it (see Fig. 2).
5. When the tubule is opened the left pipette is moved a small dis-
tance closer to the front, and step number 4 is repeated. Continue
to split open the tubule as needed for your experiment.
6. The apical membranes of cells within the tubule are cleaned
with a suction pipette under microscopic observation. With
this cleaning procedure, the success rate in obtaining gigaohm
seals increases.
350 Elena Mironova et al.
3.4 Single-Channel 1. Prepare bath (extracellular) and pipette solutions, and patch
Analysis of ENaC pipettes (see Note 11).
Activity Using the 2. The fire-polished glass patch pipette is slowly lowered to the
Patch-Clamp Method surface of the selected cell (see Note 12) and a high-resistance
(>1 gΩ) seal is formed between the pipette and plasma mem-
brane by applying negative pressure to the back of the record-
ing pipette usually through gentle suction. Seal formation is
assessed by monitoring pipette resistance with resistance going
from ~7–10 mΩ to more than 8–10 gΩ upon successfully
forming the cell-attached seal.
3. The patch membrane can then be voltage clamped (keeping
the voltage constant) to observe changes in current or current
clamped (keeping current constant) to observe changes in
membrane voltage. For our cell-attached voltage-clamp stud-
ies, currents were low-pass filtered at 100 Hz by an eight-pole
Bessel filter and digitized and stored on a PC hard drive using
the Digidata interface (see Note 3).
4 Notes
Acknowledgments
References
1. Biervert C, Schroeder BC, Kubisch C, Berkovic in the study of ion channels. J Pharmacol
SF, Propping P, Jentsch TJ, Steinlein OK Toxicol Methods 51:177–185
(1998) A potassium channel mutation in neo- 12. Robbins J, Trouslard J, Marsh SJ, Brown DA
natal human epilepsy. Science 279:403–406 (1992) Kinetic and pharmacological properties of
2. Bonny O, Hummler E (2000) Dysfunction of the M-current in rodent neuroblastoma × glioma
epithelial sodium transport: from human to hybrid cells. J Physiol 451:159–185
mouse. Kidney Int 57:1313–1318 13. Staruschenko A, Booth RE, Pochynyuk O,
3. Hummler E, Horisberger JD (1999) Genetic Stockand JD, Tong Q (2006) Functional
disorders of membrane transport. V. The epithe- reconstitution of the human epithelial Na+
lial sodium channel and its implication in human channel in a mammalian expression system.
diseases. Am J Physiol 276:G567–G571 Methods Mol Biol 337:3–13
4. Rossier BC, Pradervand S, Schild L, Hummler 14. Bugaj V, Mironova E, Kohan DE, Stockand JD
E (2002) Epithelial sodium channel and the (2012) Collecting duct-specific endothelin B
control of sodium balance: interaction between receptor knockout increases ENaC activity. Am
genetic and environmental factors. Annu Rev J Physiol Cell Physiol 302:C188–C194
Physiol 64:877–897 15. Mironova E, Peti-Peterdi J, Bugaj V, Stockand
5. Schild L (1996) The ENaC channel as the pri- JD (2011) Diminished paracrine regulation of
mary determinant of two human diseases: the epithelial Na+ channel by purinergic signal-
Liddle syndrome and pseudohypoaldoster- ing in mice lacking connexin 30. J Biol Chem
onism. Nephrologie 17:395–400 286:1054–1060
6. Schild L (2004) The epithelial sodium channel: 16. Pochynyuk O, Bugaj V, Rieg T, Insel PA,
from molecule to disease. Rev Physiol Biochem Mironova E, Vallon V, Stockand JD (2008)
Pharmacol 151:93–107 Paracrine regulation of the epithelial Na+ chan-
7. Simon DB, Bindra RS, Mansfield TA, Nelson- nel in the mammalian collecting duct by puri-
Williams C, Mendonca E, Stone R, Schurman nergic P2Y2 receptor tone. J Biol Chem
S, Nayir A, Alpay H, Bakkaloglu A, Rodriguez- 283:36599–36607
Soriano J, Morales JM, Sanjad SA, Taylor CM, 17. Roos KP, Strait KA, Raphael KL, Blount MA,
Pilz D, Brem A, Trachtman H, Griswold W, Kohan DE (2012) Collecting duct-specific
Richard GA, John E, Lifton RP (1997) knockout of adenylyl cyclase type VI causes a
Mutations in the chloride channel gene, urinary concentration defect in mice. Am J
CLCNKB, cause Bartter’s syndrome type III. Physiol Renal Physiol 302:F78–F84
Nat Genet 17:171–178 18. Rubera I, Hummler E, Beermann F (2009)
8. Snyder PM, Price MP, McDonald FJ, Adams Transgenic mice and their impact on kidney
CM, Volk KA, Zeiher BG, Stokes JB, Welsh MJ research. Pflugers Arch 458:211–222
(1995) Mechanism by which Liddle’s syn- 19. Bailey MA, Giebisch G, Abbiati T, Aronson PS,
drome mutations increase activity of a human Gawenis LR, Shull GE, Wang T (2004) NHE2-
epithelial Na+ channel. Cell 83:969–978 mediated bicarbonate reabsorption in the distal
9. Wollnik B, Schroeder BC, Kubisch C, Esperer tubule of NHE3 null mice. J Physiol 561:
HD, Wieacker P, Jentsch TJ (1997) 765–775
Pathophysiological mechanisms of dominant 20. Huang DY, Osswald H, Vallon V (2000)
and recessive KVLQT1 K+ channel mutations Sodium reabsorption in thick ascending limb of
found in inherited cardiac arrhythmias. Hum Henle’s loop: effect of potassium channel block-
Mol Genet 6:1943–1949 ade in vivo. Br J Pharmacol 130:1255–1262
10. Berjukow S, Doring F, Froschmayr M, Grabner 21. Thomson SC, Rieg T, Miracle C, Mansoury H,
M, Glossmann H, Hering S (1996) Endogenous Whaley J, Vallon V, Singh P (2012) Acute and
calcium channels in human embryonic kidney chronic effects of SGLT2 blockade on glomer-
(HEK293) cells. Br J Pharmacol 118:748–754 ular and tubular function in the early diabetic
11. Gamper N, Stockand JD, Shapiro MS (2005) rat. Am J Physiol Regul Integr Comp Physiol
The use of Chinese hamster ovary (CHO) cells 302:R75–R83
Channels in the Split-Open Tubule 353
22. Vallon V (2009) Micropuncturing the nephron. 34. Sakmann, B. and Neher, E. (1995) Single-channel
Pflugers Arch 458:189–201 recording. Springer Second Ed, 1–722.
23. Wang T, Hropot M, Aronson PS, Giebisch G 35. Bugaj V, Pochynyuk O, Mironova E, Vandewalle
(2001) Role of NHE isoforms in mediating A, Medina JL, Stockand JD (2008) Regulation
bicarbonate reabsorption along the nephron. of the epithelial Na+ channel by endothelin-1 in
Am J Physiol Renal Physiol 281:F1117–F1122 rat collecting duct. Am J Physiol Renal Physiol
24. Kirchner KA (1990) Greater loop chloride 295:F1063–F1070
uptake contributes to blunted pressure natri- 36. Karpushev AV, Levchenko V, Ilatovskaya DV,
uresis in Dahl salt sensitive rats. J Am Soc Pavlov TS, Staruschenko A (2011) Novel
Nephrol 1:180–186 role of Rac1/WAVE signaling mechanism in
25. Roman RJ, Kaldunski ML (1991) Enhanced regulation of the epithelial Na+ channel.
chloride reabsorption in the loop of Henle in Hypertension 57:996–1002
Dahl salt-sensitive rats. Hypertension 17: 37. Sun P, Lin DH, Yue P, Jiang H, Gotlinger KH,
1018–1024 Schwartzman ML, Falck JR, Goli M, Wang
26. Wagner CA, Lukewille U, Valles P, Breton S, WH (2010) High potassium intake enhances
Brown D, Giebisch GH, Geibel JP (2003) A the inhibitory effect of 11,12-EET on ENaC.
rapid enzymatic method for the isolation of J Am Soc Nephrol 21:1667–1677
defined kidney tubule fragments from mouse. 38. Palmer LG, Frindt G (1986) Amiloride-
Pflugers Arch 446:623–632 sensitive Na channels from the apical mem-
27. Schafer JA, Watkins ML, Li L, Herter P, brane of the rat cortical collecting tubule. Proc
Haxelmans S, Schlatter E (1997) A simplified Natl Acad Sci USA 83:2767–2770
method for isolation of large numbers of 39. Bugaj V, Pochynyuk O, Stockand JD (2009)
defined nephron segments. Am J Physiol Renal Activation of the epithelial Na+ channel in the
Physiol 273:F650–F657 collecting duct by vasopressin contributes to
28. Pavlov TS, Chahdi A, Ilatovskaya DV, water reabsorption. Am J Physiol Renal Physiol
Levchenko V, Vandewalle A, Pochynyuk O, 297:F1411–F1418
Sorokin A, Staruschenko A (2010) Endothelin-1 40. Chen L, Williams SK, Schafer JA (1990)
inhibits the epithelial Na+ channel through Differences in synergistic actions of vasopressin
βPix/14-3-3/Nedd4-2. J Am Soc Nephrol and deoxycorticosterone in rat and rabbit
21:833–843 CCD. Am J Physiol Renal Physiol 259:
29. Pavlov TS, Ilatovskaya DV, Levchenko V, F147–F156
Mattson DL, Roman RJ, Staruschenko A 41. Frindt G, Burg MB (1972) Effect of vasopres-
(2011) Effects of cytochrome P450 metabo- sin on sodium transport in renal cortical col-
lites of arachidonic acid on the epithelial sodium lecting tubules. Kidney Int 1:224–231
channel (ENaC). Am J Physiol Renal Physiol 42. Frindt G, Palmer LG (1996) Regulation of Na
301:F672–F681 channels in the rat cortical collecting tubule:
30. Pochynyuk O, Tong Q, Medina J, Vandewalle effects of cAMP and methyl donors. Am J
A, Staruschenko A, Bugaj V, Stockand JD Physiol Renal Physiol 271:F1086–F1092
(2007) Molecular determinants of PI(4,5)P2 43. Hawk CT, Li L, Schafer JA (1996) AVP and
and PI(3,4,5)P3 regulation of the epithelial aldosterone at physiological concentrations
Na+ channel. J Gen Physiol 130:399–413 have synergistic effects on Na + transport in rat
31. Pochynyuk O, Bugaj V, Vandewalle A, Stockand CCD. Kidney Int 57:S35–S41
JD (2008) Purinergic control of apical plasma 44. Helman SI, Grantham JJ, Burg MB (1971)
membrane PI(4,5)P2 levels sets ENaC activity Effect of vasopressin on electrical resistance of
in principal cells. Am J Physiol Renal Physiol renal cortical collecting tubules. Am J Physiol
294:F38–F46 220:1825–1832
32. Staruschenko A, Pochynyuk O, Vandewalle A, 45. Pochynyuk O, Rieg T, Bugaj V, Schroth J,
Bugaj V, Stockand JD (2007) Acute regulation Fridman A, Boss GR, Insel PA, Stockand JD,
of the epithelial Na + channel by phosphati- Vallon V (2010) Dietary Na+ inhibits the open
dylinositide 3-OH kinase signaling in native probability of the epithelial sodium channel in
collecting duct principal cells. J Am Soc the kidney by enhancing apical P2Y2-receptor
Nephrol 18:1652–1661 tone. FASEB J 24:2056–2065
33. Yu L, Helms MN, Yue Q, Eaton DC (2008) 46. Stockand JD, Mironova E, Bugaj V, Rieg T,
Single-channel analysis of functional epithelial Insel PA, Vallon V, Peti-Peterdi J, Pochynyuk
sodium channel (ENaC) stability at the apical O (2010) Purinergic inhibition of ENaC pro-
membrane of A6 distal kidney cells. Am J duces aldosterone escape. J Am Soc Nephrol
Physiol Renal Physiol 295:F1519–F1527 21:1903–1911
Chapter 28
Abstract
One of the most important functions of the kidney is the filtration of the blood that takes place in the
glomeruli. Glomerular epithelial cells (podocytes) have several functions, including regulation of the
filtration process and glomerular basement membrane turnover. Dysfunction of podocytes is a major cause
of glomerular kidney diseases. Gain-of-function mutations in the TRPC6 channel underlie a subset of
familial forms of focal segmental glomerulosclerosis (FSGS). While growing evidence supports an impor-
tant role of TRPC channels in podocytes, the regulation of these channels has yet to be investigated in
freshly isolated glomeruli. Native settings in glomeruli provide, by all means, the most appropriate as well
as one of the most challenging environments to study ion channel regulation. Thus, it is important to
develop new methods that would better reflect the native settings of the podocytes. To address this ques-
tion, we have established an experimental approach that allows studying podocytes in the freshly isolated
decapsulated intact glomeruli. Here we describe the preparation of the rat glomeruli for patch-clamping,
focusing on special conditions required for single-channel analysis of TRPC channels. Several tricks useful
for cell-attached patch-clamping of the glomerular podocytes and solutions appropriate for registration of
the TRPC channels are also provided.
Key words TRPC channels, TPC6, Glomeruli, Podocytes, Patch clamp, FSGS
1 Introduction
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_28, © Springer Science+Business Media, LLC 2013
355
356 Daria V. Ilatovskaya and Alexander Staruschenko
2 Materials
2.1 Rat Kidney 1. Eight-week-old Sprague–Dawley male rat (we used internal
Isolation Surgery MCW colony; however this rat strain is broadly commercially
and Perfusion available (for instance, Charles River, USA, strain code 400)).
2. 25 mL of the cold Hanks Balanced Salt Solution (HBSS)
(Sigma-Aldrich, USA).
3. 100 μL of pentobarbital (Sigma-Aldrich, USA).
4. 10 mL of the physiological salt solution (saline) (Sigma-
Aldrich, USA).
5. Polyethylene tubing (PE50, Sigma-Aldrich, USA).
6. Syringe pump-based perfusion system (Harvard Apparatus,
USA).
7. 1 × 2 teeth tip 12 cm Adson forceps (Fine Science Tools,
USA).
8. Straight sharp 13 cm surgical scissors (Fine Science Tools,
USA).
9. Straight 10 cm Graefe forceps (Fine Science Tools, USA).
10. Straight 12.5 cm Halsted-Mosquito hemostats (Fine Science
Tools, USA).
11. Schwartz 26 mm sharp-bend micro serrefines (Fine Science
Tools, USA).
12. Straight 11 cm Dumont #4 forceps (Fine Science Tools,
USA).
13. Surgical suture, braided silk (Surgical Specialties Corporation).
14. Temperature controlled surgical table for rodents (Harvard
Apparatus, USA).
15. Nitrile gloves (Kimberly-Clark, USA).
16. Binocular microscope (Nikon Eclipse TS100, Nikon, USA).
3 Methods
3 ml/min
catheterization
renal vein incision
mincing
isolation of cortex
mesh 100
15 ml
mesh 140
mesh 200
Fig. 2 Scheme of the glomeruli isolation protocol. Kidney cortex was isolated
from the Sprague–Dawley rat kidney and then homogenized with a blade. The
homogeneous cortex fraction was pushed through the consecutive steel sieves
of the different mesh size and then collected into a Petri dish
TRPC Channels in Podocytes 361
Fig. 3 Electron microscopy of freshly isolated decapsulated glomerulus. Fragments of the decapsulated
glomerulus were visualized with electron microscopy at 1,500×, 5,000×, and 20,000× (a, b, and c, respectively;
scale bars are shown). PB podocyte body, PN podocyte nucleus, EF endothelial fenestrations, FP foot processes
(reproduced from ref. 24 with permission from Elsevier)
3.2 Isolation 1. Before the experiment, fresh solution of the RPMI 1640 with
of the Rat Glomeruli BSA is prepared adding 5 mL of 30% BSA to 25 mL of the
media (see Note 4) (from here on referred to as solution A).
2. The kidney is taken from the HBSS solution and decapsulated
with the help of the surgical scissors. See Fig. 2 that illustrates
all the steps of the procedure for reference.
3. The decapsulated kidney is coronary cut in two halves with a
razor blade (see Note 5) and then cortex is isolated and placed
into approximately 4 mL of the ice-cold solution A.
4. The kidney is minced with a razor blade and mixed with a fine
transfer pipette to obtain a homogenous substance (see Note 6).
5. The homogenate from step 4 is transferred onto the top of the
100 mesh sieve (see Note 7 that comments on the mesh size)
362 Daria V. Ilatovskaya and Alexander Staruschenko
3.3 Single-Channel 1. Immediately after isolation, the 15 mL tube with the fraction
Analysis of the TRPC of the glomeruli (see Note 12) is left on ice for up to 20 min
Channel Activity Using to let the glomeruli settle down and concentrate at the bottom
Patch-Clamp Method of the tube (see Note 13).
2. After sedimentation the supernatant is removed and the rest of
the solution containing the concentrated fraction of the glom-
eruli is kept on ice.
3. Before the patch-clamp experiment, the solution containing
glomeruli is mixed and 50 μL of obtained solution are removed
by a pipettor and placed on the poly-D-lysine pre-coated 5 × 5
mm cover glasses (see Note 14); the chip is left at room tem-
perature for 5–10 min to let the glomeruli settle down and
attach to the surface. As demonstrated in Fig. 3, the podocytes
of isolated glomeruli are intact.
4. The glass chip placed into the recording chamber of the patch-
clamp setup is filled with the extracellular bathing solution and
the chamber is perfused with the solution for 2–3 min to
remove the unattached glomeruli and ensure the replacement
of the BSA-containing solution with the bath solution.
5. Directly before the patch-clamp experiments the pipette solu-
tion used for the single-channel recordings is complimented
with various inhibitors to prevent the registration of the unre-
lated chloride, calcium, and potassium currents: 100 μM
niflumic acid or DIDS, 10 mM TEA, 10 nM iberiotoxin,
10 nM nicardipine (see Note 15).
6. The patch pipette is slowly lowered to the surface of the glom-
erulus (see Note 16) and a high-resistance seal is formed with
application of gentle suction. The formation of a gigaohm seal
(>1 GΩ) is monitored by the pipette resistance that is increas-
ing from 7 to 10 MΩ to more than 1 GΩ after the suction is
applied. Figure 4 illustrates the decapsulated rat glomerulus in
the patch-clamp setup with a patch pipette attached to a podo-
cyte on the edge of the glomerulus.
TRPC Channels in Podocytes 363
Fig. 4 A representative image of a decapsulated rat glomerulus in the patch-clamp setup with a patch pipette
attached to a podocyte on the edge of the glomerulus (40× and a close-up image) (reproduced from ref. 24
with permission from Elsevier)
a b
o o
+40 mV c
+70 mV
c
o
c +60 mV
o
c +20 mV
o
c +40 mV
c 0 mV
c 0 mV
c
o -20 mV c
o -40 mV
c c
-40 mV o -60 mV
o
c
c
o -80 mV
-60 mV
o c
-100 mV
o
c
-80 mV c
-120 mV
o o
1 pA 1 s 1 pA 1 s
2
c d
C 1
Y1
-60 mV
Y2 Em, mV
-100 -50 50 100
C
-1
Y1
-60 mV I, pA
Y2 -2
1 pA 2s
Fig. 5 Single-channel biophysical properties of TRPC-like channels identified in podocytes of freshly isolated
rat glomeruli. All recordings were performed in cell-attached configuration in voltage-clamp mode.
Representative current traces of “big” (a) and “small” (b) channels identified in the podocytes. The holding
membrane potentials are indicated near the traces. c and o denote closed and open current levels, respec-
tively. (c) Typical current traces held at −60 mV demonstrating coexistence of the two types of the channels in
the same patch. Yi denotes the open current levels of channels with different conductances. (d) Single-channel
current–voltage relationship for two types of channels identified. Values are means ± SEM of at least four
experiments (reproduced from ref. 24 with permission from Elsevier)
4 Notes
FBS, DMEM media with FBS) and got a lower yield of the
glomeruli, and the preparation was very difficult to patch; the
glomeruli seemed to be too soft compared to original
preparation.
5. We suppose that it might be more convenient to cut the kidney
coronary in the ratio 1:3; this will allow removing the medulla
from one piece, only. The second part will consist of the cortex
only.
6. It is supposed that 2–3 mL of the solution A is added to the
fraction isolated after every step and then it is mixed with a fine
transfer pipette until homogeneity is reached.
7. The mesh size of the sieves listed in this chapter is adjusted for
the 8-week-old male Sprague–Dawley rats; if you are going to
isolate the glomeruli from mice or rats of other age, we would
recommend adjusting the mesh size accordingly. The probable
starting point for adjusting the mesh size for mice can be found
in the works of Akis et al. and Rops et al. (25, 26).
8. Steel sieves need to be soaked in the solution A. For large
sieves, a drop of the solution is to be spread on the top of the
sieve in the area you are going to use (up to 6–7 cm in diam-
eter), and the small sieve is to be fully soaked in the solution A.
The flow of the homogenate can be facilitated with the spoon-
shaped end of the spatula. The homogenate that stayed on the
reverse side of the sieve and did not get into the Petri dish can
be collected from the reverse side of the sieve with a razor
blade. Homogenate pushed through a new sieve always requires
being collected into a new Petri dish.
9. We would recommend being very careful with the last sieve as
the pure fraction of the glomeruli rests of the top of it and it is
likely to lose them at this step, so it would be reasonable to
seep the solution onto it slowly, drop by drop.
10. After the glomeruli fraction settles down to the bottom of the
tube, we recommend removing most of the supernatant and
leaving about 800 μL of the solution in the tube.
11. Takemoto et al. (27) suggested that the glomeruli of the mice
can be efficiently isolated with the Dynabeads. In brief, the kid-
ney is perfused with the solution containing magnetic Dynabeads,
then minced, digested with collagenase, pushed through the
sieve, and then the Dynabeads-containing glomeruli are sepa-
rated with a magnetic particle concentrator. However, this
approach seems to be more time-consuming and disturbs the
native setting of the glomerulus with the Dynabeads.
12. The fraction that is isolated with this preparation is highly pure
and consists mostly of the decapsulated glomeruli; however, up
to 10% of the glomeruli in the solution might still have a capsule.
TRPC Channels in Podocytes 367
Acknowledgments
References
1. Patrakka J, Tryggvason K (2009) New insights 9. Moller CC, Wei C, Altintas MM, Li J, Greka
into the role of podocytes in proteinuria. Nat A, Ohse T, Pippin JW, Rastaldi MP, Wawersik
Rev Nephrol 5:463–468 S, Schiavi S, Henger A, Kretzler M, Shankland
2. Pavenstadt H, Bek M (2002) Podocyte elec- SJ, Reiser J (2007) Induction of TRPC6 chan-
trophysiology, in vivo and in vitro. Microsc nel in acquired forms of proteinuric kidney dis-
Res Tech 57:224–227 ease. J Am Soc Nephrol 18:29–36
3. Abramowitz J, Birnbaumer L (2009) Physiology 10. Dietrich A, Chubanov V, Gudermann T (2010)
and pathophysiology of canonical transient recep- Renal TRPathies. J Am Soc Nephrol
tor potential channels. FASEB J 23:297–328 21:736–744
4. Kiselyov K, Patterson RL (2009) The integra- 11. Reiser J, Polu KR, Moller CC, Kenlan P, Altintas
tive function of TRPC channels. Front Biosci MM, Wei C, Faul C, Herbert S, Villegas I, vila-
14:45–58 Casado C, McGee M, Sugimoto H, Brown D,
5. Watanabe H, Murakami M, Ohba T, Ono K, Kalluri R, Mundel P, Smith PL, Clapham DE,
Ito H (2009) The pathological role of tran- Pollak MR (2005) TRPC6 is a glomerular slit
sient receptor potential channels in heart dis- diaphragm-associated channel required for nor-
ease. Circ J 73:419–427 mal renal function. Nat Genet 37:739–744
6. Woudenberg-Vrenken TE, Bindels RJ, 12. Winn MP, Conlon PJ, Lynn KL, Farrington
Hoenderop JG (2009) The role of transient MK, Creazzo T, Hawkins AF, Daskalakis N,
receptor potential channels in kidney disease. Kwan SY, Ebersviller S, Burchette JL, Pericak-
Nat Rev Nephrol 5:441–449 Vance MA, Howell DN, Vance JM, Rosenberg
7. Kim JY, Saffen D (2005) Activation of M1 PB (2005) A mutation in the TRPC6 cation
muscarinic acetylcholine receptors stimulates channel causes familial focal segmental glom-
the formation of a multiprotein complex cen- erulosclerosis. Science 308:1801–1804
tered on TRPC6 channels. J Biol Chem 13. Heeringa SF, Moller CC, Du J, Yue L, Hinkes
280:32035–32047 B, Chernin G, Vlangos CN, Hoyer PF, Reiser
8. Moller CC, Flesche J, Reiser J (2009) J, Hildebrandt F (2009) A novel TRPC6
Sensitizing the Slit Diaphragm with TRPC6 mutation that causes childhood FSGS. PLoS
ion channels. J Am Soc Nephrol 20:950–953 One 4:e7771
TRPC Channels in Podocytes 369
14. Winn MP, Daskalakis N, Spurney RF, 23. Savin VJ, Sharma R, Lovell HB, Welling DJ
Middleton JP (2006) Unexpected role of (1992) Measurement of albumin reflection
TRPC6 channel in familial nephrotic syn- coefficient with isolated rat glomeruli. J Am
drome: does it have clinical implications? J Am Soc Nephrol 3:1260–1269
Soc Nephrol 17:378–387 24. Ilatovskaya DV, Levchenko V, Ryan RP,
15. Eckel J, Lavin PJ, Finch EA, Mukerji N, Burch Cowley AW Jr, Staruschenko A (2011)
J, Gbadegesin R, Wu G, Bowling B, Byrd A, NSAIDs acutely inhibit TRPC channels in
Hall G, Sparks M, Zhang ZS, Homstad A, freshly isolated rat glomeruli. Biochem Biophys
Barisoni L, Birbaumer L, Rosenberg P, Winn Res Commun 408:242–247
MP (2011) TRPC6 enhances angiotensin 25. Akis N, Madaio MP (2004) Isolation, culture,
II-induced albuminuria. J Am Soc Nephrol and characterization of endothelial cells from
22:526–535 mouse glomeruli. Kidney Int 65:2223–2227
16. Chen S, He FF, Wang H, Fang Z, Shao N, 26. Rops AL, van der Vlag J, Jacobs CW, Dijkman
Tian XJ, Liu JS, Zhu ZH, Wang YM, Wang S, HB, Lensen JF, Wijnhoven TJ, van den Heuvel
Huang K, Zhang C (2011) Calcium entry via LP, van Kuppevelt TH, Berden JH (2004)
TRPC6 mediates albumin overload-induced Isolation and characterization of conditionally
endoplasmic reticulum stress and apoptosis in immortalized mouse glomerular endothelial
podocytes. Cell Calcium 50:523–529 cell lines. Kidney Int 66:2193–2201
17. Krall P, Canales CP, Kairath P, Carmona-Mora 27. Takemoto M, Asker N, Gerhardt H, Lundkvist
P, Molina J, Carpio JD, Ruiz P, Mezzano SA, A, Johansson BR, Saito Y, Betsholtz C (2002)
Li J, Wei C, Reiser J, Young JI, Walz K (2010) A new method for large scale isolation of kid-
Podocyte-specific overexpression of wild type ney glomeruli from mice. Am J Pathol
or mutant trpc6 in mice is sufficient to cause 161:799–805
glomerular disease. PLoS One 5:e12859 28. Sours S, Du J, Chu S, Ding M, Zhou XJ, Ma
18. Schlondorff J, del Camino D, Carrasquillo R, R (2006) Expression of canonical transient
Lacey V, Pollak MR (2009) TRPC6 mutations receptor potential (TRPC) proteins in human
associated with focal segmental glomeruloscle- glomerular mesangial cells. Am J Physiol Renal
rosis cause constitutive activation of NFAT- Physiol 290:F1507–F1515
dependent transcription. Am J Physiol Cell 29. Merritt JE, Armstrong WP, Benham CD,
Physiol 296:C558–C569 Hallam TJ, Jacob R, Jaxa-Chamiec A, Leigh
19. Kanda S, Harita Y, Shibagaki Y, Sekine T, BK, McCarthy SA, Moores KE, Rink TJ
Igarashi T, Inoue T, Hattori S (2011) Tyrosine (1990) SK&F 96365, a novel inhibitor of
phosphorylation-dependent activation of receptor-mediated calcium entry. Biochem J
TRPC6 regulated by PLC-gamma1 and neph- 271:515–522
rin: effect of mutations associated with focal 30. Dryer SE, Reiser J (2010) TRPC6 channels
segmental glomerulosclerosis. Mol Biol Cell and their binding partners in podocytes: role
22:1824–1835 in glomerular filtration and pathophysiology.
20. Gloy J, Henger A, Fischer KG, Nitschke R, Am J Physiol Renal Physiol 299:F689–F701
Mundel P, Bleich M, Schollmeyer P, Greger R, 31. Goel M, Sinkins WG, Zuo CD, Estacion M,
Pavenstadt H (1997) Angiotensin II depolar- Schilling WP (2006) Identification and local-
izes podocytes in the intact glomerulus of the ization of TRPC channels in the rat kidney.
Rat. J Clin Invest 99:2772–2781 Am J Physiol Renal Physiol
21. Dahly-Vernon AJ, Sharma M, McCarthy ET, 290:F1241–F1252
Savin VJ, Ledbetter SR, Roman RJ (2005) 32. Greka A, Mundel P (2011) Balancing Calcium
Transforming growth factor-beta, 20-HETE Signals through TRPC5 and TRPC6 in
interaction, and glomerular injury in Dahl salt- Podocytes. J Am Soc Nephrol 22:1969–80
sensitive rats. Hypertension 45:643–648 33. Tian D, Jacobo SM, Billing D, Rozkalne A,
22. Sharma R, Khanna A, Sharma M, Savin VJ Gage SD, Anagnostou T, Pavenstadt H, Hsu
(2000) Transforming growth factor-beta1 HH, Schlondorff J, Ramos A, Greka A (2010)
increases albumin permeability of isolated rat Antagonistic regulation of actin dynamics and
glomeruli via hydroxyl radicals. Kidney Int cell motility by TRPC5 and TRPC6 channels.
58:131–136 Sci Signal 3:ra77
Chapter 29
Abstract
Transient receptor potential (TRP) channels are expressed in almost every segment of renal nephron from
the glomerulus to the inner medullary collecting duct. Serving as a route for Ca2+ entry from the intratubu-
lar space into cells in response to external cues, TRP channels modulate water–electrolyte transport, thus
determining functional properties of the renal tubule. In this chapter, we discuss technical aspects of using
Ca2+ imaging to monitor activity of TRP channels in situ, namely, in the freshly isolated distal nephrons,
with a special emphasis on the mechanosensitive TRPV4 channel and its role in tubular flow sensing.
Key words Collecting duct, Connecting tubule, Fura-2, Mechanosensitivity, Shear stress
1 Introduction
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_29, © Springer Science+Business Media, LLC 2013
371
372 Mykola Mamenko et al.
cellular responses (14, 18). However, ICs, which lack the primary
cilia, respond to flow changes with comparable increases in [Ca2+]i
as PCs which possess primary cilia (11). Furthermore, mechanosen-
sitive Ca2+ responses are abolished upon Ca2+ removal from extra-
cellular media (14, 19). This suggests that mechanotransduction in
the distal nephron involves direct Ca2+ entry via Ca2+-permeable
channels/conduits at the apical/luminal plasma membrane.
Transient receptor potential (TRP) channels belong to one of
the most versatile superfamily of ion channels. TRP channels are
known to participate in cellular adaptations to a variety of environ-
mental stimuli, including thermo-sensation, chemo-sensation, and
mechanical forces (reviewed in ref. 20). TRPs are Ca2+-permeable
channels, but can also conduct other physiologically relevant cat-
ions (Na+, K+, Mg2+) with various degrees of selectivity (20).
Activation of these channels drives Ca2+ entry from the extracellu-
lar media, thus, likely contributing to the elevation of [Ca2+]i in
response to environmental changes (20). Several TRP channels,
including TRPC3, TRPC6, TRPV4-6, and TRPP2, can be detected
with immunohistochemistry in the native distal nephron cells and
distal nephron-derived cultures (14, 21–23). Among these chan-
nels, TRPV4 has routinely been shown to be activated by mechani-
cal stimuli in many cell types (23–27). Indeed, recent experimental
evidence points to a pivotal role of TRPV4 and likely TRPV4–
TRPP2 heteromers in responses to elevations in tubular flow in
distal nephron cells (19, 28).
Conventional patch clamp is the most direct experimental
methodology to assess functional status of ion channels at the
plasma membrane (29). This approach, though, has serious technical
difficulties when applied to study mechanosensitive TRP channels
in native epithelia, including that of the distal nephron. Electrical
coupling between cells within epithelial monolayer and/or high
constitutive background currents preclude obtaining high-resistant
(~GW) seals in both “whole-cell” and “perforated-patch”
configurations and thus, accurate voltage clamping. Furthermore,
basal activity of mechanosensitive TRP channels is generally low.
Direct mechanical stimulations to enhance channel activity are also
difficult to perform. This makes single-channel recordings of
mechanosensitive channels in isolated nephrons in “cell-attached”
configuration an inefficient and daunting task. In contrast, Fura-2-
based ratiometric imaging provides a reliable way to probe TRP
channels activity by monitoring changes of [Ca2+]i in individual
cells as a reflection of Ca2+ entry through the channels.
Since developed by M. Burg and colleagues (30, 31), the tech-
nique to perfuse isolated segments of renal tubule has been instru-
mental to study regulation of transport of water and solutes as well
as [Ca2+]i dynamics by pharmacological and mechanical stimuli. This
technique allows perfusing the tubular lumen at physiologically
relevant rates presented in the intact nephron and distinguishing
Ca2+ Imaging in Distal Nephrons 373
Fig. 1 Ca2+ imaging in freshly isolated split-opened distal nephron. Representative micrographs of the same
split-opened distal nephron taken with bright-field illumination (a), excitation at 380 nm after loading with
Fura-2 (b), the combined illumination with visible light and 380 nm (c), and after fixation and staining of the
distal nephron with anti-AQP2 antibodies (d). Nuclear DAPI staining is also shown (pseudocolor blue). Examples
of AQP2-positive principal cells (PC) and AQP2-negative intercalated cells (IC) are pointed with arrows
2 Materials
2.1 Rodent Kidney 1. A rat or a mouse (6–8 weeks of age) of wild-type (such as S/D
Extraction and C57BL/6, respectively) or a genetically modified strain of
interest.
2. CO2 gas tank and a hermetic chamber for sacrifice.
3. Dissecting scissors (Walter Stern, Port Washington, NY, USA).
4. Specimen forceps (Walter Stern, Port Washington, NY, USA).
5. Gloves (Microflex Tranquility powder-free nitrile gloves).
6. Ice-cold balanced salt solution (BSS): 150 mM NaCl, 5 mM
KCl, 2 mM MgCl2, 1 mM CaCl2, 10 mM Hepes, 5 mM
Glucose; pH = 7.4.
7. 60 × 15 mm plastic tissue culture dishes (Sarstedt, Newton,
NC, USA).
2.2 Isolation and 1. Nikon SMZ645 stereomicroscope (or equivalent from other
Split-Opening of Distal vendors).
Nephrons 2. Dumont SS fine forceps (Fine Science Tools, Foster City, CA,
USA).
3. 60 × 15 mm plastic tissue culture dishes (Sarstedt, Newton,
NC, USA).
4. Balanced salt solution (BSS, see Subheading 2.1, item 6).
5. 5 × 5 mm coverslips, cut from square 22 × 22 mm, No. 1.5
cover glasses (Thermo Fisher Scientific, Pittsburgh, PA, USA).
6. 0.01% solution of poly-L-lysine (Sigma-Aldrich Corp., St.
Louis, MO, USA).
7. Single-edge razor blades (American Line 66–0089, available
from numerous vendors).
8. Freshly isolated murine kidney.
9. Inverted microscope (Nikon Eclipse Ti-U microscope or
analogue).
10. Two micromanipulators (Sutter Instrument Company, Novato,
CA, USA; MP-285 motorized micromanipulators or analogue).
11. Glass borosilicate pipettes (World Precision Instruments,
Sarasota, FL, USA).
2.4 Immunostaining 1. 12-well microtiter plate (BD Falcon, Franklin Lakes, NJ, USA)
of Distal Nephron Cells or 35 × 10 mm diameter plastic tissue culture dishes (Sarstedt,
and Visualization of Newton, NC, USA).
the Tubules with 2. Phosphate buffer saline (PBS), pH = 7.4 (made of 10× concen-
Immunofluorescence trate, Sigma-Aldrich Corp., St. Louis, MO, USA).
3. 4% paraformaldehyde (Sigma-Aldrich Corp., St. Louis, MO,
USA) solution in PBS (pH = 7.4).
4. 0.1% Triton X-100 (Sigma-Aldrich Corp., St. Louis, MO,
USA) solution in PBS (pH = 7.4).
5. 0.05% Tween 20 in PBS (PBS-Tween buffer, also available as a
dry powder from Sigma-Aldrich, USA).
6. Normal goat serum (NGS, Jackson Immunoresearch, West
Grove, PA, USA).
7. Anti-AQP2 antibodies labeled with ATTO-550 fluorescent
dye (Alomone labs, Jerusalem, Israel).
8. 300 nM solution of 4¢,6-diamidino-2-phenylindole (DAPI,
Sigma-Aldrich Corp., St. Louis, MO, USA) in PBS.
9. Mounting medium (Thermo Scientific, Pittsburgh, PA, USA).
376 Mykola Mamenko et al.
3 Methods
3.2 Monitoring TRP 1. The major principle of monitoring [Ca2+]i in individual cells is
Channels Function based on the phenomenon that fluorescent excitation spec-
Using [Ca2+]i Imaging trum of Fura-2 shifts towards shorter wavelengths upon bind-
in Distal Nephron Cells ing Ca2+. Typically, Fura-2 emission is collected at 510 nm in
response to repetitive brief excitation at 340 and 380 nm after
certain periods of time (see Note 4). The ratio of fluorescent
intensities R = F340/F380 represents an index of relative changes
378 Mykola Mamenko et al.
Fig. 2 TRPV4 activity determines flow-mediated [Ca2+]i responses in both principal and intercalated cells.
Representative time courses of changes in [Ca2+]i in PC and IC (similar to that show in Fig. 1) in response to
application of the TRPV4 activator GSK101679A in C57BL/6 wild-type (a) and TRPV4−/− (b) mice. Changes in
[Ca2+]i in response to an abrupt 10× elevation in perfusion rate from 1.5 to 15 mL/min in PC and IC of wild-type
(c) and TRPV4−/− (d) mice. Respective scale bars for a, b and c, d are also shown
3.3 Using Parallel 1. Shear stress is the force per unit area that is generated on the
Plate Chamber to cell surface due to fluid flow over the surface. In the kidney the
Assess Shear Stress tubular fluid has a low viscosity, relative to vascular vessels, due
to protein-free tubular fluid. Nonetheless, because of high
tubular flow rates, the shear stresses in the distal nephron can
still be very high with typical values ranging from 0.3 to
25 dyn/cm2, although even higher values are likely in various
pathophysiological states (23, 34). Further, other mechanical
stresses may also be at play for a given experimental condition
380 Mykola Mamenko et al.
4 Notes
References
1. Eaton DC, Malik B, Saxena NC, Al-Khalili OK, 13. Mamenko M, Zaika O, Jin M, O’Neil RG,
Yue G (2001) Mechanisms of aldosterone’s Pochynyuk O (2011) Purinergic Activation of
action on epithelial Na+ transport. J Membr Ca-Permeable TRPV4 Channels Is Essential for
Biol 184:313–319 Mechano-Sensitivity in the Aldosterone-
2. Hummler E (2003) Epithelial sodium channel, Sensitive Distal Nephron. PLoS One 6:e22824
salt intake, and hypertension. Curr Hypertens 14. Nauli SM, Alenghat FJ, Luo Y, Williams E,
Rep 5:11–18 Vassilev P, Li X, Elia AE, Lu W, Brown EM,
3. Stockand JD (2010) Vasopressin regulation of Quinn SJ, Ingber DE, Zhou J (2003)
renal sodium excretion. Kidney Int 78:849–856 Polycystins 1 and 2 mediate mechanosensation
4. Inoue T, Nonoguchi H, Tomita K (2001) in the primary cilium of kidney cells. Nat Genet
Physiological effects of vasopressin and atrial 33:129–137
natriuretic peptide in the collecting duct. 15. Praetorius HA, Leipziger J (2009) Released
Cardiovasc Res 51:470–480 nucleotides amplify the cilium-dependent,
5. Satlin LM, Sheng S, Woda CB, Kleyman TR flow-induced [Ca2+]i response in MDCK cells.
(2001) Epithelial Na+ channels are regulated by Acta Physiol (Oxf) 197:241–251
flow. Am J Physiol 280:F1010–F1018 16. Praetorius HA, Leipziger J (2010) Intrarenal
6. Satlin LM, Carattino MD, Liu W, Kleyman R purinergic signaling in the control of renal tubu-
(2006) Regulation of cation transport in the lar transport. Annu Rev Physiol 72:377–393
distal nephron by mechanical forces. Am J 17. Woda CB, Leite M Jr, Rohatgi R, Satlin LM
Physiol 291:F923–F931 (2002) Effects of luminal flow and nucleotides
7. Woda CB, Bragin A, Kleyman TR, Satlin LM on [Ca2+]i in rabbit cortical collecting duct. Am
(2001) Flow-dependent K+ secretion in the J Physiol 283:F437–F446
cortical collecting duct is mediated by a maxi-K 18. Liu W, Murcia NS, Duan Y, Weinbaum S,
channel. Am J Physiol 280:F786–F793 Yoder BK, Schwiebert E, Satlin LM (2005)
8. Geyti CS, Odgaard E, Overgaard MT, Jensen Mechanoregulation of intracellular Ca2+ con-
ME, Leipziger J, Praetorius HA (2008) Slow centration is attenuated in collecting duct of
spontaneous [Ca2+]i oscillations reflect nucle- monocilium-impaired orpk mice. Am J Physiol
otide release from renal epithelia. Pflugers Arch 289:F978–F988
455:1105–1117 19. Berrout J, Jin M, Mamenko M, Zaika O,
9. Hovater MB, Olteanu D, Welty EA, Schwiebert Pochynyuk O, O’Neil RG (2012) Function of
EM (2008) Purinergic signaling in the lumen of TRPV4 as a mechanical transducer in flow-
a normal nephron and in remodeled PKD encap- sensitive segments of the renal collecting duct
sulated cysts. Purinergic Signal 4:109–124 system. J Biol Chem 287:8782–8791
10. Leipziger J (2003) Control of epithelial trans- 20. Song MY, Yuan JX (2010) Introduction to
port via luminal P2 receptors. Am J Physiol TRP channels: structure, function, and regula-
284:F419–F432 tion. Adv Exp Med Biol 661:99–108
11. Liu W, Xu S, Woda C, Kim P, Weinbaum S, 21. Goel M, Sinkins WG, Zuo CD, Hopfer U,
Satlin LM (2003) Effect of flow and stretch on Schilling WP (2007) Vasopressin-induced
the [Ca2+]i response of principal and interca- membrane trafficking of TRPC3 and AQP2
lated cells in cortical collecting duct. Am J channels in cells of the rat renal collecting duct.
Physiol 285:F998–F1012 Am J Physiol 293:F1476–F1488
12. Liu W, Morimoto T, Woda C, Kleyman TR, 22. Goel M, Sinkins WG, Zuo CD, Estacion M,
Satlin LM (2007) Ca2+ dependence of flow- Schilling WP (2006) Identification and local-
stimulated K secretion in the mammalian cortical ization of TRPC channels in the rat kidney. Am
collecting duct. Am J Physiol 293:F227–F235 J Physiol 290:F1241–F1252
384 Mykola Mamenko et al.
23. Wu L, Gao X, Brown RC, Heller S, O’Neil RG 30. Burg MB, Grantham J, Abramow M, Orloff J,
(2007) Dual role of the TRPV4 channel as a Schafer JA (1997) Preparation and study of
sensor of flow and osmolality in renal epithelial fragments of single rabbit nephrons. J Am Soc
cells. Am J Physiol 293:F1699–F1713 Nephrol 8:675–683
24. Strotmann R, Harteneck C, Nunnenmacher K, 31. Burg M, Grantham J, Abramow M, Orloff J
Schultz G, Plant TD (2000) OTRPC4, a nonse- (1966) Preparation and study of fragments of
lective cation channel that confers sensitivity to single rabbit nephrons. Am J Physiol 210:
extracellular osmolarity. Nat Cell Biol 2:695–702 1293–1298
25. Liedtke W, Choe Y, Marti-Renom MA, Bell AM, 32. Weinbaum S, Duan Y, Satlin LM, Wang T,
Denis CS, Sali A, Hudspeth AJ, Friedman JM, Weinstein AM (2010) Mechanotransduction
Heller S (2000) Vanilloid receptor-related osmot- in the renal tubule. Am J Physiol 299:
ically activated channel (VR-OAC), a candidate F1220–F1236
vertebrate osmoreceptor. Cell 103:525–535 33. Grynkiewicz G, Poenie M, Tsien RY (1985) A
26. Hartmannsgruber V, Heyken WT, Kacik M, new generation of Ca2+ indicators with greatly
Kaistha A, Grgic I, Harteneck C, Liedtke W, improved fluorescence properties. J Biol Chem
Hoyer J, Kohler R (2007) Arterial response to 260:3440–3450
shear stress critically depends on endothelial 34. Jin M, Berrout J, O’Neil RG (2011)
TRPV4 expression. PLoS One 2:e827 Regulation of TRP channels by osmomechani-
27. Gao X, Wu L, O’Neil RG (2003) Temperature- cal stresses. In: Zhu MX (ed) Methods in signal
modulated diversity of TRPV4 channel gating: transduction series. CRC Press, New York, NY,
activation by physical stresses and phorbol ester pp 353–373
derivatives through protein kinase C-dependent 35. Resnick N, Yahav H, Shay-Salit A, Shushy M,
and -independent pathways. J Biol Chem Schubert S, Zilberman LC, Wofovitz E (2003)
278:27129–27137 Fluid shear stress and the vascular endothelium:
28. Du J, Wong WY, Sun L, Huang Y, Yao X for better and for worse. Prog Biophys Mol
(2012) Protein Kinase G Inhibits Flow-Induced Biol 81:177–199
Ca2+ Entry into Collecting Duct Cells. J Am 36. White CR, Frangos JA (2007) The shear stress
Soc Nephrol 23:1172–1180 of it all: the cell membrane and mechanochemi-
29. Sakmann B, Neher E (2009) Single-channel cal transduction. Philos Trans R Soc Lond B
recording. Springer, New York, NY Biol Sci 362:1459–1467
Chapter 30
Abstract
Electrophysiological studies provide essential clues about the regulation and physiological function of ion
channel proteins. Probing ion channel activity in vivo, though, often is challenging. This can limit the
usefulness of such model organisms as Drosophila for electrophysiological studies. This is unfortunate
because these genetically tractable organisms represent powerful research tools that facilitate elaboration
of complex questions of physiology. Here, we describe a recently developed method for recording ion
channel activity in Drosophila sensory neurons. This approach is based on patch-clamping primary neuron
cultures from Drosophila embryos. Such cultures allow the study of ion channels in different genetic back-
grounds. In addition to describing how to prepare a primary neuronal cell culture from Drosophila embryos,
we discuss, as an example of utility, analysis of Na+ currents in cultured class IV multidendritic (md) sen-
sory neurons with the patch clamp technique. Excitability of md sensory neurons, manifested as action
potential firing, is revealed with whole-cell current-clamping. Voltage-clamping class IV md neurons
revealed the activity of the voltage-gated Na+ channel, paralytic. Moreover, challenging class IV md neu-
rons with acidic pH activates acid-sensing inward Na+ currents. Genetic manipulation of Drosophila com-
bined with this electrophysiological readout of activity identifies pickpocket1 (Ppk1), a member of the
Deg/ENaC channel family, as responsible for conducting an acid-sensing Na+ current in class IV md sen-
sory neurons.
Key words Patch clamping, Primary cell culture, Ion channel, Drosophila sensory neurons
1 Introduction
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_30, © Springer Science+Business Media, LLC 2013
385
386 Volodymyr Kucher et al.
2 Materials
2.2 Electro- 1. Inverted microscope (we use Eclipse Ti; Nikon, Melville, NY)
physiological Analysis fitted with epifluorescence and excitation and emission filters
of Class IV md for GFP (FITC/RSGFP LP emission; #21012, Chroma Tech.
Sensory Neurons Corp., Rockingham, VT) as needed to identify the cell type of
interest.
2. Patch-clamp amplifier (we use Axopatch 200B; Molecular
Devices, Sunnyvale, CA; see Note 2).
3. Digitizer, i.e., Digidata 1322A A/D board (Molecular Devices)
interfaced with a PC running an appropriate acquisition and
analysis software (we use pClamp 9.2 software suite; Molecular
Devices).
4. Micromanipulator (i.e., MP-285; Sutter Instr. Co., Novato,
CA).
5. Fast exchange recording chamber (i.e., Model RC-22, Warner
Instr., Hamden, CT).
6. Perfusion system (we use Valve Bank II pinch valve perfusion
system from AutoMate Scientific Inc., San Francisco, CA).
7. Micro-vibration isolation table with Faraday cage.
8. Micropipette puller (we use Model P-97 flaming/brown puller,
Sutter Instr. Co.)
388 Volodymyr Kucher et al.
3 Methods
3.1 Preparation 1. Fly stocks are maintained on standard food at 25°C and a 12 h
of a Primary Neuron light/dark cycle. Preparation of the primary neuron culture
Culture from from embryos is performed as previously described (2) with
Drosophila Embryos some modifications.
Patch-Clamping Drosophila Sensory Neurons 389
Fig. 2 Morphological development of primary neurons from Drosophila embryos plated in cell culture. Bright
field images of primary cells from Drosophila embryos (a) 1 h after plating and (b) 72 h after growing in culture.
Higher magnification bright field (c) and fluorescence (d) images of cultured GFP-expressing class IV md sen-
sory neurons. Scale bars are 50 μm (a, b) and 3 μm (c, d)
These cells are round and mostly dissociated. Figure 2b–d shows
the same cells after 72 h in culture. By this time, the cells have dif-
ferentiated into neurons containing branching neuronal extensions
(big arrow) and forming neuronal clusters (small arrow).
50 pA
5 ms
Amiloride
pH4.5
100 pA
Is 30 sec It
4 Notes
Amiloride Wash
It
100 pA
100 pA
100 pA
It
It It
500 pA
100 sec
Fig. 6 Identification of the current conducted by Ppk1 in Drosophila class IV md sensory neurons. Representative
acid-evoked macroscopic Na+ currents in voltage-clamped (holding potential −60 mV) class IV md neurons
from wild type (left ), ppk1−/− null mutants (second from the left ), ppk1−/− mutants rescued with expression of
the wild-type ppk1 transgene in md neurons (third from the left ), and flies with targeted expression of the
gain-of-function ppk1deg transgene in class IV md neurons (right ). In these experiments, neurons were treated
with 1 mM amiloride prior to providing the activating acid pulse with blocker and low pH subsequently washed
simultaneously to evoke It
References
1. Seecof RL, Unanue RL (1968) Differentiation 2. Sicaeros B, O’Dowd, DK (2007) Preparation
of embryonic Drosophila cells in vitro. Exp of neuronal cultures from midgastrula stage
Cell Res 50:654–660 Drosophila embryos. J Vis Exp 5:226
Patch-Clamping Drosophila Sensory Neurons 397
3. Cross D, Sang J (1978) Cell culture of indi- Welsh MJ, Johnson WA (2003) Enhanced
vidual Drosophila embryos: I. Development of Locomotion Caused by Loss of the Drosophila
wild-type cultures. J Embryol Exp Morphol DEG/ENaC Protein Pickpocket1. Curr Biol
45:161–172 13:1557–1563
4. Shields G, Sang JH (1977) Improved medium 11. Zhong L, Hwang RY, Tracey WD (2010)
for culture of Drosophila embryonic cells. Pickpocket Is a DEG/ENaC Protein Required
Drosoph Inf Serv 52:161 for Mechanical Nociception in Drosophila
5. Hodges DD, Lee D, Preston CF, Boswell K, Larvae. Curr Biol 20:1–6
Hall LM, O’Dowd DK (2002) tipE regulates 12. Kim SE, Coste B, Chadha A, Cook B,
Na+-dependent repetitive firing in Drosophila Patapoutian A (2012) The role of Drosophila
neurons. Mol Cell Neurosci 19:402–416 Piezo in mechanical nociception. Nature
6. O’Dowd DK (1995) Voltage-gated currents 483:209–212
and firing properties of embryonic Drosophila 13. Hong K, Driscoll M (1994) A transmembrane
neurons grown in a chemically defined domain of the putative channel subunit
medium. J Neurobiol 27:113–126 MEC-4 influences mechanotransduction and
7. Matthews BJ, Kim ME, Flanagan JJ, Hattori neurodegeneration in C. elegans. Nature
D, Clemens JC, Zipursky SL, Grueber WB 367:470–473
(2007) Dendrite self-avoidance is controlled 14. Waldmann R, Champigny G, Voilley N,
by Dscam. Cell 129:593–604 Lauritzen I, Lazdunski M (1996) The mam-
8. Grueber WB, Ye B, Yang CH, Younger S, malian degenerin MDEG, an amiloride-sensi-
Borden K, Jan LY, Jan Y-N (2007) Projections tive cation channel activated by mutations
of Drosophila multidendritic neurons in the causing neurodegeneration in Caenorhabditis
central nervous system: links with peripheral elegans. J Biol Chem 271:10433–10436
dendrite morphology. Development 15. Hamill OP, Marty A, Neher E, Sakmann B,
134:55–64 Sigworth FJ (1981) Improved patch-clamp
9. Adams CM, Anderson MG, Motto DG, Price techniques for high-resolution current record-
MP, Johnson WA, Welsh MJ (1998) Ripped ing from cells and cell-free membrane patches.
pocket and pickpocket, novel Drosophila Pflugers Arch 391:85–100
DEG/ENaC subunits expressed in early devel- 16. Ashburner M, Golic KG, Hawley RS (2005)
opment and in mechanosensory neurons. J Drosophila. A laboratory handbook, Secondth
Cell Biol 140:143–152 edn. Cold Spring Harbor, New York, NY
10. Ainsley JA, Pettus JM, Bosenko D, Gerstein 17. Sakmann B, Neher E (1983) Single-channel
CE, Zinkevich N, Anderson MG, Adams CM, recording. Plenum Press, New York, NY
Part VI
Abstract
Recent discovery of the light-activated ion channel, channelrhodopsin (ChR), has provided researchers a
powerful and convenient tool to manipulate the membrane potential of specific cells with light. With
genetic targeting of these channels and illumination of light to a specific location, the experimenter can
selectively activate the voltage-gated ion channels (VGICs) of ChR-expressing cells, initiating electrical
signaling in temporally and spatially precise manners. In neuroscience research, this can be used to study
electrical signal processing within one neuron at the cellular level, or the synaptic connectivity between
neurons at the circuitry level. To conduct experiments with ChRs, these exogenous channels need to be
introduced into the cells of interest, commonly through a viral approach. This chapter provides an over-
view of the design, production, and validation of recombinant adeno-associated virus (rAAV) for ChR
expression that can be used in vitro or in vivo to infect neurons. The virus produced can be used to con-
duct “optogenetic” experiments in behaving animals, in vitro preparations and cultured cells, and can be
used to study signal transduction and processing at a cellular or circuitry level.
1 Introduction
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_31, © Springer Science+Business Media, LLC 2013
401
402 John Y. Lin
Fig. 1 Schematic of the light-induced activation of voltage-gated ion channel (VGIC) with channelrhodopsin
(ChR) and examples of ChR expression in neurons. (a) Illumination of plasma membrane expressing ChR leads
to the depolarization of the membrane, and the activation of voltage-gated ion channel (VGIC). The opening and
closing of ChR is controlled by the photo-isomerization of all-trans retinal incorporated in the ChR protein.
(b) Examples of neurons infected by recombinant adeno-associated virus (rAAV) expressing ChR-Citrine (eYFP)
fusion protein as visualized with the fluorescence of the mCitrine. (c) Examples of light-induced depolarization
in cultured neurons expressing the ChR variant ChIEF. The left trace shows the cell in response to 750 ms light
pulse and the right trace showed the membrane response to two bursts of 10 Hz light stimulation
ChRs are evolved to provide algae with a simple vision, there are
many properties of these proteins that are not ideal for its use as a
research tool (7). However, many subsequent developments and
engineering of ChRs have improved kinetics, desensitization/inac-
tivation, and membrane trafficking of ChR, with the variant ChIEF
being thus far the best general purpose ChR variant with good
membrane trafficking, a low level of desensitization, and fast kinet-
ics (7–9). Two aspects that have yet to be improved are the low
conductance of ChRs (8, 10, 11), and having different variants
with selective ion permeability (2, 8). To compensate for these two
properties, a high expression level in the cells of interest is often
necessary to achieve significant depolarization.
Neuroscientists have utilized ChRs to selectively excite geneti-
cally defined neurons or neurons of a defined location. By express-
ing ChRs in specific neurons, the experimenters can trigger action
potentials in these neurons and artificially induce synaptic release
with light in in vitro and in vivo preparations (Fig. 1). The majority
of studies utilizing ChRs in rodents achieve targeted expression
with recombinant viral vectors. Expressing ChR utilizing a viral
vector is a quick, simple, and effective approach compared to alter-
natives such as generating transgenic animals and in utero elec-
troporation. Of the different recombinant viruses that have been
used with ChR, recombinant adeno-associated virus (rAAV) has
been the most successful and efficient viral vector for in vivo use.
Recombinant AAV is small in size and can be concentrated at a
high titer, which can lead to high numbers of infected cells in vivo.
At this moment, many core facilities and commercial services can
produce high-titer rAAV, although this is typically expensive (~US
$1,500/strain) and time-consuming (3–4 weeks). In this chapter,
the protocol utilized within our laboratory to generate high-titer
rAAV is described. The resulting virus is suitable for use in culture
and in vivo in rodents. The protocol typically takes 1–1.5 weeks to
complete and generate >200–300 mL of rAAV at titer >1012 genome
copy (GC)/milliliter. The simplified flow chart of rAAV produc-
tion is shown in Fig. 2. The main instruments required are cell
culture facilities approved for virus generation and ultracentrifuge
with suitable rotor, which are available in many research facilities
(see Note 1 regarding safety and precautions about handling
recombinant virus). Although there are many alternative methods
for some of the steps described here (transfection methods, cell
line used, virus purification), I have provided the methods that
have consistently generated high-titer rAAV in our laboratory in a
cost-effective manner.
Prior to generating rAAV for ChR expression, it is important to
design the appropriate viral vector for the desired experiment. In
the system described here, the maximum genetic insert that can be
placed between the standard AAV inverted terminal repeats (ITRs)
is ~4.7 kbp, which includes the genetic material for the promoter,
404 John Y. Lin
Fig. 2 The flow chart for the production and validation of rAAV for ChR expression.
This chart shows the steps and typical time requirement involved in the produc-
tion and validation of rAAV for ChR experiment. The molecular cloning of the viral
vector can be done in parallel to the preparation of cells for transfection
Fig. 3 Common AAV viral vector design for ChR expression. The top example shows the most frequently utilized
AAV vector design for direct ChR expression which contains a promoter, the transgene-encoding ChR-
fluorescent protein (FP) fusion protein, woodchuck hepatitis virus posttranscriptional regulatory element
(WPRE), and poly(A) tail between two AAV inverted terminal repeats (ITR). The examples below show alternative
designs including the omission of WPRE, utilization of intron to increase expression level, and utilization of a
2A-FP sequence for cytosolic FP expression. The bottom example shows the FLEX/DIO approach for targeted
ChR expression with a Cre-recombinase-expressing mouse strain where the ChR-FP transgene is inserted in
reverse (non-expressing) between two sets of lox sites between the promoter and WPRE. The presence of Cre-
recombinase would excise and invert the transgene between the lox sites, leading to expression of ChR-FP
and alternative viral vector designs for expressing ChR with rAAV.
The desired components can be assembled with standard molec-
ular cloning procedures into the AAV viral vectors containing the
AAV-ITR sites.
2 Materials
2.1 Production 1. Ad-helper plasmid (75 mg), AAV-helper plasmid (30 mg), and
of rAAV In Trans AAV-ChR plasmid (25.5 mg). Production of rAAV in trans
requires the transfection of three separate plasmids at 1:1:1
molar ratio. We recommend maxiprep-quality plasmid DNA.
AAV-helper plasmid contains the rep and cap genes and deter-
Optogenetic Excitation with Channelrhodopsin in Neurons 407
Table 1
Composition of the 15, 25, 40, and 58% iodixanol gradient solution
Phenol red
Optiprep (mL) 10× GB (mL) 5 M NaCl (mL) ddH2O (mL) (5 mg/mL) (mL) Total (mL)
15% 10 4 8 18 0 40
25% 11.67 2.8 0 14 70 28.47
40% 16 2.4 0 5.6 0 24
58% 24.17 0.8325 0 0 60 25
Fig. 4 The responses of ChIEF to different wavelengths of light. (a) The representative
responses of ChIEF to 570, 530, 470, and 410 nm light of the same photon flux
under voltage-clamp. (b) The maximal and steady-state (measured at 950–
1,000 ms after initiation of illumination) response spectra of ChIEF normalized to
its maximal response across the spectra. The optimal excitation wavelength is
between 440 and 490 nm (Figure modified with permission from Lin et al. (8))
3 Methods
3.2 Purification and 1. Build an iodixanol gradient in a Quick-Seal tube by adding 9.7 mL
Concentration of rAAV of 15% gradient solution to the bottom of the tube using a 10 mL
syringe syringe fitted with a long 18-guage metal cannula needle.
2. Add subsequent layers below the previous layer by extending
the syringe needle to the bottom of the tube and slowly inject-
ing the gradient solutions. The remaining compositions are
6.4 mL of 25% gradient solution, followed by 5.4 mL of 40%
gradient solution and then 5.4 mL of 58% gradient solution.
3. Add the virus-containing supernatant to the gradient by slowly
dripping the solution with an 18-guage needle fitted on a
syringe without the plunger onto the top layer of the gradient.
Minimize the disruption of the layers.
Optogenetic Excitation with Channelrhodopsin in Neurons 411
Fig. 5 Example of iodixanol gradients before and after centrifugation. (a) The iodixanol gradient preparation
showing the layers containing the 1× GB, virus solution, and 15, 25, 40, and 58% gradient solution before (A1)
and after (A2) centrifugation. (b) The placement of the needle for the extraction of virus solution after
centrifugation
4. Fill the remaining empty volume of the tube to the top using
1× GB as in step 3.
5. Weigh the tube and make a counter balance tube of identical
weight filled with the same layers (replacing the virus with
1 × GB); the differences should be <0.01 g.
6. Seal the tip of the tubes using a Tube Topper Sealer. Gently
squeeze the tube to ensure the tips are sealed and airtight. The
tube should appear as shown in Fig. 5.
7. Place the two tubes in the opposite spaces of a 70Ti Beckman
rotor. Lower the Quick-Seal Tube Spacer on top of the tube
prior to sealing the rotor lid. Centrifuge the tube in vacuum at
169,538 × g for 2 h 10 min at 18°C. The acceleration should
be set to “max” and deceleration to “9.”
8. Remove the tubes from the rotor and mount the tube contain-
ing the virus on a utility clamp (Fig. 5).
9. Insert an 18-gauge needle near the top of the tube as airflow
vent. Place a beaker directly underneath the tube to catch any
leakage (Fig. 5b).
10. Prepare a 3 mL syringe with an 18 gauge needle for extraction
of the virus. Insert the needle at the interface between the 40
and 58% gradient buffer layers with the bevel of the needle fac-
ing up (Fig. 5b). Slowly extract ~2 mL of solution, first with the
beveled needle opening facing upwards during the first half of
extraction and then facing downward for the rest (see Note 7).
11. Add the 1.5 mL of 1× GB to the 2 mL of virus solution and
load the solution into a 50 kDa MW Amicon centrifuge filter
cartridge. Spin at 1,500 × g for 15 min.
412 John Y. Lin
4 Notes
Acknowledgments
References
1. Nagel G, Ollig D, Fuhrmann M, Kateriya S, 5. Zemelman BV, Nesnas N, Lee GA, Miesenbock
Musti AM, Bamberg E, Hegemann P (2002) G (2003) Photochemical gating of heterolo-
Channelrhodopsin-1: a light-gated proton chan- gous ion channels: remote control over geneti-
nel in green algae. Science 296:2395–2398 cally designated populations of neurons. Proc
2. Nagel G, Szellas T, Huhn W, Kateriya S, Natl Acad Sci USA 100:1352–1357
Adeishvili N, Berthold P, Ollig D, Hegemann 6. Posch KC, Burns RD, Napoli JL (1992)
P, Bamberg E (2003) Channelrhodopsin-2, a Biosynthesis of all-trans-retinoic acid from reti-
directly light-gated cation-selective membrane nal. Recognition of retinal bound to cellular
channel. Proc Natl Acad Sci USA 100: retinol binding protein (type I) as substrate by
13940–13945 a purified cytosolic dehydrogenase. J Biol
3. Zhang F, Prigge M, Beyriere F, Tsunoda SP, Chem 267:19676–19682
Mattis J, Yizhar O, Hegemann P, Deisseroth K 7. Lin JY (2011) A user’s guide to channelrho-
(2008) Red-shifted optogenetic excitation: a dopsin variants: features, limitations and future
tool for fast neural control derived from Volvox developments. Exp Physiol 96:19–25
carteri. Nat Neurosci 11:631–633 8. Lin JY, Lin MZ, Steinbach P, Tsien RY (2009)
4. Szobota S, Gorostiza P, Del Bene F, Wyart C, Characterization of engineered channelrho-
Fortin DL, Kolstad KD, Tulyathan O, Volgraf dopsin variants with improved properties and
M, Numano R, Aaron HL, Scott EK, Kramer kinetics. Biophys J 96:1803–1814
RH, Flannery J, Baier H, Trauner D, Isacoff 9. Mattis J, Tye KM, Ferenczi EA, Ramakrishnan
EY (2007) Remote control of neuronal activity C, O’Shea DJ, Prakash R, Gunaydin LA, Hyun
with a light-gated glutamate receptor. Neuron M, Fenno LE, Gradinaru V, Yizhar O, Deisseroth
54:535–545 K (2012) Principles for applying optogenetic
Optogenetic Excitation with Channelrhodopsin in Neurons 415
tools derived from direct comparative analysis of 16. Dittgen T, Nimmerjahn A, Komai S, Licznerski
microbial opsins. Nat Methods 9:159–172 P, Waters J, Margrie TW, Helmchen F, Denk
10. Feldbauer K, Zimmermann D, Pintschovius V, W, Brecht M, Osten P (2004) Lentivirus-based
Spitz J, Bamann C, Bamberg E (2009) genetic manipulations of cortical neurons and
Channelrhodopsin-2 is a leaky proton pump. their optical and electrophysiological monitor-
Proc Natl Acad Sci USA 106:12317–12322 ing in vivo. Proc Natl Acad Sci USA 101:
11. Gunaydin LA, Yizhar O, Berndt A, Sohal VS, 18206–18211
Deisseroth K, Hegemann P (2010) Ultrafast 17. Beaton A, Palumbo P, Berns KI (1989)
optogenetic control. Nat Neurosci 13:387–392 Expression from the adeno-associated virus p5
12. Yizhar O, Fenno LE, Prigge M, Schneider F, and p19 promoters is negatively regulated in
Davidson TJ, O’Shea DJ, Sohal VS, Goshen I, trans by the rep protein. J Virol 63:
Finkelstein J, Paz JT, Stehfest K, Fudim R, 4450–4454
Ramakrishnan C, Huguenard JR, Hegemann 18. Nathanson JL, Yanagawa Y, Obata K, Callaway
P, Deisseroth K (2011) Neocortical excitation/ EM (2009) Preferential labeling of inhibitory
inhibition balance in information processing and excitatory cortical neurons by endogenous
and social dysfunction. Nature 477:171–178 tropism of adeno-associated virus and lentivirus
13. Atasoy D, Aponte Y, Su HH, Sternson SM vectors. Neuroscience 161:441–450
(2008) A FLEX switch targets 19. Loeb JE, Cordier WS, Harris ME, Weitzman
Channelrhodopsin-2 to multiple cell types for MD, Hope TJ (1999) Enhanced expression of
imaging and long-range circuit mapping. transgenes from adeno-associated virus vectors
J Neurosci 28:7025–7030 with the woodchuck hepatitis virus posttran-
14. Pelletier J, Sonenberg N (1988) Internal initia- scriptional regulatory element: implications for
tion of translation of eukaryotic mRNA directed gene therapy. Hum Gene Ther 10:2295–2305
by a sequence derived from poliovirus RNA. 20. Huang MT, Gorman CM (1990) Intervening
Nature 334:320–325 sequences increase efficiency of RNA 3¢ pro-
15. Osborn MJ, Panoskaltsis-Mortari A, McElmurry cessing and accumulation of cytoplasmic RNA.
RT, Bell SK, Vignali DA, Ryan MD, Wilber Nucleic Acids Res 18:937–947
AC, McIvor RS, Tolar J, Blazar BR (2005) A 21. Cardin JA, Carlen M, Meletis K, Knoblich U,
picornaviral 2A-like sequence-based tricistronic Zhang F, Deisseroth K, Tsai LH, Moore CI
vector allowing for high-level therapeutic gene (2009) Driving fast-spiking cells induces
expression coupled to a dual-reporter system. gamma rhythm and controls sensory responses.
Mol Ther 12:569–574 Nature 459:663–667
Chapter 32
Abstract
In the vibrant field of optogenetics, optics and genetic targeting are combined to commandeer cellular
functions, such as the neuronal action potential, by optically stimulating light-sensitive ion channels
expressed in the cell membrane. One broadly applicable manifestation of this approach are covalently
attached photochromic tethered ligands (PTLs) that allow activating ligand-gated ion channels with out-
standing spatial and temporal resolution. Here, we describe all steps towards the successful development
and application of PTL-gated ion channels in cell lines and primary cells. The basis for these experiments
forms a combination of molecular modeling, genetic engineering, cell culture, and electrophysiology. The
light-gated glutamate receptor (LiGluR), which consists of the PTL-functionalized GluK2 receptor, serves
as a model.
Key words Optogenetics, Optochemical genetics, Optical switch, Photochromic tethered ligand,
Azobenzene, LiGluR, Glutamate receptor
1 Introduction
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0_32, © Springer Science+Business Media, LLC 2013
417
418 Stephanie Szobota et al.
Fig. 1 Synthetic strategies for manipulating ligand-gated ion channels with light. (a) In the example of the iono-
tropic glutamate receptor GluK2, glutamate binding in the extracellular ligand binding domain triggers domain
closure and gate opening. (b) Gating after ligand uncaging. (c) Gating after reversible conversion of a photo-
chromic ligand to a binding-competent conformation. (d) Reversible light control by a PTL. (e) Chemical struc-
ture of MAG1, the prototypical maleimide-azobenzene-glutamate PTL, and 4-MG. (f, g) Crystal structure of the
ligand binding domain of GluK2 in complex with 2S, 4R-4-methylglutamate (4-MG). 4-MG, residues surround-
ing the binding site suited for PTL attachment, and exit tunnel are highlighted as described in the text
2 Materials
2.2 Molecular 1. Bacterial cell culture shaker in a 37°C room or 37°C incubator
Biology with shaker (~225 rpm).
2. LB medium: 1% bactotryptone, 0.5% yeast extract, 1% NaCl,
pH 7.0, autoclaved.
3. Bacteriological agar.
4. Ampicillin stock solution (100 mg/mL in water), stored at
−20°C in 500 mL aliquots.
420 Stephanie Szobota et al.
2.3 Cell Culture 1. Cell culture facility equipped with laminar flow hood, water
and Transfection bath (37°C), incubator (5% CO2), and cell counting chamber;
of HEK293 Cells HEK293 cells (American Tissue Culture Collection, LGC
Standards, Teddington, UK) cultured according to the pro-
vider’s recommendation in 25 cm2 tissue culture flasks.
2. 500 mL Dulbecco’s modified essential medium (DMEM) sup-
plemented with 5% fetal bovine serum (FBS), sterile filtered,
and stored at 4°C. Supplement half of the medium with 1%
penicillin/streptomycin solution, and use the supplemented
DMEM unless noted otherwise.
3. Dulbecco’s phosphate buffered saline (DPBS): 8 g NaCl,
200 mg KCl, 200 mg KH2PO4, 2.16 g Na2HPO4⋅7H2O, water
added to 1 L and sterile filtered.
4. Borate buffer: 1.55 g boric acid, 2.375 g Na+ borate, water
added to 500 mL, pH 8.5, sterile filtered, and stored at 4°C.
5. Opti-MEM I reduced serum medium (LifeTech, Vienna).
6. 0.25% Trypsin-EDTA solution (LifeTech).
7. Lipofectamine 2000 transfection reagent (LifeTech).
8. Poly-L-lysine (PLL) HBr (molecular weight 70,000–150,000).
Open PLL container in the laminar flow hood and add water
to a concentration of 10 mg/mL. Aliquot in 1.5 mL Eppendorf
tubes and store at −20°C.
9. Round cover glasses (18 mm diameter, Carolina Biological,
Burlington, NC, USA) (see Note 4).
10. Polystyrene 25 cm2 cell culture flask with filter lid (0.2 mm pore
size, sterile).
11. Polystyrene 12-well plate (sterile).
Remote Controlling Ion Channels 421
2.6 Whole-Cell Patch 1. Patch clamp amplifier with data acquisition electronics, elec-
Clamp Measurements trode holder, and software installed on a desktop computer
(e.g., AxoPatch200B, Digidata 1440A, HL-U, and pCLAMP
10, Biberach, Germany).
2. Micromanipulator on rotating base mounted to a tower (e.g.,
MP-285ROE, 285RBI, and MT-71-9, Sutter Instruments,
Science Products, Hofheim, Germany) (see Note 7).
3. Glass micropipette puller (e.g., P-97, Sutter Instruments).
4. Extracellular solution: 145 mM NaCl, 4 mM KCl, 1 mM
MgCl2, 2 mM CaCl2, 10 mM HEPES, pH 7.4 (see Note 8).
5. Intracellular solution: 135 mM K-gluconate, 10 mM NaCl, 2 mM
MgCl2, 2 mM MgATP, 1 mM EGTA, 10 mM HEPES, pH 7.4.
6. Salt bridge solution: 3 M KCl and 1% electrophoresis-grade
agarose in water.
7. Glass capillary tubes (1.5 mm outer diameter, 1.17 mm inner
diameter, 100 cm length, Warner Instruments, Hamden, CT).
8. Unplugged glass Pasteur pipettes (length >200 mm).
9. Glass bottom petri dish (glass surface: 25 mm diameter,
0.2 mm thickness, Bioptechs, Butler, PA, USA) (see Note 9).
3 Methods
3.3 Polymerase 1. Dilute the vector containing the ion channel gene to a concen-
Chain Reaction tration of 20 ng/mL.
to Generate Cysteine 2. Resuspend lyophilized mutagenesis oligonucleotides (forward
Mutants and reverse) to a concentration of 300 mM by adding X*3.33 mL
water to the original tubes (X is the amount of nanomoles
delivered in the tube). Vortex for 10 s and spin briefly every
5 min for a total time of 15 min. Store at −20°C.
424 Stephanie Szobota et al.
3.4 Amplification 1. Warm up LB agar plates (one for each PCR) at 37°C for 30 min
and Selection (see Note 11).
of Mutagenized 2. Transform PCR without further purification into competent
Receptors cells. Maintain cells on ice during the transformation and treat
cells gently without vortexing or excessive pipetting.
3. Spread bacteria evenly on plates with a sterile glass rod.
4. Place plates at 37°C upside down overnight.
5. On the following day, combine 3 mL LB medium with 3 mL
ampicillin solution in glass tubes (four for each PCR).
6. Using a pipette tip, pick four colonies of each plate by lightly
touching a single colony with the tip and dropping the pipette
tip into a glass tube.
7. Place glass tubes at 37°C with shaking (~225 rpm) overnight.
8. On the following day, extract vector from bacteria with
MiniPrep kit following the manufacturer’s instructions and
send for verification by sequencing.
3.6 Verification 1. Add 0.5 mL MAG1 stock solution to 1 mL DPBS, vortex, and
of MAG1 spin briefly.
Concentration 2. Prepare 5 tenfold serial dilutions: Add 100 mL of the first
(See Note 13) MAG1 dilution to 900 mL PBS to produce a second dilution.
Remote Controlling Ion Channels 425
3.8 Preparation 1. Warm trypsin solution, DPBS, and DMEM in water bath to
of HEK293 Cells 37°C. Warm 9 mL DMEM in a culture flask in the incubator
(see Note 15).
2. Add 1.5 mL DMEM to each cover glass prepared previously in
the 12-well plate.
3. Remove cells from incubator and aspirate media.
4. Add 10 mL DPBS, let stand for 10 s, and aspirate.
5. Add 3 mL trypsin solution and let stand for 3 min.
6. To detach cells from flask, tap flask against a vertical surface.
7. Add 7 mL media to the flask and pipet up and down with a
serological pipette (see Note 16).
8. Transfer 1 mL cell suspension to the culture flask that contains
9 mL pre-warmed media. Place flask in incubator to passage
the culture in the future.
9. Transfer the remaining 9 mL cell suspension to a 50 mL coni-
cal tube.
10. Centrifuge at 1,000 × g for 3 min and aspirate supernatant.
426 Stephanie Szobota et al.
3.11 Dissection 1. Obtain P0–P4 Sprague Dawley rat pups (local ethical permis-
of Hippocampus sion and other appropriate approvals will be required)
2. Autoclave dissection tools prior to dissection.
3. Warm saline in water bath, warm media (refer to Subheading 2.4)
in incubator and bring trypsin solution to room temperature.
4. Fill two 60 mm petri dishes halfway with warm saline.
5. Humanely sacrifice the animal in accordance to the local ethi-
cal guidance. Sever neck at base of head with large scissors and
place under dissecting scope.
6. Remove skin from back to front with forceps.
7. Slice the skull from back to front with small scissors.
Remote Controlling Ion Channels 427
8. Peel back the skull to each side with large forceps, remove brain
with curved spatula, and slide it into petri dish containing the
saline. Repeat for additional animals and then transfer all brains
into a new dish.
9. Remove cerebellum and separate hemispheres with scalpel.
10. Remove meninges from the outer surface of the brain (see
Note 19).
11. Identify the hippocampus as a curved structure that is slightly
denser and therefore has a dark contrast to the rest of the tis-
sue. Gently cut away the hippocampus being very careful not
to damage the tissue.
12. Remove remaining meninges with minimal damage to the hip-
pocampus and clean away any excess tissue still attached to it
with small scalpel.
13. Carefully transfer hippocampus to 15 mL conical tube filled
with saline.
3.12 Preparation 1. Remove all but 4.5 mL saline from the tube containing hip-
of Hippocampal Cells pocampi, add 500 mL warm trypsin, and invert to mix.
2. Place at 37°C for 8 min.
3. Add 9 mL warm saline to dilute trypsin, invert to mix, and
remove solution using serological pipette. Repeat for a total of
four washes.
4. Add 1 mL media and triturate six times with large-diameter
Pasteur pipette. Remove media with suspended cells and strain
into a 50 mL conical tube.
5. Repeat previous step with small-diameter Pasteur pipette if
more cells are desired.
6. Count cells and dilute to a concentration of 200,000 cells/mL.
7. Add 500 mL cell suspension to each well and place in
incubator.
8. After 15 min, replace media to eliminate debris that has not
attached to cover glass.
9. After 4–7 days in culture, AraC is added to a final concentra-
tion of 4 mM to prevent growth of glia.
10. To maintain cultures, replace half of the media with fresh media
once per week.
3.13 Transfection 1. For each cover glass, warm 1 mL transfection media (985 mL
of Hippocampal Cells MEM supplemented with 15 mL high-glucose MEM), 500 mL
HBSS, and 500 mL growth media in the wells of a 6-well plate
placed in the incubator.
2. Bring CaCl2 solution, BBS, and expression vectors to room
temperature.
428 Stephanie Szobota et al.
3.14 Preparation 1. Using the flame of a gas burner, bend the end of a Pasteur
of Whole-Cell Patch pipette into a “U” shape where the “sides” and the “bottom”
Clamp Measurements of the U are ~2 cm long (see Note 20).
2. Heat salt bridge solution in a microwave until homogenous.
Allow to cool to 50°C.
3. Aspirate solution in a 1 mL pipette tip and fill the U-shaped
glass to complete the salt bridge. Store the salt bridge in 3 M
KCl at 4°C.
4. Using glass capillary tubing and a multistage puller, prepare
micropipettes with taper length of ~4 mm, tip diameter of
~2 mm, and electrical resistance of 2–8 MW. Refer to Chapter
7 for further considerations regarding the patch pipette
manufacturing.
3.15 Preparation 1. Connect the light source to the data acquisition electronics
of Microscopy and configure the software such that the wavelength can be
computer controlled.
2. Connect the optical output of the light source to the back port
of the microscope.
3. To direct light of all wavelengths to the sample, fix a total
reflectance mirror with superglue in the spare filter holder at
the position reserved for the dichroic mirror. Place this filter
cube in the turret (see Note 21).
4. Place the unmodified filter cube for YFP imaging in the turret.
5. Measure the intensity of the light exiting the objective using
the power meter (see Note 6).
Remote Controlling Ion Channels 429
3.16 MAG1 Labeling 1. In the laminar flow hood, fill four wells of a 12-well plate with
of HEK293 Cells 1 mL extracellular solution and supplement one well with DTT
and Neurons to a final concentration of 1 mM. Fill one well with concanava-
(See Note 22) lin A solution.
2. To remove growth medium, transfer a cover glass with HEK293
cells or neurons from growth medium to extracellular
solution.
3. To activate surface cysteines by ensuring that they are reduced,
transfer the cover glass to extracellular solution with DTT and
incubate for 10 min.
4. To remove DTT, transfer cover glass to extracellular solution
and incubate for 2 min.
5. For concanavalin treatment, transfer cover glass to concanava-
lin A solution and incubate for 10 min (see Note 23).
6. In the meantime, thaw MAG1 stock solution and illuminate
with UV light for 1–15 min (see Note 24).
7. Dilute illuminated MAG1 in one of the wells to a final concen-
tration of 10–50 mM (see Note 25).
8. Transfer cover glass to this solution and incubate for 10–30 min.
Keep the well plate in the dark during this time.
9. Transfer cover glass to a well with extracellular solution and
keep in the dark until the experiment.
3.17 Patch Clamp 1. Add 500 mL extracellular solution to glass bottom petri dish
Measurements and and transfer the cover glass to the dish.
Photoswitching 2. Connect the glass bottom dish to a dish with the reference
electrode using the salt bridge.
3. Adjust the microscope for visualization of YFP by choosing the
appropriate filter cube and by switching the light source to
510 nm excitation light.
4. Using the eye pieces, identify a transfected cell (see Note 26).
5. Apply and clamp positive pressure to the micropipette while
lowering it into extracellular solution to prevent adsorption of
debris at the liquid–air interface.
6. Position the micropipette over the center of the cell
(see Note 27).
7. Slowly lower the micropipette until it presses down on the cell
surface, causing an indentation.
8. Rotate the modified filter cube that holds the total reflectance
mirror into the light path.
9. Adjust the holding potential to −70 mV.
10. Release the positive pressure from the micropipette, observe
GW seal formation, and use negative pressure to break through
430 Stephanie Szobota et al.
Fig. 2 Optical control of HEK293 cell currents and hippocampal neuron action potentials with LiGluR. (a) Whole-
cell current in a voltage-clamped HEK293 cells expressing LiGluR (holding potential −60 mV). Current can be
photo-controlled with maximum activation at ~380 nm and inactivation at ~500 nm. As a positive control for
ion channel expression, glutamate as a free ligand can be added using gravity flow. (b) LiGluR activation depo-
larizes and triggers action potential firing in a current-clamped hippocampal neuron (top trace). After a brief
pulse, depolarization is stable in the dark (bottom trace) (Part a is reprinted by permission from Macmillan
Publishers Ltd: Nat Chem Biol (7), copyright 2006. Part b is reprinted from Neuron, ref. 17 with permission from
Elsevier Ltd)
4 Notes
1. In our experience, all mammalian expression vectors containing
the cytomegalovirus (CMV) promoter performed well in
HEK293 cells and neurons with robust protein expression.
Increased expression of ion channels was achieved using the
hybrid CMV enhancer/chicken b-actin promoter or with
woodchuck hepatitis virus posttranscriptional regulatory ele-
ment (16). Most ion channel genes can be obtained in expres-
sion vectors from Addgene (http://www.addgene.org/).
2. Co-transfection of fluorescent proteins allows selecting trans-
fected cells in patch clamp experiments. Furthermore,
fluorescent proteins under the control of cell type-specific pro-
moters can be used to distinguish cell types. To target neuronal
cells in hippocampal cell cultures, a modified pcDNA3.1(−)
expression vector containing the neuron-specific human syn-
apsin 1 promoter is available from the authors.
3. Mutagenesis oligonucleotide primers can be reliably designed
using the PRIMERX website (http://www.bioinformatics.
org/primerx/) following the QuickChange parameters.
Sequencing oligonucleotide primers should be designed
according to the recommendations of the sequencing provider,
which also may offer universal oligonucleotide primers that
bind to many vectors.
4. In our experience, cover glasses from some other manufactur-
ers require additional cleaning in 70% EtOH solution followed
by washing in water.
5. In the past years, the polychrome has become a standard light
source for optogenetic experiments as it offers bright illumina-
tion with maximum wavelength flexibility. A good alternative
is filter-based light sources capable of producing high-intensity
illumination (>10 mW/mm2) with fast switching time between
filters (<2 ms) (e.g., DG4, Sutter Instruments, Science
Products, Hofheim, Germany).
6. For accurate optogenetic experiments, it is required to mea-
sure the light intensity produced in every individual setup. This
is commonly done at the very end of the light path by quanti-
fying the light exciting the objective. The total intensity is
measured using the power meter, while the illuminated area
can be estimated to be the field of view. Latter can be deter-
mined using a micrometer-sized grid. For a more elaborate
measurement, the illuminated area is controlled using an aper-
ture or pinhole. Most optogenetic applications work well with
intensities of >1 mW/mm2, and MAG1 can be reliably con-
verted with as little intensity as 0.1 mW/mm2 (11).
432 Stephanie Szobota et al.
14. While 2–8 h are recommended for treatment, cover glasses can
be left in PLL solution for several weeks, and PLL solution can
be reused at least six times if kept sterile.
15. Warming media in the incubator is preferred over warming it
in the water bath as equilibration with CO2 and consequently
proper pH are ensured.
16. Use of serological pipettes is recommended as narrow 1 mL
pipette tips can damage cells.
17. For transfection of most glutamate receptor vectors, the
amount of Lipofectamine and vector can be scaled down sub-
stantially (up to fivefold) compared to the recommendation in
the manufacturer’s protocol. It is critical to use miniscule
amounts of fluorescent protein vector compared to ion chan-
nel vector (typically 10–20 times less fluorescent protein vector
than the ion channel vector).
18. For whole-cell patch clamp measurements, HEK293 cells
should be sparse at the time of experiment to prevent gap junc-
tion formation between adjacent cells. For many other pur-
poses (e.g., imaging) a confluent layer works well and gives the
best transfection efficiency and cell health.
19. Meninges will cause microglial growth in the cell culture that
is harmful to neuronal growth. When removing meninges, dis-
card damaged hippocampal tissue.
20. In the first step, hold the pipette horizontally and heat it 2 cm
from the end until the terminal piece drops by gravity to pro-
duce a right angle. Repeat to complete the U shape. Using a
glass cutter and appropriate safety equipment, separate the U
shape from the rest of the Pasteur pipette.
21. It is not safe to look into the eye pieces when using this modified
filter cube as unfiltered stray light may reach to the eye piece.
It is at no time safe to expose eyes to unfiltered light from the
light source, to light in the UV range, or to bright light of any
wavelength: Consider local safety regulations.
22. While this procedure describes MAG1 labeling of cells express-
ing LiGluR, it is generally applicable to other PTLs and ion
channels. Labeling and wash steps can be executed either at
room temperature or at 37°C. DTT treatment and UV preil-
lumination are optional steps that yield the greatest efficiency
of MAG1 labeling and thus photoswitching.
23. Concanavalin A blocks desensitization of GluK2 and is required
for experiments in HEK293 cells but not neurons.
24. Efficiency of MAG1 labeling is increased if MAG1 is converted
to the cis-isoform in UV light before the labeling. This can be
achieved with any handheld UV source, e.g., a UV LED
pointer, and at any light intensity. Indicative values: at an
434 Stephanie Szobota et al.
Acknowledgments
References
1. VanEngelenburg SB, Palmer AE (2008) with an optical switch. Nat Chem Biol
Fluorescent biosensors of protein function. 2:47–52
Curr Opin Chem Biol 12:60–65 8. Banghart M, Borges K, Isacoff E, Trauner D,
2. Fenno L, Yizhar O, Deisseroth K (2011) The Kramer RH (2004) Light-activated ion chan-
development and application of optogenetics. nels for remote control of neuronal firing. Nat
Annu Rev Neurosci 34:389–412 Neurosci 7:1381–1386
3. Szobota S, Isacoff EY (2010) Optical control 9. Chambers JJ, Banghart MR, Trauner D,
of neuronal activity. Annu Rev Biophys Kramer RH (2006) Light-induced depolariza-
39:329–348 tion of neurons using a modified Shaker K+
4. Fehrentz T, Schonberger M, Trauner D (2011) channel and a molecular photoswitch.
Optochemical genetics. Angew Chem Int Ed J Neurophysiol 96:2792–2796
Engl 50:12156–12182 10. Janovjak H, Sandoz G, Isacoff EY (2011) A
5. Chabala LD, Lester HA (1986) Activation of modern ionotropic glutamate receptor with a
acetylcholine receptor channels by covalently K+ selectivity signature sequence. Nat Commun
bound agonists in cultured rat myoballs. 2:232
J Physiol 379:83–108 11. Janovjak H, Szobota S, Wyart C, Trauner D,
6. Tochitsky I, Banghart MR, Mourot A, Yao JZ, Isacoff EY (2010) A light-gated, potassium-
Gaub B, Kramer RH, Trauner D (2012) selective glutamate receptor for the optical
Optochemical control of genetically engi- inhibition of neuronal firing. Nat Neurosci
neered neuronal nicotinic acetylcholine recep- 13:1027–1032
tors. Nat Chem 4:105–111 12. Hsin J, Arkhipov A, Yin Y, Stone JE, Schulten
7. Volgraf M, Gorostiza P, Numano R, Kramer K (2008) Using VMD: an introductory tuto-
RH, Isacoff EY, Trauner D (2006) Allosteric rial. Curr Protoc Bioinformatics Chapter
control of an ionotropic glutamate receptor 5:Unit 5.7
Remote Controlling Ion Channels 435
13. Mayer ML (2005) Crystal structures of the of an ionotropic glutamate receptor. Proc Natl
GluR5 and GluR6 ligand binding cores: Acad Sci USA 104:10865–10870
molecular mechanisms underlying kainate 16. Donello JE, Loeb JE, Hope TJ (1998) Woodchuck
receptor selectivity. Neuron 45:539–552 hepatitis virus contains a tripartite posttranscrip-
14. Stawski P, Janovjak H, Trauner D (2010) tional regulatory element. J Virol 72:5085–5092
Pharmacology of ionotropic glutamate recep- 17. Szobota S, Gorostiza P, Del Bene F, Wyart C,
tors: A structural perspective. Bioorg Med Fortin DL, Kolstad KD, Tulyathan O, Volgraf M,
Chem 18:7759–7772 Numano R, Aaron HL, Scott EK, Kramer RH,
15. Gorostiza P, Volgraf M, Numano R, Szobota S, Flannery J, Baier H, Trauner D, Isacoff EY (2007)
Trauner D, Isacoff EY (2007) Mechanisms of Remote control of neuronal activity with a light-
photoswitch conjugation and light activation gated glutamate receptor. Neuron 54:535–545
INDEX
A Chromatography
affinity ......................................................12, 37–38, 281
Action potential (AP) ..................................4, 159, 166, 175, FPLC .......................................................................... 39
185, 201, 277, 392, 403, 430 size exclusion ................................................... 46, 48, 53
Adeno-associated virus ............................................ 401–414 ClC-2 ............................... 321, 328–329, 331–332, 335, 336
Adenovirus .......................................................23, 24, 28–32 Cloning
Affinity purification...................................... 39, 66, 251, 254 differential ................................................................. 5–9
Agarose gel electrophoresis...............................260, 262, 263 expression .......................................................... 8–10, 12
Amiloride ................................. 324, 331, 334, 388, 393–395 functional ............................................................. 5, 8–14
Amphotericin B...............................................150, 151, 153, subtractive.................................................................. 5–7
154, 156, 313 Confocal microscopy ............................................... 202, 210
Angiotensin II AT1 receptors .................................. 214, 215 Cortical collecting duct ....................................345, 347, 351
Antibodies Cover-slip preparation ....................................... 61, 234–235
blocking antibodies .....................................246, 247, 254 Cre–lox system .................................................................... 4
protein detection........................................................ 220 Crystallization .................................... 49, 278, 279, 282–283
AP. See Action potential (AP) Current clamp ......................................... 106, 162, 167, 175,
Automated patch clamp recording .......................... 181, 182 180, 185, 350, 388, 412, 430
Azobenzene ............................................................. 402, 432 Cysteine-based cross-linking approach ................... 267–275
B D
Bilayer recording ............................................................... 92 Dorsal root ganglion (DRG)
Biolistic particle delivery. See Transfection neuron culture .......................................56, 160–162, 312
Biotinylation ........................................................................ 7 slice preparation ................................................. 312, 319
BK channels .............................................134–144, 146, 277 transfection of ................................... 9, 11, 56, 60, 61, 64
Black lipid membranes .................................................... 114 Drosophila melanogaster
Bradykinin receptors........................................................ 378 embryos ............................................................. 386–387
Brain slice patch-clamping sensory neurons from ............... 385–396
preparation................................................................. 304 Drug screening .................................................................. 91
recording............................................................ 104, 192
E
C
Electrophysiology
Calcium-activated K+ channels ................. 248, 277, 334, 367 patch clamp ........................................ 137, 149, 189, 269
Calcium channels ....................................... 14, 183, 191, 197 single channel recording ............................................ 137
Calmodulin voltage clamp ............................................................. 110
dansyl-calmodulin (D-CaM)............................. 217–230 Electroporation.................................................. 24, 403, 413
interaction with ion channels............................. 217–230 Epithelial Na+ channel ............................................. 267, 342
Carboxy-terminal epitope tagging ............................... 34, 39 Erythrocyte
Cardiomyocyte .................................................192, 193, 196 ATP release from ....................................................... 336
Channelrhodopsin ................................................... 401–414 electrophysiology ....................................................... 173
Chemiluminescence.................................. 234, 236, 239, 240 ion channels ....................................................... 321–337
Chinese hamster ovary (CHO) cells........................9, 11, 24, isosmotic haemolysis.......................................... 327, 328
25, 151, 152, 155, 173, 212, 214, 215 tracer flux ................................................... 332–333, 337
Nikita Gamper (ed.), Ion Channels: Methods and Protocols, Methods in Molecular Biology, vol. 998,
DOI 10.1007/978-1-62703-351-0, © Springer Science+Business Media, LLC 2013
437
438 Index
F I
Fluorescence Immunochemistry ................................................... 189, 234
microscopy ............................................ 25, 70, 201–207, Immunofluorescence ....................................... 253, 375–376
209, 356, 421 Inflammatory mediator .................................................... 64
spectroscopy ....................................................... 217, 219 Integral membrane protein ....................................... 91, 133,
Fluorescent dye 203, 257, 385
Fluo3/4 ..............................................................329, 335 Intracellular perfusion.............................................. 119, 380
Fura2 .........................................................................378 Ionomycin....................................................... 324, 326, 327,
Lucifer yellow ............................................................317 329, 331, 334, 378
Fluorescent protein (FP) .................................. 22, 136, 202, Isolated renal tubules ............................................... 346, 373
203, 206, 211, 404–406, 409, 412, 431, 433 Isosmotic haemolysis ....................................... 327, 328, 331
Focal segmental glomerulosclerosis (FSGS) ....................356
Forster resonance energy transfer (FRET) K
efficiency.................................................... 210–212, 214
FRET pair .................................................................210 KATP channels...........................................................233–241
principles ........................................................... 209, 210 K+ channel
quantification.....................................................214–216 endogenous ................................................................247
FPLC. See Chromatography, FPLC heterologous expression of .........................................151
FRET. See Forster resonance energy transfer (FRET) patch clamp recording of ...........................................312
FSGS. See Focal segmental glomerulosclerosis (FSGS) KCNQ channels. See Kv7 channels
Kv7 channels
G interaction with calmodulin ............................... 218, 219
recording from DRG slices ........................................312
GABA
two-electrode voltage clamp recording of ................... 81
currents ......................................................................270
receptors ....................................................................267 L
Gardos channels ...............................326, 327, 331, 334, 336
Gel electrophoresis ............................................ 44, 260–263 Laminin coating ........................................ 61, 103, 194, 195
Gene delivery. See Transfection Lentivirus ................................................ 67, 71, 73, 74, 405
GFP. See Green fluorescent protein (GFP) Ligand-gated ion channels ................................. 79, 99, 102,
Giant axon ................................................................... 79, 80 173, 179, 417–434
Giant patch. See Macropatch Lipid bilayer ............................................... 52, 92, 109–118,
Glutamate receptors 201, 385
glutamate binding ......................................................418 Lipofectamine ...................................................... 24–27, 31,
use for optogenetics ...................................................418 135, 139, 145, 202, 204, 269, 272,
G protein coupled receptor ........................................ 56, 378 274, 420, 426, 433
Gramicidin ......................................................................150 Liposomes ......................................................... 31, 110, 118
Green fluorescent protein (GFP) ......................... 29, 31, 61, Live-cell imaging ............................................. 190, 209–216
152, 182, 205, 207, 211, 269, 272–274, 387–389, L-type calcium channels ..................................................191
391, 394, 404 Lucifer yellow. See Fluorescent dye
H M
HEK293 cells. See Human embryonic kidney Macropatch ....................................... 92, 106, 119, 126, 127
(HEK293) cells Mammalian expression systems ............................... 9, 21–32
HERG potassium channels .............................................174 M channels. See Kv7 channels
Heterologous expression system ........................ 34, 174, 262 M current ................................................................311–319
Hippocampal neurons Mechanosensitive ion channel .......................................... 62
culture ........................................................................421 Membrane protein purification ...................................33–53
dendritic recordings ...................................................303 Microinjection .................................................................. 82
pyramidal neurons .............................................306–308 MthK channel ......................................................... 277, 278
Index 439