7
Please cite this article in press as: Lebbe EKM, et al. Ala-7, His-10 and Arg-12 are crucial amino acids for activity of a synthetically engineered -conotoxin. Peptides (2013), http://dx.doi.org/10.1016/j.peptides.2013.07.005 ARTICLE IN PRESS G Model PEP 69015 1–7 Peptides xxx (2013) xxx–xxx Contents lists available at SciVerse ScienceDirect Peptides jo ur nal homep age: www.elsevier.com/locate/peptides Ala-7, His-10 and Arg-12 are crucial amino acids for activity of a synthetically engineered -conotoxin Eline K.M. Lebbe a , Steve Peigneur a , Ward Brullot b , Thierry Verbiest b , Jan Tytgat a,Q1 a Toxicology and Pharmacology, University of Leuven (KU Leuven), Campus Gasthuisberg O&N2, Herestraat 49, P.O. Box 922, 3000 Leuven, Belgium b Laboratory for Molecular Electronics and Photonics, Division Molecular Imaging and Photonics, Department of Chemistry, University of Leuven, Celestijnenlaan 200D, P.O. Box 2425, 3001 Heverlee, Belgium a r t i c l e i n f o Article history: Received 13 May 2013 Received in revised form 1 July 2013 Accepted 1 July 2013 Available online xxx Keywords: (-)Conotoxin Voltage-gated sodium channel Midi R2 Circular dichroism Cone snail Peptidomimetics a b s t r a c t Cone snail toxins or conotoxins are often small cysteine-rich peptides which have shown to be highly selective ligands for a wide range of ion channels such as voltage-gated sodium channels (Na V s). Na V s participate in a wide range of electrophysiological processes. Consequently, their malfunction has been associated with numerous diseases. The development of subtype-selective modulators of Na V s remains highly important in the treatment of such disorders. In order to expand our knowledge in the search for novel therapeutics to treat Na V -related diseases, we explored the field of peptide engineering. In the current study, the impact of well considered point mutations into a bioactive peptide that was found to be a very potent and selective inhibitor of Na V s (i.e. Midi R2) was examined. We designed two pep- tides, named Midi R2[A7G] and Midi R2[H10A, R12A] which have mutations at position 7, and both 10 and 12, respectively. Electrophysiological recordings indicated that an Ala to Gly mutation at position 7 increased IC 50 -values from the nanomolar range to the micromolar range. For Midi R2[H10A, R12A] at a concentration of 10 M, activity is even reduced to 0–10% for all of the tested Na V -channels. Circular dichroism measurements proved that overall structural conformations did not change. These findings suggest that the minimal space between the second and the third intercysteine loop of Midi R2 is the sequence RRWARDHSR and that His at position 10 and Arg at position 12 are crucial amino acids for the potency and specificity of Midi R2. In this way, new insights into the structure–activity relationships of -conotoxins were found. © 2013 Published by Elsevier Inc. 1. Introduction Venoms from cone snails (genus Conus) can be seen as an untapped cocktail of biologically active compounds, being increas- ingly recognized as a new emerging source of peptide-based therapeutics [21]. These creatures (family Conidae) are unique because of their possibility to use an incredibly diverse arse- nal of small (<5 kDa), cysteine-rich peptides (called conopeptides or conotoxins) to hunt for preys [1]. So far, about 500 differ- ent cone snail species are known and each species produces an unprecedented molecular diversity of pharmacologically active components. Up to date, only a few hundred venom components Abbreviations: NaV-channel, voltage-gated sodium channel; CD, circular dichro- ism. Corresponding author. Tel.: +32 16 32 34 03; fax: +32 16 32 34 05. E-mail addresses: [email protected], eline [email protected] (E.K.M. Lebbe), [email protected] (S. Peigneur), [email protected] (W. Brullot), [email protected] (T. Verbiest), [email protected] (J. Tytgat). of over potentially 100,000 have been functionally and structurally investigated [26]. Furthermore, the consideration of Conus venoms as gold mines for the discovery of new therapeutics is validated by the knowledge that, out of the limited number studied conopep- tides, already six peptides have reached human clinical trials, and one was approved as analgesic in 2004 [2,14,27]. Conotoxins display a great molecular diversity which is mir- rored by the impressive diversity of their targets and even more in the specific recognition of different binding sites within one tar- get [3]. Exactly this unparalleled specificity enables conotoxins to be highly selective and extremely potent ligands of several sub- types of voltage-gated ion channels, while others are considered as impressive selective and potent blockers of neuronal and muscle subtypes of nicotine-type acetylcholine receptors (nAChRs) [2,14]. Voltage-gated sodium channels (Na V s) are important trans- membrane proteins regarding generation as well as propagation of action potentials in excitable cells such as neuronal and muscular cells [7,16,20]. Nine channel isoforms of the Na V 1 subfamily have been identified up to now, having similar functional properties but different distribution in muscle and nerve cells [8]. Consequently, defective Na V s cause several diseases or channelopathies like 0196-9781/$ see front matter © 2013 Published by Elsevier Inc. http://dx.doi.org/10.1016/j.peptides.2013.07.005 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53

Ala-7, His-10 and Arg-12 are crucial amino acids for activity of a synthetically engineered μ-conotoxin

Embed Size (px)

Citation preview

G

P

As

EQ1

a

b

C

ARRAA

K(VMCCP

1

uitbnoeuc

i

(w(

0h

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

ARTICLE IN PRESS Model

EP 69015 1–7

Peptides xxx (2013) xxx– xxx

Contents lists available at SciVerse ScienceDirect

Peptides

jo ur nal homep age: www.elsev ier .com/ locate /pept ides

la-7, His-10 and Arg-12 are crucial amino acids for activity of aynthetically engineered �-conotoxin

line K.M. Lebbea, Steve Peigneura, Ward Brullotb, Thierry Verbiestb, Jan Tytgata,∗

Toxicology and Pharmacology, University of Leuven (KU Leuven), Campus Gasthuisberg O&N2, Herestraat 49, P.O. Box 922, 3000 Leuven, BelgiumLaboratory for Molecular Electronics and Photonics, Division Molecular Imaging and Photonics, Department of Chemistry, University of Leuven,elestijnenlaan 200D, P.O. Box 2425, 3001 Heverlee, Belgium

a r t i c l e i n f o

rticle history:eceived 13 May 2013eceived in revised form 1 July 2013ccepted 1 July 2013vailable online xxx

eywords:�-)Conotoxinoltage-gated sodium channelidi R2

ircular dichroismone snaileptidomimetics

a b s t r a c t

Cone snail toxins or conotoxins are often small cysteine-rich peptides which have shown to be highlyselective ligands for a wide range of ion channels such as voltage-gated sodium channels (NaVs). NaVsparticipate in a wide range of electrophysiological processes. Consequently, their malfunction has beenassociated with numerous diseases. The development of subtype-selective modulators of NaVs remainshighly important in the treatment of such disorders. In order to expand our knowledge in the searchfor novel therapeutics to treat NaV-related diseases, we explored the field of peptide engineering. In thecurrent study, the impact of well considered point mutations into a bioactive peptide that was foundto be a very potent and selective inhibitor of NaVs (i.e. Midi R2) was examined. We designed two pep-tides, named Midi R2[A7G] and Midi R2[H10A, R12A] which have mutations at position 7, and both 10and 12, respectively. Electrophysiological recordings indicated that an Ala to Gly mutation at position 7increased IC50-values from the nanomolar range to the micromolar range. For Midi R2[H10A, R12A] at

a concentration of 10 �M, activity is even reduced to 0–10% for all of the tested NaV-channels. Circulardichroism measurements proved that overall structural conformations did not change. These findingssuggest that the minimal space between the second and the third intercysteine loop of Midi R2 is thesequence RRWARDHSR and that His at position 10 and Arg at position 12 are crucial amino acids for thepotency and specificity of Midi R2. In this way, new insights into the structure–activity relationships of�-conotoxins were found.

33

34

35

36

37

38

39

40

41

. Introduction

Venoms from cone snails (genus Conus) can be seen as anntapped cocktail of biologically active compounds, being increas-

ngly recognized as a new emerging source of peptide-basedherapeutics [21]. These creatures (family Conidae) are uniqueecause of their possibility to use an incredibly diverse arse-al of small (<5 kDa), cysteine-rich peptides (called conopeptidesr conotoxins) to hunt for preys [1]. So far, about 500 differ-

Please cite this article in press as: Lebbe EKM, et al. Ala-7, His-10 and Arg�-conotoxin. Peptides (2013), http://dx.doi.org/10.1016/j.peptides.2013.07

nt cone snail species are known and each species produces annprecedented molecular diversity of pharmacologically activeomponents. Up to date, only a few hundred venom components

Abbreviations: NaV-channel, voltage-gated sodium channel; CD, circular dichro-sm.∗ Corresponding author. Tel.: +32 16 32 34 03; fax: +32 16 32 34 05.

E-mail addresses: [email protected], eline [email protected]. Lebbe), [email protected] (S. Peigneur),[email protected] (W. Brullot), [email protected]

T. Verbiest), [email protected] (J. Tytgat).

196-9781/$ – see front matter © 2013 Published by Elsevier Inc.ttp://dx.doi.org/10.1016/j.peptides.2013.07.005

42

43

44

45

46

47

48

49

© 2013 Published by Elsevier Inc.

of over potentially 100,000 have been functionally and structurallyinvestigated [26]. Furthermore, the consideration of Conus venomsas gold mines for the discovery of new therapeutics is validated bythe knowledge that, out of the limited number studied conopep-tides, already six peptides have reached human clinical trials, andone was approved as analgesic in 2004 [2,14,27].

Conotoxins display a great molecular diversity which is mir-rored by the impressive diversity of their targets and even more inthe specific recognition of different binding sites within one tar-get [3]. Exactly this unparalleled specificity enables conotoxins tobe highly selective and extremely potent ligands of several sub-types of voltage-gated ion channels, while others are considered asimpressive selective and potent blockers of neuronal and musclesubtypes of nicotine-type acetylcholine receptors (nAChRs) [2,14].

Voltage-gated sodium channels (NaVs) are important trans-membrane proteins regarding generation as well as propagation ofaction potentials in excitable cells such as neuronal and muscular

-12 are crucial amino acids for activity of a synthetically engineered.005

cells [7,16,20]. Nine channel isoforms of the NaV1 subfamily havebeen identified up to now, having similar functional properties butdifferent distribution in muscle and nerve cells [8]. Consequently,defective NaVs cause several diseases or channelopathies like

50

51

52

53

ING Model

P

2 eptide

eot

hhcai

pkfpoimtaSivcp

2

2

LwPatalisaw

2

9wwmomL

2

srrmBiTe

54

55

56

57

58

59

60

61

62

63

64

65

66

67

68

69

70

71

72

73

74

75

76

77

78

79

80

81

82

83

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

100

101

102

103

104

105

106

107

108

109

110

111

112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

127

128

129

130

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

153

154

155

156

157

158

159

160

161

162

163

164

165

166

167

168

169

170

ARTICLEEP 69015 1–7

E.K.M. Lebbe et al. / P

pileptic disorders [12], neuromuscular diseases, and cardiomy-pathies [6]. Blocking the aberrant Na+ current can be effective inreating these disorders [23].

To date, four families of conotoxins which target NaV-channelsave been described: �-conotoxins [31] and �-conotoxins [4,13]aving agonistic effects, whereas �O-conotoxins [25] and �-onotoxins [10,33] have antagonistic effects. The small size as wells their high potency and selectivity turn �-conotoxins into verynteresting compounds [20,29].

In previous research from our lab, miniaturized peptides thatotently and selectively block NaVs, were designed based on twonown �-conotoxins (KIIIA from Conus kinoshitai [5,24] and BuIIICrom Conus bullatus [17,36]) in order to create potent and selectiveeptides that can be synthesized cost-effectively [34]. In a studyf structure-activity relationships in KIIIA, key residues that aremportant for activity on mammalian neuronal NaV1.2 and skeletal

uscle NaV1.4 subtypes were identified. The authors demonstratedhat the engineering of KIIIA could provide subtype-selective ther-peutics against NaVs for the potential treatment of pain [24].imilarly, the design of two new Midi-analogous was investigatedn this study. Based on the most promising compound from pre-ious studies, namely Midi R2, we explored the effect of wellonsidered mutations into the pharmacophore of this miniaturizedeptide.

. Materials and methods

In this study, the following materials and methods were used.

.1. Chemical synthesis and analysis of synthetic peptides

The following peptides were synthesized by ChinaPeptides Co.,td. (Shanghai, China): Midi R2[A7G] and Midi R2[H10A,R12A]ith molecular masses of 1752.0 Da and 1614.9 Da, respectively.

urity was confirmed to be >98% by reversed-phase HPLC on annalytical Vydac C18 column (218MS54, 4.6 × 250 mm, 5-�m par-icle size; Grace, Deerfield, IL) with a flow rate of 1 ml min−1. UVbsorbance was monitored at 214 and 280 nm with a dual wave-ength absorbance detector. A linear gradient of 0–40% acetonitrilen 40 min was used at a flow rate of 1 ml min−1. The gradient waset by means of a mixture of solvent A (0.085% (v/v) trifluoroaceticcid (TFA) in acetonitrile) and solvent B (0.1% TFA (v/v) in water)ith an initial concentration of 0% solvent A.

.2. Folding of the peptides

Peptides were dissolved in the physiological buffer solution ND-6. Folding mixtures were retained at room temperature for twoeeks. At different time points following dissolution, aliquots wereithdrawn. Aliquots were analyzed with reversed-phase HPLC byeans of an analytical Vydac C18 column as described in the previ-

us section. The concentration of the folded synthetic peptides waseasured using a Nanodrop ND-1000 spectrophotometer (Isogen

ife Science, De Meern, the Netherlands).

.3. Heterologous expression in Xenopus oocytes

Complementary DNA encoding the NaV-channels wasubcloned into the corresponding vector: the �-subunitsNaV1.1/pLCT1(NotI), rNaV1.2/pLCT1(NotI), rNaV1.3/pNa3T(NotI),NaV1.4/pUI-2(NotI), hNaV1.5/pcDNA3.1(XbaI),NaV1.6/pLCT(NotI), rNaV1.7/pBSTA.rPN1(SacII), cockroach

Please cite this article in press as: Lebbe EKM, et al. Ala-7, His-10 and Arg�-conotoxin. Peptides (2013), http://dx.doi.org/10.1016/j.peptides.2013.0

latella germanica BgNaV1.1/pGH19(NotI), and the correspond-ng �-subunits r�1/pSP64T(EcoRI) and Drosophila melanogasteripE/pGH19(NotI). The linearized plasmids – respective restrictionnzymes are indicated in parentheses – were transcribed using

171

PRESSs xxx (2013) xxx– xxx

the T7 (for rNaV1.1, rNaV1.2, rNaV1.3, rNaV1.4, mNaV1.6, rNaV1.7,BgNaV1.1, TipE) or the SP6 (for hNaV1.5 and r�1) mMESSAGE-mMACHINE transcription kit (Ambion, Austin, TX). The harvestingof stage V–VI oocytes from anaesthetized female Xenopus lae-vis frogs was previously described [35]. Oocytes were injectedwith 50–70 nl of cRNA at a concentration of 1–3 ng/nl using amicro-injector (Drummond Scientific, Broomall, PA). The oocyteswere incubated in a ND-96 solution containing: NaCl, 96 mM;KCl, 2 mM; CaCl2, 1.8 mM; MgCl2, 2 mM and HEPES, 5 mM (pH7.4), supplemented with 50 mg/l gentamycin sulfate and 0.5 mMtheophylline. Oocytes were stored for 1–5 days at 16 ◦C untilsufficient expression of NaVs was achieved.

2.4. Electrophysiology

Whole-cell currents from oocytes were recorded at room tem-perature (18–22 ◦C) by the two-electrode voltage clamp techniqueusing a GeneClamp 500 amplifier (Molecular Devices, Sunnyvale,CA) controlled by a pClamp data acquisition system (MolecularDevices). Oocytes were placed in a bath containing ND-96 solu-tion. Voltage and current electrodes were filled with 3 M KCl, andthe resistances of both electrodes were maintained as low as possi-ble (between 0.5 and 1.5 M�). The elicited currents were sampledat 20 kHz and filtered at 2 kHz using a four-pole, low pass Besselfilter. To eliminate the effect of the voltage drop across the bathgrounding electrode, the bath potential was actively controlled bya two-electrode bath clamp. Leak subtraction was performed usinga – P/4 protocol.

Whole-cell current traces were evoked every 5 s by a 100-msdepolarization to the voltage corresponding to the maximal acti-vation of the NaV-subtype in control conditions, starting from aholding potential of −90 mV. Concentration-response curves wereconstructed by adding different toxin concentrations directly to thebath solution. The percentage of NaV blockade was plotted againstthe logarithm of the applied concentrations and fitted with the Hillequation: y = 100/[1 + (EC50/[toxin])h], where y is the amplitude ofthe toxin-induced effect, EC50 is the toxin concentration at halfmaximal efficacy, [toxin] is the toxin concentration and h is the Hillcoefficient. To investigate the effects on the voltage dependence ofactivation, current traces were induced by 100-ms depolarizationsfrom a holding potential of −90 to 65 mV with 5-mV increments.The Na+ conductance was calculated from the currents using Ohm’slaw: gNa = INa/(V − Vrev), where INa represents the Na+ current peakamplitude at a given test potential V, and Vrev is the reversal poten-tial. The values of gNa were plotted as a function of voltage and fittedusing the Boltzmann equation: gNa/gmax = [1 + (exp(Vg − V)/k)]−1,where gmax represents maximal gNa, Vg is the voltage correspond-ing to half-maximal conductance and k is the slope factor. Toinvestigate the effects on the steady-state inactivation process,oocytes were depolarized using a standard two-step protocol.From a holding potential of −90 mV, 100-ms prepulses weregenerated, ranging from −90 to 65 mV with 5-mV increments,immediately followed by a 100-ms test pulse to −10 mV. Datawere normalized to the maximal Na+ current amplitude, plottedagainst prepulse potential and fitted using the Boltzmann equa-tion: INa/Imax = [(1 − C)/(1 + exp((V − Vh)/k))] + C, where Imax is themaximal INa, Vh is the voltage corresponding to half-maximal inac-tivation, V is the test voltage, k is the slope factor, and C is a constantrepresenting a non-inactivating persistent fraction (close to zero incontrol). Comparison of two sample means was made using a pairedStudent’s t test (p < 0.05). All data are presented as mean ± standarderror (SEM) of at least 3 independent experiments (n ≥ 3). All data

-12 are crucial amino acids for activity of a synthetically engineered7.005

was analyzed using pClamp Clampfit 10.0 (Molecular Devices®,Downingtown, PA) and Origin 7.5 software (Originlab®, Northamp-ton, MA).

172

173

174

ARTICLE IN PRESSG Model

PEP 69015 1–7

E.K.M. Lebbe et al. / Peptides xxx (2013) xxx– xxx 3

Fig. 1. Sequences of naturally occurring �-conotoxins (KIIIA en BuIIIC) and synthetically designed peptides.Shown are the sequences of two known, naturally occurring �-conotoxins, KIIIA from Conus kinoshitai (23, 24) and BuIIIC from Conus bullatus (25, 26) from which the Minipeptide and analogs Midi and Midi R2 were derived by Stevens et al. (27). Midi R2[A7G] and Midi R2[H10A, R12A] are investigated in this study. Dashes are put in thes k lines above the sequences represent the disulfide bridges. Cysteine amino acids are redc

2

i1

3

3

aoapRfsi

3

rtdi

3

Rmos

3

mts

Fig. 2. CD spectrum of Midi, Midi R2 and its analogous.

and BgNa 1.1). From these Na -subtypes, Midi R2[H10A, R12A]

175

176

177

178

179

180

181

182

183

184

185

186

187

188

189

190

191

192

193

194

195

196

197

198

199

200

201

202

203

204

205

206

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

223

224

equences to make all sequences and intercysteine loops of comparable length. Blacolored and mutated amino acids are indicated in bold.

.5. CD measurements

CD spectra were recorded on a JASCO J-820 spectropolarimetern H2O solution (MQ-water, pH 7.0) at 20 ◦C, with a quartz cell of-mm path length. The results are expressed in deg/(cm mol).

. Results

.1.1. �-Conotoxin engineering

Synthetic analogs of Midi and Midi R2, namely Midi R2[A7G]nd Midi R2[H10A, R12A] were engineered based on the studyf Stevens et al., 2012 [34] and McArthur et al., 2011 [24] Wettempted to design with Midi R2[A7G] a shorter peptide than theotent inhibitor Midi R2. The rationale for the double mutant, Midi2[H10A, R12A], was to strive for a more selective NaV1.7 inhibitor

unded on the study from [24] with KIIIA. Peptides were synthe-ized and folded in ND-96 solution. Peptide sequences are shownn Fig. 1.

.2. Folding of the peptides

To investigate the folding of the Midi analogs, changes in HPLCetention time were investigated and compared with the reten-ion time of the reduced peptide (data are not shown). As was alsoescribed in Stevens et al., 2012 [34], the Midi R2-analogs, dissolved

n ND-96 solution, were maximally folded after one week.

.3. CD measurements

The CD spectra of Midi R2, Midi R2[A7G] and Midi R2[H10A,12A] were strongly superimposable to each other with a mini-um around 202 nm (Fig. 2). A difference in signal strength can be

bserved between Midi and Midi R2 since Midi R2 seems to give atronger signal.

.4. Electrophysiological characterization

Using the two electrode voltage clamp technique our two

Please cite this article in press as: Lebbe EKM, et al. Ala-7, His-10 and Arg�-conotoxin. Peptides (2013), http://dx.doi.org/10.1016/j.peptides.2013.07

utants, Midi R2 [A7G] and Midi R2[H10A, R12A], were charac-erized by electrophysiological recordings in order to determineubtype selectivity profiles.

The peptides Midi, Midi R2, Midi R2[A7G] and Midi R2[H10A, R12A] were examinedby circular dichroism spectroscopy. This figure shows that the overall structure ofthe mutated peptides did not change.

3.4.1. Midi R2[A7G]Midi R2[A7G] was screened against a panel of seven mam-

malian NaV-channel isoforms at a concentration of 10 �M(Fig. 3). Among these isoforms Midi R2[A7G] inhibited all ofthe NaV-subtypes (NaV1.1 to NaV1.7) with inhibition percent-ages of 8.5 ± 3.3%, 25.3 ± 7.4%, 29.1 ± 4.2%, 23.8 ± 4.5%, 22.7 ± 6.2%,31.1 ± 8.2%, 12.4 ± 2.6% respectively. The IC50 values of MidiR2[A7G] on NaV1.2 and NaV1.4 are 14.7 ± 3.2 �M and 15.3 ± 3.9 �Mrespectively. Dose-response curves from NaV1.2 and NaV1.4 canbe found in Fig. 4 The NaV-subtypes NaV1.3 and NaV1.6 are mostsensitive for Midi R2[A7G].

Midi R2[A7G] was also tested on BgNaV1.1, an isoform from B.germanica, cockroach, representative for an insect NaV-channel, butno remarkable activity was observed at a concentration of 10 �M.

3.4.2. Midi R2[H10A, R12A]Midi R2[H10A, R12A] was submitted to a screening on the

same panel of voltage-gated sodium channels (NaV1.1 to NaV1.7

-12 are crucial amino acids for activity of a synthetically engineered.005

V V(10 �M) does not target any of them (Fig. 5). Remarkably, thecombination of the point mutations H10A and R12A completelyabolishes the activity of this peptide on the tested NaV-channels.

225

226

227

ARTICLE IN PRESSG Model

PEP 69015 1–7

4 E.K.M. Lebbe et al. / Peptides xxx (2013) xxx– xxx

Fig. 3. Electrophysiological measurements of Midi R2[A7G].Current traces were obtained by two-electrode voltage clamp on X. laevis oocytes heterologously expressing the NaV-isoforms as described under “Materials and Methods”.Currents were evoked by a depolarizing pulse starting from a holding potential of −90 mV to the voltage corresponding to the maximal activation in control conditions.T . The

p d NaV

AM

4

tarop

FAM1F

228

229

230

231

232

233

234

235

236

237

238

239

240

241

242

243

244

races shown are representative of at least three independent experiments (n ≥ 3)eak amplitude of the currents after exposure to 10 �M of Midi R2[A7G]. NaV1.3 an

comparison of the blocking percentages of the peptides Midi R2,idi R2[A7G] and Midi R2[H10A, R12A] is made in Fig. 6.

. Discussion

In the current study, we investigated the impact of point muta-ions into the pharmacophore of a bioactive peptide onto the

Please cite this article in press as: Lebbe EKM, et al. Ala-7, His-10 and Arg�-conotoxin. Peptides (2013), http://dx.doi.org/10.1016/j.peptides.2013.0

ctivity and potency of blocking NaV-channels. We integrated theesults of Stevens et al. [34] and those of McArthur et al. [24] intour design strategy. Stevens et al. [34] designed several bioactiveeptides (Mini and Midi) derived from two naturally occurring

ig. 4. Concentration-response curves on NaV1.2 and NaV1.4.. Midi R2 (�) and Midi R2[A7G] (©) were tested on NaV1.2, which was shown to be most

idi R2[A7G] are 34.1 ± 2.4 nM and 14.7 ± 3.2 �M respectively. B. Midi R2 (�) and Midi R2[5.3 ± 3.9 �M. Currents were obtained as described under “Materials and Methods”. The por both NaV-isoforms, results were fit with the Hill equation.

dotted line indicates the zero-current level. Asterisks (*) indicate the steady-state1.6 are most sensitive to Midi R2[A7G].

�-conotoxins, namely KIIIA from Conus kinoshitai, and BuIIIC fromConus bullatus. They illustrated that an extra Ala residue added tothe sequence of the Mini peptide restores the spacing (in compar-ison with the original �-conotoxins) which led to a peptide thatis active in the range of nanomolar concentrations onto NaV1.2,NaV1.4 and NaV1.6. They called this peptide Midi (sequence isshown in Fig. 1). The difference in activity between Mini (in a micro-molar range) and Midi (in a nanomolar range) was attributed to

-12 are crucial amino acids for activity of a synthetically engineered7.005

a difference in spacing whereupon they concluded that at leastsix residues are required to set up a fully functional bioactivescaffold that can interact properly with a binding area on the NaV-channel (see also [30]). Interestingly, the NMR structure of the

potently inactivated by Midi R2 (27). The corresponding IC50 values for Midi R2 andA7G] (©) were tested on NaV1.4 with corresponding IC50 values of 330 ± 570 nM andercentage of block was plotted against the logarithm of the tested concentrations.

245

246

247

248

ARTICLE IN PRESSG Model

PEP 69015 1–7

E.K.M. Lebbe et al. / Peptides xxx (2013) xxx– xxx 5

Fig. 5. Electrophysiological measurements of Midi R2[H10A, R12A]T ytes hr icatesa

Mtmla

FTwv

249

250

251

252

253

254

he current traces were obtained by two-electrode voltage clamp on X. laevis oocepresentative of at least three independent experiments (n ≥ 3). The dotted line indpplication of 10 �M Midi R2[H10A, R12A].

idi peptide does not show a complete alpha-helix which was

Please cite this article in press as: Lebbe EKM, et al. Ala-7, His-10 and Arg�-conotoxin. Peptides (2013), http://dx.doi.org/10.1016/j.peptides.2013.07

hought to be crucial for �-conotoxin biological activity [19,30]. Theutant Midi R2 in which an extra positive charge was integrated,

ed to lower IC50 values compared to Midi. The mutant exhibits very potent and selective block on NaV1.2 [34]. The overall

ig. 6. Overview of block by Midi R2, Midi R2[A7G] and Midi R2[H10A, R12A]he three peptides, Midi R2, Midi R2[A7G] and Midi R2[H10A, R12A], were tested on NaV1as seen at that concentration, this is represented by an arbitrary value of 1% for the clar

alues are discussed under “Results”. Data are represented as the means ± SE (indicated b

eterologously expressing a single type of cloned NaV-isoforms. Traces shown are the zero-current level. The asterisk (*) distinguishes the steady-state current after

higher potency is attributed to the larger electrostatic repulsion,

-12 are crucial amino acids for activity of a synthetically engineered.005

which was also previously proposed for BuIIIC [18]. Consequently,we decided to continue our research with this Midi R2-mutantin order to create smaller and/or more NaV-isoform selectivepeptides.

.1-NaV1.7 and BgNaV1.1 at a concentration of 10 �M. When no blocking activity (%)ity of the figure (this is the case for Midi R2[H10A, R12A] on NaV1.4). Other specificy error bars); each experiment was performed at least three times (n ≥ 3).

255

256

257

258

ING Model

P

6 eptide

tobhmltvNRtNws

ipRo

bm[KvNtfisowantmidN

tiaotoaas

iacaAtpLtfeI

Ras

259

260

261

262

263

264

265

266

267

268

269

270

271

272

273

274

275

276

277

278

279

280

281

282

283

284

285

286

287

288

289

290

291

292

293

294

295

296

297

298

299

300

301

302

303

304

305

306

307

308

309

310

311

312

313

314

315

316

317

318

319

320

321

322

323

324

325

326

327

328

329

330

331

332

333

334

335

336

337

338

339

340

341

342

343

344

345

346

347

348

349

350

351

352

353

354

355

356

357

358

359

360

361

362

363

364

365

366

367

368

369

370

371

372

373

374

375

376

377

378

379

380

381

382

383

384

ARTICLEEP 69015 1–7

E.K.M. Lebbe et al. / P

The first mutant we created was Midi R2[A7G]. The purpose ofhis mutation was to investigate the minimal spacing of the sec-nd intercysteine loop. As Gly is an amino acid comparable to Ala,ut without a –CH3 group, we wanted to see if a smaller peptideolds or loses its activity. Our observations suggest that the smallerutant, Midi R2[A7G], is not only less active and potent, but it

oses also its selectivity. The IC50 value for NaV1.2 increased fromhe nanomolar to the micromolar range (34.1 ± 2.4 nM for Midi R2ersus 14.7 ± 3.2 �M for Midi R2[A7G]) as was also observed foraV1.4 (330 ± 570 nM for Midi R2 versus 15.2 ± 1.2 �M for Midi2[A7G]). Presumably, key residues are in a less favorable posi-ion because of the smaller spacing and thus interactions with theaV-channels are partially hindered. Further structural researchith site-directed mutagenesis or NMR or ideally co-crystallization

hould be conducted to verify if this is correct.From our observations, we can conclude that the minimal spac-

ng between the second and the third cysteine residues for aotent and selective inhibitor analog of Midi is not the sequenceRWGRDHSR, but RRWARDHSR, as CD measurements proved thatverall structural conformations did not change.

The second mutant, called Midi R2[H10A, R12A], has a dou-le mutation on positions 10 and 12. The idea behind these pointutations is based on the results from the work of McArthur et al.

24]. In their study, they described that the substitution H12A inIIIA severely reduced its affinity for NaV1.2 and NaV1.4 as IC50alues increased >2000-fold. In contrast, the increase in IC50 foraV1.7 was only 133-fold, which indicated that selectivity shifted

o NaV1.7. The substitution R14A in KIIIA also changed target speci-city for NaV1.7 over both NaV1.2 and NaV1.4. Unfortunately, bothubstitutions were found to substantially reduce KIIIA affinity. Inur study, we wanted to test if double substitutions [R10A, H12A]ould give an explicit shift toward NaV1.7 selectivity. The results

re dissimilar with what was suggested by McArthur et al., butonetheless very interesting. With the double mutant as toxin pep-ide we could not block any of the tested NaV-subtypes whereas CD

easurements proved that overall structural conformations stayedntact. Thus, compared to Midi R2, potency and activity decreasedramatically rendering Midi R2[H10A, R12A] inactive to the testedaV-subtypes.

Other studies investigating these mutation sites in several pep-ides, like for instance the �-conotoxin SIIIA from Conus striatus,ndicated that His-16 replacement caused large reductions in SIIIAffinity. NMR measurements pointed out that this altered affinityccurred from structural changes in the C-terminal half of the pep-ide which potentially disrupted the alpha-helix. Consequently, therientation of the major binding determinants required for highffinity interactions at NaVs was disturbed. It was suggested that anrginine and tyrosine (but not Ala) can replace His-16 as a structuraltabilizer [30].

Replacement of Arg-18 was found to be important for the block-ng effect of SIIIA. This mutation contributes significantly to theffinity at NaV1.4, and to a lesser extent at NaV1.2. Hereupon theyoncluded that the pharmacophoric residues important for the highffinity of SIIIA at NaVs are located along the alpha-helical motif plusrg-18 [30]. Li et al. (2001) investigated Arg-19 substitution into

he �-conotoxin GIIIA from Conus geographus. They suggested thatositively charged toxin residues (like for instance Arg-1, Lys-11,ys-16 and Arg-19) are critical for its biological activity. In this way,he charges, as well as their precise location within the pore, wereound to be critical for toxin-channel interaction [22]. Choudharyt al. showed that Arg-19 of GIIIA strongly interacts with domainV of the NaV-channels [9].

Please cite this article in press as: Lebbe EKM, et al. Ala-7, His-10 and Arg�-conotoxin. Peptides (2013), http://dx.doi.org/10.1016/j.peptides.2013.0

These and our observations suggest that the C-terminus of Midi2 and other smaller �-conotoxins like KIIIA or BuIIIC consist of

crucial epitope where charged residues cannot be changed. Ourtudy on Midi R2[H10A, R12A] is new in the fact that the double

[

PRESSs xxx (2013) xxx– xxx

mutant has never been analyzed before. We expected that potencywould be less, but we hoped that we could find a significant increasein affinity to NaV1.7. This NaV-subtype is namely an important anal-gesic drug target because of its role in human as well as animalpain perception [11,28]. However, our findings support the devel-opment of peptides with novel subtype specificity and analgesicpotential.

The importance of testing cockroach B. germanica BgNaV1.1 canbe seen in the world of the agro industry and the application ofinsecticides because voltage gated sodium channels are well rec-ognized targets in the development of novel insecticides [32].

5. Conclusion

In this study we investigated the influence of point mutationsto the potency and activity – i.e. the blocking effect on NaVs – ofa �-conotoxin, named Midi R2. As stated in previous research, anArg on position 2 (peptide Midi R2), leads to lower IC50 values andexhibits a very potent and selective block on NaV1.2 [34]. Whenthe Ala on position 7 in Midi R2 is mutated to a Gly, the IC50 valueincreases from the nanomolar range to the micromolar range. Thisindicates that for a potent and specific peptide, the minimal spacebetween the second and the third cysteine residues is the sequenceRRWARDHSR. For the double mutant, Midi R2[H10A, R12A], whereHis (position 10) and Arg (position 12) are mutated to Ala, the activ-ity of the peptide (measured at 10 �M) is even reduced to 0–10% forall NaV-subtypes. Circular dichroism measurements indicated thatstructural conformations of the mutants did not change. The muta-tions we made are thus critical for the function of the peptide. Thesefindings are important in the future engineering of �-conotoxin-based lead compounds in the development of novel therapeuticsthat can be used to treat NaV-related diseases such as epilepsy [38]and pain [15,37].

Acknowledgements

The authors would like to thank Beatrice Garcia Mille forher work in molecular biology. This work was supported by thefollowing grants: G.0433.12, G.A071.10N and G.0257.08 (F.W.O.Vlaanderen), EU-FP7-MAREX, IUAP 7/10 (Inter-University Attrac-tion Poles Program, Belgian State, Belgian Science Policy) andOT/12/081 (KU Leuven). W. Brullot received funding from theAgency for Innovation by Science and Technology (IWT) Flanders.

References

[1] Armishaw CJ, Alewood PF. Conotoxins as research tools and drug leads. CurrProtein Pept Sci 2005;6:221–40.

[2] Basus VJ, Nadasdi L, Ramachandran J, Miljanich GP. Solution structure of omega-conotoxin MVIIA using 2D NMR spectroscopy. FEBS Lett 1995;370:163–9.

[3] Becker S, Terlau H. Toxins from cone snails: properties, applications andbiotechnological production. Appl Microbiol Biotechnol 2008;79:1–9.

[4] Buczek O, Wei D, Babon JJ, Yang X, Fiedler B, Chen P, et al. Structure andsodium channel activity of an excitatory I1-superfamily conotoxin. Biochem-istry 2007;46:9929–40.

[5] Bulaj G, West PJ, Garrett JE, Watkins M, Zhang MM, Norton RS, et al. Novel cono-toxins from Conus striatus and Conus kinoshitai selectively block TTX-resistantsodium channels. Biochemistry 2005;44:7259–65.

[6] Campuzano O, Beltran-Alvarez P, Iglesias A, Scornik F, Perez G, Brugada R.Genetics and cardiac channelopathies. Genet Med 2010;12:260–7.

[7] Catterall WA. From ionic currents to molecular mechanisms: the structure andfunction of voltage-gated sodium channels. Neuron 2000;26:13–25.

[8] Catterall WA, Goldin AL, Waxman SG. International Union of PharmacologyXLVII. Nomenclature and structure–function relationships of voltage-gatedsodium channels. Pharmacol Rev 2005;57:397–409.

[9] Choudhary G, Aliste MP, Tieleman DP, French RJ, Dudley Jr SC. Docking of

-12 are crucial amino acids for activity of a synthetically engineered7.005

mu-conotoxin GIIIA in the sodium channel outer vestibule. Channels (Austin)2007;1:344–52.

10] Cruz LJ, Gray WR, Olivera BM, Zeikus RD, Kerr L, Yoshikami D, et al. Conusgeographus toxins that discriminate between neuronal and muscle sodiumchannels. J Biol Chem 1985;260:9280–8.

385

386

387

388

389

ING Model

P

eptide

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[ture/function characterization of micro-conotoxin KIIIA, an analgesic, nearly

390

391

392

393

394

395

396

397

398

399

400

401

402

403

404

405

406

407

408

409

410

411

412

413

414

415

416

417

418

419

420

421

422

423

424

425

426

427

428

429

430

431

432

433

434

435

436

437

438

439

440

441

442

443

444

445

446

447

448

449

450

451

452

453

454

455

456

457

458

459

460

461

ARTICLEEP 69015 1–7

E.K.M. Lebbe et al. / P

11] Eijkelkamp N, Linley JE, Baker MD, Minett MS, Cregg R, Werdehausen R,et al. Neurological perspectives on voltage-gated sodium channels. Brain2012;135:2585–612.

12] Escayg A, Goldin AL. Sodium channel SCN1A and epilepsy: mutations and mech-anisms. Epilepsia 2010;51:1650–8.

13] Fiedler B, Zhang MM, Buczek O, Azam L, Bulaj G, Norton RS, et al. Specificity,affinity and efficacy of iota-conotoxin RXIA, an agonist of voltage-gated sodiumchannels Na(V)1.2, 1.6 and 1.7. Biochem Pharmacol 2008;75:2334–44.

14] Han TS, Teichert RW, Olivera BM, Bulaj G. Conus venoms – a rich source ofpeptide-based therapeutics. Curr Pharm Des 2008;14:2462–79.

15] Han TS, Zhang MM, Walewska A, Gruszczynski P, Robertson CR, Cheatham3rd TE, et al. Structurally minimized mu-conotoxin analogues as sodiumchannel blockers: implications for designing conopeptide-based therapeutics.ChemMedChem 2009;4:406–14.

16] Hille B, Armstrong CM, MacKinnon R. Ion channels: from idea to reality. NatMed 1999;5:1105–9.

17] Holford M, Zhang MM, Gowd KH, Azam L, Green BR, Watkins M, et al. Prun-ing nature: biodiversity-derived discovery of novel sodium channel blockingconotoxins from Conus bullatus. Toxicon 2009;53:90–8.

18] Hui K, Lipkind G, Fozzard HA, French RJ. Electrostatic and steric contributionsto block of the skeletal muscle sodium channel by �-conotoxin. J Gen Physiol2002;119:45–54.

19] Khoo KK, Feng ZP, Smith BJ, Zhang MM, Yoshikami D, Olivera BM, et al. Structureof the analgesic �-conotoxin KIIIA and effects on the structure and function ofdisulfide deletion. Biochemistry 2009;48:1210–9.

20] Knapp O, McArthur JR, Adams DJ. Conotoxins targeting neuronal voltage-gated sodium channel subtypes: potential analgesics? Toxins (Basel)2012;4:1236–60.

21] Lewis RJ, Dutertre S, Vetter I, Christie MJ. Conus venom peptide pharmacology.Pharmacol Rev 2012;64:259–98.

22] Li RA, Ennis IL, French RJ, Dudley Jr SC, Tomaselli GF, Marban E. Clockwisedomain arrangement of the sodium channel revealed by (�)-conotoxin (GIIIA)docking orientation. J Biol Chem 2001;276:11072–7.

23] Mantegazza M, Curia G, Biagini G, Ragsdale DS, Avoli M. Voltage-gated sodium

Please cite this article in press as: Lebbe EKM, et al. Ala-7, His-10 and Arg�-conotoxin. Peptides (2013), http://dx.doi.org/10.1016/j.peptides.2013.07

channels as therapeutic targets in epilepsy and other neurological disorders.Lancet Neurol 2010;9:413–24.

24] McArthur JR, Singh G, McMaster D, Winkfein R, Tieleman DP, French RJ. Interac-tions of key charged residues contributing to selective block of neuronal sodiumchannels by �-conotoxin KIIIA. Mol Pharmacol 2011;80:573–84.

[

PRESSs xxx (2013) xxx– xxx 7

25] McIntosh JM, Hasson A, Spira ME, Gray WR, Li W, Marsh M, et al. A newfamily of conotoxins that blocks voltage-gated sodium channels. J Biol Chem1995;270:16796–802.

26] Menez A, Stocklin R, Mebs D. ‘Venomics’ or: the venomous systems genomeproject. Toxicon 2006;47:255–9.

27] Miljanich GP. Ziconotide: neuronal calcium channel blocker for treating severechronic pain. Curr Med Chem 2004;11:3029–40.

28] Nassar MA, Stirling LC, Forlani G, Baker MD, Matthews EA, Dickenson AH, et al.Nociceptor-specific gene deletion reveals a major role for Nav1.7 (PN1) in acuteand inflammatory pain. Proc Natl Acad Sci USA 2004;101:12706–11.

29] Olivera BM, Rivier J, Clark C, Ramilo CA, Corpuz GP, Abogadie FC, et al. Diversityof Conus neuropeptides. Science 1990;249:257–63.

30] Schroeder CI, Ekberg J, Nielsen KJ, Adams D, Loughnan ML, Thomas L, et al.Neuronally micro-conotoxins from Conus striatus utilize an alpha-helical motifto target mammalian sodium channels. J Biol Chem 2008;283:21621–8.

31] Shon KJ, Grilley MM, Marsh M, Yoshikami D, Hall AR, Kurz B, et al. Purifica-tion, characterization, synthesis, and cloning of the lockjaw peptide from Conuspurpurascens venom. Biochemistry 1995;34:4913–8.

32] Smith JJ, Herzig V, King GF, Alewood PF. The insecticidal potential of venompeptides. Cell Mol Life Sci 2013. Doi: 10.1007/s00018-013-1315-3.

33] Spence I, Gillessen D, Gregson RP, Quinn RJ. Characterization of the neurotoxicconstituents of Conus geographus (L.) venom. Life Sci 1977;21:1759–69.

34] Stevens M, Peigneur S, Dyubankova N, Lescrinier E, Herdewijn P, Tytgat J. Designof bioactive peptides from naturally occurring �-conotoxin structures. J BiolChem 2012;287:31382–92.

35] Van Der Haegen A, Peigneur S, Tytgat J. Importance of position 8 in �-conotoxin KIIIA for voltage-gated sodium channel selectivity. FEBS J 2011;278:3408–18.

36] Wilson MJ, Yoshikami D, Azam L, Gajewiak J, Olivera BM, Bulaj G, et al.�-Conotoxins that differentially block sodium channels NaV1.1 through 1.8identify those responsible for action potentials in sciatic nerve. Proc Natl AcadSci USA 2011;108:10302–7.

37] Zhang MM, Green BR, Catlin P, Fiedler B, Azam L, Chadwick A, et al. Struc-

-12 are crucial amino acids for activity of a synthetically engineered.005

irreversible blocker of mammalian neuronal sodium channels. J Biol Chem2007;282:30699–706.

38] Zuliani V, Rivara M, Fantini M, Costantino G. Sodium channel blockers for neu-ropathic pain. Expert Opin Ther Pat 2010;20:755–79.

462

463

464

465