commonmoleculardeterminantsoftarantulahuwentoxin-iv ... · the ability of the tarantula...

11
Common Molecular Determinants of Tarantula Huwentoxin-IV Inhibition of Na Channel Voltage Sensors in Domains II and IV * S Received for publication, April 4, 2011, and in revised form, May 9, 2011 Published, JBC Papers in Press, June 9, 2011, DOI 10.1074/jbc.M111.246876 Yucheng Xiao ‡§ , James O. Jackson II § , Songping Liang §1 , and Theodore R. Cummins ‡2 From the Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202 and the § Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China The voltage sensors of domains II and IV of sodium channels are important determinants of activation and inactivation, respectively. Animal toxins that alter electrophysiological excit- ability of muscles and neurons often modify sodium channel activation by selectively interacting with domain II and inacti- vation by selectively interacting with domain IV. This suggests that there may be substantial differences between the toxin- binding sites in these two important domains. Here we explore the ability of the tarantula huwentoxin-IV (HWTX-IV) to inhibit the activity of the domain II and IV voltage sensors. HWTX-IV is specific for domain II, and we identify five residues in the S1–S2 (Glu-753) and S3–S4 (Glu-811, Leu-814, Asp-816, and Glu-818) regions of domain II that are crucial for inhibition of activation by HWTX-IV. These data indicate that a single residue in the S3–S4 linker (Glu-818 in hNav1.7) is crucial for allowing HWTX-IV to interact with the other key residues and trap the voltage sensor in the closed configuration. Mutagenesis analysis indicates that the five corresponding residues in domain IV are all critical for endowing HWTX-IV with the ability to inhibit fast inactivation. Our data suggest that the tox- in-binding motif in domain II is conserved in domain IV. Increasing our understanding of the molecular determinants of toxin interactions with voltage-gated sodium channels may per- mit development of enhanced isoform-specific voltage-gating modifiers. Voltage-gated sodium channels (VGSCs) 3 play critical roles in the generation and propagation of action potentials. Nine VGSC subunit subtypes (Nav1.1–1.9) have been cloned and characterized from mammals (1). These subtypes are expressed in different excitable tissues and are involved in distinct physi- ological functions such as neurotransmitter release, muscle contraction, secretion, and pain sensation (2– 4). The nine subunits share a common four-domain (DI–DIV) structure, in which each domain has six transmembrane segments (S1–S6) (1). Based on distinct functional behaviors during channel activity, the six transmembrane segments are generally sepa- rated into two structural components (5). The central pore module is the basis for the ion conduction pathway and is formed by the S5–S6 region of the channel. The voltage sensor modules are formed by the S1–S4 regions and are essentially independent structures that directly respond to changes in the transmembrane potential with conformational alterations that are coupled to opening and closing of the pore module. VGSCs undergo voltage-dependent activation subsequently followed by fast inactivation through successive activities of the voltage sensors in the four domains. The S4 segments in the voltage sensors of DI, DII, and DIII are determinants of channel activa- tion, whereas that of DIV is predominantly involved in channel inactivation (5– 8). The four membrane-spanning segments (S1–S4) of the voltage sensors of mammal VGSCs exhibit high sequence similarity in the four domains, but the amino acids and sequence length of the intracellular and extracellular link- ers are quite divergent. Because crystal structures of sodium channels are lacking, it remains unknown whether the voltage sensor differences result in altered conformations of the mem- brane-spanning segments or whether the orientations of amino acid side chains are structurally conserved at corresponding positions in the four different voltage sensors. Because of their high binding affinity and subtype-specific selectivity, animal toxins are powerful tools for investigation of the structure-function relationship of VGSCs (9). The voltage sensors of DII and DIV are the two most common toxin-bind- ing sites in VGSCs (see Fig. 1A). Scorpion -toxins bind to the DII voltage sensor to enhance voltage-dependent activation by trapping DIIS4 in the activated state (10 –12). Scorpion -tox- ins bind to the DIV voltage sensor and inhibit fast inactivation by trapping DIVS4 in the closed state (13, 14). These findings seem to suggest that there are substantial differences between the toxin-binding sites on the voltage sensors of DII and DIV. However, we recently demonstrated that ProTx-II, another tarantula toxin, can both inhibit hNav1.7 activation by interact- * This work was supported, in whole or in part, by National Institutes of Health Grants NS054642 and NS053422 (to T. R. C.). This work was also supported by the 973 Research Program of China under Contract 2010CB529800 (to S. L.); the Scientific Research Fund of Hunan Provincial Education Depart- ment (07A035) and the Program for New Century Excellent Talents in Uni- versity under Contract NCET-07-0279 (to Y. X.). S The on-line version of this article (available at http://www.jbc.org) contains supplemental Tables S1–S4 and Figs. S1 and S2. 1 To whom correspondence may be addressed: Life Sciences College, Hunan Normal University, Changsha, Hunan 410081, China. E-mail: [email protected]. 2 To whom correspondence may be addressed: 950 W. Walnut St, R2-Rm. 468, Dept. of Pharmacology and Toxicology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202. E-mail: [email protected]. 3 The abbreviations used are: VGSC, voltage-gated sodium channel; DI, DII, DIII, and DIV, domains I, II, III and IV; HWTX-IV, huwentoxin-IV; Kv, voltage gated potassium channel; VSD, voltage sensing domain; Sn, transmem- brane segment n. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 286, NO. 31, pp. 27301–27310, August 5, 2011 © 2011 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A. AUGUST 5, 2011 • VOLUME 286 • NUMBER 31 JOURNAL OF BIOLOGICAL CHEMISTRY 27301 by guest on December 28, 2019 http://www.jbc.org/ Downloaded from

Upload: others

Post on 07-Sep-2019

5 views

Category:

Documents


0 download

TRANSCRIPT

Common Molecular Determinants of Tarantula Huwentoxin-IVInhibition of Na� Channel Voltage Sensors in Domains IIand IV*□S

Received for publication, April 4, 2011, and in revised form, May 9, 2011 Published, JBC Papers in Press, June 9, 2011, DOI 10.1074/jbc.M111.246876

Yucheng Xiao‡§, James O. Jackson II§, Songping Liang§1, and Theodore R. Cummins‡2

From the ‡Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202 andthe §Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, College of Life Sciences, HunanNormal University, Changsha, Hunan 410081, China

The voltage sensors of domains II and IV of sodium channelsare important determinants of activation and inactivation,respectively. Animal toxins that alter electrophysiological excit-ability of muscles and neurons often modify sodium channelactivation by selectively interacting with domain II and inacti-vation by selectively interacting with domain IV. This suggeststhat there may be substantial differences between the toxin-binding sites in these two important domains. Here we explorethe ability of the tarantula huwentoxin-IV (HWTX-IV) toinhibit the activity of the domain II and IV voltage sensors.HWTX-IV is specific for domain II, andwe identify five residuesin the S1–S2 (Glu-753) and S3–S4 (Glu-811, Leu-814, Asp-816,andGlu-818) regions of domain II that are crucial for inhibitionof activation by HWTX-IV. These data indicate that a singleresidue in the S3–S4 linker (Glu-818 in hNav1.7) is crucial forallowing HWTX-IV to interact with the other key residues andtrap the voltage sensor in the closed configuration.Mutagenesisanalysis indicates that the five corresponding residues indomain IV are all critical for endowing HWTX-IV with theability to inhibit fast inactivation. Our data suggest that the tox-in-binding motif in domain II is conserved in domain IV.Increasing our understanding of the molecular determinants oftoxin interactions with voltage-gated sodium channelsmay per-mit development of enhanced isoform-specific voltage-gatingmodifiers.

Voltage-gated sodium channels (VGSCs)3 play critical rolesin the generation and propagation of action potentials. Nine

VGSC � subunit subtypes (Nav1.1–1.9) have been cloned andcharacterized frommammals (1). These subtypes are expressedin different excitable tissues and are involved in distinct physi-ological functions such as neurotransmitter release, musclecontraction, secretion, and pain sensation (2–4). The nine �subunits share a common four-domain (DI–DIV) structure, inwhich each domain has six transmembrane segments (S1–S6)(1). Based on distinct functional behaviors during channelactivity, the six transmembrane segments are generally sepa-rated into two structural components (5). The central poremodule is the basis for the ion conduction pathway and isformed by the S5–S6 region of the channel. The voltage sensormodules are formed by the S1–S4 regions and are essentiallyindependent structures that directly respond to changes in thetransmembrane potential with conformational alterations thatare coupled to opening and closing of the pore module. VGSCsundergo voltage-dependent activation subsequently followedby fast inactivation through successive activities of the voltagesensors in the four domains. The S4 segments in the voltagesensors of DI, DII, and DIII are determinants of channel activa-tion, whereas that of DIV is predominantly involved in channelinactivation (5–8). The four membrane-spanning segments(S1–S4) of the voltage sensors of mammal VGSCs exhibit highsequence similarity in the four domains, but the amino acidsand sequence length of the intracellular and extracellular link-ers are quite divergent. Because crystal structures of sodiumchannels are lacking, it remains unknown whether the voltagesensor differences result in altered conformations of the mem-brane-spanning segments or whether the orientations of aminoacid side chains are structurally conserved at correspondingpositions in the four different voltage sensors.Because of their high binding affinity and subtype-specific

selectivity, animal toxins are powerful tools for investigation ofthe structure-function relationship of VGSCs (9). The voltagesensors of DII and DIV are the two most common toxin-bind-ing sites in VGSCs (see Fig. 1A). Scorpion �-toxins bind to theDII voltage sensor to enhance voltage-dependent activation bytrapping DIIS4 in the activated state (10–12). Scorpion �-tox-ins bind to the DIV voltage sensor and inhibit fast inactivationby trapping DIVS4 in the closed state (13, 14). These findingsseem to suggest that there are substantial differences betweenthe toxin-binding sites on the voltage sensors of DII and DIV.However, we recently demonstrated that ProTx-II, anothertarantula toxin, can both inhibit hNav1.7 activation by interact-

* This work was supported, in whole or in part, by National Institutes of HealthGrants NS054642 and NS053422 (to T. R. C.). This work was also supportedby the 973 Research Program of China under Contract 2010CB529800 (toS. L.); the Scientific Research Fund of Hunan Provincial Education Depart-ment (07A035) and the Program for New Century Excellent Talents in Uni-versity under Contract NCET-07-0279 (to Y. X.).

□S The on-line version of this article (available at http://www.jbc.org) containssupplemental Tables S1–S4 and Figs. S1 and S2.

1 To whom correspondence may be addressed: Life Sciences College,Hunan Normal University, Changsha, Hunan 410081, China. E-mail:[email protected].

2 To whom correspondence may be addressed: 950 W. Walnut St, R2-Rm. 468,Dept. of Pharmacology and Toxicology, Stark Neurosciences ResearchInstitute, Indiana University School of Medicine, Indianapolis, IN 46202.E-mail: [email protected].

3 The abbreviations used are: VGSC, voltage-gated sodium channel; DI, DII,DIII, and DIV, domains I, II, III and IV; HWTX-IV, huwentoxin-IV; Kv, voltagegated potassium channel; VSD, voltage sensing domain; Sn, transmem-brane segment n.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 286, NO. 31, pp. 27301–27310, August 5, 2011© 2011 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.

AUGUST 5, 2011 • VOLUME 286 • NUMBER 31 JOURNAL OF BIOLOGICAL CHEMISTRY 27301

by guest on Decem

ber 28, 2019http://w

ww

.jbc.org/D

ownloaded from

ing with DII and impair hNav1.7 inactivation by interactingwith DIV (15). This raises the possibility that there might beconserved molecular determinants of toxin binding in the DIIand DIV voltage sensors.Here we used electrophysiology and point mutagenesis to

extensively explore the molecular determinants involved inhuwentoxin-IV (HWTX-IV) interactions with hNav1.7.HWTX-IV is a voltage sensormodifier from the tarantulaOrni-thoctonus huwena that interacts with the DII voltage sensor.However, in contrast to scorpion �-toxins, HWTX-IV trapsDIIS4 in the closed state and specifically inhibits channel acti-vation (16, 17). Our results indicate that HWTX-IV partiallyshares the binding site on DII voltage sensor with scorpion�-toxin CssIV, although these two toxins trap DIIS4 in distinctstates. We determined that interaction with Glu-818 is key forHWTX-IV to access other important residues in theDII voltagesensor. Interestingly, we find that each of the five residues thatwe identified in the voltage sensor of DII as being critical for theinteraction with HWTX-IV also regulate the ability of the DIVvoltage sensor to interact with HWTX-IV.

EXPERIMENTAL PROCEDURES

Molecular Biology—All of the hNav1.7 mutations were con-structed using theQuikChange II XL site-directedmutagenesiskit according to themanufacturer’s instruction. The constructswere sequenced to confirm that the appropriate mutationswere made.Electrophysiological Recording—WT and mutant hNav1.7

channels were transiently transfected into HEK293 cells usingthe standard calcium phosphate precipitationmethod as previ-ously described (15).Whole cell patch clamp recordings were carried out at room

temperature (�21 °C) using an EPC-10 amplifier (HEKA, Lam-brecht, Germany). Fire-polished electrodes were fabricatedfrom 1.7-mm capillary glass (VWR, West Chester, PA) using aP-97 puller (Sutter, Novato, CA). The standard pipette solutioncontained 140 mM CsF, 1 mM EGTA, 10 mM NaCl, and 10 mM

HEPES, pH 7.3. The standard bathing solution was 140 mM

NaCl, 3 mM KCl, 1 mMMgCl2, 1 mM CaCl2, and 10mMHEPES,pH 7.3. After filling with pipette solution, the access resistanceof electrode pipette ranged from 0.7 to 1.3 M�. The liquid junc-tion potential for these solutions was �8 mV; data were notcorrected to account for this offset. Voltage errors were mini-mized using 80% series resistance compensation, and thecapacitance artifact was canceled using the computer-con-trolled circuitry of the patch clamp amplifier. Linear leak sub-traction, based on resistance estimates from four to five hyper-polarizing pulses applied before the depolarizing test potential,was used for all voltage clamp recordings. Membrane currentswere usually filtered at 5 kHz and sampled at 20 kHz.The stock solutions for 50 �M CssIV and 1 mM HWTX-IV

weremade using bathing solution containing 1mg/mlBSA, andaliquots were stored at �20 °C. Before use, the solution wasdiluted to the concentrations of interest with fresh bathingsolution. Toxin was diluted into the recording chamber (vol-ume of 300 �l) and mixed by repeatedly pipetting 30 �l toachieve the specified final concentration. The extent of the

inhibitory effect of the toxin was typically assessed around 20min after toxin treatment.HomologyModeling—Three-dimensionalmodels of hNav1.7

DII-VSD (Phe-728 to Lys-846) were created based on the com-plete closed state and open state structures of the mammalianKv1.2 (Ser-158 to Ile-316) provided by Pathak et al. (18).Sequence alignment was conducted using the ClusterW1.8program, and the alignment of the four transmembrane seg-ments (S1–S4) was then refinedmanually. Homologymodelingwas performed using Accelrys Discovery Studio 1.6 software(Accelrys, San Diego, CA). The models were further refined byenergy “minimization” protocol using a CHARMm force field,in which both the minimization algorithm “steepest descent”and “conjugated gradient” were set to 500 steps.Data Analysis—The data were analyzed using the Pulsefit

(HEKA) and GraphPad Prism 4 (GraphPad Software) pro-grams. All of the data points are shown as the means � S.E. n ispresented as the number of experimental cells. Statistical anal-ysis was carried out by Student’s t test, and p � 0.05 indicated asignificant difference. Steady-state activation and inactivationcurves were fitted using the Boltzmann equation: y � 1/(1 �exp((V1⁄2 � V)/k), in which V1⁄2, V, and k represent midpointvoltage of kinetics, test potential, and slope factor, respectively.Dose-response curves to determine IC50 values were fittedusing the Hill equation: y� 1/(1� exp((logIC50 �X)nH), whereX is the toxin dose, nH is the Hill coefficient, and IC50 is thehalf-maximal inhibitory concentration. In this study, the nHwas set to 1 because our mutagenesis data have shown that thetoxin had a single high affinity binding site in sodium channels.For HWTX-IV action on fast inactivation of WT and mutantNav1.7, the nH was also set to 1 because only sodium channelDIV is involved in channel inactivation gating.

RESULTS

Electrophysiological Properties of WT and Mutant Nav1.7Channels—To explore hNav1.7-HWTX-IV interactions andidentify the crucial residues in hNav1.7, we mutated each resi-due in the extracellular portions of the DII S1–S2 and S3–S4regions. Mutations were principally designed using the follow-ing rules: 1) charged residues were mutated to be neutral, 2)unique residues in the spider VGSC (19), which is presumablyresistant to HWTX-IV, were introduced into hNav1.7, and 3) ifan uncharged residue is conserved in hNav1.7 and the spiderVGSC, it was substituted by a smaller side chain residue, eitherAla or Cys (Fig. 1).We first characterized the electrophysiological properties of

WT and mutant Nav1.7 channels, expressed in HEK293 cells,using the whole cell recording configuration and compared thevoltage-dependent properties of steady-state activation andinactivation to WT Nav1.7. Consistent with previous reportsthat the voltage sensor in DII is an important determinant ofchannel activation (7, 20), mutations of most amino acid resi-dues significantly altered channel activation (Fig. 2A and sup-plemental Table S1). In particular, both hydrophilic and hydro-phobic residues in the DIIS4 segment are involved in favoringhNav1.7 in the closed state. Positively charged residues (typi-cally Arg) embedded within the S4 segment are primary con-tributors to gating charge and voltage sensitivity of ion channels

Conserved Toxin-binding Motif in Na� Channel Voltage Sensors

27302 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 • NUMBER 31 • AUGUST 5, 2011

by guest on Decem

ber 28, 2019http://w

ww

.jbc.org/D

ownloaded from

(21, 22). Among themutations in the voltage sensor of DII, onlyE753Q substantially shifted the voltage dependence of steady-state inactivation (by �17.3 mV; Fig. 2B and supplementalTable S1).Critical Residues in hNav1.7 DII Interacting with HWTX-IV—

Inour previouswork, we demonstrated that an acidic residue inthe extracellular DIIS3–S4 region (Glu-818) is critical for inter-

acting with HWTX-IV. The mutation of Glu-818 to Gln andCys reduced toxin binding affinity for hNav1.7 by 63- and 400-fold, respectively (15, 16). Because the DII voltage sensor inVGSCs is a mobile structure, we hypothesized that interactionwith onlyGlu-818would not be sufficient forHWTX-IV to traptheDII voltage sensor in the closed state and that other residuesmust be involved in the toxin-channel interaction that under-lies channel inhibition. In the current study, we found thatmutation of four additional amino acid residues (Glu-753 in theDIIS1-S2 region and Glu-811, Leu-814, and Asp-816 in theDIIS3–S4 region) produced substantial decreases inHWTX-IVinhibition of hNav1.7 activation (Fig. 3). Fig. 3A shows typicalcurrent traces of WT and these four mutant Nav1.7 channelselicited by a 20-ms depolarization of �10 mV from a holdingpotential of�100mV. As reported previously, 1�MHWTX-IVcompletely blocksWTNav1.7 (16). However, in the presence of1 �MHWTX-IV, the amount of current that still could be acti-vated ranged from 6.2 � 1.3 to 31.1 � 2.1% of control for thesefour mutant channels (n � 4–8). The IC50 values forHWTX-IV blocking WT and all 39 mutant hNav1.7 channelstested are summarized in Fig. 3B. These data indicate that theE753Q, E811Q, L814C, and D816A mutations reduced toxinbinding affinity by 5-, 18-, 9-, and 13-fold, respectively (Fig. 3B).Although three mutations (V817A, G819P, and S825A) slightlyincreasedHWTX-IV sensitivity for hNav1.7, the IC50 values forthese mutations were only �2-fold smaller than that for WTchannels.Seven of the eight mutations that altered HWTX-IV inhibi-

tion (with V817A being the exception) significantly shifted thevoltage dependence of activation of hNav1.7 to more positivepotentials (Fig. 2B). However, it seems unlikely that changes in

FIGURE 1. Amino acid sequence alignment of the DIIS1–S2 and S3–S4linker regions of VGSC � subunit subtypes. A, schematic diagram ofsodium channel � subunit. The voltage sensor (segment 4) of each domain isshaded in gray and marked with ��. B, amino acid sequence alignment ofDIIS1–S2 and S3–S4 regions of eight VGSC subtypes. The subtypes Nav1.1,Nav1.2, Nav1.3, and Nav1.4 are from rat (r). The subtypes Nav1.5, Nav1.6, andNav1.7 are from human (h). The subtype sNav is a spider (s) VGSC cloned fromthe tarantula O. huwena (17). Compared with hNav1.7, the identical aminoacid residues are marked with a dot (.) in other VGSC subtypes. The five aminoacid residues of interest identified in this study are shaded in gray.

FIGURE 2. Functional effects of mutations of amino acid residues in hNav1.7 DII-VSD. WT and mutant channels are expressed in HEK293 cells. A, effects ofmutations of amino acid residues in DII-VSD on hNav1.7 activation. The cells were held at �100 mV. To assess the voltage dependence of activation, familiesof currents were induced by 50-ms depolarizing steps to various potential ranging from �80 to �40 mV. Channel conductances were calculated with theequation: G(V) � I/(V � Vrev), in which I, V, and Vrev represented inward, test potential, and reversal potential, respectively. B, effects of mutations of amino acidresidues in hNav1.7 DII-VSD on steady-state inactivation. The cells were held at �100 mV. The voltage dependence of steady-state inactivation was estimatedusing a standard double-pulse protocol, in which a 20-ms depolarizing test potential of 0 mV followed a 500-ms prepulse at potentials that ranged from �130to �10 mV with a 10-mV increment. The data points are shown as the means � S.E. The half-activation potential (V1⁄2) and slope factor (k) were determined withBoltzmann fits. In both A and B, V1⁄2 values of channel activation were yielded by the fit of the Boltzmann equation as indicated under “ExperimentalProcedures.” The original data were summarized in supplemental Table S1. *, p � 0.05; **, p � 0.001 significance between WT and mutant channels.

Conserved Toxin-binding Motif in Na� Channel Voltage Sensors

AUGUST 5, 2011 • VOLUME 286 • NUMBER 31 JOURNAL OF BIOLOGICAL CHEMISTRY 27303

by guest on Decem

ber 28, 2019http://w

ww

.jbc.org/D

ownloaded from

HWTX-IV binding affinity result from altering the voltage-de-pendent gating of the mutant channels, because there was nodirect correlation between changes in the voltage dependenceof activation and changes in sensitivity to HWTX-IV when allof the mutations are considered. We also examined whetherexposure to HWTX-IV had obvious effects on the voltage-de-pendent properties of the various mutant channels. For WTNav1.7 and the majority of mutant channels, in the presence of0.1 (or 1) �M HWTX-IV, there was no apparent change in thevoltage dependence of activation and inactivation (supplemen-tal Fig. S1 and Tables S2 and S3). This lack of an effect mostlikely reflects that in the presence of HWTX-IV, we are mainlymeasuring the activity of residual currents from channels thatdid not interact with HWTX-IV. This is consistent with ourprevious data showing that in the presence of saturating con-centrations of HWTX-IV, no Nav1.7 current can be detectedunless the channels are depolarized beyond �100 mV, reflect-ing a depolarizing shift of �200 mV in the voltage dependenceof activation of hNav1.7 by HWTX-IV (16). However, for threemutations (R827Q/C and L828A) in DIIS4, 0.1 �M HWTX-IVnot only shifted the voltage dependence of channel activation(by �7.2, �8.7, and �6.2 mV, respectively), but it also doubledthe slope factors for the voltage dependence of channel activa-tion (Fig. 4 and supplemental Table S2). HWTX-IV did notaffect inactivation properties of either WT or these mutantchannels (supplemental Table S3). None of these mutations(R827Q/C or L828A) altered the IC50 for HWTX-IV (Fig. 3B),indicating that neitherArg-827 nor Leu-828 is directly involvedin the toxin-channel interaction. Therefore, these three muta-tions most likely enhance the ability of hNav1.7 channels toactivate with HWTX-IV bound by altering the stability of theclosed configuration of the DII voltage sensor.

HWTX-IVDoesNot Bind toWTNav1.4—Our previous stud-ies have shown thatWT rNav1.4 (the skeletal muscle VGSC) isresistant toHWTX-IV and thatmutation ofGln-657 in rNav1.4(which corresponds to Glu-818 in hNav1.7; see Fig. 1) to gluta-mate greatly increases toxin sensitivity of rNav1.4 (16). Becausethree of the other crucial residues that we identified in thisstudy are highly conserved in rNav1.4 and hNav1.7 (Fig. 1), wenext asked whether HWTX-IV still binds toWT rNav1.4, eventhough inhibition of current amplitude is not observed. To testthis hypothesis, we designed a simple competitive bindingexperiment with CssIV (11), a typical scorpion �-toxin. CssIVpartially shares the binding site on the DII voltage sensor ofVGSCs with HWTX-IV because mutations of three residues inrNav1.2a (Glu-779, Glu-837, and Leu-840, corresponding toGlu-753, Glu-811, and Leu-814 in hNav1.7, respectively; Fig. 1)have previously been shown to additively reduce CssIV bindingaffinity for rNav1.2a by 175-fold (10). However, in contrast toHWTX-IV, CssIV traps the DII voltage sensor in the activatedstate, thereby shifting the voltage dependence of VGSC activa-tion to more negative potentials. Therefore, we reasoned thatif HWTX-IV was able to “silently” bind to WT Nav1.4,HWTX-IV pretreatment would be expected to substantiallyinterfere with CssIV binding to rNav1.4. To test this, a triple-pulse protocol (described in the legend of Fig. 5) was used toassess the fraction of rNav1.4 channels modified by CssIV. Inthe absence of CssIV and HWTX-IV, the rNav1.4 channelswere closed at potentials less than�60mV, and no current wasrecorded by the 20-ms depolarizing pulse to �65 mV from aholding potential of �100 mV (Fig. 5A; n � 6). In the presenceof 0.5 �M CssIV, inward rNav1.4 current, resulting from thenegative shift of channel activation, was evidently induced atthe subthreshold potential (�65 mV), and the ratio of the cur-

FIGURE 3. HWTX-IV binding affinity for hNav1.7 channels. A, inhibition of hNav1.7 currents by 1 �M HWTX-IV. Current traces for WT and five mutant (E753Q,E811Q, L814C, D816A, and E818C) hNav1.7 channels before (filled circle) and after (open circle) toxin treatment were elicited by depolarization to �10 mV froma holding potential of �100 mV. B, IC50 values of HWTX-IV on WT and mutant hNav1.7 channels. IC50 values are yielded by the fit of Hill equation as describedunder “Experimental Procedures,” in which the slope factor was set to 1. The IC50 value for E818C was adapted from our previous work (15). *, p � 0.05; **, p �0.001 significant difference between WT and mutant channels.

Conserved Toxin-binding Motif in Na� Channel Voltage Sensors

27304 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 • NUMBER 31 • AUGUST 5, 2011

by guest on Decem

ber 28, 2019http://w

ww

.jbc.org/D

ownloaded from

rent elicited at �65 mV (I�65 mV) to that elicited at �10 mV(I�10 mV) was 0.035 � 0.003 (n � 3). This ratio was not alteredby additional application of 1 �M HWTX-IV (Fig. 5A, upperpanels), the concentration that blocks 77.8� 2.0%of theQ657Emutant rNav1.4 channels in our previous work (16). In Fig. 5A(lower panels), 1�MHWTX-IV by itself did not induce rNav1.4current at �65 mV, consistent with our previous observationon hNav1.7 (16). Additional application of 0.5 �M CssIV in thepresence ofHWTX-IV still induced inward current at�65mV,and the ratio of I�65 mV to I�10 mV(control) was 0.035 � 0.004(n � 3), which is identical to the value caused by CssIV withoutHWTX-IV (Fig. 5B). These results demonstrate thatHWTX-IV did not affect CssIV binding to WT rNav1.4, indi-

cating that HWTX-IV does not bind silently to rNav1.4 in DII.Together with our previous results on specific point mutationsin rNav1.4 (Q657E) and in hNav1.7 (E818Q/C) (15, 16), theseresults suggest that interaction with the acidic residue Glu atposition 818 in the DIIS3–S4 linker of hNav1.7 is not only thekey determinant of voltage sensor trapping and channel inhibi-tion but is also the key factor for allowing HWTX-IV to accessand interact with the other crucial residues.Molecular Determinants of HWTX-IV Binding Are Con-

served on DII and DIV—Scorpion toxins that target VGSCs aretypically classified as either �-toxins that specifically inhibitinactivation by selectively binding to the extracellular portionof the DIVS3-S4 region or �-toxins that specifically enhanceactivation by selectively binding to the extracellular portion ofthe DIIS3–S4 region (10–14). This indicates that the extracel-lular portion of the S3–S4 region of the DII and DIV voltagesensors, also referred to as the voltage sensor paddles, may bestructurally distinct. However, the tarantula toxin ProTx-II canreportedly interact with multiple voltage sensor paddles (23),and we have recently demonstrated that ProTx-II can inhibitboth activation and inactivation of Nav1.7 (15). This suggeststhat either ProTx-II is relatively promiscuous in its interactionswith the voltage sensors of sodium channels or that the DII andDIV voltage sensors are more similar than might be predictedbased on the specificity of scorpion toxins. Nav1.7 DIV is resis-tant to HWTX-IV because the toxin does not inhibit fast inac-tivation of either WT or E818C mutant channels (15, 16, 24)(Fig. 3A). However, sequence alignment indicates that three ofthe five critical residues in Nav1.7 DII are conserved in DIV(Fig. 6A). Although the other two critical residues, Glu-811 andGlu-818, are correspondingly replaced by Gly-1581 and Ile-1588, respectively (Fig. 6A), the four membrane-spanning seg-ments (S1–S4) in DII show relatively high sequence similarity(�66%) with those in DIV. Therefore, we next asked whetherthe molecular determinants of toxin binding on the Nav1.7 DIIvoltage sensor might be conserved in the DIV voltage sensor.To address this question, we constructed six Nav1.7 DIV con-

FIGURE 4. Effects of HWTX-IV on the voltage-dependent activation offour hNav1.7 mutants expressed in Hek293 cells. Four mutations of inter-est (R824Q (A), R827Q (B), L828A (C), and R830Q (D)) were located in the DIIS4segment. The cells were held at �100 mV. Families of currents before andafter 100 nM HWTX-IV treatment (left panels) were induced by 50-ms depolar-izing steps to various potential ranging from �80 to �100 mV. Channel con-ductance values before and after 100 nM HWTX-IV treatment (right panels)were calculated with the equation as described for Fig. 2A.

FIGURE 5. HWTX-IV did not affect the interaction of scorpion �-toxinCssIV with WT rNav1.4. A, representative rNav1.4 current traces were elic-ited by a triple-pulse protocol in which a 20-ms moderate depolarizing poten-tial of �10 mV was applied from a holding potential of �100 mV followed61.2 ms later by 1-ms strong conditioning depolarizing voltage of �70 mV,and then after a second period of 61.2 ms at �100 mV, a 20-ms depolarizingpotential of �65 mV was given. This protocol was applied every 5 s. Thecurrent traces elicited by the conditioning pulse were not shown. Upperpanel, rNav1.4 current (black trace) was pretreated with 500 nM CssIV (greentrace) and then applied with 1 �M HWTX-IV (red trace). Bottom panel, rNav1.4current (black trace) was pretreated with 1 �M HWTX-IV (green trace) and thenapplied with 500 nM CssIV (red trace). B, effects of HWTX-IV (or CssIV) on theratio of the current elicited at �65 mV (I�65 mV) to that elicited at �10 mV (I�10mV) in the absence and in the presence of CssIV (or HWTX-IV). n � 3– 6; errorbars, S.E. **, p � 0.001; N.S, no significance.

Conserved Toxin-binding Motif in Na� Channel Voltage Sensors

AUGUST 5, 2011 • VOLUME 286 • NUMBER 31 JOURNAL OF BIOLOGICAL CHEMISTRY 27305

by guest on Decem

ber 28, 2019http://w

ww

.jbc.org/D

ownloaded from

structs, containing systemic substitutions of the five residues inDIV that correspond to the five crucial residues in DII (Fig. 6).These channel constructs were generated in the Nav1.7-E818Cchannel variant because this construct is substantially resistantto inhibition of activation byHWTX-IV, allowing investigationof the effect of HWTX-IV on the DIV mutants.Of the five corresponding residues in DIV (Glu-1524, Gly-

1581, Leu-1584, Asp-1586, and Ile-1588), only the second andlast amino acids differ from those in DII (Fig. 6A). To simplifythe description of the various mutant channels, we will refer toeach construct by the five-letter code for the five key residues.In addition, we will capitalize the amino acid residue if it isidentical to that found in DII. Thus, the native DIV construct isEgLDi, and the construct where all five match those in DIIis EELDE.The electrophysiological properties of these mutant chan-

nels, expressed in HEK293 cells, were determined (Fig. 6B andsupplemental Table S4). Comparedwith E818C, additional sin-gle mutations shifted the voltage dependence of activation byless than �7 mV, but the voltage dependence of inactivationwas more substantially affected (ranging from �13.9 to �12.2mV). This result is consistentwith the general principle that theDIV voltage sensor is predominantly involved in channel inac-tivation (5, 6, 8).Studies on scorpion �-toxins have demonstrated that stabi-

lizing the DIV voltage sensor in the closed state impairs fastinactivation of VGSCs (13). To estimate HWTX-IV efficacy forinhibiting fast inactivation, we measured the I5ms/Ipeak ratio,providing an estimate of the probability that the channel is notinactivated after 5 ms. The EELDi construct did not exhibitaltered sensitivity to inhibition of fast inactivation byHWTX-IV (Fig. 7A), despite the fact that in this construct fourof the five crucial residues identified in DII are now present inDIV. In contrast, the introduction of Glu at the last position

(creating the EgLDE construct) made Nav1.7 DIV substantiallymore sensitive to HWTX-IV. This is consistent with our previ-ous observation that the corresponding Glu residue in the DIIsofNav1.4 andNav1.7 is absolutely crucial for high affinity inter-action of the voltage sensor with HWTX-IV (16). In the EgLDEchannels, 10 �M HWTX-IV inhibited fast inactivation by56.1 � 7.3% (n � 4; Fig. 7A). This effect on inactivation wasalmost doubled by addition of the final difference, producingthe EELDE construct. In this channel construct, where all fivecritical residues in DII are present at the corresponding posi-tions in DIV, HWTX-IV is able to substantially inhibit inacti-vation of Nav1.7 channels (Fig. 7, A and B).We next asked whether substitutions of the three residues in

DIV that were originally conserved compared with DII couldmodulate the ability of HWTX-IV to inhibit inactivation ofchannels containing the critical I1588E mutation. We foundthat substitutions at all three of these conserved residues pro-duced a decrease in the toxin sensitivity of the DIV voltagesensor. In comparison with the EgLDE construct, 10 �M

HWTX-IV inhibited fast inactivation of the EgcDE constructby only 16.9 � 1.9% (n � 3). In comparison with the EELDEconstruct, 10 �M HWTX-IV inhibited fast inactivation of theqELDE and EELaE constructs by only 46.3 � 4.8% (n � 4) and7.6 � 1.9% (n � 5), respectively. In Fig. 7C, the IC50 values forHWTX-IV inhibition of inactivation were estimated from thedata shown in Fig. 7C and are reported in Table 1. From thesedata, it is clear that DIV substitutions that mimicked critical

FIGURE 6. Functional effects of mutations of amino acid residues inhNav1.7 DIV-VSD. A, amino acid sequence alignment of S1–S2 and S3–S4regions of Nav1.7 DII and DIV. Compared with DII, the identical residues aremarked with a dot (.) in DIV. Five crucial residues in DII and DIV are shaded ingray. B, effects of mutations of amino acid residues in DIV-VSD on steady-stateactivation (left) and inactivation (right) of the E818C mutant hNav1.7 channel.The cells were held at �100 mV. Channel conductance and steady-state inac-tivation were determined using the method as described for Fig. 2. The V1⁄2

values and slope factor are summarized in supplemental Table S4. Each DIVconstruct is identified by the five-letter code representing the amino acidresidues present at the five shaded positions in DIV. The uppercase lettersindicate that the residue is identical to that of the corresponding crucial res-idue in DII.

FIGURE 7. Mutations in DIV-VSD altered the ability of HWTX-IV to slowfast inactivation of the E818C-mutant hNav1.7 channel expressed inHek293 cells. A, typical current traces from seven mutant hNav1.7 channelsbefore (filled circle) and after (open circle) application of 10 �M HWTX-IV. Cur-rent traces were elicited by the 20-ms depolarizing potential, rangingbetween �10 and 0 mV, from a holding potential of �100 mV. B, families ofrepresentative current traces through the triple mutant hNav1.7 E818C/G1581E/I1588E induced by 50-ms depolarizing steps to various potentialsranging from �100 to �150 mV. The cells were held at �100 mV. C, dose-response curves of HWTX-IV inhibiting fast inactivation of mutant hNav1.7channels. The data points (means � S.E.) come from three to seven cells andare fit to the Hill equation as described under “Experimental Procedures.”

Conserved Toxin-binding Motif in Na� Channel Voltage Sensors

27306 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 • NUMBER 31 • AUGUST 5, 2011

by guest on Decem

ber 28, 2019http://w

ww

.jbc.org/D

ownloaded from

residues in DII (G1581E and I1588E) enhanced the ability ofHWTX-IV to inhibit inactivation and, conversely, DIV muta-tions that mimic disruptivemutations of critical residues in DII(E1524Q, L1584C, and D1586A) decreased the ability ofHWTX-IV to inhibit inactivation. Furthermore, as withHWTX-IV inhibition of activation (16), inhibition of fast inac-tivation of theDIV construct containing all five crucial residueswas voltage-dependent (Fig. 7B and supplemental Fig. S2A).The inhibition was reversible but required substantial depolar-izations (� �100 mV), suggesting that interaction ofHWTX-IV with the DIV voltage sensor is relatively strong.However, 10 �M HWTX-IV did not significantly shift thesteady-state activation and inactivation for the unblocked DIVmutant channels (supplemental Fig. S2B).In our previous work (15, 16), the IC50 value for HWTX-IV is

estimated to be 26 nM for WT Nav1.7 DII, which is �100-foldsmaller than that for modulation of inactivation with theEELDE channel, the construct having all five of the critical res-idues identified in DII present in DIV. As can be seen in Fig. 8A,there are eight additional amino acid residues that are not con-served between the S3 segments of DII and DIV, and theDIVS3–S4 linker is eight residues longer than the DIIS3–S4linker. Therefore, we next asked whether these differences alsocontributed to the sensitivity of DIV for HWTX-IV. We con-structed fourDII/DIV chimeras as shown in Fig. 8A. In chimeraI, the whole DIIS3 segment (Trp-797 to Glu-818) was trans-planted into DIV. In chimeras II and III, the ETYFV andETYFVSPT residues were additionally deleted from chimera I,respectively. In chimera IV, the extracellular portion of S3 andthe S3–S4 linker regions of DIV were fully replaced by the cor-responding part of DII (Fig. 8A). The parameters of the voltagedependence of channel activation and steady-state inactivationare summarized in supplemental Table S4. Each chimerashifted the voltage-dependent activation of the Nav1.7-E818Cchannel variant by less than �10 mV. Although chimera I didnot evidently shift the voltage dependence of steady-state inac-tivation, chimeras II–IV did so bymore than�30mV (Fig. 8B).Again, this finding is consistent with the general principle thatthe DIV voltage sensor is predominantly involved in channelinactivation (5, 6, 8).The chimeric channels all contained the five crucial residues

from DII, and they successively increased the ability ofHWTX-IV to inhibit fast inactivation. HWTX-IV (10 �M)inhibited fast inactivation of chimera I channels by 42.1 � 8.7%(n� 4; Fig. 8C). The IC50 valuewas estimated to be 12.8�M (Fig.

8D), which was 6-fold larger than the value for the startingEELDE construct. This may be partially caused by the sub-stitution L1587V because the mutation V817A in DIIinduced a 2-fold increase in toxin binding affinity. ChimerasII and III are more sensitive to HWTX-IV than chimera I.Their IC50 values were 1.4 and 0.51 �M, respectively, indicat-ing that the deletion of the additional residues in the S3–S4linker (ETYFV in chimera II and ETYFVSPT in chimera III)produced additional 8- and 25-fold increases in HWTX-IVbinding affinity, respectively (Fig. 8, C and D). Finally, inchimera IV, two more residues in the S3–S4 linker (Leu andPhe) were replaced by the corresponding residues in Nav1.7DII (Gly and Leu) (Fig. 8A). This replacement was found todouble toxin sensitivity with an IC50 value of 0.27 �M (Fig. 8,C and D). This result indicated that the longer amino acidsequence in the DIVS3–S4 linker, compared with theDIIS3–S4 linker, could interfere to some extent with thebinding of HWTX-IV. Therefore, this finding, together withthe data from point mutation experiments, demonstratedthat the molecular determinants of HWTX-IV binding areconserved in the VSDs of both DII and DIV.

FIGURE 8. Effects of HWTX-IV on hNav1.7 DII/DIV chimeras expressed inHek293 cells. A, amino acid sequence alignment of S3–S4 regions of hNav1.7DII (red) and DIV (black). The S3 segment and S3–S4 linker in DII that wastransplanted into DIV is highlighted in red in the four chimeras. Positivelycharged residues (Arg) in S4 segment are shaded in black bars. B, character-ization of functional properties of the DII/DIV chimeras. The cells were held at�100 mV. Channel conductance (left) and steady-state inactivation (right)were determined using the method as described in Fig. 2. C, typical currenttraces from four hNav1.7 DII/DIV chimeras before and after application of 10�M HWTX-IV. D, dose-response curves of HWTX-IV slowing fast inactivation offour Nav1.7 DII/DIV chimeras. The data points (means � S.E.) from three tofour cells are fit to a Hill equation. Note that all DII/DIV chimeras were con-structed based on the E818C mutant hNav1.7 channel.

TABLE 1Inhibition of inactivation of the E818C mutant hNav1.7 channels byHWTX-IVThe identity of the amino acid residues at the five key positions in DIV are shown.Those that differ from the key residues in DII are shown as lowercase letters.

Identity of five keyresidues in DIV

Inhibition ofinactivation (10 �M)

EstimatedIC50

% �MEgLDi None detectedEELDi None detectedEgLDE 56.1 � 7.3% 7.2EELDE 80.6 � 1.1% 2.2EgcDE 16.9 � 1.9% 47.4qELDE 46.3 � 4.8% 9.7EELaE 7.6 � 1.9% 115.7

Conserved Toxin-binding Motif in Na� Channel Voltage Sensors

AUGUST 5, 2011 • VOLUME 286 • NUMBER 31 JOURNAL OF BIOLOGICAL CHEMISTRY 27307

by guest on Decem

ber 28, 2019http://w

ww

.jbc.org/D

ownloaded from

DISCUSSION

In this study, we investigated the molecular determinants ofVGSC voltage sensor trapping by the tarantula toxin HWTX-IV. We first identified the specific residues in the extracellularportions of the S1–S2 and S3–S4 regions of DII from hNav1.7that are crucial for the action HWTX-IV. Based on theseresults, we determined that some of the essential moleculardeterminants of theHWTX-IV receptor inDII are conserved inDIV. These results contribute to our fundamental understand-ing of how tarantula toxins interact withVGSCs andmodify theactivity of specific voltage sensors.Previouslywe showed thatHWTX-IV is aVGSCgatingmod-

ifier that inhibits activation by trapping DIIS4 in the closedstate (16). Interestingly, our current data indicate that none ofthe amino acid residues in the N-terminal part (extracellularhalf) of DIIS4 directly interact withHWTX-IV (Fig. 3B). The S4segment is a mobile amphipathic �-helical structure composedof positive, polar, and hydrophobic residues. Consistent withthe proposal that positive residues serve as the primary origina-tor of voltage sensitivity, charge-neutralizing mutations of thethree outermost Arg residues in hNav1.7 DIIS4 segmentshift the voltage dependence of activation to more positivepotentials. Other residues in DIIS4 are also involved in voltagesensor activation (Fig. 2A). Among them, Leu-828 is uniquebecause the Ala mutation negatively shifts voltage dependenceof activation, suggesting that Leu-828 favorsDIIS4 in the closedstate. In our previous work, HWTX-IV action greatly increasesthe energy required for DIIS4 activation. Only toxin-unboundWTNav1.7 channels can activate within the physiological volt-age range (16). Of all the mutants examined, HWTX-IV onlysignificantly shifted the voltage-dependent activation of theR827Q/C and L828Amutants to more positive potentials. Thisbehavior can be explained by either activation of toxin-boundVGSCs or dissociation of toxin-VGSCs at depolarized voltageswithin the physiological range for these three mutants. Thisresult indicates that interaction of Arg-827 and Leu-828 withother residues in the voltage sensor may provide a higherenergy barrier for DIIS4 activation than with other residues inDIIS4.Our data, together with earlier work (15, 16), identify five

residues that are critically involved in the interaction ofHWTX-IV with hNav1.7. One residue, Glu-753, is in theDIIS1-S2 linker, and the others, Glu-811, Leu-814, Asp-816,andGlu-818, are located in the extracellular portion of DIIS3 orthe DIIS3-S4 linker. Glu-818 is the most crucial because theE818C mutation eliminated the sensitivity of hNav1.7 toHWTX-IV (15, 16). However, interaction of HWTX-IV withonly Glu-818 is not sufficient to stabilize DIIS4 in the closedstate. Mutations of the other four residues are predicted to col-lectively reduce toxin binding affinity by 10,530-fold (assumingtheir effects are additive). Their cumulative effects would the-oretically increase the IC50 value from 26 nM to 274 �M.

Scorpion �-toxin CssIV is another DII voltage sensor modi-fier that has been extensively characterized (10, 11). CssIV par-tially shares the binding site on the DII voltage sensor withHWTX-IV. Three of the critical residues involved inHWTX-IV action have been identified as molecular determi-

nants of CssIV activity. This finding is surprising because incontrast to HWTX-IV, which stabilizes the DII voltage sensorin the closed state, CssIV traps the DII voltage sensor in theactivated state. However, Asn-842 and Glu-844 in rNav1.2a,which correspond to Asp-816 and Glu-818 in hNav1.7, are notinvolved in forming the CssIV-binding receptor (11). Con-versely, mutation of rNav1.2a Gly-845 to Asn decreases CssIV-binding affinity by 13-fold in rNav1.2a, whereas the corre-sponding mutation in hNav1.7 (G819N) does not significantlychange HWTX-IV sensitivity. Thus, although the interactionsites of CssIV and HWTX-IV overlap, the distinct confirma-tions of the DII voltage sensor in the closed and activated statesare likely to be important in the formation of the divergenttoxin-binding motifs for these two voltage-gating modifiers.Three of the critical residues in hNav1.7 are conserved in

rNav1.4 (the exceptions being Asp-816 and Glu-818). Our datashow that HWTX-IV does not interact with wild-type rNav1.4.However, mutation of rNav1.4 Gln-657 (corresponding to Glu-818 in hNav1.7) to Glu makes rNav1.4 sensitive to HWTX-IV(16), consistent with the seemingly crucial role that Glu-818 inhNav1.7 plays in trapping the voltage sensor in the closed con-figuration. We constructed closed state and activated statemodels of the Nav1.7 DII voltage sensor based on three-dimen-sional structures of the Kv1.2 channel in the closed and acti-vated states (18, 24) to gain some additional insight into the roleof Glu-818 (Fig. 9). In the closed state homologymodel, the sidechain of Glu-818 is located at the extracellular surface of thevoltage sensor with a vertical orientation, but the side chains ofthe other crucial residues (Glu-753, Glu-811, Leu-814, andAsp-816) are predicted to be embedded within the voltage sen-sor structure. Therefore, we propose that the action ofHWTX-IV trapping hNav1.7 DIIS4 in the closed statemight beseparated into two successive steps: HWTX-IV first interactswith Glu-818 and then interacts with the other four residuesdeeper in the voltage sensor structure. Interestingly, Glu-818 inNav1.7 corresponds to Glu-276 in Kv1.2 and Asn-283 in Kv2.1,respectively (18, 25), and based on the proposed orientation ofthese potassiumchannel residues in the open configuration, theside chain of hNav1.7 Glu-818 rotates into the protein interiorof the DII voltage sensor (or possibly into the lipid membrane)in the model of the activated state. Therefore, our interactionmodel predicts that HWTX-IV would not be able to favorably

FIGURE 9. Structural models of sodium channel DII-VSD at resting andactivated states. Closed state and activated state models of the Nav1.7 DIIvoltage sensor are based on three-dimensional structures of the Kv1.2 chan-nel in the closed and activated states (18, 24). The side chains of five residuescrucial for hNav1.7 binding HWTX-IV are shown in ball and stick representa-tion. Note that the model indicates that the accessibility of the side chain ofhNav1.7 Glu-818 to extracellular compounds is reduced in the activated state.

Conserved Toxin-binding Motif in Na� Channel Voltage Sensors

27308 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 • NUMBER 31 • AUGUST 5, 2011

by guest on Decem

ber 28, 2019http://w

ww

.jbc.org/D

ownloaded from

interact with Glu-818 in the activated state and would be likelyto dissociate from sodium channels following activation of thevoltage sensor. This prediction from homology modeling isconsistent with our previous experimental work, in which theNav1.7-HWTX-IV complex could be dissociated by strongdepolarization, after which reinhibition of hNav1.7 occurredwith the same time course as the initial inhibition (16).The S3b-S4 region in the voltage sensors of voltage-gated ion

channels, referred to as the voltage sensor paddle, has beendeemed to be a major determinant of toxin sensitivity (23,26–28). The voltage sensor paddle regions of domains II and IVare only 30% identical (Fig. 6A). One possibility is that toxinsthat interactwithmultiple paddles do so throughdistinct toxin-paddle interfaces, perhaps with different toxin residues inter-acting with distinct motifs on the different paddles (28). How-ever, our data show that the ability of HWTX-IV to interactwithDIV can be substantiallymodified by simplymodifying thefive residues in DIV that correspond to the five residues identi-fied as criticalmolecular determinants of HWTX-IV binding toDII. Each of the five critical residues in the DII voltage sensorand the corresponding residues in DIV modulate the interac-tion of their respective voltage sensor with HWTX-IV in anidentical manner. Moreover, consistent with the observationfor the DII voltage sensor, the amino acid at position 1588,corresponding to Glu-818 in DII, directly determines the “all-or-none” toxin sensitivity of the DIV voltage sensor. As can beseen in Fig. 6, only two of the five critical residues (Glu-811 andGlu-818) are not conserved in hNav1.7 DIV. The first corre-sponds to Gly-1588. However, the mutation G1588E by itselffailed to significantly alter the resistance of hNav1.7 DIV toHWTX-IV (Fig. 7) and only is able to impact the effects ofHWTX-IV if the crucial glutamine corresponding toGlu-818 ispresent inDIV. These results suggest that theHWTX-IV-bind-ing motifs on DII and DIV are likely to have the same basicconfiguration. Our data also show that replacement of theentire extracellular portion of the S3–S4 region in DIV withthat of DII can further increase the sensitivity of DIV toHWTX-IV, consistentwith the proposal that the voltage sensor“paddle” is one of the determinants of toxin sensitivity (23, 28).Our findings may help explain the cross-activities of some

tarantula toxins (e.g. ProTx-II) on different domains of sodiumchannels (15, 23, 29, 30). Furthermore, some tarantula toxins,such as HWTX-I and Jingzhaotoxin-XI, exhibit cross-activitieson sodium, potassium, and calcium channels (29, 31), suggest-ing that the basic toxin-binding motif might be conserved indifferent ion channels. Among the nineVGSC subtypes, Nav1.7is of special interest because this subtype is an important con-tributor to pain sensation (32, 33). Nav1.7 blockers have beenconsidered as ideal candidates for producing analgesia (4).However, toxins binding to the DIV voltage sensor are pre-dicted to impair fast inactivation. Indeed, we recently showedthat ProTx-II can simultaneously inhibit hNav1.7 activationand inactivation, by interacting with DII and DIV (15). Gener-ally, inhibition of inactivation can increase neuronal excitabilityby prolonging action potential duration, and persistent sodiumcurrents can lead to axonal damage (34). Therefore, if the volt-age sensor structures and toxin-binding motifs in DII and DIVare similar, this could complicate the development of voltage-

gating modifiers that specifically target DII of Nav1.7. Indeed,the residue Phe-813, which plays a key role in the subtype selec-tivity of ProTx-II (35), is conserved in the DIV voltage sensor(Fig. 6) and substantially modulates the interaction of ProTx-IIwith the voltage sensor paddles of both DII and DIV (15, 23).Our data indicate that this residue is not important forHWTX-IV interactions (15). However, it might be feasible todevelop a HWTX-IV/ProTx-II chimeric toxin that requiresinteraction with both Phe-813 and Glu-818 in the toxin-bind-ingmotif of theDII voltage sensor, possibly generating aNav1.7isoform-specific modifier that selectively inhibits activation ofNav1.7 channels. Our increased understanding of the molecu-lar determinants of toxin interactions with Nav1.7 and otherVGSCs should aid the rationale design of enhanced voltage-gating modifiers.

Acknowledgment—We thankDr.Marie-FranceMartin-Eauclaire forgenerously providing the scorpion �-toxin CssIV.

REFERENCES1. Catterall, W. A., Goldin, A. L., andWaxman, S. G. (2005) Pharmacol. Rev.

57, 397–4092. Catterall, W. A. (2000) Neuron 26, 13–253. Drenth, J. P., and Waxman, S. G. (2007) J. Clin. Invest. 117, 3603–36094. Cummins, T. R., Sheets, P. L., and Waxman, S. G. (2007) Pain 131,

243–2575. Goldin, A. L. (2003) Curr. Opin. Neurobiol. 13, 284–2906. Cha, A., Ruben, P. C., George, A. L., Jr., Fujimoto, E., and Bezanilla, F.

(1999) Neuron 22, 73–877. Cestele, S., Scheuer, T., Mantegazza, M., Rochat, H., and Catterall, W. A.

(2001) J. Gen. Physiol. 118, 291–3028. Sheets, M. F., and Hanck, D. A. (2007) J. Physiol. 582, 317–3349. Blumenthal, K. M., and Seibert, A. L. (2003) Cell Biochem. Biophys. 38,

215–23810. Cestele, S., Yarov-Yarovoy, V., Qu, Y., Sampieri, F., Scheuer, T., and Cat-

terall, W. A. (2006) J. Biol. Chem. 281, 21332–2134411. Cestele, S., Qu, Y., Rogers, J. C., Rochat, H., Scheuer, T., and Catterall,

W. A. (1998) Neuron 21, 919–93112. Karbat, I., Cohen, L., Gilles, N., Gordon, D., Gurevitz, M., Izhar, K.,

Lior, C., Nicholas, G., Dalia, G., and Michael, G. (2004) FASEB J. 18,683–689

13. Rogers, J. C., Qu, Y., Tanada, T.N., Scheuer, T., andCatterall,W.A. (1996)J. Biol. Chem. 271, 15950–15962

14. Kahn, R., Karbat, I., Ilan, N., Cohen, L., Sokolov, S., Catterall, W. A., Gor-don, D., and Gurevitz, M. (2009) J. Biol. Chem. 284, 20684–20691

15. Xiao, Y., Blumenthal, K., Jackson, J. O., 2nd, Liang, S., and Cummins, T. R.(2010)Mol. Pharmacol. 78, 1124–1134

16. Xiao, Y., Bingham, J. P., Zhu, W., Moczydlowski, E., Liang, S., and Cum-mins, T. R. (2008) J. Biol. Chem. 283, 27300–27313

17. Peng, K., Shu, Q., Liu, Z., and Liang, S. (2002) J. Biol. Chem. 277,47564–47571

18. Pathak, M. M., Yarov-Yarovoy, V., Agarwal, G., Roux, B., Barth, P., Ko-hout, S., Tombola, F., and Isacoff, E. Y. (2007) Neuron 56, 124–140

19. Zuo, X. P., He, H. Q., He,M., Liu, Z. R., Xu, Q., Ye, J. G., and Ji, Y. H. (2006)FEBS Lett. 580, 4508–4514

20. Mantegazza, M., and Cestele, S. (2005) J. Physiol. 568, 13–3021. Kontis, K. J., Rounaghi, A., and Goldin, A. L. (1997) J. Gen. Physiol. 110,

391–40122. Struyk, A. F., Scoggan, K. A., Bulman, D. E., and Cannon, S. C. (2000)

J. Neurosci. 20, 8610–861723. Bosmans, F., Martin-Eauclaire, M. F., and Swartz, K. J. (2008)Nature 456,

202–20824. Xiao, Y., Luo, X., Kuang, F., Deng, M., Wang, M., Zeng, X., and Liang, S.

(2008) Toxicon 51, 230–239

Conserved Toxin-binding Motif in Na� Channel Voltage Sensors

AUGUST 5, 2011 • VOLUME 286 • NUMBER 31 JOURNAL OF BIOLOGICAL CHEMISTRY 27309

by guest on Decem

ber 28, 2019http://w

ww

.jbc.org/D

ownloaded from

25. Long, S. B., Campbell, E. B., and Mackinnon, R. (2005) Science 309,897–903

26. Lee, S. Y., and MacKinnon, R. (2004) Nature 430, 232–23527. Bosmans, F., and Swartz, K. J. (2010) Trends Pharmacol. Sci. 31, 175–18228. Li-Smerin, Y., and Swartz, K. J. (1998) Proc. Natl. Acad. Sci. U.S.A. 95,

8585–858929. Liao, Z., Yuan, C., Deng, M., Li, J., Chen, J., Yang, Y., Hu,W., and Liang, S.

(2006) Biochemistry 45, 15591–1560030. Corzo, G., Gilles, N., Satake, H., Villegas, E., Dai, L., Nakajima, T., and

Haupt, J. (2003) FEBS Lett. 547, 43–5031. Wang,M., Guan, X., and Liang, S. (2007)Biochem. Biophys. Res. Commun.

357, 579–58332. Dib-Hajj, S. D., Cummins, T. R., Black, J. A., and Waxman, S. G. (2007)

Trends Neurosci. 30, 555–56333. Cox, J. J., Reimann, F., Nicholas, A. K., Thornton, G., Roberts, E., Springell,

K., Karbani, G., Jafri, H., Mannan, J., Raashid, Y., Al-Gazali, L., Hamamy,H., Valente, E. M., Gorman, S., Williams, R., McHale, D. P., Wood, J. N.,Gribble, F. M., and Woods, C. G. (2006) Nature 444, 894–898

34. Bechtold, D. A., and Smith, K. J. (2005) J. Neurol. Sci. 233, 27–3535. Schmalhofer, W. A., Calhoun, J., Burrows, R., Bailey, T., Kohler, M. G.,

Weinglass, A. B., Kaczorowski, G. J., Garcia, M. L., Koltzenburg, M., andPriest, B. T. (2008)Mol. Pharmacol. 74, 1476–1484

Conserved Toxin-binding Motif in Na� Channel Voltage Sensors

27310 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 • NUMBER 31 • AUGUST 5, 2011

by guest on Decem

ber 28, 2019http://w

ww

.jbc.org/D

ownloaded from

Yucheng Xiao, James O. Jackson II, Songping Liang and Theodore R. CumminsChannel Voltage Sensors in Domains II and IV

+Common Molecular Determinants of Tarantula Huwentoxin-IV Inhibition of Na

doi: 10.1074/jbc.M111.246876 originally published online June 9, 20112011, 286:27301-27310.J. Biol. Chem. 

  10.1074/jbc.M111.246876Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

Supplemental material:

  http://www.jbc.org/content/suppl/2011/06/15/M111.246876.DC2

Supplemental material:

  http://www.jbc.org/content/suppl/2011/06/15/M111.246876.DC3

Supplemental material:

  http://www.jbc.org/content/suppl/2011/06/15/M111.246876.DC4

Supplemental material:

  http://www.jbc.org/content/suppl/2011/06/06/M111.246876.DC1

  http://www.jbc.org/content/286/31/27301.full.html#ref-list-1

This article cites 35 references, 13 of which can be accessed free at

by guest on Decem

ber 28, 2019http://w

ww

.jbc.org/D

ownloaded from