ION CHANNELS A pharmacological masterkey mechanism that … · 2019-04-11 · ION CHANNELS A...

7
ION CHANNELS A pharmacological master key mechanism that unlocks the selectivity filter gate in K + channels Marcus Schewe 1 *, Han Sun 2 *, Ümit Mert 1 , Alexandra Mackenzie 3,4,5 , Ashley C. W. Pike 3 , Friederike Schulz 1 , Cristina Constantin 6,7 , Kirsty S. Vowinkel 8 , Linus J. Conrad 4,5 , Aytug K. Kiper 8 , Wendy Gonzalez 9,10 , Marianne Musinszki 1 , Marie Tegtmeier 1 , David C. Pryde 11 , Hassane Belabed 12 , Marc Nazare 12 , Bert L. de Groot 13 , Niels Decher 8 , Bernd Fakler 6,7 , Elisabeth P. Carpenter 3,4 , Stephen J. Tucker 4,5 , Thomas Baukrowitz 1 Potassium (K + ) channels have been evolutionarily tuned for activation by diverse biological stimuli, and pharmacological activation is thought to target these specific gating mechanisms. Here we report a class of negatively charged activators (NCAs) that bypass the specific mechanisms but act as master keys to open K + channels gated at their selectivity filter (SF), including many two-pore domain K + (K 2P ) channels, voltage-gated hERG (human ether-à-go-gorelated gene) channels and calcium (Ca 2+ )activated big-conductance potassium (BK)type channels. Functional analysis, x-ray crystallography, and molecular dynamics simulations revealed that the NCAs bind to similar sites below the SF, increase pore and SF K + occupancy, and open the filter gate.These results uncover an unrecognized polypharmacology among K + channel activators and highlight a filter gating machinery that is conserved across different families of K + channels with implications for rational drug design. D ampening cellular electrical activity by pharmacological activation of specific types of K + channels has therapeutic potential for the treatment of a variety of disease states, including epilepsy, arrhythmias, vascular constriction, and various pain condi- tions (1, 2). Consequently, screening efforts have identified a number of agents that open dif- ferent types of K + channels (2), presumably by targeting their respective channel-specific acti- vation mechanisms. Distinct structural mechanisms enable K + chan- nels to respond to a plethora of physiological stimuli, including voltage, temperature, mechan- ical force, and various second messengers, such as adenosine triphosphate (ATP), Ca 2+ , and H + , as well as bioactive lipids such as phosphatidyl- inositol 4,5-bisphosphate (PIP 2 ) and arachidonic acid (3, 4). However, despite this complexity, these activation pathways seem to converge on the two principal mechanisms known to gate K + channels open: dilation of the lowergate at the intracellular pore entrance used by inwardly rectifying (K ir )(5) and voltage-gated (K v )K + channels (6), and activation of the selectivity filter (SF) gate used by most two-pore domain K + (K 2P ) channels (4, 7, 8) and Ca 2+ -activated big-conductance K + (BK Ca ) channels (9, 10). In voltage-gated hERG (human ether-à-go-gorelated gene) channels, both mechanisms coexist, with voltage opening the lower gate but rapid in- activation occurring through closure of the SF gate (11, 12). Here we identify a common mech- anism for drug-induced channel opening that bypasses these physiological activation mechan- isms in SF-gated K + channels. For the mechanosensitive K 2P channels TREK-1 and TREK-2, the voltage-gated hERG channel, and the Ca 2+ -activated BK Ca channel, a series of small-molecule activators all harboring a nega- tively charged group (tetrazole or carboxylate) have been proposed to act as selective channel openers [i.e., BL-1249 for TREK-1/-2 (13); PD- 118057 for hERG (14); and NS11021 for BK Ca (15)]. However, application of these compounds to their respective nontargetchannels revealed an unexpected polypharmacology: All three open- ers displayed equal efficiency in opening TREK-1 channels (Fig. 1A) and hERG channels (Fig. 1B), as well as BK Ca channels (Fig. 1C), whose acti- vation curve is strongly shifted to more negative voltages (fig. S1C). This suggests that these acti- vators may not target channel-specific activation mechanisms and may instead share a common mechanism. In all cases, the compound-mediated effect was effectively antagonized by large quater- nary ammonium ions (QA L + ) such as tetra-pentyl- ammonium (TPenA) or tetra-hexyl-ammonium (THexA) that are known to block K + channels at a site immediately below the inner entrance to the SF (16) (Fig. 1, A and B, and fig. S1C). Like- wise, all these activators reduced the QA L + - mediated inhibition in these different K + chan- nels (Fig. 1C and figs. S1, A and B, and S7, A and B). Furthermore, extended screening with BL-1249 also revealed potent activation of several other K 2P channels (TREK-2, TRAAK, TALK-1, TALK-2, THIK-1, and THIK-2; fig. S1D). Together, these data suggest that these negatively charged acti- vators (NCAs) (BL-1249, PD-118057, and NS11021) act on a gating mechanism that is shared among these different classes of K + channels and that their action involves a site that overlaps with the conserved QA L + -binding site located below the SF. A distinctive feature of all NCA-responsive K + channels is their gating by the SF, a mech- anism that is intimately coupled to ion per- meation (17, 18). In K 2P channels, this coupling leads to pronounced activation by Rb + , which displays an ion occupancy distinct to K + at the four SF K + binding sites (S1 to S4) that stabi- lizes the activated state of the SF gate (17). Interestingly, Rb + not only activated all NCA- responsive K 2P channels but also led to robust activation of BK Ca and hERG channels (Fig. 1D). By contrast, Rb + failed to exert any activatory effect on K + channels gated at the helix-bundle crossing (i.e., K ir and most K v channels), as was observed for K v 1.1, K v 1.5, K v 3.1, and K ir 1.1 (Fig. 1D); consistent with this, these channels were also not activated by BL-1249 (fig. S2, A to E). Furthermore, cyclic nucleotidegated channels that are also gated at the SF were not acti- vated by BL-1249, indicating that the NCA mech- anism may be specific to SF-gated K + channels (fig. S2F). To gain further mechanistic insight into chan- nel opening by the NCAs, we next investigated their binding by x-ray crystallography, cysteine- scanning mutagenesis, and atomistic molecular dynamics (MD) simulations. First, anomalous diffraction data were collected from TREK-2 channels cocrystallized with a brominated deriv- ative of BL-1249 (BL-1249 Br ) (Fig. 2A; fig. S3, A to C; and supplementary materials and methods). Although no discrete electron density was visible for BL-1249 itself, in anomalous difference maps, two bromine peaks were clearly visible per TREK-2 dimer (fig. S3, A and B) and the main-chain protein backbone showed excellent agreement with a previously crystallized high-resolution structure of TREK-2 [Protein Data Bank (PDB) 4XDJ] (19). Both bromine anomalous difference peaks were located at the entrance of the side fenestrations branching off the central pore ca- vity below the SF. Comparison with a structure RESEARCH Schewe et al., Science 363, 875880 (2019) 22 February 2019 1 of 6 1 Institute of Physiology, Christian-Albrechts University of Kiel, 24118 Kiel, Germany. 2 Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Department of Structural Biology, 13125 Berlin, Germany. 3 Structural Genomics Consortium, University of Oxford, Oxford OX3 7DQ, UK. 4 OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford OX1 3PN, UK. 5 Clarendon Laboratory, Department of Physics, University of Oxford, Oxford OX1 3PU, UK. 6 Institute of Physiology II, Albert-Ludwigs University of Freiburg, 79104 Freiburg, Germany. 7 Centers for Biological Signaling Studies CIBSS and BIOSS, 79104 Freiburg, Germany. 8 Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, 35037 Marburg, Germany. 9 Centro de Bioinformatica y Simulacion Molecular, Universidad de Talca, 3465548 Talca, Chile. 10 Millennium Nucleus of Ion Channels- Associated Diseases (MiNICAD), Universidad de Talca, 3465548 Talca, Chile. 11 Pfizer Worldwide Medicinal Chemistry, Neuroscience and Pain Research Unit, Portway Building, Granta Park, Great Abington, Cambridgeshire CB21 6GS, UK. 12 Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Department of Medicinal Chemistry, 13125 Berlin, Germany. 13 Computational Biomolecular Dynamics Group, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany. *These authors contributed equally to this work. Corresponding author. Email: [email protected]. de (M.S.); [email protected] (T.B.) Present address: Curadev Pharma Ltd., Sandwich Kent, UK. on April 11, 2019 http://science.sciencemag.org/ Downloaded from

Transcript of ION CHANNELS A pharmacological masterkey mechanism that … · 2019-04-11 · ION CHANNELS A...

Page 1: ION CHANNELS A pharmacological masterkey mechanism that … · 2019-04-11 · ION CHANNELS A pharmacological masterkey mechanism that unlocks the selectivity filter gate in K+ channels

ION CHANNELS

A pharmacological master keymechanism that unlocks theselectivity filter gate in K+ channelsMarcus Schewe1*†, Han Sun2*, Ümit Mert1, Alexandra Mackenzie3,4,5,Ashley C. W. Pike3, Friederike Schulz1, Cristina Constantin6,7, Kirsty S. Vowinkel8,Linus J. Conrad4,5, Aytug K. Kiper8, Wendy Gonzalez9,10, Marianne Musinszki1,Marie Tegtmeier1, David C. Pryde11‡, Hassane Belabed12, Marc Nazare12,Bert L. de Groot13, Niels Decher8, Bernd Fakler6,7, Elisabeth P. Carpenter3,4,Stephen J. Tucker4,5, Thomas Baukrowitz1†

Potassium (K+) channels have been evolutionarily tuned for activation by diverse biologicalstimuli, and pharmacological activation is thought to target these specific gating mechanisms.Here we report a class of negatively charged activators (NCAs) that bypass the specificmechanisms but act as master keys to open K+ channels gated at their selectivity filter (SF),includingmany two-pore domainK+ (K2P) channels,voltage-gatedhERG (humanether-à-go-go–related gene) channels and calcium (Ca2+)–activated big-conductance potassium (BK)–typechannels. Functional analysis, x-ray crystallography, and molecular dynamics simulationsrevealed that the NCAs bind to similar sites below the SF, increase pore and SF K+ occupancy,and open the filter gate.These results uncover an unrecognized polypharmacology amongK+ channel activators and highlight a filter gating machinery that is conserved across differentfamilies of K+ channels with implications for rational drug design.

Dampening cellular electrical activity bypharmacological activation of specific typesof K+ channels has therapeutic potentialfor the treatment of a variety of diseasestates, including epilepsy, arrhythmias,

vascular constriction, and various pain condi-tions (1, 2). Consequently, screening efforts haveidentified a number of agents that open dif-ferent types of K+ channels (2), presumably bytargeting their respective channel-specific acti-vation mechanisms.Distinct structuralmechanisms enable K+ chan-

nels to respond to a plethora of physiological

stimuli, including voltage, temperature, mechan-ical force, and various second messengers, suchas adenosine triphosphate (ATP), Ca2+, and H+,as well as bioactive lipids such as phosphatidyl-inositol 4,5-bisphosphate (PIP2) and arachidonicacid (3, 4). However, despite this complexity,these activation pathways seem to converge onthe two principal mechanisms known to gate K+

channels open: dilation of the “lower” gate atthe intracellular pore entrance used by inwardlyrectifying (Kir) (5) and voltage-gated (Kv) K+

channels (6), and activation of the selectivityfilter (SF) gate used by most two-pore domainK+ (K2P) channels (4, 7, 8) and Ca2+-activatedbig-conductance K+ (BKCa) channels (9, 10). Involtage-gatedhERG (humanether-à-go-go–relatedgene) channels, both mechanisms coexist, withvoltage opening the lower gate but rapid in-activation occurring through closure of the SFgate (11, 12). Here we identify a common mech-anism for drug-induced channel opening thatbypasses these physiological activation mechan-isms in SF-gated K+ channels.For themechanosensitive K2P channels TREK-1

and TREK-2, the voltage-gated hERG channel,and the Ca2+-activated BKCa channel, a seriesof small-molecule activators all harboring a nega-tively charged group (tetrazole or carboxylate)have been proposed to act as selective channelopeners [i.e., BL-1249 for TREK-1/-2 (13); PD-118057 for hERG (14); and NS11021 for BKCa (15)].However, application of these compounds totheir respective “nontarget” channels revealedan unexpected polypharmacology: All three open-ers displayed equal efficiency in opening TREK-1channels (Fig. 1A) and hERG channels (Fig. 1B),as well as BKCa channels (Fig. 1C), whose acti-

vation curve is strongly shifted to more negativevoltages (fig. S1C). This suggests that these acti-vators may not target channel-specific activationmechanisms and may instead share a commonmechanism. In all cases, the compound-mediatedeffect was effectively antagonized by large quater-nary ammonium ions (QAL

+) such as tetra-pentyl-ammonium (TPenA) or tetra-hexyl-ammonium(THexA) that are known to block K+ channels ata site immediately below the inner entrance tothe SF (16) (Fig. 1, A and B, and fig. S1C). Like-wise, all these activators reduced the QAL

+-mediated inhibition in these different K+ chan-nels (Fig. 1C and figs. S1, A and B, and S7, A andB). Furthermore, extended screening with BL-1249also revealed potent activation of several other K2P

channels (TREK-2, TRAAK, TALK-1, TALK-2,THIK-1, and THIK-2; fig. S1D). Together, thesedata suggest that these negatively charged acti-vators (NCAs) (BL-1249, PD-118057, andNS11021)act on a gatingmechanism that is shared amongthese different classes of K+ channels and thattheir action involves a site that overlaps withthe conserved QAL

+-binding site located belowthe SF.A distinctive feature of all NCA-responsive

K+ channels is their gating by the SF, a mech-anism that is intimately coupled to ion per-meation (17, 18). In K2P channels, this couplingleads to pronounced activation by Rb+, whichdisplays an ion occupancy distinct to K+ at thefour SF K+ binding sites (S1 to S4) that stabi-lizes the activated state of the SF gate (17).Interestingly, Rb+ not only activated all NCA-responsive K2P channels but also led to robustactivation of BKCa and hERG channels (Fig. 1D).By contrast, Rb+ failed to exert any activatoryeffect on K+ channels gated at the helix-bundlecrossing (i.e., Kir and most Kv channels), as wasobserved for Kv1.1, Kv1.5, Kv3.1, and Kir1.1 (Fig.1D); consistent with this, these channels werealso not activated by BL-1249 (fig. S2, A to E).Furthermore, cyclic nucleotide–gated channelsthat are also gated at the SF were not acti-vated by BL-1249, indicating that the NCA mech-anism may be specific to SF-gated K+ channels(fig. S2F).To gain further mechanistic insight into chan-

nel opening by the NCAs, we next investigatedtheir binding by x-ray crystallography, cysteine-scanning mutagenesis, and atomistic moleculardynamics (MD) simulations. First, anomalousdiffraction data were collected from TREK-2channels cocrystallized with a brominated deriv-ative of BL-1249 (BL-1249Br) (Fig. 2A; fig. S3, A toC; and supplementary materials and methods).Although no discrete electron density was visiblefor BL-1249 itself, in anomalous difference maps,two bromine peakswere clearly visible per TREK-2dimer (fig. S3, A and B) and the main-chainprotein backbone showed excellent agreementwith a previously crystallized high-resolutionstructure of TREK-2 [Protein Data Bank (PDB)4XDJ] (19). Both bromine anomalous differencepeaks were located at the entrance of the sidefenestrations branching off the central pore ca-vity below the SF. Comparison with a structure

RESEARCH

Schewe et al., Science 363, 875–880 (2019) 22 February 2019 1 of 6

1Institute of Physiology, Christian-Albrechts University of Kiel,24118 Kiel, Germany. 2Leibniz-Forschungsinstitut fürMolekulare Pharmakologie (FMP), Department of StructuralBiology, 13125 Berlin, Germany. 3Structural GenomicsConsortium, University of Oxford, Oxford OX3 7DQ, UK.4OXION Initiative in Ion Channels and Disease, University ofOxford, Oxford OX1 3PN, UK. 5Clarendon Laboratory,Department of Physics, University of Oxford, Oxford OX13PU, UK. 6Institute of Physiology II, Albert-LudwigsUniversity of Freiburg, 79104 Freiburg, Germany. 7Centersfor Biological Signaling Studies CIBSS and BIOSS, 79104Freiburg, Germany. 8Institute of Physiology andPathophysiology, Vegetative Physiology, Philipps-Universityof Marburg, 35037 Marburg, Germany. 9Centro deBioinformatica y Simulacion Molecular, Universidad de Talca,3465548 Talca, Chile. 10Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Talca,3465548 Talca, Chile. 11Pfizer Worldwide MedicinalChemistry, Neuroscience and Pain Research Unit, PortwayBuilding, Granta Park, Great Abington, Cambridgeshire CB216GS, UK. 12Leibniz-Forschungsinstitut für MolekularePharmakologie (FMP), Department of Medicinal Chemistry,13125 Berlin, Germany. 13Computational BiomolecularDynamics Group, Max Planck Institute for BiophysicalChemistry, 37077 Göttingen, Germany.*These authors contributed equally to this work.†Corresponding author. Email: [email protected] (M.S.); [email protected] (T.B.) ‡Presentaddress: Curadev Pharma Ltd., Sandwich Kent, UK.

on April 11, 2019

http://science.sciencem

ag.org/D

ownloaded from

Page 2: ION CHANNELS A pharmacological masterkey mechanism that … · 2019-04-11 · ION CHANNELS A pharmacological masterkey mechanism that unlocks the selectivity filter gate in K+ channels

that included QAL+ (16) showed that these bro-

mine positions reside within the spherical vol-ume of THexA but outside that of the smallertetra-ethyl-ammonium (TEA) ion. Consistentwith this, BL-1249 activation of TREK-2 channels

was antagonized by THexA but not by TEA (Fig.2, B and C).These structural data were complemented by

cysteine-scanningmutagenesis of the pore-liningM2 and M4 helices of TREK-1. Six residues, in-

cluding the highly conserved Pro183 (P183) andLeu304 (L304) (also investigated in TREK-2, fig.S3D), were identified where mutations markedlyreduced the apparent affinity of BL-1249. Theseresidues cluster around the bromine densities

Schewe et al., Science 363, 875–880 (2019) 22 February 2019 2 of 6

500 ms

-0.5

nA

basal

+ 25 µMBL-1249500 ms

-1n

A

basal

+ 25 µMNS11021

-1 0 1Log PD-118057 [µM]

0.0

0.2

0.4

0.6

0.8

1.0

I tail/

I tail-

max

hERG channel activatorB 120 K+

120 K+PD-118057in TPenA

PD-118057

100 ms

2n

A

HN CF3

Br

S

NH

CF3

N NN

N

NS11021

100 ms

2n

A

100 ms

5n

A1.0

0.8

0.6

0.4

0.2

0.0

I/Im

ax

-2 -1 0 1 2Log THexA [µM]

BKCa channel activatorC

basal

+ 50 µM

+ 10 µMPD-118057

+ 50 µMBL-1249

120 K+

120 K+

THexAin NS11021

THexA

TREK K2P channel activator

NH

NN

NN

BL-1249

-80 -40 +40 +80

V [mV]

I[n

A]

8

-8

16

basal

+ 50 µMNS11021

-2 -1 0 1 2 3Log BL-1249 [µM]

1.0

0.8

0.6

0.4

0.2

0.0

I/Im

ax

BL-1249

BL-1249in THexA

A

-80 -40 +40 +80

V [mV]

I[n

A] 30

15

-15

120 K+

120 K++ 10 µM

basal

+40 +80

V [mV]

I[n

A]

20

-20

40

basal

+ 10 µMPD-118057

TREK-1

TREK-2

TRAAK

100

10

1

TALK-1TALK-2

BK CahERG

K v1.1

K v1.5

67 22 22 15 35 17 11

K v3.1

I Rb

+/I K

+

0.1

14 25 11

K ir1.110

SF-gated/NCA-activated HBC-gated

3n

A

100 ms120 K+

120 K+/120 Rb+

BKCa

5n

A

TREK-1

100 ms

hERG

20 ms

0.4

nA

-80 mV

+100 mV-80 mV

+60 mV

-100 mV

D E

τ = 12.5 ± 0.9 ms

τ = 22.2 ± 1.4 ms

basal basal

basal

+ 10 µM500 ms

-1n

A

NH

PD-118057

O

O

Cl

Cl

Fig. 1. NCAs open SF-gated K+ channels via a similar site.(A) Representative TREK-1 channel currents recorded in inside-out(i-o) patches evoked by voltage ramps in the absence (basal) andpresence of the indicated compounds. BL-1249 (compound structureshown) dose-response curves represent currents at +40 mV and withor without 5 mM THexA that produced 77 ± 6% inhibition of basal currents(n ≥ 8). I, current; V, voltage. (B) hERG channel currents (voltage stepsfrom −80 to +60 mV) in i-o patches with or without the indicatedcompounds; arrows indicate peak tail current amplitudes at −100 mV.PD-118057 (compound structure shown) dose-response curves representnormalized tail currents with or without 1 mM TPenA that produced 91 ± 1%inhibition of basal currents (n ≥ 6). (C) BKCa channel currents [voltagesteps from a holding potential of −80 to +100 mV (zero Ca2+)] in i-o

patches with or without the indicated compounds. THexA inhibitionrepresents currents at +100 mV and with or without 50 mM NS11021(compound structure shown; n ≥ 11). (D) Rb+ effects on different K+

channels measured in i-o patches. Bars ± SEM represent fold change ofoutward currents upon exchange of intracellular K+ by Rb+ for K2P andBKCa channels (+100 mV); for hERG, Kv1.1, Kv1.5, and Kv3.1 channels(+60 mV); and for Kir1.1 channels (+40 mV). The channels are grouped aseither SF-gated and NCA-activated or helix-bundle crossing (HBC)–gated. (E) Representative traces of Rb+ activation for BKCa, TREK-1, andhERG channels using the indicated protocols [arrow indicates thestarting point of hERG inactivation after inactivation recovery(at −100 mV)]. Time constant (t) values from monoexponential fits toinactivation time course (n ≥ 12). For (A) to (C), errors bars indicate SEM.

RESEARCH | REPORTon A

pril 11, 2019

http://science.sciencemag.org/

Dow

nloaded from

Page 3: ION CHANNELS A pharmacological masterkey mechanism that … · 2019-04-11 · ION CHANNELS A pharmacological masterkey mechanism that unlocks the selectivity filter gate in K+ channels

detected in the TREK-2 cocrystal with BL-1249Br

(Fig. 2D and fig. S3, C and D). A role for L304 inthis presumed binding site was further supportedby cysteine-modification protection experimentsin which the time course of irreversible poreblockade induced by application of the cysteine-modifying agent MTS-TBAO [8-(tributylam-monium)octyl methanethiosulfonate] (20) toTREK-1 L304C (Leu304→Cys) channels wasmark-edly slowed by the presence of BL-1249 (Fig. 2, Eand F). This effect was specific for BL-1249, astwo further channel activators with distinct bind-ing sites [2-APB at the C terminus (21) andML335behind the SF (22)] both failed to slow this rate(Fig. 2, E and F). Furthermore, TREK-1 activation

with2-APBorML335wasnot antagonizedbyQAL+

inhibition, and mutations at the BL-1249 site didnot affect 2-APB activation (fig. S4, A, B, and E).In addition, we performed MD simulations to

examine the orientation of BL-1249Br within itsproposed binding site (Fig. 2G). The favored bind-ing pose oriented the negatively charged tetrazolegroup of BL-1249 toward the S6 “cavity bindingsite” for K+ just below the SF. The remainder ofthe BL-1249 molecule engaged with residues inM2 and M4 consistent with our scanning muta-genesis data (Fig. 2D and fig. S3D).Moreover, thebromine atom in these simulations was found tobe within 3 to 4 Å of the bromine densities de-termined by crystallography (fig. S3E). Together,

these data indicate that BL-1249 binds to a sitebelow the SF and reveal a critical role of thenegative charge of the acidic tetrazole ring (pKa

around 5, where Ka is the dissociation constant),implying a pH-dependent compound efficacy.Indeed, when tested with the K2P channel TALK-2[exhibiting little intrinsic intracellular pH (pHi)sensitivity], BL-1249 potency dropped stronglywith a lowering of the solution pHi to 5, whereascontrol experiments with 2-APB lacked this pHdependence (Fig. 3F).We have recently used atomistic MD simula-

tions and a double-bilayer setup to study ion per-meation in the TRAAKK2P channel (17). Therefore,we carried out simulations of ion permeation in

Schewe et al., Science 363, 875–880 (2019) 22 February 2019 3 of 6

Fig. 2. Identificationof the BL-1249binding site in TREKK2P channels. (A) Thestructure depictsTREK-2 (PDB 4XDJ),with pink spheresrepresenting thepositions of Br atoms ina brominated BL-1249derivate (BL-1249Br)obtained by cocrystalli-zation of TREK-2 andBL-1249Br (see alsofig. S3). With thismedium-resolution data,only the Br atoms wereidentified, because theygave peaks in anomalousdifference maps.N, N terminus; C, C termi-nus. (B) The same struc-ture also showing sphericalrepresentations ofTHexA (green) and TEA(dark blue) with theircentral nitrogen atoms(yellow).Their positionsare based on the crystalstructures of KcsA withQA+s (16). Note that theBr atoms [pink spheresin (A)] are within thesphere of THexA but notof TEA. (C) BL-1249dose-response curves forTREK-2 with or without100 mM TEA (n ≥ 12)or 5 μM THexA (n ≥ 13)(TEA andTHexAproduce74 ± 3 and 83 ± 2%basalcurrent inhibition, respectively). Error bars indicate SEM. (D) Scanningmutagenesis of M2 and M4 helices showing BL-1249 median effectiveconcentration (EC50) values ± SEM determined at +40 mV; the inset showsa K2P channel alignment for channels strongly activated by BL-1249 (see alsofig. S1D) with residues homologous to TREK-1 P183 and L304 highlighted.(E) Time courses of 10 μM MTS-TBAO cysteine modification of L304C inTREK-1 before and after maximal activation by BL-1249 (50 μM), ML335(50 μM), and 2-APB (1 mM) (left panel).The graphic depicts TREK-1 withpredicted drug binding sites relative to the position of residue L304C.

(F) Time values ± SEM for half-maximal MTS-TBAO modification inhibition(t1/2) in the presence of different agonists. ***P ≤ 0.001; n.s., not significant.(G) Representation of favored binding pose of BL-1249 (pink) in TREK-2 alongwith the location of the TREK-1 corresponding mutations (green) that reduceBL-1249 activation. For (A) and (G), the A and B superscripts indicate thesubunit of the TREK-2 dimer. Single-letter abbreviations for amino acidresidues are as follows: A, Ala; C, Cys; D, Asp; E, Glu; F, Phe; G, Gly;H, His; I, Ile; K, Lys; L, Leu; M, Met; N, Asn; P, Pro; Q, Gln; R, Arg; S, Ser;T,Thr; V, Val; W,Trp; and Y,Tyr.

selectivity filter

out-

inside

M3AM1A

M4A

C

NN

CM2A

Basal

+ BL-1249

19 9

t 1/2

of

MT

S-T

BA

Om

od

ific

atio

nat

L30

4[s

] 3.0

2.5

0.0

2.0

1.5

1.0

+ 2-APB

+ ML335

8 7

0.5

ML335

I/Im

ax

1.0

0.0

0.2

0.4

0.6

0.8

0 2 4

Time [s]

MTS-TBAO

6

BL-1249

2-APB

basal

1 3 5

L304C

ML335

MTS-TBAO

BL-1249

2-APB

7

TREK-1 L304C F

D

26

M2

0

10

20

30

BL

-124

9E

C50

[µM

]

TREK-1

L189CA301C

L188CA302C

V303CL304C

S305CM306C

I307CG308C

D309CW310C

F187CP183C

I182CG181C

L180CL184C

F185CWTG186C

P198A

L320B

I323B

BL-1249

M2A

M4B

S321B

E TREK-2G

(central nitrogen)

TEA

THexA

NH

NH

N

NN

Br

BL-1249BrA B

LLLLL

PPPPP

FFNNF

IIIII

183 304

GGGGG

TREK-1

TALK-1TALK-2

TRAAK

TREK-2

ASLLA

VVIIV

MTLLM

STPKS

LLLIL

M2 M4

198 320

18 15 18 6 19 5 16 6 88 101317 86 102018231426

BL-1249in THexA

BL-1249in TEA

BL-1249

Log BL-1249 [µM]

3210-1

TREK-2C1.0

0.8

0.6

0.4I/Im

ax

0.2

0.0

M4

K+

* * *

n.s

. n.s

.

M1A

M4A M3AM2A

RESEARCH | REPORTon A

pril 11, 2019

http://science.sciencemag.org/

Dow

nloaded from

Page 4: ION CHANNELS A pharmacological masterkey mechanism that … · 2019-04-11 · ION CHANNELS A pharmacological masterkey mechanism that unlocks the selectivity filter gate in K+ channels

TRAAK with BL-1249 modeled into the equiva-lent site in the TRAAK channel structure (PDB4I9W) (Fig. 3A). This indicated several changesinduced by BL-1249: (i) K+ occupancy at the S6site located adjacent to the negatively chargedtetrazole group of BL-1249 increased ~16-fold(Fig. 3, A and B), (ii) K+ occupancy of the S1 andS4 sites increased (Fig. 3B), and (iii) the rate ofK+ permeation increased by 1.6-fold (24 ± 2 ions/ms compared to 15 ± 2 ions/ms without BL-1249;Fig. 3C).The effect of BL-1249 on ion permeation was

further investigated with single-channel record-ings of TREK-2 expressed in human embryonickidney (HEK) 293 cells. Besides an increase inopen probability, an increase in the measuredsingle-channel amplitude was also observedin both the inward (from −29.3 ± 1.5 to −34.1 ±1.9 pA at −100 mV; n = 7) and outward (from17.7 ± 1.3 to 21.7 ± 1.4 pA at +100 mV; n = 7)directions in response to BL-1249 (Fig. 3, D andE). This result is consistent with the observedincrease in SF ion occupancy at S1 and S4 thatis expected to enhance ion permeation via adirect knock-on effect for ions entering the SFfrom either side (23). A similar increase in uni-tary conductance was also observed for TREK-1channels recorded in patches from Xenopusoocytes (fig. S4, A to C). Notably, increases insingle-channel conductance have not been ob-served upon activation of TREK-1, TREK-2, orTRAAK K2P channels by other physiologicalstimuli (24, 25).Collectively, these results indicate that BL-1249

increases ion permeation and channel-open prob-ability by influencing K+ occupancy at sites belowand within the SF. In line with this notion, muta-tions in the SF that change filter ion occupancyat the S1 and S4 sites (17, 26) and induce the ac-tivated “leak mode” in K2P channels (17) alsorender them insensitive to BL-1249 (and var-ious other activators discussed below; fig. S6,A to D).The negatively chargedmoiety identifiedwith-

in BL-1249, PD-118057, andNS11021 is also foundin a series of known activators of TREK-1 andTREK-2 K2P channels [ML67-33 (27), tetrazole;DCPIB (28), carboxylate], hERG channels [PD-307243 (29), carboxylate; NS3623 (30), tetrazole],and BKCa channels [GoSlo-SR-5-6 (31), sulpho-nate], and its requirement for channel activationhas been demonstrated for ML67-33 and GoSlo-SR-5-6 (27, 31). Indeed, these compounds alsoshare all the hallmark features of BL-1249 action,including polypharmacology [i.e., mutual activa-tion of K2P, BKCa, and hERG channels (Fig. 4, Cand D), sensitivity to QAL

+ (Fig. 4A; fig. S7, A toC; and tables S2 and S3] and mutations thatreduce BL-1249 activation in TREK-1 (fig. S4, Cand D). In addition, MD simulations of theirinteraction with structures of the TREK-2, BKCa,and hERG channel pores identified similar stablebinding poses below the SF with orientation ofthe negative moiety toward the cavity and a con-comitant increase in K+ occupancy at cavity andSF ion binding sites (Fig. 4, C and D, and figs.S7D and S8, A to C). Notably, this assumed NCA

binding site overlaps with the “promiscuous in-hibitor binding site” in the hERG channel, whichunderlies drug-induced long QT syndrome (12, 32).This site is thought to accommodate many hy-drophobic molecules (e.g., terfenadine), andconsistent with this, we found that activation byPD-118057 strongly reduced inhibition by ter-fenadine (Fig. 4B).The molecular features of the NCA compounds

define a common pharmacophore that, besidesthe negatively charged group, comprises both aro-matic and hydrophobic moieties (Fig. 4E). As acontrol, we tested tetrazole-containing compoundsthat do not fit this common pharmacophore onTREK-1, BKCa, and hERG channels and foundthat they were unable to promote channel acti-vation (fig. S9, A to C).Our results uncover a class of K+ channel

openers, the NCAs, that act as a universal masterkey to unlock the SF gate. Mechanistically, theseNCAs bind below the SF, where their negativecharge promotes K+ binding to the pore cavity,and thereby also alter the ion occupancy in theSF in a way that is known to promote activa-tion of the filter gate (17). We hypothesize that,

in particular, the increase at the S1 and S4 sitesis responsible for activating the SF gate becauseall NCA-responsive channels are also activated byRb+ permeation, which is thought to increaseion occupancy at these sites, whereas mutationsknown to reduce S1 and S4 ion occupancy inK2P channels abolish NCA activation. Further-more, a loss of K+ binding to the S1 site has beenimplicated in SF inactivation in Kv channels (33),hERG channels (32), and TREK-2 K2P channels(19). However, at this time, we cannot excludethe possibility that nonelectrostatic interactionsof the NCAs with their respective binding sitesalso contribute to the stabilization of the activeSF state because these sites involve gating-sensitiveregions [i.e., the TM4 (8, 19) and S6 segments(6, 34)]. In any case, our results support the viewthat many K2P channels, as well as BKCa channels,adopt a low-activity (i.e., inactivated) state of theirSF at rest and that the various physiologicalstimuli induce structural changes that drive theSF into an active (open) state. The NCAs appearto operate by means of bypassing these activa-tion mechanisms to directly stabilize the SF inits active state.

Schewe et al., Science 363, 875–880 (2019) 22 February 2019 4 of 6

A

TREK-2Dbasal activity

+ 3 µM BL-1249

50 ms10

pA

0 10 20 30 40

104

103

102

101

100

I [pA]C

ou

nts

10p

A

5 ms

oo

Basal

+ BL-1249

40

30

0

20

10

Sin

gle

chan

nel

amp

l.[p

A]

E TREK-2

Basal

+ BL-1249

0

-10

-40

-20

-30

* *

+100 mV -100 mV

* *

2

6

10

14

-2

-10

z-ax

isal

on

gS

F[Å

]

K+ permeation K+ permeation with BL-1249

BL-1249

2.9 Å

S0

S1

S2

S3

S4

S5

S6

TRAAK

0 2500 5000Frequency/µs

8.0 7.4

Fo

ldac

tiva

tio

n

1

100

10

F TALK-2

6.0 5.0

pHi

2421

14

14

5.07.4

BL-1249

2-APB

17 17

Per

mea

tio

nev

ents

/µs

25

20

0

30

15

10

5

Basal

+ BL-1249

C TRAAK* *

basal BL-1249

TRAAKB

Ion occupancy sites

20

1.5

1.0S0

S6

S2

S3

S4

S5

S1

// //

Fo

ldch

ang

ein

ion

occ

up

ancy

wit

hB

L-1

249

-6

Fig. 3. Effects of BL-1249 on pore K+ occupancy and permeation. (A) Ion occupancy(frequency/ms) of K+ binding sites (S0 to S6) obtained from permeation MD simulations ofTRAAK in the presence or absence of BL-1249, with BL-1249 adopting the identical position inTREK-2 (see Fig. 2G), that is, the negatively charged tetrazole ring (blue) in close proximityto the S6 K+ ion. Coordinates were saved every 40 ps. The black arrow points to the increasein S6 K+ occupancy. (B) Fold change in ion occupancy for the S0 to S6 sites. (C) MD simulations ofK+ permeation in TRAAK. Bars represent permeation events/ms from independent 200-ns MDsimulations without (n = 50) and with BL-1249 (n = 30). **P ≤ 0.01. (D) Single-channel TREK-2currents recorded at +80 mV from i-o patches before (basal) and after addition of 3 mM BL-1249,with arrows pointing to expanded scales of framed sections. The lower-middle panel depictscurrent amplitude histograms with or without 3 mM BL-1249 from i-o patches with ≤2 activechannels (the right-hand peak represents the amplitude of two BL-1249–activated channels).Dotted lines indicate the single-channel amplitude maxima with or without BL-1249. (E) Pairedsingle-channel current amplitudes before and after addition of 3 mM BL-1249 (n = 7). **P ≤ 0.01.(F) Fold activation of TALK-2 currents in i-o patches with 50 mM BL-1249 or 1 mM 2-APB applied atthe indicated pHi values. Error bars indicate SEM.

RESEARCH | REPORTon A

pril 11, 2019

http://science.sciencemag.org/

Dow

nloaded from

Page 5: ION CHANNELS A pharmacological masterkey mechanism that … · 2019-04-11 · ION CHANNELS A pharmacological masterkey mechanism that unlocks the selectivity filter gate in K+ channels

In addition, our findings have important im-plications for the development of drugs that tar-get K+ channels, because they reveal the bindingsites and the mechanism of action for many es-tablished activators in various K+ channels. Ourfindings have also identified the first activatorsfor several K2P channels (e.g., TALK-1/-2 andTHIK-1/-2). Notably, the NCA binding site over-laps with the promiscuous inhibitor site in hERG,and thus, targeting this NCA sitemight represent

a promising approach to circumvent the drug-induced longQT syndrome, which is, as of now, aserious burden in drug development (12). How-ever, the identified polypharmacology also rep-resents a challenge for the development of anyNCA-based compound into a highly subtype-specific K+ channel agonist. Nevertheless, struc-tural differences between K2P, hERG, and BKCa

channels may still permit a rational drug designthat reduces this promiscuity. However, in some

acute situations such as ischemic stroke or statusepilepticus, exploiting the polypharmacology ofNCAs to promote simultaneous opening of mul-tiple neuroprotective K+ channels (e.g., BKCa,TREK-1, TREK-2, TRAAK, THIK-1, and THIK-2)may even be beneficial.

REFERENCES AND NOTES

1. S. I. V. Judge, P. J. Smith, P. E. Stewart, C. T. Bever Jr., RecentPat. CNS Drug Discov. 2, 200–228 (2007).

Schewe et al., Science 363, 875–880 (2019) 22 February 2019 5 of 6

A TREK-1

DCPIB12 19

* * *

24

BL-1249

NC

AE

C50

[µM

]

1000

100

10

1

K2P activators hERG activators BKCa activators

8

* * *+

QA

L+

ML67-3310 9

* * *

9 12

* * *

+Q

AL

+

PD-3072439 5

* * *

+Q

AL

+

PD-11805711 5

* * *

+Q

AL

+

NS3623

+Q

AL

+

+Q

AL

+

7 5

* * *

NS11021X9

+Q

AL

+

8

* * *

+Q

AL

+

O

O

S

HN CF3

O

OO

NH2

NH

NN

NN

OO

O

ClO

Cl

NHHN

F3C

S

Br

CF3

NN

NN

NS11021

Cl

Cl

O

O

HN

PD-118057

N

O O

N

NHCl

Cl

PD-307243

NHHN

F3C

O

Br

NN

NN

NS3623

Cl

N

Cl

N N

NN

ML67-33

DCPIB

BL-1249

NCA Pharmacophore

81.5˚

33.5˚

5.3 Å

aromatic

negativelycharged

hydrophobic5.9 Å

3.2 Å

65˚

hERG activatorsK2P activatorsE BKCa activators

GoSlo-SR-5-6

Cchannel

NS11021

PD-118057

KCa

channelK+

D

B

Log Terfenadine [nM]

3210-1

1.0

0.8

0.6

0.4

I tail/

I tail-

ma x

0.2

0.0-2

Terfenadine+ PD-118057

Terfenadine

120 K+

120 K+

hERG

K+

K+

GoSlo-SR-5-6

Y652

F656

1

NS11021

BL-1249

NS3623

PD-307243

10

Fo

ldch

ang

eo

fac

tiva

tio

n

PD-118057

14 10 6 65

GoSlo-SR-5-61 10

NS11021

NS3623

12

ML67-33

10

DCPIB

10

BL-1249

PD-118057

PD-307343

12 714

10

100F

old

chan

ge

of

acti

vati

on

13

GoSlo-SR-5-6

P309C

I301B

A305C

A305B I308C

I308B

BhERG

Fig. 4. NCA binding sites and common pharmacophores. (A) EC50

values (at +40 mV) for TREK-1 activation with compounds described asactivators of either K2P, hERG, or BKCa channels. Competitive antagonismis seen in the presence of QAL

+ (either THexA or TPenA, which produce~70 to 80% inhibition of respective basal K+ currents). ***P ≤ 0.001; error barsindicate SEM. (B) Terfenadine inhibition of hERG channels with or without10 mM PD-118057. Error bars indicate SEM. (C) Structure of the hERGchannel (PDB 5VA1) with the pore region expanded. This region was used formolecular docking and MD simulations to obtain the favored binding pose ofPD-118057 (orange). Terfenadine-interacting residues are highlighted. Thecarboxylate group (red) interacts with a K+ ion below the SF (see also fig. S7D);

the bar chart below represents the fold activation of hERG tail currents at−100 mV with 10 mM of the indicated compounds. Error bars indicate SEM.(D) Same as in (C) but for the BKCa channel (PDB 5TJI), showing the favoredbinding pose of NS11021 (orange) where the tetrazole group (red) interactswith a K+ ion below the SF and the residues in proximity to NS11021highlighted. The bar chart below shows the fold activation of BKCa at +100 mVwith 10 mM of the indicated compounds (at zero Ca2+). Error bars indicateSEM; the B and C superscripts indicate the subunit of the tetramer.(E) Representation of the K2P, hERG, and BKCa activators used to generate acommon NCA pharmacophore consisting of aromatic (blue), hydrophobic(gray), and acidic (red) moieties, with distances and angles as shown.

RESEARCH | REPORTon A

pril 11, 2019

http://science.sciencemag.org/

Dow

nloaded from

Page 6: ION CHANNELS A pharmacological masterkey mechanism that … · 2019-04-11 · ION CHANNELS A pharmacological masterkey mechanism that unlocks the selectivity filter gate in K+ channels

2. V. K. Vyas, P. Parikh, J. Ramani, M. Ghate, Curr. Med. Chem.10.2174/0929867325666180430152023 (2018).

3. S. Hou, S. H. Heinemann, T. Hoshi, Physiology 24, 26–35 (2009).4. M. I. Niemeyer, L. P. Cid, W. González, F. V. Sepúlveda,

Mol. Pharmacol. 90, 309–317 (2016).5. F. V. Sepúlveda, L. Pablo Cid, J. Teulon, M. I. Niemeyer,

Physiol. Rev. 95, 179–217 (2015).6. G. Yellen, Nature 419, 35–42 (2002).7. P. L. Piechotta et al., EMBO J. 30, 3607–3619 (2011).8. S. N. Bagriantsev, R. Peyronnet, K. A. Clark, E. Honoré,

D. L. Minor Jr., EMBO J. 30, 3594–3606 (2011).9. C. M. Wilkens, R. W. Aldrich, J. Gen. Physiol. 128, 347–364

(2006).10. Y. Zhou, X.-M. Xia, C. J. Lingle, Proc. Natl. Acad. Sci. U.S.A.

108, 12161–12166 (2011).11. P. L. Smith, T. Baukrowitz, G. Yellen, Nature 379, 833–836

(1996).12. J. I. Vandenberg, E. Perozo, T. W. Allen, Trends Pharmacol. Sci.

38, 899–907 (2017).13. L. Pope et al., ACS Chem. Neurosci. 9, 3153–3165 (2018).14. J. Zhou et al., Mol. Pharmacol. 68, 876–884 (2005).15. B. H. Bentzen et al., Mol. Pharmacol. 72, 1033–1044 (2007).16. M. J. Lenaeus, D. Burdette, T. Wagner, P. J. Focia, A. Gross,

Biochemistry 53, 5365–5373 (2014).17. M. Schewe et al., Cell 164, 937–949 (2016).18. J. G. McCoy, C. M. Nimigean, Biochim. Biophys. Acta 1818,

272–285 (2012).19. Y. Y. Dong et al., Science 347, 1256–1259 (2015).20. M. Rapedius et al., Channels 6, 473–478 (2012).21. R.-G. Zhuo et al., Front. Cell. Neurosci. 10, 127 (2016).22. M. Lolicato et al., Nature 547, 364–368 (2017).23. W. Kopec et al., Nat. Chem. 10, 813–820 (2018).24. M. V. Clausen, V. Jarerattanachat, E. P. Carpenter,

M. S. P. Sansom, S. J. Tucker, Proc. Natl. Acad. Sci. U.S.A. 114,E8343–E8351 (2017).

25. D. Kang, C. Choe, D. Kim, J. Physiol. 564, 103–116 (2005).

26. M. Zhou, R. MacKinnon, J. Mol. Biol. 338, 839–846(2004).

27. S. N. Bagriantsev et al., ACS Chem. Biol. 8, 1841–1851(2013).

28. L. Minieri et al., Br. J. Pharmacol. 168, 1240–1254 (2013).29. E. Gordon et al., Mol. Pharmacol. 73, 639–651 (2008).30. R. S. Hansen et al., Mol. Pharmacol. 70, 1319–1329 (2006).31. S. Roy et al., ChemMedChem 7, 1763–1769 (2012).32. W. Wang, R. MacKinnon, Cell 169, 422–430.e10 (2017).33. V. Pau, Y. Zhou, Y. Ramu, Y. Xu, Z. Lu, Nat. Struct. Mol. Biol.

24, 857–865 (2017).34. H. Yang, G. Zhang, J. Cui, Front. Physiol. 6, 29 (2015).

ACKNOWLEDGMENTS

We thank F. Lesage for the THIK-1 and THIK-2 K2P channel clones;M. A. Hollywood for providing the BKCa activator GoSlo-SR-5-6;and J. Kusch for initially testing the BL-1249 effect on CNGA1channels. We thank all members of our respective laboratoriesfor technical support and comments on the manuscript. We thankDiamond Light Source Ltd. and its staff for access to themacromolecular crystallography beamlines. We also acknowledgethe North-German Supercomputing Alliance (HLRN) for providingHigh Performance Computing (HPC) resources that havecontributed to the research results reported in this paper. Funding:T.B. and B.L.d.G. were supported by the DFG; B.F. was supportedby the DFG (SFB746, TRR 152, and EXC 294 and 2189); S.J.T.and E.P.C were supported by a BBSRC Industrial PartnershipAward (BB/N009274/1); and L.J.C. was supported by a WellcomeTrust (OXION) Ph.D. studentship. A.M. was funded by the EPSRCLife Sciences Interface Doctoral Training Centre. A.C.W.P. andE.P.C. are members of the SGC (charity reference no. 1097737)funded by AbbVie, Bayer Pharma AG, Boehringer Ingelheim, theCanada Foundation for Innovation, Genome Canada, Janssen,Merck KGaA, MSD, Novartis, the Ontario Ministry of EconomicDevelopment and Innovation, Pfizer, São Paulo ResearchFoundation-FAPESP, and Takeda, as well as the Innovative

Medicines Initiative Joint Undertaking ULTRA-DD grant 115766 andthe Wellcome Trust 106169/Z/14/Z. D.C.P. was an employee ofPfizer at the time of the research. Author contributions: M.S. andT.B. conceived the study and designed the electrophysiologicalexperiments. M.S., Ü.M., F.S., and M.T. performed all inside-outpatch-clamp experiments in Xenopus oocytes. M.S. and T.B.analyzed the data. M.M. designed K+ channel mutations. C.C.performed whole-cell recordings in CHO cells and analyzed thedata together with B.F. Single-channel recordings in Xenopusoocytes were carried out and analyzed by K.S.V. and A.K.K.,supervised by N.D. Single-channel recordings in HEK293 cells wereperformed and analyzed by L.J.C., supervised by S.J.T. A.M.purified TREK-2 and cocrystallized TREK-2 with BL-1249Br. A.M.and A.C.W.P. obtained and analyzed the x-ray data, supervisedby E.P.C. and S.J.T. D.C.P. synthesized the brominated BL-1249.H.B. synthesized ML67-33, supervised by M.N. H.S. designed,performed, and analyzed all molecular dockings and MDsimulations with critical comments of B.L.d.G. W.G. calculatedand analyzed the NCA pharmacophore together with M.S. andN.D. M.S. and M.M. prepared and edited all figures. T.B., M.S., B.F.,E.P.C., and S.J.T. contributed to the writing and editing of themanuscript and approved the manuscript. Competing interests:The authors declare no competing interests. Data and materialsavailability: All data are available in the main text or thesupplementary materials. Requests for materials should beaddressed to the corresponding authors.

SUPPLEMENTARY MATERIALS

www.sciencemag.org/content/363/6429/875/suppl/DC1Materials and MethodsFigs. S1 to S9Tables S1 to S3References (35–67)

9 August 2018; accepted 28 January 201910.1126/science.aav0569

Schewe et al., Science 363, 875–880 (2019) 22 February 2019 6 of 6

RESEARCH | REPORTon A

pril 11, 2019

http://science.sciencemag.org/

Dow

nloaded from

Page 7: ION CHANNELS A pharmacological masterkey mechanism that … · 2019-04-11 · ION CHANNELS A pharmacological masterkey mechanism that unlocks the selectivity filter gate in K+ channels

channels+A pharmacological master key mechanism that unlocks the selectivity filter gate in K

Thomas BaukrowitzHassane Belabed, Marc Nazare, Bert L. de Groot, Niels Decher, Bernd Fakler, Elisabeth P. Carpenter, Stephen J. Tucker andS. Vowinkel, Linus J. Conrad, Aytug K. Kiper, Wendy Gonzalez, Marianne Musinszki, Marie Tegtmeier, David C. Pryde, Marcus Schewe, Han Sun, Ümit Mert, Alexandra Mackenzie, Ashley C. W. Pike, Friederike Schulz, Cristina Constantin, Kirsty

DOI: 10.1126/science.aav0569 (6429), 875-880.363Science 

, this issue p. 875Sciencethe channels. Targeting this NCA site might be exploited in rational drug design.molecular dynamics simulations showed that the NCAs bind below the selectivity filter to open the filter gate and activatesimilar mechanism to activate many types of potassium channels. X-ray crystallography, functional analysis, and

report a class of negatively charged activators (NCAs) with a defined pharmacore that use aet al.and pain. Schewe Using drugs to activate potassium channels has the potential to treat conditions like epilepsy, heart arrhythmias,

A key to potassium channel activation

ARTICLE TOOLS http://science.sciencemag.org/content/363/6429/875

MATERIALSSUPPLEMENTARY http://science.sciencemag.org/content/suppl/2019/02/20/363.6429.875.DC1

REFERENCES

http://science.sciencemag.org/content/363/6429/875#BIBLThis article cites 64 articles, 15 of which you can access for free

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Terms of ServiceUse of this article is subject to the

is a registered trademark of AAAS.Sciencelicensee American Association for the Advancement of Science. No claim to original U.S. Government Works. The title Science, 1200 New York Avenue NW, Washington, DC 20005. 2017 © The Authors, some rights reserved; exclusive

(print ISSN 0036-8075; online ISSN 1095-9203) is published by the American Association for the Advancement ofScience

on April 11, 2019

http://science.sciencem

ag.org/D

ownloaded from