Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2025 Apr 28;122(18):e2425494122. doi: 10.1073/pnas.2425494122

Design and structural basis of selective 1,4-dihydropyridine inhibitors of the calcium-activated potassium channel KCa3.1

Seow Theng Ong a,1, Young-Woo Nam b, Joshua A Nasburg c, Alena Ramanishka b, Xuan Rui Ng a, Zhong Zhuang a, Stephanie Shee Min Goay a, Hai M Nguyen c, Latika Singh c, Vikrant Singh c, Alicia Rivera d,e, M Elaine Eyster f, Yang Xu g, Seth L Alper d,e, Heike Wulff c, Miao Zhang b, K George Chandy a,1
PMCID: PMC12067266  PMID: 40294255

Significance

An approach to turn old drugs into new drugs is “selective optimization of a side activity (SOSA).” Here, we modified the 1,4-dihydropyridine nucleus and transformed its side activity (weak blockade of the calcium-activated potassium channel KCa 3.1) into the main activity while diminishing the original main activity (blockade of L-type voltage-gated calcium channels). The new 1,4-dihydropyridines block KCa 3.1 channels with single-digit nanomolar potency and excellent selectivity, binding in a water-filled cavity in KCa 3.1’s pore to impede ion permeation. In proof-of-concept studies, the 1,4-dihydropyridines blocked mutant KCa 3.1 channels that cause hereditary xerocytosis and effectively treated ischemic stroke in a rat model. KCa 3.1-selective 1,4-dihydropyridines have considerable potential as therapeutics.

Keywords: KCa3.1/KCNN4; 1,4-dihydropyridine; cryo-EM; hereditary xerocytosis; acute ischemic stroke

Abstract

The 1,4-dihydropyridines, drugs with well-established bioavailability and toxicity profiles, have proven efficacy in treating human hypertension, peripheral vascular disorders, and coronary artery disease. Every 1,4-dihydropyridine in clinical use blocks L-type voltage-gated calcium channels. We now report our development, using selective optimization of a side activity (SOSA), of a class of 1,4-dihydropyridines that selectively and potently inhibit the intermediate-conductance calcium-activated K+ channel KCa3.1, a validated therapeutic target for diseases affecting many organ systems. One of these 1,4-dihydropyridines, DHP-103, blocked KCa3.1 with an IC50 of 6 nM and exhibited exquisite selectivity over calcium channels and a panel of >100 additional molecular targets. Using high-resolution structure determination by cryogenic electron microscopy together with mutagenesis and electrophysiology, we delineated the drug binding pocket for DHP-103 within the water-filled central cavity of the KCa3.1 channel pore, where bound drug directly impedes ion permeation. DHP-103 inhibited gain-of-function mutant KCa3.1 channels that cause hereditary xerocytosis, suggesting its potential use as a therapeutic for this hemolytic anemia. In a rat model of acute ischemic stroke, the second leading cause of death worldwide, DHP-103 administered 12 h postischemic insult in proof-of-concept studies reduced infarct volume, improved balance beam performance (measure of proprioception) and decreased numbers of activated microglia in infarcted areas. KCa3.1-selective 1,4-dihydropyridines hold promise for the many diseases for which KCa3.1 has been experimentally confirmed as a therapeutic target.


1,4-dihydropyridines were first introduced as therapeutics in 1975 and now rank among the most prescribed medicines in the world (1, 2). They are widely used to treat human hypertension, peripheral vascular disorders, and coronary artery disease (1, 2). Every 1,4-dihydropyridine approved for therapeutic use in the past five decades blocks L-type voltage-gated calcium channels (CaV1.x subfamily) (1, 2). High-resolution structural studies using cryogenic electron microscopy (cryo-EM) reveal that 1,4-dihydropyridines bind in a lipid bilayer-accessible hydrophobic fenestration between repeats III and IV of mammalian CaV1.x channels (3, 4). The 1,4-dihydropyridine nucleus has been decorated by diverse substituents to generate analogues against non-Cav1.x targets including receptors (e.g., adenosine receptors) (5) and transporters (e.g., multidrug resistance protein 4) (6). However, these 1,4-dihydropyridines exhibit significantly lower potency and/or selectivity for their specific targets than do 1,4-dihydropyridine CaV1.x inhibitors. Here, we describe a class of 1,4-dihydropyridines that block the intermediate-conductance Ca2+-activated K+ channel KCa3.1 (KCNN4, IKCa1, SK4, Gardos channel) (711) with single-digit nanomolar potency and remarkable selectivity.

The KCa3.1 channel functions as a cation counterbalancer, sustaining calcium signaling in B and T lymphocytes, proliferating myofibroblasts, microglia, macrophages, glioblastoma cells, and other cell types through K+ efflux-driven hyperpolarization of the plasma membrane (12). The channel is also the primary conduit for conductive K+ efflux in red blood cells (10, 11). Despite KCa3.1’s broad tissue distribution, the KCa3.1-specific triarylmethane inhibitor, Senicapoc, was well tolerated in human trials, suggesting that selective blockade of KCa3.1 is safe in vivo (13). Moreover, knockout of the gene encoding KCa3.1 in mice does not cause a grossly apparent deleterious phenotype, further supporting the safety in vivo of specific KCa3.1 blockade (12). KCa3.1 has been experimentally verified in animal models as a therapeutic target for varied diseases including neuroinflammatory diseases (acute ischemic stroke, Alzheimer’s disease, perioperative neurocognitive disorders, spinal cord injury, traumatic brain injury), fibrosis (lung, liver, kidney, heart), inflammatory bowel disease, atherosclerosis, vascular restenosis, hemolytic anemias (hereditary xerocytosis, sickle cell disease), allergic airway disease, secretory diarrheas, and glioblastoma (12, 14). KCa3.1-selective 1,4-dihydropyridines may therefore have wide therapeutic use. One of the 1,4-dihydropyridines, DHP-103, blocked KCa3.1 potently (IC50 6 nM) and with >500-fold selectivity over CaV1.x channels and >100 additional molecular targets. High-resolution cryo-EM structural studies combined with mutagenesis and electrophysiology revealed that DHP-103 binds in the water-filled central cavity beneath KCa3.1’s selectivity filter to block K+ permeation through the channel pore. In proof-of-concept efficacy studies, DHP-103 inhibited gain-of-function mutant KCa3.1 channels that cause hereditary xerocytosis, a hemolytic anemia, and showed efficacy in a rat model of acute ischemic-reperfusion stroke.

Results and Discussion

Identification of KCa3.1 Channel-Selective 1,4-Dihydropyridine Inhibitors.

More than two decades ago, the 1,4-dihydropyridines nifedipine, nitrendipine, and nimodipine were reported to block the recombinant human KCa3.1 channel, albeit at >200-fold lower potency than for blockade of CaV1.x channels (15). A few years earlier, a single nitrendipine derivative (compound 29) was shown to inhibit the human erythrocyte KCa3.1 channel more potently than the L-type voltage-gated calcium channel of guinea pig ileal longitudinal muscle (16). These studies suggested that KCa3.1-selective 1,4-dihydropyridines could be developed. Old drugs can be turned into new drugs through “selective optimization of a side activity (SOSA).” We have now used a SOSA approach (17) to modify the 1,4-dihydropyridine scaffold so as to transform its side activity (weak blockade of KCa3.1) into its main activity (potent blockade of KCa3.1) while reducing the original main activity (blockade of L-type voltage-gated calcium channels).

We started with commercially available 1,4-dihydropyridines known to inhibit CaV1.x channels, some used clinically as drugs, and designed and synthesized a series of 1,4-dihydropyridines. Using patch clamp recordings, we identified 1,4-dihydropyridines that block KCa3.1 channels with single-digit nanomolar potency. Fig. 1 AC and G shows the effects of exemplar 1,4-dihydropyridines on human KCa3.1-mediated K+ currents in transfected HEK293 cells elicited by voltage ramps (−120 mV to 40 mV from holding potential −80 mV) with 1 µM free Ca2+ in the pipette solution. SI Appendix, Fig. S1 A and B lists the analyzed 1,4-dihydropyridines, their moieties at positions R1-R8, and their IC50 values for block of KCa3.1. The 1,4-dihydropyridines cluster into four groups based on IC50 values: group 1 (>5 µM), group 2 (1-5 µM), group 3 (<200 nM), and group 4 (<30 nM) (SI Appendix, Fig. S1B). We attribute the order-of-magnitude improvement in the potency of group 3 vs. group 1 to the presence of methyl acetate or propan-2-one at R5 and R6 (SI Appendix, Fig. S1B). Further increases in potency of group 4 1,4-dihydropyridines reflect removal of NO2 (from either R3 or R4) or a halogen (from R4), and introduction of a halogen (F or Cl) at R1 and CF3 at R3 (Fig. 1A and SI Appendix, Fig. S1B).

Fig. 1.

Fig. 1.

Novel 1,4-dihydropyridines block KCa3.1 channels significantly more potently than L-type voltage-gated CaV1.2 calcium channels. Blockade by three exemplar 1,4-dihydropyridines from groups 4a (DHP-101), 4b (DHP-104), and 4c (DHP-103) on human KCa3.1 currents (AC) and human CaV1.2 currents (DF). For CaV1.2 current measurements, we used 0.5 µM nifedipine at the end of each experiment to determine complete block. (G) Concentration–response curves of DHP-101, DHP-104, DHP-103 on KCa3.1 and CaV1.2 currents are compared (n ≥ 3, mean ± SD).

Since 1,4-dihydropyridines are potent inhibitors of L-type voltage-gated CaV1.x channels, we compared block of CaV1.2 and KCa3.1 currents by these compounds (SI Appendix, Fig. S1C). The group 2 1,4-dihydropyridine DHP-07 blocked CaV1.2 channels far more potently than the KCa3.1 channel, while 1,4-dihydropyridines from groups 3, 4a, 4b, and 4c inhibited KCa3.1 significantly more potently than CaV1.2 (Fig. 1 DG and SI Appendix, Fig. S1C). Group 4b and 4c 1,4-dihydropyridines showed 600- to 2,000-fold selectivity for KCa3.1 over CaV1.2 (Fig. 1 EG and SI Appendix, Fig. S1C). To further evaluate selectivity, we examined the ability of group 4c compound DHP-103 (SI Appendix, Fig. S1 A and B) to inhibit [3H]nitrendipine binding to the 1,4-dihydropyridine binding site on CaV1.x subfamily channels expressed in the rat cerebral cortex. At 3 µM, DHP-103 caused a ~45% inhibition of [3H]nitrendipine binding (SI Appendix, Fig. S2), which is roughly 500-fold higher than its IC50 (6 nM) for KCa3.1 channel block. These data derived from electrophysiological and radionuclide-labeled antagonist binding assays demonstrate that groups 4b and 4c 1,4-dihydropyridines exhibit significant selectivity for KCa3.1 channels over CaV1.x channels. While both H and CH3 are tolerated at positions R7 and R8 for potent inhibition of KCa3.1, H at both R7 and R8 (group 4c) is preferred for KCa3.1-selectivity over CaV1.2 (Fig. 1G and SI Appendix, Fig. S1 AC).

We next measured the ability of group 4c compound, DHP-103, to block other ion channels. DHP-103 showed ≥ 1,000-fold selectivity for KCa3.1 over a panel of 13 ion channels, including closely related calcium-activated K+ channels (KCa1.1, KCa2.2, KCa2.3), and channels critical for normal cardiac function (KV11.1/hERG, KV1.4, KV1.5, KV4.2, NaV1.5) (SI Appendix, Table S1). As further assessment of selectivity, we tested DHP-103 on a panel of 97 molecular targets including channels, transporters, receptors, and kinases (Eurofins pharmacological P9 diversity panel safety screen), and on 5 cytochrome P450 enzymes. DHP-103 (3 µM) had minimal effect on 94 targets, while it caused 40 to 60% inhibition of three targets (progesterone receptor, glucocorticoid receptor, and the DHP-binding site on CaV1.x subfamily channels) (SI Appendix, Fig. S2). Since the concentration of DHP-103 tested (3 µM) is 500-fold higher than its IC50 (6 nM) for KCa3.1 blockade (Fig. 1G and SI Appendix, Fig. S1), we conclude that DHP-103 exhibits significant selectivity for KCa3.1 over all 97 molecular targets. DHP-103 also exhibited >800-fold selectivity for KCa3.1 over the five tested cytochrome P450 enzymes (SI Appendix, Table S2). These studies demonstrate that DHP-103 blocks KCa3.1 with excellent potency and exquisite selectivity over 116 other therapeutic targets. We attribute DHP-103’s single-digit nanomolar potency and remarkable KCa3.1-selectivity to large lipophilic groups (F, CF3) at R1 and R3, ketoesters at R5 and R6, and H at R4, R7, and R8 (Fig. 1 and SI Appendix, Fig. S1).

Ketoesters such as those occupying positions R5 and R6 are generally considered to negatively impact drug stability. We therefore compared the metabolic stability of group 4c 1,4-dihydropyridines in mouse and human liver microsomes to that of verapamil, a calcium channel blocking drug used to treat human hypertension, cardiac arrhythmias, and angina. DHP-103, DHP-110, and DHP-113 exhibited significantly greater metabolic stability than verapamil based on measured t1/2 and Cl’int (SI Appendix, Table S3). In vitro biotransformation of DHP-103 in liver microsomes from the mouse, rat, dog, and monkey (1-h duration) resulted in one major metabolite and ten minor metabolites (SI Appendix, Fig. S3 A and B). Furthermore, DHP-103’s pharmacokinetic properties in mice (see later) are similar to Felodipine’s (18), a CaV1.x-channel-blocking 1,4-dihydropyridine with ketoesters at R5 and R6 (like DHP-103), which is used to treat human hypertension and stroke.

In summary, we successfully transformed a canonical blocker of L-type voltage-gated calcium channels (CaV1.x) into a class of potent and selective inhibitors targeting the intermediate-conductance calcium-activated KCa3.1 channel.

DHP-103 Binds in a Water-Filled Cavity in the KCa3.1 Pore.

A recent high-resolution cryo-EM structure defined the mechanism of Ca2+-calmodulin-dependent activation of KCa3.1 (19). However, no high-resolution KCa3.1 structure in complex with an inhibitor or activator has been reported. We used cryo-EM structure-determination to identify the DHP-103 binding site in full-length human KCa3.1 complexed with calmodulin (CaM) in the presence of 2 mM Ca2+. Through cryo-EM single particle reconstruction we obtained a cryo-EM map of the Ca2+- and DHP-103-bound KCa3.1/CaM complex (henceforth referred to as 103_KCa3.1) at 3.45 Å resolution with C4 symmetry and 3.84 Å resolution with C1 symmetry, respectively (SI Appendix, Fig. S4 and Table S4). Our drug-bound 103_KCa3.1 structure shows better alignment with the Ca2+-bound apo_KCa3.1/CaM activation state I (apo_KCa3.1_I; Protein Data Bank [PDB]: 6cnn; RMSD: 1.335 Å) than apo_KCa3.1 activation state II (apo_KCa3.1_II; PDB: 6cno; RMSD: 2.21 Å) (19). The superimposition of the drug-bound 103_KCa3.1 and apo_KCa3.1_I structures is shown in Fig. 2A. DHP-103 is not displayed in this superimposition, but is shown in the channel pore structure of 103_KCa3.1 in Fig. 2B. Four K+ (K1 to K4) occupy the selectivity filter in the three structures with an additional K+ (K5) in the central cavity of the drug-bound 103_KCa3.1 structure (Fig. 2A). The closely spaced ions within the selectivity filter are likely a result of integrated electron densities from multiple K+-bound channels, since molecular dynamics simulation of individual channels suggest that water molecules may occupy positions between two K+ ions.

Fig. 2.

Fig. 2.

Binding site for 1,4-dihydropyridine in KCa3.1. (A) Comparison of our drug-bound 103_KCa3.1 structure (KCa3.1, gold; CaM, pink) with apo_KCa3.1_1 (pdb:6cnn) (KCa3.1, cyan; CaM, gray). The additional K+ in the central cavity of our drug-bound 103_KCa3.1 structure is identified by a red arrow. (B) EM density of DHP-103 (pink) is shown in the central cavity sandwiched between the innermost K+ (K4, green) in the selectivity filter and the additional K+ (K5, green) in the central cavity of the drug-bound 103_KCa3.1 structure. Sidechains of Thr250, Val275, Thr278, and Val275 (yellow) are labeled. (C) Effect of DHP-103 on KCa3.1_wild-type, KCa3.1_T250S-V275A, and KCa3.1_T212F-V272F K+ currents. (D) Concentration–response curves of DHP-103 on KCa3.1_wild-type vs. KCa3.1_T250S-V275A and KCa3.1_T212F-V272F K+ currents.

DHP-103’s EM density was seen in maps refined with both C4 and C1 symmetry (Fig. 2B and SI Appendix, Fig. S4) in the water-filled central cavity below the selectivity filter, sandwiched between the innermost K+ in the selectivity filter (K4) and the additional K+ in the central cavity (K5). DHP-103’s interactions with these two K+ ions seem a key determinate of its central cavity binding and action on the KCa3.1 channel. Negatively charged activator drugs have been reported to bind below the selectivity filters of KV11.1, K2P, and KCa1.1 channels and promote K+-binding to their central cavities (20). Our drug-bound 103_KCa3.1 structure shows direct interactions of permeant ions with a channel-bound inhibitor drug.

DHP-103’s EM density was seen in close proximity to and between the sidechains of Thr250 and Val275 in all four subunits of the KCa3.1 homotetramer (Fig. 2B). The four Thr250 sidechains contribute to the binding site for the innermost K+ (K4) in KCa3.1’s selectivity filter (19, 21). We used mutagenesis and electrophysiology to test DHP-103’s interactions with Thr250 and Val275 as predicted by the structure. Since DHP-103 is 1000-fold less effective vs. KCa2.2 (IC50 >10 µM) and KCa2.3 (IC50 6 µM) (SI Appendix, Table S1), we replaced Thr250 and Val275 in KCa3.1 with the corresponding residues from KCa2.2/KCa2.3. DHP-103 blocked the KCa3.1_T250S-V275A mutant channel (IC50 2 µM) with ~333-fold lower potency than the wild-type KCa3.1 channel (IC50 6 nM), confirming that Thr250 and Val275 are essential for the high-affinity binding of DHP-103 to KCa3.1 (Fig. 2 C and D). DHP-103’s EM density is also in proximity to Thr278, while ion K5 is near Val282 (Fig. 2B). Although we have not tested the effects of mutating these two residues, it is possible that DHP-103’s interactions with residues Thr250, Val275, Thr278, and the two K+ ions K4 and K5, and K5’s interaction with Val282, contribute to both stabilization of DHP-103 binding within the central cavity and to blockade of ion permeation through the pore. Interestingly, earlier mutagenesis and molecular modeling studies predicted that three other potent small molecule inhibitors of KCa3.1, the triarylmethane TRAM-34 (22), the benzothiazinone NS6180 (23), and the 4-phenyl-pyran-3 rac-11 (24), each directly blocked K+ permeation by interacting with the sidechains of Thr250 and Val275 in the central cavity (21, 25). In contrast, nifedipine, a CaV1.x-selective 1,4-dihydropyridine with micromolar potency for KCa3.1, was predicted to bind in a lipid bilayer-accessible hydrophobic fenestration between adjacent subunits (21), analogous to the binding site of CaV.1x-selective 1,4-dihydropyridines in the L-type CaV1.x channel (3, 4). Interaction with Thr250 and Val275 in the central cavity and blockade of ion permeation thus appears to be a feature of small molecule inhibitors (1,4-dihydropyridines, triarylmethanes, benzothiazinones, 4-phenyl-pyran-3) that block KCa3.1 with low nanomolar potency.

DHP-103 may reach its binding site within the central cavity in the fully activated (apo_KCa3.1_II) channel either by crossing the entire membrane and then entering through the inner gate (10.2 Å diameter), or by traversing half the membrane and accessing the binding site through the lipid bilayer-accessible hydrophobic fenestration between adjacent subunits (9.3 Å diameter) (SI Appendix, Fig. S5). To distinguish between these two possibilities, we narrowed the fenestrae by replacing Thr212 and Val272 with bulky phenylalanines (21), and then measured block of the mutant channel by DHP-103. Narrowing the fenestration with the KCa3.1_T212F-V272F mutation caused a small (~4-fold) reduction in DHP-103’s potency (IC50: 23 nM) compared to KCa3.1_wild-type (IC50 6 nM), whereas the KCa3.1_T250S-V275A pore mutation reduced DHP-103 potency (IC50: 2 µM) by ~333-fold Fig. 2 C and D). This result potentially excludes the fenestration as DHP-103’s point-of-entry and suggests that the open inner gate in the fully activated state (10.2 Å wide) may be the passage through which DHP-103 reaches it binding site within the central cavity. Interestingly, the inner gate is narrower (7 Å wide) in drug-bound 103_KCa3.1 (SI Appendix, Fig. S5), perhaps suggesting that the inner gate narrows after DHP-103 fits into its binding site in the central cavity. These ideas need to be tested by molecular dynamics simulation and kinetic studies.

Since KCa3.1 is a calcium-activated K+ channel, we examined whether the internal calcium concentration affected DHP-103 potency. Changing the internal free calcium concentration from 0.5 to 10 µM did not affect block by 50 nM DHP-103, a concentration that causes >80% block in the presence of 1 μM Ca2+ (Fig. 1 C and G and SI Appendix, Fig. S6). Increasing the external K+ concentration from 4.5 to 140 mM did not alter the IC50 for DHP-103 (compare Fig. 1 and Fig. 3). This result excludes displacement of DHP-103 from its binding site in the central cavity by the permeant ion.

Fig. 3.

Fig. 3.

DHP-103 blocks gain-of-function mutations of human KCa3.1 that cause hereditary xerocytosis. (A and B) Side view (A) and extracellular view (B) of apo-KCa3.1_1 (PDB: 6cnn) (CaM, gray; KCa3.1, light blue) with locations of gain-of-function mutations shown as purple spheres in one subunit. (C) Effect of DHP-103 on KCa3.1_wild-type and gain-of-function KCa3.1 mutants KCa3.1_S314P, KCa3.1_A322V, and KCa3.1_R352H. (D and E) Concentration–response curves (D) and IC50 values of DHP-103 (E) on KCa3.1_wild-type channels and gain-of-function KCa3.1 mutant channels KCa3.1_S314P, KCa3.1_A322V, and KCa3.1_R352H (n ≥ 6, mean ± SD; **P < 0.001 vs. KCa3.1_wild-type; one-way ANOVA followed by Tukey’s test). (F and G) Block by DHP-103 (F) and TRAM-34 (G) of A23187-stimulated 83Rb+ influx into red blood cells from a healthy human donor and HX patients with the heterozygous KCa3.1 gain-of-function mutations R352H (F) and V282M (F and G).

Collectively, our cryo-EM, mutagenesis, and electrophysiology data show that binding of DHP-103 in KCa3.1’s water-filled central cavity impedes ion permeation. We could not delineate the precise orientation of DHP-103 and the positioning of its moieties (at R1 to R8) within its KCa3.1 binding site, as symmetry-mismatch between the fourfold symmetrical KCa3.1 channel and the multiple rotational orientations of the flat DHP-103 phenyl ring prevented modeling of DHP-103 into its cryo-EM density.

DHP-103 Blocks KCa3.1 Mutants that Cause Hereditary Xerocytosis.

Hereditary xerocytosis (HX) (a.k.a dehydrated hereditary stomatocytosis; MIM: 194380) is a disease characterized by erythrocyte dehydration, generally well compensated hemolytic anemia, and variable iron overload, perinatal edema, and susceptibility to thrombotic events (2635). The disease is linked to mutations of two genes, PIEZO1 (encoding the mechanosensitive nonselective cation channel, PIEZO1) and KCNN4 (encoding KCa3.1) (2635). Gain-of-function mutations of either channel are thought to result in erythrocyte dehydration via excessive loss of K+ and water (2635). Senicapoc, a triarylmethane inhibitor of KCa3.1, reduces red blood cell K+ loss and dehydration due to gain-of-function mutations of KCa3.1 or PIEZO1 (32, 33) and is being evaluated in a clinical trial in HX patients with the V282M mutation (ClinicalTrial.gov: NCT04372498). KCa3.1-selective 1,4-dihydropyridines might therefore be useful for treatment of HX if they block disease-causing gain-of-function mutant KCa3.1 channels. We tested DHP-103 on HX-causing gain-of-function mutations of KCa3.1 located either in the CaM-binding region of KCa3.1 (S314P, A322V, R352H) or at the inner gate in the ion conduction pathway (V282M) (Fig. 3 A and B). In patch-clamp experiments, DHP-103 blocked recombinant wild-type and mutant (S314P, A322V, and R352H) KCa3.1 channels with low nanomolar potency (IC50 < 20 nM), though mutant A322V was significantly less sensitive than the wild-type KCa3.1 channel (Fig. 3 CE). In calcium ionophore-initiated (A23187) 83Rb+ (surrogate for K+) flux studies, we compared red blood cells from two HX patients with the heterozygous gain-of-function KCa3.1 mutant R352H versus healthy control red blood cells. DHP-103 inhibited Rb+ influx into the heterozygous mutant red blood cells as effectively as into wild-type red blood cells, with an IC50 value (Fig. 3 F and G) comparable to the patch-clamp data (Fig. 3 CE). In contrast, DHP-103 was less effective in inhibiting Rb+ influx into red blood cells from two HX patients with the heterozygous gain-of-function KCa3.1 mutant V282M than into healthy control red blood cells, whereas a control KCa3.1 inhibitor (triarylmethane TRAM-34) (22) was roughly equipotent on mutant and wild-type channels (Fig. 3 F and G). These results from patch-clamp and Rb+ flux studies suggest that DHP-103 may have use in the treatment of HX, particularly HX caused by the KCa3.1_R352H mutation.

DHP-103 Is Efficacious in a Rat Model of Acute Ischemia–Reperfusion Stroke.

Acute ischemic stroke is the second leading cause of death worldwide (36) and fifth-most common cause of death in the United States (3740). Large vessel occlusion (LVO) accounts for 24 to 46% of acute ischemic stroke (~80,000 people annually in the United States) and is associated with significantly higher mortality rates than non-LVO ischemic stroke (37, 38, 40, 41). Within hours following the ischemic insult, a strong, sustained inflammatory response develops in the infarcted area and peri-infarct zone, driven primarily by activated microglia/macrophages with contributions from infiltrating T lymphocytes later in the process (37, 38). This neuroinflammation exacerbates the severity of stroke and impairs recovery (38). Endovascular thrombectomy, the standard-of-care treatment for LVO stroke (42), restores tissue-saving blood supply to the compromised area but does little to dampen the initial severity or subsequent progression of neuroinflammation (38). Since KCa3.1 blockers suppress ischemia-triggered microglial activation and inhibit infiltration of T cells into infarcted areas (37, 38), we assessed DHP-103’s potential as a therapeutic in a rat model of acute ischemic LVO stroke that recapitulated ischemia followed by restoration of blood flow. We initiated treatment 12 h after ischemia–reperfusion, at the peak of neuroinflammation, to dampen neuroinflammation.

Of particular relevance, DHP-103 crosses the blood–brain barrier in mice and achieves brain concentrations sufficient for KCa3.1 target engagement with a t1/2 of 2.24 to 2.48 h following 25 mg/kg intraperitoneal administration (Fig. 4A and SI Appendix, Table S5A). Note, the brain Cmax of 10,458 ng/mL (29 μM) corresponds to an estimated free DHP-103 brain concentration of 1.7 μM (based on mouse plasma protein binding of 94.18%; SI Appendix, Table S6), which ensures maximal, specific KCa3.1 blockade with minimal inhibition of brain CaV1.x channels. Moreover, both mice and rats show similar pharmacokinetic profiles for DHP-103 following intravenous (2 or 5 mg/kg) or oral administration (50 mg/kg), making it likely that DHP-103 enters the brain in rats and reaches concentrations adequate for substantial blockade of KCa3.1 channel activity as it does in mice (Fig. 4B and SI Appendix, Table S5B). After intravenous or oral administration, the plasma Cmax ranges from 2 to 8 μM in both mice and rats, with estimated free DHP-103 plasma concentrations of 0.06 to 0.48 μM (based on mouse and human plasma protein binding of 94.18% and 97.76%, respectively; SI Appendix, Table S6) and a respective t1/2 of 1.15 to 5.15 h (SI Appendix, Table S5B). Furthermore, DHP-103 was well tolerated in a 2-wk repeat-dose toxicology study in mice (25 and 50 mg/kg twice daily, oral administration) where it caused no significant changes in body weight, food consumption, clinical assessment, clinical chemistry, hematological parameters, organ weights, gross organ pathology, or organ histopathology at plasma concentrations sufficient to cause significant blockade of KCa3.1 channel activity (SI Appendix, Tables S5B and S7–S12 and Fig. S7). Given the similarity in pharmacokinetic properties of DHP-103 in both mice and rats, and therefore in vivo drug exposure, it is likely that rats tolerate DHP-103 similarly to mice.

Fig. 4.

Fig. 4.

DHP-103 shows efficacy in a rat model of large vessel occlusion [middle cerebral artery occlusion (MCAO)] acute ischemic-reperfusion stroke. (A) Concentration-time profile of DHP-103 in the plasma (red) and brain (black) of mice following a single intraperitoneal administration of DHP-103 (25 mg/kg; n = 2, mean ± SD) highlighting its brain penetration. Key parameters are shown in SI Appendix, Table S5A. (B) Plasma concentration-time profile of DHP-103 in mice (red) and rats (black) following a single intravenous injection (open circles) or oral administration (filled circles) (n = 3 for each, mean ± SD). Key parameters are shown in SI Appendix, Table S5B. (C) Mean % infarct volume in respective groups (n = 24 for each). Horizontal bars show statistically significant differences between DHP-103- or edaravone-treated MCAO ischemia–reperfusion rats vs. control MCAO ischemia–reperfusion rats (without treatment or administered vehicle) using one-way ANOVA (*P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001). See SI Appendix, Fig. S6A for TTC-stained brain sections. (D) Balance beam test (proprioception) scores for each group on day 0_1 h (n = 8), day 1 (n = 7), day 2 (n = 8), and day 7 (n = 5) after ischemia–reperfusion (mean ± SD; two-way ANOVA of MCAO ischemia–reperfusion rats treated with DHP-103 or edaravone compared to MCAO ischemia–reperfusion control rats that were administered vehicle; *P < 0.1, **P < 0.01, ***P < 0.001, ****P < 0.0001). See SI Appendix, Fig S5 for details about the balance beam test. (E) % of Iba1+CD11b+ activated microglia in the hippocampus (CA1), caudate-putamen (CPu), primary motor cortex (M1), and somatosensory cortex (S1) in each of three experimental groups: MCAO ischemia–reperfusion without treatment, MCAO ischemia–reperfusion administered vehicle and MCAO ischemia–reperfusion administered DHP-103 (10 mg/kg); (n = 5, mean ± SD; one-way ANOVA; *P < 0.1, **P < 0.01). See SI Appendix, Fig. S6B for representative immunofluorescence micrographs.

We occluded the middle cerebral artery (MCAO) with a thread-plug for 60 min (43), then allowed reperfusion for 7 d (SI Appendix, Fig. S8 A and B). We used seven groups in our study, each group consisting of 29 male rats. Control groups included sham-operated rats (blunt dissection, exposure of right common carotid artery, then suture-closure), and rats subjected to MCAO ischemia followed by restoration of blood flow (MCAO ischemia–reperfusion) without further treatment or followed by administration of vehicle (twice daily by intraperitoneal injection for 7 d starting 12 h after reperfusion). Note, the two MCAO ischemia–reperfusion control groups are models for LVO ischemic stroke-treated with standard-of-care approaches to restore tissue-saving blood flow to compromised ischemic brain regions. A fourth control group, received the free-radical quencher edaravone (5 mg/kg, once daily by intraperitoneal injection for 7 d) starting 12 h after MCAO ischemia–reperfusion. Edaravone-Dexborneol, the first innovative drug globally to receive FDA’s breakthrough therapy designation for acute ischemic stroke, is administered to patients within 48 h of onset of acute ischemic stroke after standard-of-care treatment has been administered to restore blood flow to the ischemic brain (44, 45). DHP-103 was administered by intraperitoneal injection (2, 5, or 10 mg/kg twice daily for 7 d) starting 12 h after MCAO ischemia–reperfusion, at the height of neuroinflammation (SI Appendix, Fig. S8 A and B).

Infarct volume was significantly reduced by DHP-103 (2, 5, or 10 mg/kg), with 10 mg/kg decreasing infarct volume by nearly 50% compared to control rats that underwent MCAO ischemia–reperfusion and received either no further treatment or vehicle (Fig. 4C and SI Appendix, Fig. S9A). All three doses of DHP-103 were more effective than edaravone (Fig. 4C). Treatment with the two highest doses of DHP-103 improved balance beam performance (a measure of proprioception, SI Appendix, Fig. S8C) on days 1 and 2 compared to the MCAO-reperfusion vehicle-group, with efficacy exceeding that of edaravone treatment (Fig. 4D). By day 7, proprioception scores had almost returned to baseline, and we were unable to distinguish statistical differences between controls and treated groups (Fig. 4D). We cannot explain why the proprioception scores on day 7 returned to baseline levels (Fig. 4D) in MCAO ischemia–reperfusion control groups, since these rats showed significant infarct size (Fig. 4C) and the presence of activated microglia (Fig. 4E) in infarcted brain regions (Fig. 4E) on day 8.

KCa3.1 plays a critical role in calcium signaling, the respiratory burst, migration, proliferation, and nitric oxide production of activated microglia, and inhibitors of the channel suppress microglia-driven neuroinflammation (37, 40, 46). In our studies on a mouse microglial cell line, DHP-103 blocked endogenous KCa3.1 channels in the potency range that blocks the recombinant human KCa3.1 channel (SI Appendix, Fig. S10 A and B). Moreover, DHP-103 suppressed thapsigargin-induced store-operated calcium entry (SI Appendix, Fig. S10 CE) at a concentration (1 µM) comparable to the free brain concentration of DHP-103 after IP administration (Fig. 4A and SI Appendix, Table S5A). Guided by these results, we examined whether treatment with DHP-103 reduced the numbers of activated microglia in infarcted regions. We used two markers to identify activated microglia in infarcted brain sections, CD11b (highly expressed on myeloid-lineage cells including microglia) and Iba1 (upregulated during microglial activation). We compared three groups (five animals in each group): 1) MCAO ischemia–reperfusion without treatment, 2) MCAO ischemia–reperfusion administered vehicle, and 3) MCAO ischemia– reperfusion administered DHP-103 (10 mg/kg). Treatment with DHP-103 (10 mg/kg) significantly reduced the number of Iba1+CD11b+ activated microglia in infarcted regions of the hippocampus (CA1), caudate putamen and striatum (CPu), primary motor cortex (M1), and primary somatosensory cortex (S1) compared to negative controls (Fig. 4E and SI Appendix, Fig. S9B). We did not assess T cell numbers in infarcted regions but suspect that suppression of T cell infiltration contributes to DHP-103’s efficacy, based on the established physiological role of KCa3.1 in T lymphocytes (15, 22, 37, 38).

In summary, DHP-103 treatment initiated 12 h after ischemic insult and reestablishment of blood flow to infarcted brain regions reduces infarct volume, improves proprioception scores, and reduces the number of activated microglia in the infarcted brain compared to the MCAO ischemia–reperfusion control groups (without further treatment or with treatment with vehicle). Although our study was done on male rats, our findings should be applicable to females, as other KCa3.1 blockers suppress microglia-mediated neuroinflammation equally effectively in males and females in rodent models of Alzheimer’s disease and Parkinson’s disease (47, 48).

Conclusion

We reveal four important new findings in this study. 1) Using a SOSA approach (17), we transformed 1,4-dihydropyridines, canonical blockers of L-type voltage-gated calcium channels, into potent and selective inhibitors targeting the intermediate-conductance calcium-activated K+ channel, KCa3.1, a therapeutic target in small and large animals for a diverse list of diseases. One of these, DHP-103 (logP: 3.45), a 1,4-dihydropyridine defined as a New Chemical Entity (NCE) by the criteria of the Chemical Abstract Services (CAS), is marginally more potent (IC50: 6 nM) than other KCa3.1 inhibitors including TRAM-34 (25 nM), Senicapoc (11 nM), NS6180 (9 nM), 4-phenyl-4H-pyran rac-11 (8 nM), cyclohexadiene rac-16 (8 nM), tri-fluoro trivanillic ester 13b (19 nM) and fused thiazin-3-ones (30 to 100 nM) (2123, 49, 50). DHP-103’s >500-fold selectivity for KCa3.1 over a panel of 116 other targets, including channels, transporters, receptors, and enzymes, is better characterized than that for the other inhibitors. Furthermore, DHP-103 belongs to a chemical class of compounds (1,4-dihydropyridines) used safely for five decades with well-established bioavailability and toxicity in humans. 2) We used cryo-EM, mutagenesis, and electrophysiology to delineate the binding site of DHP-103 within the KCa3.1 channel pore. DHP-103’s direct interactions with residues in the cavity (Thr250, Val275, Thr278) and with permeant ions (K4: innermost K+ in the selectivity filter; K5: additional K+ in central cavity), and K5’s interaction with Val282, stabilizes the inhibitor at the binding site and impedes ion permeation through the pore. Increasing the external K+ concentration does not displace DHP-103 from its binding site, and the potency of DHP-103 is not affected by the internal Ca2+ concentration. Our studies support the idea that DHP-103 traverses the entire lipid bilayer, then passes through the open inner gate in the fully activated channel (10.2 Å diameter) to reach KCa3.1’s central cavity, and perhaps narrows the inner gate (7 Å diameter) after occupying its binding site. Further testing with kinetic studies and molecular dynamics simulation is required to determine the veracity of this idea. 3) Using electrophysiology and Rb+ flux measurements, we showed that DHP-103 inhibits disease-relevant gain-of-function mutant KCa3.1 channels (particularly the R352H mutation) that cause hereditary xerocytosis, highlighting its possible use to treat this hemolytic anemia. 4) In proof-of-concept studies in a rat LVO-ischemia–reperfusion stroke model, we demonstrated that DHP-103 effectively reduced infarct size and dampened microglia-mediated neuroinflammation compared to control rats that underwent LVO-ischemia followed by renewal of tissue-saving blood supply to the ischemic brain (modeling standard-of-care therapy). DHP-103’s excellent brain penetration, pharmacokinetic properties, metabolic stability, and tolerability in rodents suggests its possible use as an adjunct treatment for acute LVO ischemic stroke following standard-of-care endovascular thrombectomy to restore blood flow to compromised brain regions. Additional detailed pharmacokinetic, pharmacodynamic, and toxicological studies will be required to develop this series of 1,4-dihydropyridines into therapeutics for KCa3.1-related diseases.

Methods

1,4-Dihydropyridines and Other Compounds Used in This Study.

Amlodipine, Manidipine, Nimodipine, Nilvadipine, Cilnidipine, Nifedipine, and Nitrendipine were purchased from Sigma-Aldrich; Levamlodipine, from MedChemExpress; and Edaravone from TargetMol. The methods of synthesis of 1,4-dihydropyridines DHP-56, DHP-57, DHP-79, DHP-84, DHP-87, DHP-101, DHP-102, DHP-103, DHP-104, DHP-105, DHP-106, DHP-108, DHP-109, DHP-110, and DHP-113 are described in SI Appendix, Supplementary Methods.

Screening of 1,4-Dihydropyridines by Electrophysiology.

1,4-dihydropyridines were screened for block of KCa3.1 and other channels by patch-clamp using a QPatch HTX automated electrophysiology platform (Sophion, Denmark) and by manual patch-clamp. For QPatch experiments, the giga-seal and whole-cell requirements for the automated electrophysiology were the following: minimum seal resistance of 0.1 GΩ, holding potential −80 mV, holding pressure −20 mbar, positioning pressure −70 mbar. For KCa3.1 experiments, the external solution was Na+-Ringer’s containing (in mM): 160 NaCl, 10 HEPES, 4.5 KCl, 1 MgCl2, 2 CaCl2 (pH 7.2, 310 mOsm). The internal solution contained (in mM): 120 KCl, 10 HEPES, 1.75 MgCl2, 1 Na2ATP, 10 EGTA, 8.6 CaCl2 (1 μM free Ca2+), (pH 7.4, 300 mOsm). Following establishment of whole-cell configuration, cells were held at −80 mV and KCa3.1 currents elicited by a voltage protocol that held at −80 mV for 20 ms, stepped to −120 mV for 20 ms, ramped from −120 to +40 mV over 200 ms, and then stepped back to −120 mV for 20 ms. This pulse protocol was applied every 10 s. Current slopes (in amperes per second) were measured using the Sophion QPatch software and exported to Microsoft Excel and GraphPad Prism 9 for analysis. Decreases in slope between −85 and −65 mV were used to determine the IC50 for KCa3.1 inhibition. Cells with membrane capacitances of 10 to 30 pF were analyzed. Manual patch-clamp studies on KCa3.1 (Figs. 1, 2 C and D, and 3 CE) and other ion channels are described in SI Appendix, Supplementary Methods.

Electrophysiological Studies on Gain-of-Function KCa3.1 Mutations that Cause Hereditary Xerocytosis.

Mutations were introduced into human KCa3.1 channels using the QuickChange II site-directed mutagenesis kit (Agilent) or through molecular cloning services (Genscript). Wild-type and mutant channel cDNAs constructed in pIRES2-AcGFP1 (Clontech) were transfected into HEK293 cells by the calcium–phosphate method. KCa3.1 currents shown in Fig. 3 were recorded 1 to 2 d after transfection, using an inside-out patch configuration with an Axon200B amplifier (Molecular Devices) at room temperature. pClamp 10.5 (Molecular Devices) was used for data acquisition and analysis. Resistance of the patch electrodes ranged from 3 to 5 MΩ. The pipette solution contained (in mM): 140 KCl, 10 HEPES (pH 7.4), 1 MgSO4. The bath solution contained (in mM): 140 KCl, 10 HEPES (pH 7.2), 1 EGTA, 0.1 Dibromo-BAPTA, and 1 HEDTA, mixed with Ca2+ to yield desired free Ca2+ concentrations, calculated using Maxchelator (Chris Patton: https://somapp.ucdmc.ucdavis.edu/pharmacology/bers/maxchelator/webmaxc/webmaxcS.htm). The intracellular face of the inside-out patch was initially exposed to bath solution containing 1 μM free Ca2+. Channel responses to a range of concentrations of tested compounds were measured in the presence of 1 μM free Ca2+. Currents were recorded by repetitive 1-s-voltage ramps from −100 to 100 mV from a holding potential of 0 mV. To construct concentration-dependent inhibition curves, we normalized current amplitudes at −90 mV in the presence of DHP-103 to that in its absence. Normalized currents were plotted as a function of DHP-103 concentration. IC50 values and Hill coefficients were determined by fitting data points to a Hill equation.

Erythrocytes Used for 83Rb Influx Assay.

Human blood samples were obtained with written informed consent from patients with dehydrated stomatocytosis (hereditary xerocytosis; HX). Blood collected in heparin-containing vacutainer tubes was transported by courier from local collection centers to Beth Israel Deaconess Medical Center. All procedures were HIPAA-compliant and approved by the Institutional Review Board of Beth Israel Deaconess Medical Center. Erythrocyte preparation and erythrocyte 83Rb influx assay are described in SI Appendix, Supplementary Methods.

Middle Cerebral Artery Occlusion (MCAO) Ischemia–Reperfusion Stroke Model in Rats.

MCAO ischemia–reperfusion model studies in rats were conducted by ICE Bioscience (China) with approval of the Ethics and Moral Management Committee of Experimental Animals at Xuzhou Medical University. Male Sprague-Dawley rats weighing 240 to 270 g were purchased from Jinan Pengyue Experimental Animal Breeding Company (China). Focal cerebral ischemia was induced by occlusion of the right middle cerebral artery according to the Longa method (43). Animals were anesthetized with 10% chloral hydrate and a median incision in the neck was made to expose the right common carotid artery, internal carotid artery, and external carotid artery. A slipknot on the common carotid artery, a dead knot on the proximal side of the external carotid artery, and a slipknot on the external carotid artery near the common carotid bifurcation were tied with a silk thread. A small opening between the two knots of the external carotid artery was cut to insert a thread plug into the carotid artery, which was advanced inward into the MCA. The plug was kept in place for 60 min, then removed from the blood vessel to restore blood supply. Reperfusion continued for 7 d and the rats were sacrificed using carbon dioxide asphyxiation on day 8.

We used seven groups for our study, each group consisting of 29 rats: sham-operated rats (blunt dissection, exposure of right common carotid artery, then suture-closure), MCAO ischemia–reperfusion without treatment, MCAO ischemia–reperfusion administered vehicle, MCAO-reperfusion administered edavarone (5 mg/kg), MCAO ischemia–reperfusion administered DHP-103 (2 mg/kg), MCAO ischemia–reperfusion administered DHP-103 (5 mg/kg), and MCAO ischemia–reperfusion administered DHP-103 (10 mg/kg). Treatment began 12 h after reperfusion and continued until day 7. Rats received intraperitoneal injections of vehicle (5% v/v N-Methylpyrrolidone (NMP), 7.5% v/v Solutol HS 15, 50% v/v PEG-400, and 37.5% v/v TPGS [10% w/v]) twice daily, or DHP-103 formulated in vehicle (2, 5, 10 mg/kg, twice daily) or edaravone (5 mg/kg once daily; dissolved in DMSO before formulation in normal saline).

Immediately after reperfusion we randomized 24 of the 29 rats from each group for proprioception assessment. Proprioception was assessed individually using the balance beam test on day 0 (1 h after reperfusion) (n = 8 from each group), day 1 (n = 7), day 2 (n = 8), and day 7 (n = 5) (Fig. 4 D and E). To reduce stress, we used different rats for assessment on different days.

All 24 rats from each group that were used for proprioception assessment were euthanized on day 8 after reperfusion. Their brain slices (2 to 16 mm from frontal pole to occipital pole) were stained with 2,3,5 triphenyltetrazolium (TTC) to determine % infarct volume (Fig. 4F and SI Appendix, Fig. S6A). Infarct area was assessed by ImageJ analysis of scanned TTC-stained whole brain sections.

Immediately after reperfusion we randomized 5 of the 29 rats from each group for assessment of activated microglia. These rats were euthanized on day 8 after reperfusion and their brain sections (hippocampus-CA1, caudate-putamen, primary cortex, and somatosensory cortex) were immunostained for markers of activated microglia (Iba1 and CD11b) (n = 5) (Fig. 4H and SI Appendix, Fig. S6B). Details of immunohistochemical analysis of microglia and leukocyte markers are described in http://www.pnas.org/lookup/doi/10.1073/pnas.2425494122#supplementary-materialsSI Appendix, Supplementary Methods.

Other Methods.

http://www.pnas.org/lookup/doi/10.1073/pnas.2425494122#supplementary-materialsSI Appendix, Supplementary Methods include details of compound synthesis, electrophysiology studies using the manual patch clamp system (Figs. 13), electrophysiological studies on the KCa3.1 T250S-V275A and KCa3.1_T212F-V272F mutants, metabolite stability in mouse and human liver microsomes, biotransformation of DHP-103 in mouse, rat, dog, monkey, and human liver microsomes, purification of KCa3.1/CaM protein, cryo-EM structure determination, erythrocyte preparation, erythrocyte Rb+ flux, selectivity analysis by competitive binding and enzyme uptake assays, cytochrome P450 enzyme inhibition assays, immunohistochemistry analysis on microglia, electrophysiology studies on mouse microglia, measurement of thapsigargin-induced store-operated calcium entry into mouse microglia, pharmacokinetics, and brain uptake of DHP-103 in mouse and rat.

Supplementary Material

Appendix 01 (PDF)

Acknowledgments

This work was supported by grants from the Singapore Ministry of Health under its National Medical Research Council Clinical Scientist Individual Research Grant (CIRG1427-2015) to K.G.C.; Lee Kong Chian School of Medicine Startup Funds and Strategic Academics Initiative Grant to K.G.C., Lee Kong Chian School of Medicine Research Administration and Support Services to Lee Kong Chian School of Medicine-Innovative CRO Explorer Collaborative Platform; and National Institutes of Health Grants to M.Z. (4R33NS101182-03 and 1R15NS130420-01A1), H.W. (CounterACT Program U54NS079202) and American Heart Association Grants to M.Z. (23AIREA1039423) and Y.-W.N. (24CDA1260237). J.A.N. was supported by a National Institute of General Medical Sciences funded Pharmacology Training Program (T32GM099608). Cryogenic Electron Microscopy studies were performed at the Stanford Linear Accelerator Center Cryogenic Electron Microscopy Center (S2C2), which is supported by the National Institutes of General Medical Sciences (1R24GM154186). The Cryogenic Electron Microscopy data included in this manuscript is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. We thank Dr. L. Michael Snyder (formerly of Quest Diagnostics, Inc. and University of Massachusetts Medical Center) and Dr. Carlo Brugnara (Boston Children’s Hospital and Harvard Medical School) for Hereditary Xerocytosis blood samples. We thank Dr. Mahmood Ahmed and Xingying Chew (formerly of Nanyang Technological University) for synthesis of DHP-57, DHP-79, DHP-84, and DHP-87. We are grateful to Dr. George Augustine for his thoughtful and valuable comments.

Author contributions

S.T.O., Y.X., S.L.A., H.W., M.Z., and K.G.C. designed research; S.T.O., Y.-W.N., J.A.N., A. Ramanishka, X.R.N., Z.Z., S.S.M.G., H.M.N., L.S., V.S., and A. Rivera performed research; M.E.E. contributed new reagents/analytic tools; S.T.O., Y.-W.N., J.A.N., A. Ramanishka, X.R.N., Z.Z., S.S.M.G., H.M.N., A. Rivera, M.E.E., Y.X., S.L.A., H.W., M.Z., and K.G.C. analyzed data; and S.T.O., A. Rivera, S.L.A., H.W., M.Z., and K.G.C. wrote the paper.

Competing interests

S.T.O. and K.G.C. are co-inventors of a patent on KCa3.1-selective 1,4-dihydropyridines that has been filed by the Nanyang Technological University, Singapore (PCT WO2022115043A1). None of the other co-authors have any competing interests.

Footnotes

This article is a PNAS Direct Submission.

Contributor Information

Seow Theng Ong, Email: st.ong@ntu.edu.sg.

K. George Chandy, Email: gchandy@ntu.edu.sg.

Data, Materials, and Software Availability

The atomic coordinates have been deposited in the Protein Data Bank (PDB) under accession codes 9ED1 (https://doi.org/10.2210/pdb9ED1/pdb) (103_KCa3.1) (51). The cryo-EM map has been deposited in the Electron Microscopy Data Bank (EMDB) under accession codes EMD-47930 (https://www.ebi.ac.uk/pdbe/entry/emdb/EMD-47930) (103_KCa3.1) (51). All study data are included in the article and/or SI Appendix.

Supporting Information

References

  • 1.Fleckenstein G. G., Byon K. Y., Doring H. J., Tritthart H., “The basic Ca antagonistic actions of nifedipine on cardiac energy metabolism and vascular smooth muscle tone” in New Therapy of Ischemic Heart Disease. 1st International Nifedipine "Adalat" Symposium, Hashimoto K., Kimura E., Kobayashi T., Eds. (University of Tokyo Press, 1975), pp. 31–44. [Google Scholar]
  • 2.Triggle D. J., The 1,4-dihydropyridine nucleus: A pharmacophoric template part 1. Actions at ion channels. Mini Rev. Med. Chem. 3, 215–223 (2003). [DOI] [PubMed] [Google Scholar]
  • 3.Gao S., Yan N., Structural basis of the modulation of the voltage-gated calcium ion channel CaV 1.1 by dihydropyridine compounds. Angew. Chem. Int. Ed. Engl. 60, 3131–3137 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Zhao Y., et al. , Molecular basis for ligand modulation of a mammalian voltage-gated Ca2+ channel. Cell 177, 1495–1506.e1412 (2019). [DOI] [PubMed] [Google Scholar]
  • 5.van Rhee A. M., et al. , Interaction of 1,4-dihydropyridine and pyridine derivatives with adenosine receptors: Selectivity for A3 receptors. J. Med. Chem. 39, 2980–2989 (1996). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Doring H., Kreutzer D., Ritter C., Hilgeroth A., Discovery of novel symmetrical 1,4-dihydropyridines as inhibitors of multidrug-resistant protein (MRP4) efflux pump for anticancer therapy. Molecules 26, 18 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Fanger C. M., et al. , Calmodulin mediates calcium-dependent activation of the intermediate conductance KCa channel, IKCa1. J. Biol. Chem. 274, 5746–5754 (1999). [DOI] [PubMed] [Google Scholar]
  • 8.Ishii T. M., et al. , A human intermediate conductance calcium-activated potassium channel. Proc. Natl. Acad. Sci. U.S.A. 94, 11651–11656 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Logsdon N. J., Kang J., Togo J. A., Christian E. P., Aiyar J., A novel gene, hKCa4, encodes the calcium-activated potassium channel in human T lymphocytes. J. Biol. Chem. 272, 32723–32726 (1997). [DOI] [PubMed] [Google Scholar]
  • 10.Vandorpe D. H., et al. , cDNA cloning and functional characterization of the mouse Ca2+-gated K+ channel, mIK1. Roles in regulatory volume decrease and erythroid differentiation. J. Biol. Chem. 273, 21542–21553 (1998). [DOI] [PubMed] [Google Scholar]
  • 11.Hoffman J. F., et al. , The hSK4 (KCNN4) isoform is the Ca2+-activated K+ channel (Gardos channel) in human red blood cells. Proc. Natl. Acad. Sci. U.S.A. 100, 7366–7371 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Brown B. M., Shim H., Christophersen P., Wulff H., Pharmacology of small- and intermediate-conductance calcium-activated potassium channels. Annu. Rev. Pharmacol. Toxicol. 60, 219–240 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ataga K. I., et al. , Improvements in haemolysis and indicators of erythrocyte survival do not correlate with acute vaso-occlusive crises in patients with sickle cell disease: A phase III randomized, placebo-controlled, double-blind study of the Gardos channel blocker senicapoc (ICA-17043). Br. J. Haematol. 153, 92–104 (2011). [DOI] [PubMed] [Google Scholar]
  • 14.Hausmann D., et al. , Autonomous rhythmic activity in glioma networks drives brain tumour growth. Nature 613, 179–186 (2023). [DOI] [PubMed] [Google Scholar]
  • 15.Ghanshani S., et al. , Up-regulation of the IKCa1 potassium channel during T-cell activation. Molecular mechanism and functional consequences. J. Biol. Chem. 275, 37137–37149 (2000). [DOI] [PubMed] [Google Scholar]
  • 16.Ellory J. C., Culliford S. J., Smith P. A., Wolowyk M. W., Knaus E. E., Specific inhibition of Ca-activated K channels in red cells by selected dihydropyridine derivatives. Br. J. Pharmacol. 111, 903–905 (1994). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wermuth C. G., Selective optimization of side activities: The SOSA approach. Drug Discov. Today 11, 160–164 (2006). [DOI] [PubMed] [Google Scholar]
  • 18.Siddiqi F. H., et al. , Felodipine induces autophagy in mouse brains with pharmacokinetics amenable to repurposing. Nat. Commun. 10, 1817 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Lee C. H., MacKinnon R., Activation mechanism of a human SK-calmodulin channel complex elucidated by cryo-EM structures. Science 360, 508–513 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Schewe M., et al. , A pharmacological master key mechanism that unlocks the selectivity filter gate in K+ channels. Science 363, 875–880 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Nguyen H. M., et al. , Structural insights into the atomistic mechanisms of action of small molecule inhibitors targeting the KCa3.1 channel pore. Mol. Pharmacol. 91, 392–402 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Wulff H., et al. , Design of a potent and selective inhibitor of the intermediate-conductance Ca2+-activated K+ channel, IKCa1: A potential immunosuppressant. Proc. Natl. Acad. Sci. U.S.A. 97, 8151–8156 (2000). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Strobaek D., et al. , NS6180, a new KCa3.1 channel inhibitor prevents T-cell activation and inflammation in a rat model of inflammatory bowel disease. Br. J. Pharmacol. 168, 432–444 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Mauler F., et al. , Selective intermediate-/small-conductance calcium-activated potassium channel (KCNN4) blockers are potent and effective therapeutics in experimental brain oedema and traumatic brain injury caused by acute subdural haematoma. Eur. J. Neurosci. 20, 1761–1768 (2004). [DOI] [PubMed] [Google Scholar]
  • 25.Wulff H., Gutman G. A., Cahalan M. D., Chandy K. G., Delineation of the clotrimazole/TRAM-34 binding site on the intermediate conductance calcium-activated potassium channel, IKCa1. J. Biol. Chem. 276, 32040–32045 (2001). [DOI] [PubMed] [Google Scholar]
  • 26.Andolfo I., et al. , Multiple clinical forms of dehydrated hereditary stomatocytosis arise from mutations in PIEZO1. Blood 121, 3925–3935 (2013). [DOI] [PubMed] [Google Scholar]
  • 27.Andolfo I., et al. , Novel Gardos channel mutations linked to dehydrated hereditary stomatocytosis (xerocytosis). Am. J. Hematol. 90, 921–926 (2015). [DOI] [PubMed] [Google Scholar]
  • 28.Fermo E., et al. , Gardos channelopathy: Functional analysis of a novel KCNN4 variant. Blood Adv. 4, 6336–6341 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Glogowska E., Lezon-Geyda K., Maksimova Y., Schulz V. P., Gallagher P. G., Mutations in the Gardos channel (KCNN4) are associated with hereditary xerocytosis. Blood 126, 1281–1284 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Orfali R., et al. , Channelopathy-causing mutations in the S45A/S45B and HA/HB helices of KCa2.3 and KCa3.1 channels alter their apparent Ca2+ sensitivity. Cell Calcium 102, 102538 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Rapetti-Mauss R., et al. , A mutation in the Gardos channel is associated with hereditary xerocytosis. Blood 126, 1273–1280 (2015). [DOI] [PubMed] [Google Scholar]
  • 32.Rapetti-Mauss R., et al. , Red blood cell Gardos channel (KCNN4): The essential determinant of erythrocyte dehydration in hereditary xerocytosis. Haematologica 102, e415–e418 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Rapetti-Mauss R., Soriani O., Vinti H., Badens C., Guizouarn H., Senicapoc: A potent candidate for the treatment of a subset of hereditary xerocytosis caused by mutations in the Gardos channel. Haematologica 101, e431–e435 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Rivera A., et al. , Erythrocyte ion content and dehydration modulate maximal Gardos channel activity in KCNN4 V282M/+ hereditary xerocytosis red cells. Am. J. Physiol. Cell Physiol. 317, C287–C302 (2019). [DOI] [PubMed] [Google Scholar]
  • 35.Shmukler B. E., et al. , Dehydrated stomatocytic anemia due to the heterozygous mutation R2456H in the mechanosensitive cation channel PIEZO1: A case report. Blood Cells Mol. Dis. 52, 53–54 (2014). [DOI] [PubMed] [Google Scholar]
  • 36.Feigin V. L., et al. , World Stroke Organization (WSO): Global stroke fact sheet 2022. Int. J. Stroke 17, 18–29 (2022). [DOI] [PubMed] [Google Scholar]
  • 37.Chen Y. J., et al. , The potassium channel KCa3.1 constitutes a pharmacological target for neuroinflammation associated with ischemia/reperfusion stroke. J. Cereb. Blood Flow Metab. 36, 2146–2161 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Lee R. D., et al. , Repurposing the KCa3.1 blocker senicapoc for ischemic stroke. Transl. Stroke Res. 15, 518–532 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Martin S. S., et al. , 2024 heart disease and stroke statistics: A report of US and global data from the American Heart Association. Circulation 149, e347–e913 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Nguyen H. M., Blomster L. V., Christophersen P., Wulff H., Potassium channel expression and function in microglia: Plasticity and possible species variations. Channels (Austin) 11, 305–315 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Rennert R. C., et al. , Epidemiology, natural history, and clinical presentation of large vessel ischemic stroke. Neurosurgery 85, S4–S8 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Palaniswami M., Yan B., Mechanical thrombectomy is now the gold standard for acute ischemic stroke: Implications for routine clinical practice. Interv. Neurol. 4, 18–29 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Longa E. Z., Weinstein P. R., Carlson S., Cummins R., Reversible middle cerebral artery occlusion without craniectomy in rats. Stroke 20, 84–91 (1989). [DOI] [PubMed] [Google Scholar]
  • 44.Enomoto M., Endo A., Yatsushige H., Fushimi K., Otomo Y., Clinical effects of early edaravone use in acute ischemic stroke patients treated by endovascular reperfusion therapy. Stroke 50, 652–658 (2019). [DOI] [PubMed] [Google Scholar]
  • 45.Fu Y., et al. , Sublingual Edaravone dexborneol for the treatment of acute ischemic stroke: The TASTE-SL randomized clinical trial. JAMA Neurol. 81, 319–326 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Kaushal V., Koeberle P. D., Wang Y., Schlichter L. C., The Ca2+-activated K+ channel KCNN4/KCa3.1 contributes to microglia activation and nitric oxide-dependent neurodegeneration. J. Neurosci. 27, 234–244 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Jin L. W., et al. , Repurposing the KCa3.1 inhibitor senicapoc for Alzheimer’s disease. Ann. Clin. Transl. Neurol. 6, 723–738 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lu J., Dou F., Yu Z., The potassium channel KCa3.1 represents a valid pharmacological target for microgliosis-induced neuronal impairment in a mouse model of Parkinson’s disease. J. Neuroinflammation 16, 273 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Wulff H., Castle N. A., Therapeutic potential of KCa3.1 blockers: Recent advances and promising trends. Expert Rev. Clin. Pharmacol. 3, 385–396 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Olivan-Viguera A., et al. , Novel phenolic inhibitors of small/intermediate-conductance Ca2+-activated K+ channels, KCa3.1 and KCa2.3. PLoS ONE 8, e58614 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Nam Y. W., Zhang M., Data from “Cryo-EM structure of the human KCa3.1/calmodulin channel in complex with Ca2+ and 1,4-dihydropyridine (DHP-103).” Protein Data Bank. 10.2210/pdb9ED1/pdb. Deposited 18 November 2024. [DOI]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Appendix 01 (PDF)

Data Availability Statement

The atomic coordinates have been deposited in the Protein Data Bank (PDB) under accession codes 9ED1 (https://doi.org/10.2210/pdb9ED1/pdb) (103_KCa3.1) (51). The cryo-EM map has been deposited in the Electron Microscopy Data Bank (EMDB) under accession codes EMD-47930 (https://www.ebi.ac.uk/pdbe/entry/emdb/EMD-47930) (103_KCa3.1) (51). All study data are included in the article and/or SI Appendix.


Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

RESOURCES