Abstract
Polyphenolic compounds are widely explored for health benefits, including hypertension, but their active ingredients, molecular targets, and mechanisms remain poorly defined. We identify the xanthone Mangostin from Garcinia mangostana as a potent modulator of several potassium channels, with large-conductance K+ (BK) channels as its primary target for vasorelaxation. Mangostin activated BK channels as α subunits alone, in complexes with vascular β1 subunits, and in reconstituted BKα/β1–Cav nanodomains. It shifted BK voltage activation to more negative potentials by antagonizing channel closure and promoting channel opening without markedly altering Ca2+ sensitivity. Docking, competition, single channel analysis and mutagenesis localized the binding site in the pore cavity below the SF, involving gating-critical S6 residues I308, L312, and A316, and suggest that Mangostin stays bound in closed and open states. These findings establish BK channel activation as the core molecular mechanism driving Mangostin’s vascular effects and define its structural mode of action, informing nutraceutical safety assessment and BK-targeted drug design.
Introduction
Nutraceuticals (superfoods, dietary supplements, and traditional remedies) promise a plethora of health benefits that are usually not definitely proven by clinical studies. Well-known examples are resveratrol present in grapes and red wine (Jang et al., 1997; Brown et al., 2024), carotenes and tocopherols (Alpha-Tocopherol, Beta Carotene Cancer Prevention Study Group, 1994; Hennekens et al., 1996; Xin et al., 2022), or the recent resurgence of quercetin (Lee et al., 2021; Di Pierro et al., 2023). In evaluating clinical relevance and explore pharmacological opportunities of such phytochemicals, obstacles are often the identification of the biologically active substance in complex matrices of plant secondary metabolites and the lack of a definite cellular determinant that could link a biologically active substance to a physiological process. However, a clear molecular target and mechanism of action that can explain its effects is the prerequisite to further drug development.
Natural polyphenols are known to modulate diverse potassium (K+) channel families. For example, various members of the flavonoid subgroup activate KCa (BK) channels as well as several Kv, Kir, and K2P channels, including TREK-1 channels ((Gierten et al., 2008; Nardi and Olesen, 2008; Kim, Kang and Han, 2011); for a comprehensive review see (Richter-Laskowska et al., 2023)). A recent study suggested that α-Mangostin, a xanthone from Garcinia mangostana, modulates ion channels and binds in the pore cavity of TREK K2P channels (Kim et al., 2023). α- and γ-Mangostins are the main xanthones found in the mangostane fruit pericarp, whose ethanolic extracts are used as traditional medicine and consumed as nutraceuticals. They are thought to exert a plethora of health-promoting effects, such as cardioprotective (Eisvand et al., 2022), analgesic (Kim et al., 2023), antioxidant (Kong, Jia and Jia, 2022), antiinflammatory (Kim et al., 2021), antidiabetic (John et al., 2022), antifibrotic (Li et al., 2019), antimicrobial (Tatiya-Aphiradee, Chatuphonprasert and Jarukamjorn, 2016), and antiproliferative effects (Majdalawieh et al., 2024). Interestingly, very recent research reported that α-Mangostin has antihypertensive properties, and it decreased systolic and diastolic blood pressure in a rat model (Xu et al., 2024). Earlier studies had demonstrated the relaxation of aortic rings upon Mangostin incubation, but remained somewhat contradictory as to the site of action and the molecular target (Chairungsrilerd et al., 1996, 1997; Tep-Areenan and Suksamrarn, 2012). Hence, the molecular mechanism of this antihypertensive effect of Mangostin remains unclear.
Major potassium channels present in vascular smooth muscle are Ca2+-activated BK channels, also known as MaxiK, Slo1 or KCa1.1 channels (Wu and Marx, 2010; Tykocki, Boerman and Jackson, 2017; Pereira da Silva et al., 2022). They are synergistically activated by intracellular calcium (Cai2+) and voltage, and they play a crucial role in the regulation of vascular smooth muscle tone, with implications for blood pressure regulation. BK channels act as feedback regulators which counteract depolarization and balance the elevation of Ca 2+ through voltage-dependent L-type Ca2+ channels. The pore-forming BKα subunit is expressed ubiquitously, and tissue specific properties are conveyed by regulatory β-, γ-, and LINGO subunits (Latorre et al., 2017; Gonzalez-Perez and Lingle, 2019; Dudem et al., 2020). In vascular smooth muscle cells, BKα assembles with the β1 subunit, which enhances its apparent Cai2+ sensitivity, decreases voltage sensitivity (Dworetzky et al., 1996; Brenner et al., 2000; Orio and Latorre, 2005; Li and Yan, 2016), and modulates its pharmacological properties (McManus et al., 1995; Hanner et al., 1997).
Structurally, BK channels are homotetramers consisting of the pore-forming α subunits with 7 transmembrane segments (S0-S6), a linker region, and two intracellular RCK domains. The RCK domains and their interface to the transmembrane segments contain different Cai2+ binding sites, S1-S4 comprise the voltage sensing domain, and S5 and S6 together with the pore helix constitute the pore domain. Both depolarization or Cai2+ elevation can open the pore via allosteric mechanisms, and this regulation as well as the modulation by mutations, small molecules, and regulatory subunits can be described well by an allosteric gating model ((Horrigan and Aldrich, 2002; Latorre et al., 2017)). However, the structural determinants that underlie this allosteric modulation remain unclear. The S6 segment has been shown to undergo conformational changes during activation, and several gating sensitive residues as well as modulator binding sites for molecules of surprisingly different chemical structure are known in this pore region or the adjacent linker to the cytoplasmic C-terminus (Nardi and Olesen, 2008; Zhou, Xia and Lingle, 2011; Roy et al., 2012; Hoshi, Xu, et al., 2013; Chen, Yan and Aldrich, 2014; Webb et al., 2015; Hoshi and Heinemann, 2016; Schewe et al., 2019; Rockman, Vouga and Rothberg, 2020a; Gonzalez-Sanabria et al., 2025). Investigations of the activation mechanism indicated that these substances act without directly affecting the voltage- or Ca2+ sensory domains, probably by modifying the close interactions of S5 and S6 with the voltage sensor bundle and the gating ring that enable opening of the channel.
Small molecule BK channel activators are promising to treat diverse diseases like fragile X syndrome, overactive bladder, pulmonary hypertension, chronic obstruction, erectile dysfunction, and reperfusion injury; however, no prospective drug successfully completed a phase III trial yet (Bentzen et al., 2014; Barenco-Marins et al., 2023; Ferraguto et al., 2024). Therefore, adding a pharmacophore derived from the xanthone group may aid in developing future drug candidates.
We used a combination of functional studies in cultured cells and vascular tissue as well as molecular docking to show that BK channels are a molecular target of Mangostins and that they mediate vascular relaxation observed upon Mangostin application. BK channels were opened most prominently among the K+ channel representatives tested and were activated by α-Mangostin as well as by an extract of the mangosteen pericarp marketed as a nutraceutical. Investigation of the activation mechanism showed that α-Mangostin essentially facilitates BK channel activation by shifting its voltage dependence, modulating gating kinetics, and enhancing the open probability, without changing the Cai2+ sensitivity. These effects were due to direct binding to the pore-forming BKα subunit. Competition experiments, molecular docking, and scanning mutagenesis identified the binding region in S6 just below the selectivity filter (SF). Mangostin activation was preserved in reconstructed nanodomains with Cav channels, which cause local elevation of intracellular Ca 2+, mimicking the physiological situation in smooth muscle cells. Finally, we show that BK channels specifically mediate relaxation of aortic preparations from mice. Our study explains the molecular activation of BK channels by a natural xanthone and sheds light on the mechanism underlying one of its claimed health benefits.
Results
α-Mangostin particularly activates BKα and BKα/β1 channels
In previous studies, α-Mangostin was shown to activate members of the K2P channel family, while other K+ channels were inhibited (Kim et al., 2021, 2023). We aimed to obtain a broader pharmacological profile of α-Mangostin in K+ channels and we investigated members of all six subfamilies of K2P channels as well as representatives of the Kv and Kir channel families. Confirming the previous studies, we observed a strong activation of TREK-1 K2P channels (fold change 10.33 ± 1.64; Fig. 1A and Fig. S1) and inhibition of TRESK K2P channels as well as Kv1.3 channels by ≈50% and ≈60 %, respectively (Fig. 1A, B and Fig. S1). Furthermore, we identified three additional targets of α-Mangostin: strongly inhibited were the K2P members TWIK-1m (≈88 %), TWIK-2m (≈50 %), and TASK-3 (≈80 %) (Fig. 1A, B and Fig. S1). In contrast, BK channels were most potently activated, with a fold change of 20.41 ± 2.39 for BKα channels and of 24.42 ± 3.85 for BKα/β1 channels, which is the complex predominantly present in smooth muscle cells of the vasculature (Fig. 1A, C). This suggested that BK channels, especially the BKα/β1 heteromer, could be the molecular target mediating the vasorelaxant Mangostin effects reported in above studies.

Mangostin potently activates BKα and BKα/β1 channels compared to other potassium channel representatives.
(A) Current fold change ± SEM of currents of representatives from different potassium channel families upon application of 10 µM α-Mangostin. K2P and KCa channel currents were recorded with ramp protocols from -100 mV to +50 mV and analyzed at +40 mV; Kv1.1 and Kv1.3 channel currents were evoked using a rectangle pulse to +40 mV, and hERG currents were recorded with a rectangle pulse to +60 mV followed by hyperpolarization to -120 mV, which was analyzed. Kir channels were recorded using ramp protocols from -150 mV to +50 mV, and currents were analyzed at -140mV. All measurements were made in transiently transfected HEK293 cells in physiological potassium gradients with 100 nM intracellular free Ca2+ with a holding potential of -80 mV. TWIK-1m and TWIK-2m denote channels where the retrieval motif was removed to improve membrane expression, and intracellular K+ was exchanged for Rb+ to enhance currents. (B) Representative current traces of channels that were inhibited more than 60 % by 10 µM α-Mangostin. (C) Representative current traces of BKα and BKα/β1 channels activated by 10 µM α-Mangostin (α-M). (D) Current fold change ± SEM after application of α-Mangostin, γ-Mangostin, and a dietary supplement to TREK-1, BKα and BKα/β1 channels. Currents were recorded and analyzed as in (A). (E) Representative time course of the dose-dependent activation of BKα channels by increasing concentrations of α-Mangostin (left) and the resulting dose-response-relationships for BKα and BKα/β1 channels (right). Currents were recorded and analyzed as in (A); the grey data point at 12.5 µM was not included in the Hill fit. Data and statistics see Tables S1 and S2.
The preparations from Garcinia mangostana plants that are consumed as ‘nutraceuticals’ contain an unstandardized mixture of secondary plant metabolites, among them other Mangostin derivatives. Therefore, we also investigated their modulatory effect on TREK-1, BKα and BKα/β1 currents (Fig. 1D). The very similar γ-Mangostin activated BKα channels with slightly higher efficacy (fold change of 38.54 ± 4.8) than TREK-1 and BKα/β1 channels (fold change of 11.35 ± 1 and 29.32 ± 5.03). Interestingly, the dietary supplement (solubilized equivalent to 10 µM α-Mangostin) activated TREK1, BKα and BKα/β1 channels comparably to pure α-/γ-Mangostin (fold change of 9.18 ± 2.74, 19.98 ± 2.99 and 20.92 ± 3.49; Fig. 1D), highlighting that mangostane xanthones retain their activity in such extracts.
The application of different α-Mangostin concentrations resulted in a fast and dose-dependent activation of BKα and BKα/β1 channels, with a similar apparent EC50 of 3.16 ± 1.13 µM and 2.57 ± 1.32 µM, suggesting that the potency of α-Mangostin is not affected by the presence of the β1-subunit (Fig. 1E). However, we frequently noticed a decline of currents at concentrations >12 µM and limited solubility of higher Mangostin concentrations, which may imply that the actual maximal activation could not be reached.
α-Mangostin shifts the voltage activation of BKα and BKα/β1 channels to more negative values
Having established the prominent activation of BKα and BKα/β1 channels by α-Mangostin, we wanted to gain more insight into its activation mechanism. The open probability of BK channels is controlled by voltage signals, calcium signals, or shift of the closed-open equilibrium (e.g. by mutations or modulators), and the current further depends on the single channel amplitude and the time the channel adopts an open conductive state. We therefore investigated these aspects of BK channel gating in macroscopic and single-channel measurements.
As we already observed in the ramp measurements, the threshold of BK voltage activation was shifted to more negative potentials upon α-Mangostin application (Fig. 1C). Therefore, we first examined if α-Mangostin affects the voltage activation of BK channels. We used a step protocol over a range of potentials followed by a repolarization step to elicit tail currents in symmetrical bi-ionic conditions where intracellular K+ was replaced with Cs+ to reduce the strong outward currents in whole-cell experiments (Fig. 2A; Cs+ does not alter voltage activation (Piskorowski and Aldrich, 2006)). We quantified the change in voltage activation induced by α-Mangostin by calculating the voltage of half-maximal activation (V½) from the Boltzmann fit of conductance-voltage (GV) relationships (Fig. 2B). The application of 10 µM α-Mangostin caused a substantial left-shift of the V½ by 53.08 ± 4.9 mV in BKα channels (from 110.45 ± 2.69 mV to 57.37 ± 3.6 mV). This effect was even more pronounced in smooth muscle-like BKα/β1 channels, where V½ shifted by 82.42 ± 4.96 mV to more hyperpolarized voltages (from 147.25 ± 5.66 mV to 64.83 ± 4.25 mV). The slope of the Boltzmann fit was not different before and after α-Mangostin activation in both channels (Fig. 2B, insets). Hence, BKα and BKα/β1 channels already opened at less depolarized voltages in the presence of α-Mangostin, whereas voltage activation itself was not affected.

Effects of α-Mangostin on BKα and BKα/β1 channel gating.
(A) Representative current traces for BKα and BKα/β1 channels before and after activation by 10 µM α-Mangostin. Cells were measured in symmetrical 140 mM bi-ionic conditions with 140 mM Cs+ as the intracellular ion and 100 nM free Ca 2+. Currents were elicited by a family of rectangle pulses from -100 mV to up to +300 mV in 20 mV increments from a holding potential of -80 mV, followed by repolarization to -50 mV to elicit inward tail currents. (B) GV-relationships for BKα and BKα/β1 channels in the basal state and activated by 10 µM α-Mangostin, derived from tail current analysis of recordings as in (A). The grey arrow illustrates the left-shift of voltage activation caused by α-Mangostin and the inset shows the slope ± SEM of the Boltzmann fits. (C) Activation and deactivation kinetics of BKα and BKα/β1 channels in the basal state and after activation by 10 µM α-Mangostin. The τ ± SEM of activation/deactivation was determined from exponential fits to the current traces at +100 mV, as shown by the green and purple colored lines in panel (A). (D) GV-relationships of BKα channels in different free Ca 2+ concentrations before and after activation by 10 µM α-Mangostin, derived from tail current analysis as above. Pulse voltages were -100 mV to +300 mV/ repolarization to -50 mV from a holding potential of -80 mV for 100 nM free Ca 2+, -120 mV to +200 mV/ repolarization to -50 mV from a holding potential of -80 mV for 1 µM free Ca 2+, and -160 mV to +100 mV/ repolarization to -80 mV from a holding potential of -120 mV for 10 µM free Ca 2+, in symmetrical 140 mM bi-ionic conditions with 140 mM Cs+ as the intracellular ion. The V½ values ± SEM before and after α-Mangostin application and the resulting shifts (Δ V½ ± SEM) are shown in the bar graphs. Data and statistics see Tables S3 – S5.
α-Mangostin predominantly affects deactivation of BKα and BKα/β1 channels
An inspection of the currents elicited by the families of rectangle pulses shows that gating kinetics are altered in the α-Mangostin activated state (Fig. 2A). The shift in V½ towards more negative potentials upon α-Mangostin activation could be caused either by an acceleration of channel activation, or by a slowing down of channel deactivation, or a combination of both. We therefore analyzed the kinetics of activation and deactivation by fitting monoexponential functions to the time course of outward and tail currents for the +100 mV pulse to obtain their time constants (τ). BKα channels are characterized by fast activating current and a fast deactivation visible as decay of the tail current, while the presence of the β1 subunit slows these kinetics down (Fig. 2A, bottom; (Dworetzky et al., 1996)). After application of 10 µM α-Mangostin, activation of BKα channels was moderately accelerated (1.7-fold) with a τ of activation of 7.96 ± 1.64 ms before and 4.71 ± 0.71 ms afterwards, while the τ for deactivation at -50 mV increased ≈7-fold from 0.9 ± 0.04 ms to 6.85 ± 1.11 ms (Fig. 2C). Again, this effect was more pronounced in the smooth muscle-like BKα/β1 channels, where activation was more that 5-fold faster with τ values of 63.76 ± 16.03 ms before and 12.36 ± 1.20 ms after α-Mangostin application. Deactivation kinetics were most affected with a substantial ≈27-fold increase in τ from 3.60 ± 0.16 ms to 95.6 ± 13.99 ms (Fig. 2C). These results suggest that the mechanism of α-Mangostin action differentially affects gating transitions that are associated with activation and deactivation.
As BK channels can be massively activated by Cai2+ under physiological conditions, we further obtained GV-relationships in the basal and in the activated state for higher Cai2+ concentrations relevant in smooth muscle cells to test if α-Mangostin activation is independent of Cai2+ concentration (Fig. 2D). α-Mangostin shifted the V½ in 10 µM free Cai2+ by 50.55 ± 9.13 mV to more negative potentials, comparable to the resting state with 0.1 µM free Cai2+. However, in 1 µM Cai2+ α-Mangostin the V½ shift was slightly higher, resulting in a shift by 82.74 ± 10.17 mV. This is consistent with previous studies, as BKα channel gating is primarily voltage-driven at very low Cai2+ concentrations and Cai2+ driven at high concentrations, while in the low-micromolar range (i.e., around 1 µM), Cai2+ binding sites are partially occupied, resulting in nonlinear gating where enhanced modulator effects can occur (Cui, Cox and Aldrich, 1997; Magleby, 2003; Clay, 2017).
α-Mangostin activation mechanism on single-channel and macroscopic current level
The marked slowing of deactivation visible as the slow tail current decay in macroscopic current recordings suggests that individual channels are open for longer periods of time in the presence of α-Mangostin. To investigate the effect of α-Mangostin on individual channels, we recorded single channel currents in excised inside-out patches from HEK293 cells in the presence of 100 nM free Cai2+ (Fig. 3A). Under these conditions, the open probability (Po) of BK channels at a potential of +40 mV was very low (0.002 ± 0.0008), but rose to 0.77 ± 0.08 after activation by 10 µM α-Mangostin (Fig. 3B) with only very brief closings (Fig 3A, 1s inset). The single channel amplitude derived from the all-points-histogram was not different between basal and α-Mangostin activated states (9.18 ± 0.29 pA and 9.76 ± 0.26 pA; Fig. 3B). We constructed dwell time distributions to determine the differences in the duration that a single channel resides in the closed or open state (Fig. 3C). The Log closed dwell time distributions showed a large shift of the main component to shorter dwell times, from 2.38 ± 0.23 (237 ms) to -1.39 ± 0.36 (less than 0.1 ms). The open dwell time distribution revealed that the log open time was markedly reduced from -0.84 ± 0.45 to 0.8 ± 0.24 (i.e. from 0.14 ms to 6.28 ms). The fact that the open and closed times were both affected suggests that α-Mangostin can bind to the open and the closed state. Accordingly, application of α-Mangostin on closed BK-channels (at – 80mV) resulted in maximal activation with the first depolarization pulse, indicating that α-Mangostin reached its site of action during the closed period (Fig. 3D).

Activation mechanism of α-Mangostin.
(A) Exemplary current traces of a single BKα channel in the basal state and after activation by 10 µM α-Mangostin (1 min recordings; inset shows 1 s; O and C denote open and closed levels). Single-channel currents were recorded at +40 mV in inside-out patches from transiently transfected HEK293 cells in symmetrical potassium gradients with 100 mM free Cai2+ (n=4-7). (B) All-points histograms with Gaussian fits for the basal and α-Mangostin activated state, and bar graphs of the derived mean ± SEM open probabilities (Po) and amplitudes. The inset magnifies the open peak in the basal state. (C) Closed and open dwell time histograms with fits for channels in the basal and in the α-Mangostin activated state derived after event detection in single-channel measurements as shown in (A). (D) Normalized mean ± SEM currents of BKα channels before and after application of 3 µM or 10 µM α-Mangostin in the closed state. Channels were held closed at -80 mV and only very shortly pulsed to +60 mV after 5 min incubation to assess the current size/activation state of the first and the following pulses. Measurements were done in transiently transfected HEK293 cells in whole-cell mode in physiological potassium gradients with 100 nM Cai2+ as shown by the representative current traces to the right and steady-state currents were analyzed (grey arrow). Data and statistics see Table S6.
Localization of the α-Mangostin binding site
We recently described the polypharmacology of the class of negatively charged activators (NCA) in different potassium channels, specifically in TREK-1 and BK channels. The BK channel opener GoSlo-SR-5-6 also activated TREK-1 channels, while BL-1249 known as TREK/TRAAK channel opener, also activated BK channels. We identified their common binding site in the pore close to the fenestration of TREK-1 channels and Molecular Dynamics (MD) simulations predicted the equivalent binding site in the pore of BK channels (Schewe et al., 2019). The question arose if α-Mangostin could also occupy this binding site, as in part suggested in a docking by (Kim et al., 2023), who proposed that P183 and L304, which both are part of the NCA binding site, interact with α-Mangostin in TREK-1 channels. Therefore, we measured the dose-dependent α-Mangostin activation of TREK-1 wildtype channels and its ‘signature’ mutant of the fenestration, L304C. As expected, the apparent affinity is markedly reduced in the L304C mutant in the measurable concentration range (Fig. S2A). Quaternary ammonium ions are known to occupy the central cavity of K+ channels as TREK-1 and BK just below the SF near the NCA binding site (Li and Aldrich, 2004; Piechotta et al., 2011; Fan et al., 2023). In a competition experiment, we recorded dose-response relationships and determined theIC50 value for Tetrapentylammonium (TPA) with and without preactivation by 10 µM α-Mangostin. The TPA IC50 value increased ≈9-fold from 0.72 ± 0.64 µM to 6.33 ± 0.86 µM in the presence of α-Mangostin (Fig. S2B), showing that α-Mangostin was present in the pore and hindered the access of TPA to its binding site. Finally, cysteine scanning mutagenesis of residues in M2 and M4 revealed that mainly G181, I182, P182 on M2 and G308 in M4 were involved, while most M4 residues tested had intermediary effects (Fig. S2C), suggesting that the binding region of α-Mangostin between M2 and M4 overlaps with the NCA binding site identified with the help of BL-1249, but is not completely identical (Fig. S2D).
We next probed if α-Mangostin accesses the pore cavity of BKα channels, as in TREK channels, or binds elsewhere in the protein. We conducted a competition experiment as above and measured the dose-dependent inhibition by 0.1, 1 and 10 µM Tetrahexylammonium (THexA; Fig. 4A). In the presence of α-Mangostin, the inhibition by THexA was clearly reduced, and the apparent THexA affinity (as estimated IC50) was steeply decreased ≈21-fold from 77.51 ± 5.53 nM to 1.64 ± 0.58 µM, showing that the presence of α-Mangostin hindered the access of THexA to its binding site. This is consistent with previous findings that GoSlo-RS-5-6, which binds in the BK pore, also competes with THexA (Roy et al., 2012; Webb et al., 2015; Schewe et al., 2019). In contrast, the presence of BC5, an activator shown to bind at the interface between the transmembrane and intracellular domains (Zhang et al., 2022), did not interfere with THexA binding (estimated THexA IC50 45.60 ± 8.05 nM; Fig. 4A).

Investigation of the binding site of α-Mangostin in BKα channels.
(A) Competition experiment showing a reduction of the block caused by THexA in the presence of α-Mangostin. Left, representative current traces for different THexA concentrations in the presence of 10 µM α-Mangostin; middle, competition experiment analysis showing the relative current of BKα channels in different THexA concentrations in the absence and in the presence of 10 µM α-Mangostin or 100 µM BC5, which does not bind in the pore; and right, estimated IC50 values of THexA alone and in the presence of α-Mangostin or BC5. Whole-cell currents were recorded from transiently transfected HEK293 cells with a ramp protocol (-100 mV to +50 mV) in a physiological potassium gradient with 100 nM free Cai2+ and data are shown as mean ± SEM at +40 mV. (B) Molecular docking of α-Mangostin to the human BK channel structure (PDB ID 6v3g). The full-length structure (green) was reduced to the inner pore region (pink) for the docking, and the zoom-in shows the best pose for α-Mangostin with interacting residues in stick representation; green residues mark hits from the following functional assay. Protein chain B was removed for clarity. (C) Voltage of half-maximal activation (V½) before and after activation by 10 µM α-Mangostin in different pH and the resulting shifts in V½ (Δ V½). The pH was changed intra- and extracellularly. (D) Voltage of half-maximal activation (V½) before and after activation by 10 µM α-Mangostin, and the resulting shifts in V½ (Δ V½). (E) GV-relationships for the six BKα mutants in the S6 segment. (F) Voltage of half-maximal activation (V½) before and after activation by 1 µM GoSlo-SR-5-6, the resulting shifts in V½ (Δ V½), and the GV-relationships for the wildtype and two BKα mutants. GV-relationships were measured as in Fig. 2, and all data represent mean ± SEM. Data and statistics see Tables S9 – S12.
At least one hydroxyl group of α-Mangostin is predicted to be negatively charged in physiological pH (National Center for Biotechnology Information, 2025), therefore we tested the impact of solution pH on the activation potency. Indeed, the shift in V½ induced by 10 µM α-Mangostin was pH-dependent (Fig. 4C). In pH 6, the shift was reduced to 34.71 ± 5.63 mV, while it increased to 75.97 ± 2.02 mV in pH 8.5, indicating that the negative charge is critical for effective activation and α-Mangostin might resemble an NCA-like compound.
In addition, we used molecular docking to determine the location of a possible binding site in the BKα inner pore. We obtained 20 poses which were all located at the cavity wall in middle S6 between residues I308 and V319, in a pocket below the SF, without obstructing the central passageway for K+ ions (Fig. S3). Three poses were clustered with a binding energy of -8.58 kcal mol-1, where the hydrophobic core was wedged in between S6 and S6 of the adjacent subunit. For the best pose, possible molecular interactions were predicted for the residues I308, L312, F315 and A316 in S6 (Fig. 4B; Fig. S3).
To functionally investigate the predicted binding site, we mutated residues in the pore-lining S6 helix with side chains facing into the cavity. Many substitutions in this region affect gating of BK channels and are involved in intersubunit S6-S6 contact (Wu et al., 2009; Zhou, Xia and Lingle, 2011). Therefore, we chose only substitutions that were most WT-like with respect to Cai2+ and voltage sensitivity, i.e., I308A, L312M, A316P for the hits from the docking, and additionally S317R and Y318S as internal controls that were not predicted to be part of the binding site (Chen, Seebohm and Sanguinetti, 2002; Chen, Yan and Aldrich, 2014). We first assessed the shift in voltage activation induced by 10 µM α-Mangostin (Fig. 4D, E). V1/2 shifts of Y318S and S317R were not or only moderately different compared to the wildtype channel (41.46 ± 5.17 mV and 34.33 ± 3.37 mV). In contrast, the three mutants I308A, L312M, and in particular A316P had a markedly reduced shift in V1/2 (19.97 ± 3.12 mV, 27.89 ± 5.42 mV, and 4.56 ± 1.23 mV, respectively). To ensure that the almost absent shift in A316P was not caused by a general disruption of allosteric signal transduction, we also included A316G, which showed a reduction in V1/2 comparable to I308A and L312M (23.57 ± 2.05 mV).
Furthermore, we tested GoSlo-SR-5-6 as alternative activator, which was shown to bind in the same pore region involving A316 (Webb et al., 2015; Schewe et al., 2019; Zhang et al., 2022). Like for α-Mangostin, I308A and A316P reduced the shift in V½ induced by 1 µM GoSlo-SR-5-6 from 42.71 ± 3.05 mV to 25.89 ± 4.05 and 25.0 ± 2.8 mV; however, A316P did not abolish GoSlo-SR-5-6 activation, showing that the complete loss of activation for α-Mangostin was likely caused primarily by a loss of binding and not by an inference of the mutation with the drug transduction mechanism (Fig. 4F). Hence, we conclude that α-Mangostin binds to and activates BKα channels via the upper S6 segment, critically involving residues I308, L312, and A316.
α-Mangostin activation in BK-Cav nanodomains
In resting smooth muscle cells, there is no BK channel activation within their physiological voltage range. However, in native tissue, BK channels are organized in nanodomains together with Cav channels that can generate an increase in Cai2+ concentration in the range of several orders of magnitude in their vicinity, allowing BK channels to open (Shah, Guan and Yan, 2022). This BK-Cav complex can be restored in heterologous expression systems, and, as the intracellular solution contains 5 mM EGTA to suppress an overall increase of Cai2+, BK channels will activate only when nearby Cav channels cause a high local Cai2+ concentration (Berkefeld et al., 2006; Berkefeld and Fakler, 2008). BKα/β1 channels expressed alone therefore produced virtually no current upon stepwise depolarization to +50 mV (Fig. 5A, B), but could be activated by α-Mangostin (Fig. 5A, B). In BKα/β1-Cav complexes, Ca2+ inward currents were elicited, which allowed BKα/β1 channels to open in a more negative voltage range, and these BKα/β1 currents were further enhanced by 10 µM α-Mangostin (Fig. 5A, B). The α-Mangostin-activated current through BKα/β1 channels in the Cav complexes (as difference between BKα/β1-Cav currents before and after activation) was activated at more negative potentials than in BKα/β1 channels alone (Fig. 5B). Hence, α-Mangostin could potentiate BKα/β1 channel currents upon local Cai2+ increase through nearby Cav-channels, as it could occur upon a Cai2+ spike in a cell.

α-Mangostin activation of BK channels in physiological settings.
(A) Representative whole-cell current traces of BKα/β1 channels alone and BKα/β1 coexpressed with Cav1.2 channels before and after application of 10 µM α-Mangostin. Currents were measured in Cai2+-free conditions in a physiological potassium gradient with a family of voltage steps from -50 mV to +50 mV in 10 mV increments. The inset shows voltage activation with a family protocol up to +200 mV to show the presence of BKα/β1 channels. (B) Currents of BKα/β1 channels and BKα/β1 – Cav complexes before and after application of 10 µM α-Mangostin plotted against voltage. The last panel shows the α-Mangostin-activated currents for the range -50 to 10 mV obtained by subtracting the current before α-Mangostin application from the current after application for each potential (mean ± SEM, n=8-11 for each condition). (C) Representative contraction force recordings of aortic preparations from mice. 10 µM α-Mangostin were either applied directly to aortic preparations precontracted with 100 nM Noradrenaline (NA; top), or the precontracted preparations were incubated with 100 nM Iberiotoxin (IbTx) before α-Mangostin application and the contraction force was analyzed 10 min after α-Mangostin addition (dotted lines in recordings). The bar graph shows the normalized contraction force of preparations as mean ± SEM together with the median (orange). Data and statistics see Table S13.
α-Mangostin relaxes vascular tissue in aortic preparations via BK channels
Cardiovascular benefits of Mangostins could arise from vasodilatory effects leading to a reduction of blood pressure. Years ago, a study reported that γ-Mangostin, in which the methoxy group of α-Mangostin is exchanged for a hydroxyl group, induced relaxation of rat aortic rings, but no molecular target was found (Tep-Areenan and Suksamrarn, 2012). We sought to demonstrate that Mangostin-induced vasorelaxation of native vascular tissue is indeed mediated by BK channels. Aortic preparations from mice were equilibrated at a contraction force of 2-3 mN and subsequently precontracted half-maximally with 100 nM noradrenaline. The application of 10 µM α-Mangostin quickly and efficiently induced relaxation, while relaxation was absent or greatly attenuated when the specific BK channel inhibitor Iberiotoxin (IbTx) was applied for 5-6 minutes before α-Mangostin was administered (Fig. 5C). In total, α-Mangostin reduced the normalized contraction force of the preparations by more than 85 % (to 0.16 ± 0.08) compared to the precontracted state, while it did not change after IbTx application alone (1.04 ± 0.002) or after preblock of BK channels (0.78 ± 0.12), proving that BK channels must mediate the vasodilative effect seen upon Mangostin treatment.
Discussion
We provide mechanistic insight into how a natural xanthone compound modulates BK channels, which are major players in vascular function. α-Mangostin activates BK channels by shifting their voltage sensitivity and slowing deactivation, with lesser effects on activation and marked effects on Cai2+ sensitivity. We show that binding of α-Mangostin is state-independent, and we provide evidence for a binding site in the gating-sensitive S6 segment.
Mangostin binds to closed and open channels and promotes the open state
BK channel Po is controlled by Cai2+, voltage, a shift of the closed-open equilibrium of the pore domain, or a combination of those stimuli (Cui, Cox and Aldrich, 1997). Mangostin shifted the voltage activation curve to more negative potentials, meaning an increased open probability at negative voltages. The slope of the GV-relationship was not changed neither in homomeric BKα nor in the smooth muscle BKα/β1 channels, indicating that voltage sensitivity itself was not directly affected by α-Mangostin. Shifts in the V½ values induced by α-Mangostin were present over a wide range of Cai2+ concentrations (0.1, 1 and 10 µM), which implies that the closed-open equilibrium is shifted towards the open state by a mechanism distinct from Cai2+ sensing, or that at least the Cai2+ sensing mechanism is not likely to contribute significantly to α-Mangostin activation. The higher shift observed in in 1 µM Cai2+ likely reflects the known non-linear gating features that result from pronounced allosteric coupling of voltage and Cai2+ signals in this concentration range (Cui, Cox and Aldrich, 1997; Magleby, 2003; Clay, 2017; Sun and Horrigan, 2022).
The activation can be mainly explained by the marked slowing of the deactivation time constants. Consistently, the mean open dwell time was increased in the single channel measurements, while we observed no change in amplitude with and without α-Mangostin (corresponding to a conductance of ≈240 pS). The impact on open as well as closed dwell time distributions and the instantaneous activation of macroscopic currents from the closed state suggest that the molecule binds state-independently.
In the presence of the β1 subunit, which is present in vascular BK channels (Knaus et al., 1994), the activation characteristics were enhanced. The activation time course was little affected in BKα channels, but in BKα/β1 channels an acceleration of the activation caused by α-Mangostin became more prominent, counteracting the slow activation kinetics brought by the β1 subunit (McManus et al., 1995). Additionally, the slow deactivation time course in the presence of the β1 subunit was further slowed down remarkably. As the affinity (as estimated EC50) was very similar and the binding site is located in the S6 segment in the inner pore, the stronger shift in the V½ value is likely not due to changes in binding, but caused by an enhanced shift of the closed-open equilibrium toward the open state, such as the stabilization of the voltage sensor in an active conformation. Such β-subunit dependent effects have also been reported for other modulators, e.g., DHS-I (McManus et al., 1993; Giangiacomo et al., 1998), DHA (Hoshi, Tian, et al., 2013), arachidonic acid (Martín et al., 2021),17β-Estradiol (Valverde et al., 1999), and some GoSlo compounds (Large et al., 2015).
The binding site of α-Mangostin includes residues in the gating-sensitive S6 segment
We demonstrated the presence of α-Mangostin in the channel pore by competition for binding with quaternary ammonium ions known to bind below the SF (Li and Aldrich, 2004; Fan et al., 2023), and, predicted by molecular docking, identified residues in the S6 segment that are part of the binding site.
α-Mangostin binds in the cleft formed by the S6 helices of adjacent subunits, and substitution of the nonpolar residues I308, L312, and especially A316 most strongly reduced the shift in V½ induced by α-Mangostin. All residues are known to participate in gating: they are part of a hydrophobic network within S6 that acts as hinge upon channel opening, and polar substitutions strongly favor the open state (Zhou, Xia and Lingle, 2011; Chen, Yan and Aldrich, 2014; Hite, Tao and MacKinnon, 2017). L312 has further been shown to stabilize intersubunit interaction with F315, whose destruction opens the channel (Wu et al., 2009).
The α-Mangostin binding region overlaps with the reported binding site of the NCA GoSlo-SR-5-6, where L312, A316, S317 and V319 are involved (Webb et al., 2015; Schewe et al., 2019), but it is not identical. When we functionally investigated the α-Mangostin binding site close to the fenestration in TREK-1 channels, we also found only a partial overlap with key residues involved in the NCA binding site. Interaction with P183 and L304 was consistent with an earlier molecular docking (Kim et al., 2023), but the strongest reduction of activation was found for residues in M2 rather than M4.
Mechanism of Mangostin activation
We expect that small structural or electrostatic changes in this gating-sensitive region strongly affect the closed–open equilibrium (Wu et al., 2009; Zhou, Xia and Lingle, 2011; Chen, Yan and Aldrich, 2014). Previous studies suggest several alternative mechanisms by which α-Mangostin could induce channel opening. We have shown that NCAs bind to a region below the selectivity filter (SF) in several K⁺ channels, such as K2P (e.g., TREK), hERG, or BK channels to induce channel opening (Schewe et al., 2019). We speculated that the negative charge common to these activators might alter SF ion occupancy via an electrostatic mechanism, thereby promoting SF opening. Our finding that the potency of α-Mangostin depends on pH suggests that a negative charge may also be critical for its activation mechanism. However, the importance of the SF for BK gating remains unresolved. Furthermore, α-Mangostin also did not increase the single-channel conductance as seen for NCAs in TREK-2 channels, which are known to be gated at the SF (Schewe et al., 2019). Therefore, the contribution of the SF to the Mangostin effect requires further investigation. Alternatively, α-Mangostin binding to the gating-critical S6 region could induce gating at a putative lower gate; however, such a gate is currently speculative, as the available BK structures lack clear evidence for a lower gate (but the final closed state may still be elusive) (Hite, Tao and MacKinnon, 2017; Tao, Hite and MacKinnon, 2017).
In addition to the concepts of an SF gate or a lower gate, hydrophobic gating has been proposed as an alternative gating mechanism in BK channels. MD simulations suggest that the pore of metal-free (presumably closed) BK structures undergoes dewetting, whereas the pore of metal-bound (presumably open) structures remains hydrated (Jia et al., 2018; Gu and de Groot, 2023). Thus, binding of negatively charged α-Mangostin might prevent or antagonize dewetting of the hydrophobic cavity and thereby stabilize the conductive pore state. Indeed, a recent MD study suggested that the NCA NS11021, a smooth muscle relaxant with a biarylthiourea structure, can enter the dewetted BK pore to promote hydration (Rockman, Vouga and Rothberg, 2020b; Nordquist, Jia and Chen, 2024). However, the same MD simulations also indicated that NS11021 has no stable binding pose but engages in various hydrophobic interactions with pore-cavity residues. In contrast, our results suggest a more stable binding pose with specific interactions, as inferred from our mutagenesis data. Clearly, further work is required to elucidate the mechanism of BK channel gating and the exact way Mangostin and other NCAs modulate this process to promote channel activation.
Activation of BK channels in nanodomains and relaxation of aortic tissue
Our data demonstrate that Mangostin is able to potentiate Cav-induced BK currents, and that Mangostin-activated BK channels are responsible for relaxation of mouse vascular tissue, explaining the reduction of blood pressure reported in a recent animal study (Xu et al., 2024).
We reconstructed nanodomains of BK and Cav1.2 channels, as e.g. present in arterioles (Berkefeld et al., 2006; Tykocki, Boerman and Jackson, 2017). In smooth muscle cells, BK activation is strictly coupled to changes in Cai2+. Physiological Cai2+ concentrations lie between 100 nM in the resting state, 0.4-1 µM upon activation of a Ca2+ mobilizing receptor (Savineau and Marthan, 2000; Hill-Eubanks et al., 2011), and up to several ten micromolar (10-40 µM) when sparks form locally (ZhuGe et al., 2002; Berkefeld, Fakler and Schulte, 2010). Transient outward currents through activated BK channels then act as negative feedback, repolarize the cell and terminate Ca2+ influx (Shah, Guan and Yan, 2022). We demonstrated that the α-Mangostin activated current is potentiated by the coupling to Cav channels. BKα/β1 currents in in the physiological voltage range of smooth muscle cells (-50 - -30 mV) would be very small in resting Cai2+, while the Po would be already very high when exceeding 10 µM Ca2+, leaving less room for activation. α-Mangostin would particularly impact potentiation of BK currents upon physiological Ca2+ elevation (i.e. between 1 and 10 µM) and would synergistically lower the BKα/β1 activation threshold to enhance the negative feedback mechanism.
Accordingly, our investigation of precontracted aortic tissue from mice showed a remarkable and robust relaxation after application of α-Mangostin. Such relaxation of aortic rings after application of α- or γ-Mangostin were reported before (Chairungsrilerd et al., 1996, 1997; Tep-Areenan and Suksamrarn, 2012) but the exact molecular target remained inconsistent. Given the strong activation of BK channels and as the relaxation was absent upon preincubation with the selective inhibitor IbTx in our experiments, BK channels must represent the molecular target of Mangostins in vascular smooth muscle cells.
Interestingly, relaxation was also reported for other xanthone derivatives (Cheng and Kang, 1997; Wang et al., 2002; Capettini et al., 2009; Câmara et al., 2010; Diniz et al., 2013), which legitimates further research of substituted xanthones as pharmacophore. Robust data of bioavailability and the existence and nature of active metabolites are currently lacking. A small study claimed that α-Mangostin was bioavailable in humans after supplement ingestion (Kondo et al., 2009). Animal research analyzed metabolites and showed that maximal plasma concentrations are reached 1 hour after oral intake in mice (Ramaiya et al., 2012; Petiwala et al., 2014; Han et al., 2015). However, no metabolites have been functionally investigated to help in optimization of the Mangostin pharmacophore; however, our finding that γ-Mangostin has a slightly higher potency in activating TREK-1 and BKα channels may be a starting point.
Our screen across different K+ channel families revealed that other representatives of the K2P (TWIK-1 and TASK-3) were particularly inhibited by more than 60 %. This could also link other claimed benefits of Mangostin to their molecular targets. Importantly, hERG currents were unaffected, mitigating the cardiotoxicity risk upon consumption of Mangostin nutraceuticals. BK channel function is decreased in chronic conditions that are often associated with the lack of a balanced diet and age, such as metabolic syndrome, diabetes, and obesity (Tykocki, Boerman and Jackson, 2017). Consequences as increased vascular tone and hypertension, ultimately leading to reduced cardiovascular health and premature death, may therefore be addressed with Mangostin xanthones as a potential new class of BK channel activators.
Methods and materials
Substances
α-Mangostin, γ-Mangostin, TPA, THexA, MgATP, NaGTP and noradrenaline were purchased from Merck (Darmstadt, Germany). BC5 (Arg-4-methoxy-2-naphthylamine) was obtained from MP Biomedicals (Irvine, USA), and Iberiotoxin from Alomone Labs (Jerusalem, Israel). GoSlo-SR-5-6 (sodium 1-amino-4-((3trifluoromethylphenyl)amino)-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate)) was provided by Mark Hollywood. Mangostane food supplement stating a content of 50 mg α-Mangostin per capsule was ordered online (Swanson.com). Aiming for a 10 mM stock solution with respect to α-Mangostin, 205 mg of the powder found in the capsules was dispersed in 5 ml DMSO, brought into solution by vortexing and ultrasonication, and filtered through a 0.45 µM PTFE membrane to remove remaining particulate matter. TPA, THexA, NA, BC5, and Iberiotoxin were prepared as 1:1000 stocks in H2O, and the Mangostins and GoSlo-RS-5-6 in DMSO at 50 mM. Aliquots were stored at - 20°C and diluted to the final concentration in extracellular solution.
Molecular biology, cell culture and transfection
Coding sequences for channels and subunits listed in Table 1 were subcloned in pFAW or pcDNA3.1 vectors containing a CMV promotor for expression in HEK293 cells. Amino acid substitutions were introduced by site-directed mutagenesis PCR according to the QuikChange protocol (Stratagene, La Jolla, USA) with custom primers containing the desired base exchange. All constructs were verified by Sanger sequencing. To enhance plasma membrane expression, the retrieval motif was removed in hTWIK-1 and hTWIK-2 channels by the substitutions I293A, I294A (Feliciangeli et al., 2010) and I289A, L290A (Bobak et al., 2017), respectively, yielding hTWIK-1m and hTWIK-2m channels.


Channels and subunits used in this study
HEK293 cells were cultivated in Dulbecco’s Modified Eagles Medium (DMEM) supplemented with 10 % FCS and penicillin/streptomycin (100 U ml-1/100 µg ml-1) in 5 % CO2 at 37 °C. For cultivation of CHO-K1 cells, the medium was additionally supplemented with non-essential amino acid mix (Gibco MEM-NEAA; Thermo Fisher, Schwerte, Germany) and 10 mM Hepes.
HEK293 cells were transiently transfected with Lipofectamine 2000 (Invitrogen, Thermo Fisher, Schwerte, Germany) or FuGENE (Promega, Walldorf, Germany) as to the manufacturer’s instructions and incubated overnight. pEYFP was included as transfection marker. In BKα/β1 coexpressions, the β1 subunit was used in 10-fold excess to ensure a uniform channel population. Cells were trypsinized and seeded onto glass coverslips 2-3 h prior to electrophysiological experiments.
Electrophysiology
Voltage clamp experiments were conducted with a HEKA EPC10 amplifier controlled by Patchmaster Software (v2.78; HEKA Elektronik, Lambrecht, Germany). Standard measurements were done in transiently transfected HEK293 cells in the whole-cell configuration in physiological potassium gradients. Pipettes were pulled from thin-walled borosilicate glass, fire-polished, and had resistances of 1.4 – 2 MΩ. Series resistance was compensated to at least 70 %. The extracellular solution contained (in mM): 135 NaCl, 5 KCl, 2 CaCl2, 2 MgCl2, 10 D(+)-glucose, 10 Hepes (pH 7.3). The intracellular solution was (in mM): 140 KCl, 2 MgCl2, 1 CaCl2, 2.5 EGTA, 10 Hepes (pH 7.3), corresponding to ≈100 nM free Ca2+. For Kir 1.1 and 2.1 channel recordings, 3 mM MgATP and 0.3 mM NaGTP were included. Where indicated, K+ was substituted for Cs+ to reduce the outward currents of BK channels. Cells were held at -80 mV and a voltage ramp from -100 mV to +60 mV of 1 s duration was applied every 5 s. For Kv channels a rectangle pulse was used as indicated in the figure legends.
The activation of BK channels was measured as conductance 𝐺 in symmetrical potassium solutions to ensure a unitary conductance across the voltage range. Cs+ was used as intracellular ion to reduce outward currents. The extracellular solution contained (in mM): 140 KCl, 2 CaCl2, 2 MgCl2, 10 D(+)-glucose, 10 Hepes (pH 7.3). The intracellular solution was (in mM): 140 CsCl, 2 MgCl2, 1 CaCl2, 2.5 EGTA, 10 Hepes (pH 7.3), corresponding to ≈100 nM free Ca2+ (calculated with WEBMAXC (https://somapp.ucdmc.ucdavis.edu/pharmacology/bers/maxchelator/webmaxc/webmaxcS.htm). For recordings with 1 µM and 10 µM free Cai2+, CaCl2 and EGTA were raised to 2 mM / 2.2 mM and 2 mM / 4.04 mM, respectively. Cells were held at negative holding potentials and currents were elicited with a family of rectangle voltage pulses in 20 mV increments to positive potentials as needed for saturation of the tail currents, followed by a repolarization step to elicit the tail currents as detailed in the figure legends. The currents were analyzed with Fitmaster (HEKA Elektronik, Lambrecht, Germany), and conductance-voltage (G-V) relationships were generated from normalized tail currents and fit to a standard Boltzmann relationship (1) to obtain 𝑉1⁄2 values. Individual fits were then averaged for mean 𝑉1⁄2 values.

where 𝐺𝑚𝑎𝑥 is the maximal tail current, 𝑉1⁄2 is the voltage of half-maximal activation of the current, and 𝑘 is the slope factor.
Activation in different pH was measured using the following solutions: Extracellular solution pH 8.5 contained (in mM): 140 KCl, 2 CaCl2, 2 MgCl2, 10 D(+)-glucose, 10 Ampso; intracellular solution pH 8.5 contained (in mM): 140 CsCl2, 10 Ampso, 2 MgCl2, 1 CaCl2, 2.05 EGTA. Extracellular solution pH 6 was (in mM): 140 KCl, 2 CaCl2, 2 MgCl2, 10 D(+)-glucose, 10 MES; intracellular solution pH 6 contained (in mM): 140 CsCl2, 10 MES, 2 MgCl2, 0.5 CaCl2, 11 EDTA. The pH was changed intra- and extracellularly.
In TPA competition experiments for TREK-1 channels a dose-response relationship in the absence and presence of α-Mangostin was recorded and the IC50 was obtained from a Hill fit. The THexA competition experiments for BKα channels were conducted as 3-point determinations using 0.1 µM and 1 µM THexA as concentrations close to half-maximal inhibition without and with α-Mangostin and 10 µM THexA for full block. To estimate the IC50, a simplified 4-parameter logistic regression was used, assuming 𝑎 = 0 and 𝑏 = 1.

where a: lower asymptote, b: slope, c: IC50, d: upper asymptote, x: THexA concentration, and y: current.
Single-channel recordings
Single channels were recorded at room temperature from excised inside-out patches of HEK293 cells transiently transfected with BKα in a continuous voltage protocol at +40 mV. To increase the chances of obtaining patches with only one channel, a pBF vector with a suboptimal beta globin promotor yielding only low expression was used. Pipette resistances were 12-15 MΩ, and symmetrical intra- and extracellular solution contained (in mM) 140 KCl, 2 MgCl2, 1 CaCl2, 2.5 EGTA,10 Hepes (pH 7.3 with KOH), corresponding to ≈100 nM free Ca2+. Currents were recorded at a sampling rate of 100 kHz with a final bandwith 𝑓𝑐 of 7.4 kHz using an EPC10 amplifier controlled by Patchmaster software (v2.78; HEKA Elektronik, Lambrecht, Germany). Traces were analyzed in Clampfit (v11.2.2.17; Molecular Devices, San Jose, USA). The filter rise time 

BKα/β1 – Cav complexes were restored as described by (Berkefeld et al., 2006) with the following modification: For nanodomain formation of Cav 1.2 and BKα/β1, CHO-K1 cells were seeded onto coverslips and microinjected (InjectMan 4, Eppendorf, Hamburg, Germany) with a DNA mixture of Cavα 1C, Cav β1, Cav α2δ1, BKα, β1 and EYFP in the ratio 10:10:10:10:100:1 to ensure the presence of all subunits. Cells were incubated overnight and currents were recorded from EYFP-positive cells 14-18 h after injection. Cells were measured in a physiological potassium gradient with a voltage step protocol from -50 mV to +60 mV in 10 mV increments. The extracellular solution contained (in mM): 135 NaCl, 5 KCl, 2 CaCl2, 2 MgCl2, 10 D(+)-glucose, 10 Hepes (pH 7.3). The intracellular solution was (in mM): 140 KCl, 2 MgCl2, 5 EGTA, 10 Hepes (pH 7.3). α-Mangostin was added to the bath solution at a final concentration of 10 µM.
Molecular Docking
The input structure for molecular docking was generated by truncating the structure of the human BKα channel (PDB ID 6vg3) to only the pore region (res T229-R329) using PyMOL (Schrodinger LLC, 2015). Docking was performed with AutoDock4 (v4.2.6) using MGL Tools (v1.5.7) (Morris et al., 2009). A run with lower resolution grid was done and after α-Mangostin was seen in the pore, the docking was repeated with a gridbox restricted to the cavity below the SF. Clustering and interactions were analyzed with the built-in functions of MGL Tools.
Aortic smooth muscle preparations
1 female and 7 male CD1/CHR2 mice were killed by decapitation following isoflurane-narcosis and the aorta was removed. The aortic wall was dissected and tissue strips of ≈15x2 mm were obtained using the spiral cut technique (Peiper and Schmidt, 1972). Aortic preparations were tethered to glass holders and transferred to temperature-controlled 37 °C bath chambers filled with Krebs solution (pH 7.4; in mM: 112 NaCl, 4.7 KCl, 2.5 CaCl2, 1.2 MgCl2, 1.2 KH2PO4 25 NaHCO3 and 11.5 Glucose). The bath solution was bubbled with 95 % O2/ 5 % CO2. The contraction force was recorded with mechanoelectric transducers (Fort100, WPI, Sarasota, USA), connected to a PowerLab controlled by LabChart (AD Instruments, Mannheim, Germany). A prestrain of 2.5-3.9 mN was applied, depending on the thickness of preparations, and strips equilibrated for ca. 45 minutes. The NA concentration necessary to achieve half-maximal contraction was determined by measuring a dose-response relationship and 100 nM NA were subsequently used for precontraction. After a plateau was reached (2-4 minutes), α-Mangostin and IbTx were diluted into the bath solution to their final concentrations of 10 µM and 100 nM, respectively, and the contraction force was recorded for ca. 20 minutes. Recordings were analyzed with LabChart.
Data analysis and statistics
All data are given as mean ± SEM. Mean currents were analyzed with Patchmaster (HEKA Elektronik, Lambrecht, Germany). Fitmaster (HEKA Elektronik, Lambrecht, Germany) was used to generate GV-relationships and fits with the Boltzmann function, and for the monoexponential fits to obtain activation and deactivation time constants. EC50 / IC50 values were obtained by fitting a Hill equation to the dose-response data using IgorPro (v6.3.7; WaveMetrics, Portland, USA) or Prism (v8.4.3; Graphpad Software Inc., San Diego, USA).

With I0, basal current; Imax, activated current; c, concentration; c½ EC50 / IC50; h, Hill coefficient. Statistical analysis was conducted with the built-in functions of Prism. If data were not normally distributed, nonparametric tests were used. Variances were compared with F-tests. Two means were then compared with a paired or unpaired t-test. Three or more groups were compared using one-way ANOVA when variances and standard deviation were considered equal by Bartlett’s test, otherwise the Brown-Forsythe and Welch ANOVA was applied. Dunnett’s T3 post-hoc test was used to account for multiple comparisons in ANOVA, and Dunn’s post-hoc test was used for non-parametric tests. Data and statistical tests are reported in supplementary tables for each figure. Symbols for P values are used in the figures as follows: *** (P <0.001), ** (P ≤0.01), * (P ≤0.05), ns (P ≥0. 05).
Structures were visualized with PyMOL (Schrodinger LLC, 2015) and figures were assembled with Inkscape v1.3 (GNU General Public License, v3).
Data availability
All analyzed data are included in supplementary tables to the manuscript. Electrophysiological source data are available from the corresponding author upon request.
Acknowledgements
Parts of this study were supported by the DFG grant 506373940 to MM and TB.
Plasmids containing the BK β1 subunit and the different Cav1.2 subunits were a gift of Bernd Fakler (Universität Freiburg). Kv1.3 was kindly provided by Heinrich Terlau (Christian-Albrechts-Universität zu Kiel). GoSlo-SR-5-6 was received by courtesy of Mark Hollywood (Dundalk Institute of Technology). We thank Michaela Unmack, Sandra Grüssel, Henning Janssen and Petra Breiden for excellent technical assistance.
Additional information
Ethics declaration
Breeding, housing and procedures to obtain aortic tissue from mice were licensed and performed according to German animal protection law, the regulations of the state authorities of Mecklenburg-West Pomerania, and the standards of the University of Rostock.
The authors declare no competing interest.
Abbreviations
β1: regulatory β1-subunit of BK channels
BKα: Big Potassium (BK) channel pore-forming α-subunit (Slo1, KCa1.1, MaxiK)
BKα/β1: complex of BKα channel and β1 subunit
Cav: voltage-gated Calcium channel
hERG: human Ether-a-go-go Related Gene potassium channel
IbTx: Iberiotoxin
K2P: Tandem-Pore-Domain Potassium channel
Kir: inward-rectifier Potassium channel
Kv: voltage-gated Potassium channel
MD: Molecular Dynamics (simulations)
NCA: Negatively Charged Activator, e.g., BL-1249
ROMK: Renal Outer Medullary Potassium channel
SF: Selectivity Filter
SUR: Sulfonylurea Receptor regulatory subunit of KATP channels
THexA: Tetrahexylammonium ion
TPA: Tetrapentylammonium ion
Funding
Deutsche Forschungsgemeinschaft (DFG) (506373940)
Marianne A Musinszki
Thomas Baukrowitz
Additional files
References
- 1.The effect of vitamin E and beta carotene on the incidence of lung cancer and other cancers in male smokersThe New England Journal of Medicine 330:1029–1035https://doi.org/10.1056/NEJM199404143301501Google Scholar
- 2.Pulmonary Circulation Under Pressure: Pathophysiological and Therapeutic Implications of BK ChannelCardiovascular Drugs and Therapy https://doi.org/10.1007/s10557-023-07503-7Google Scholar
- 3.BK channel activators and their therapeutic perspectivesFrontiers in Physiology 5https://www.frontiersin.org/articles/10.3389/fphys.2014.00389
- 4.BKCa-Cav Channel Complexes Mediate Rapid and Localized Ca2+-Activated K+ SignalingScience 314:615–620https://doi.org/10.1126/science.1132915Google Scholar
- 5.Repolarizing Responses of BKCa–Cav Complexes Are Distinctly Shaped by Their Cav SubunitsJournal of Neuroscience 28:8238–8245https://doi.org/10.1523/JNEUROSCI.2274-08.2008Google Scholar
- 6.Ca2+-Activated K+ Channels: From Protein Complexes to FunctionPhysiological Reviews 90:1437–1459https://doi.org/10.1152/physrev.00049.2009Google Scholar
- 7.Recombinant tandem of pore-domains in a Weakly Inward rectifying K+ channel 2 (TWIK2) forms active lysosomal channelsScientific Reports 7:649https://doi.org/10.1038/s41598-017-00640-8Google Scholar
- 8.Vasoregulation by the β1 subunit of the calcium-activated potassium channelNature 407:870–876https://doi.org/10.1038/35038011Google Scholar
- 9.Resveratrol for the Management of Human Health: How Far Have We Come? A Systematic Review of Resveratrol Clinical Trials to Highlight Gaps and OpportunitiesInternational Journal of Molecular Sciences 25:747https://doi.org/10.3390/ijms25020747Google Scholar
- 10.Mechanism of the vasodilator effect of Euxanthone in rat small mesenteric arteriesPhytomedicine 17:690–692https://doi.org/10.1016/j.phymed.2009.12.003Google Scholar
- 11.Vasodilator and Antioxidant Effect of Xanthones Isolated from Brazilian Medicinal PlantsPlanta Medica 75:145–148https://doi.org/10.1055/s-0028-1088388Google Scholar
- 12.Pharmacological properties of alpha-mangostin, a novel histamine H1 receptor antagonistEuropean Journal of Pharmacology 314:351–356https://doi.org/10.1016/s0014-2999(96)00562-6Google Scholar
- 13.γ-Mangostin, a novel type of 5-hydroxytryptamine 2A receptor antagonistNaunyn-Schmiedeberg’s Archives of Pharmacology 357:25–31https://doi.org/10.1007/PL00005134Google Scholar
- 14.Position of aromatic residues in the S6 domain, not inactivation, dictates cisapride sensitivity of HERG and eag potassium channelsProceedings of the National Academy of Sciences of the United States of America 99:12461–12466https://doi.org/10.1073/pnas.192367299Google Scholar
- 15.BK channel opening involves side-chain reorientation of multiple deep-pore residuesProceedings of the National Academy of Sciences 111:E79–E88https://doi.org/10.1073/pnas.1321697111Google Scholar
- 16.Mechanism of vasorelaxation of thoracic aorta caused by xanthoneEuropean Journal of Pharmacology 336:23–28https://doi.org/10.1016/S0014-2999(97)01224-7Google Scholar
- 17.Novel description of the large conductance Ca2+-modulated K+ channel current, BK, during an action potential from suprachiasmatic nucleus neuronsPhysiological Reports 5:e13473https://doi.org/10.14814/phy2.13473Google Scholar
- 18.Fitting and Statistical Analysis of Single-Channel RecordsIn:
- Sakmann B.
- Neher E.
- 19.Intrinsic Voltage Dependence and Ca2+ Regulation of mslo Large Conductance Ca-activated K+ ChannelsJournal of General Physiology 109:647–673https://doi.org/10.1085/jgp.109.5.647Google Scholar
- 20.Quercetin as a possible complementary agent for early-stage COVID-19: Concluding results of a randomized clinical trialFrontiers in Pharmacology 13https://doi.org/10.3389/fphar.2022.1096853Google Scholar
- 21.Mechanism of the Vasodilator Effect of Mono-oxygenated Xanthones: A Structure-Activity Relationship StudyPlanta Medica 79:1495–1500https://doi.org/10.1055/s-0033-1350803Google Scholar
- 22.LINGO1 is a regulatory subunit of large conductance, Ca2+-activated potassium channelsProceedings of the National Academy of Sciences 117:2194–2200https://doi.org/10.1073/pnas.1916715117Google Scholar
- 23.Phenotypic alteration of a human BK (hSlo) channel by hSlobeta subunit coexpression: changes in blocker sensitivity, activation/relaxation and inactivation kinetics, and protein kinase A modulationThe Journal of Neuroscience: The Official Journal of the Society for Neuroscience 16:4543–4550https://doi.org/10.1523/JNEUROSCI.16-15-04543.1996Google Scholar
- 24.Cardioprotective effects of alpha-mangostin on doxorubicin-induced cardiotoxicity in ratsPhytotherapy Research 36:506–524https://doi.org/10.1002/ptr.7356Google Scholar
- 25.Calcium-gated potassium channel blockade via membrane-facing fenestrationsNature Chemical Biology :1–10https://doi.org/10.1038/s41589-023-01406-2Google Scholar
- 26.Potassium channel silencing by constitutive endocytosis and intracellular sequestrationThe Journal of Biological Chemistry 285:4798–805https://doi.org/10.1074/jbc.M109.078535Google Scholar
- 27.Therapeutic efficacy of the BKCa channel opener chlorzoxazone in a mouse model of Fragile X syndromeNeuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology 49:2032–2041https://doi.org/10.1038/s41386-024-01956-6Google Scholar
- 28.Mechanism of Maxi-K Channel Activation by Dehydrosoyasaponin-IJournal of General Physiology 112:485–501https://doi.org/10.1085/jgp.112.4.485Google Scholar
- 29.Regulation of two-pore-domain (K2P) potassium leak channels by the tyrosine kinase inhibitor genisteinBritish Journal of Pharmacology 154:1680–90https://doi.org/10.1038/bjp.2008.213Google Scholar
- 30.Regulation of BK Channels by Beta and Gamma SubunitsAnnual review of physiology 81:113–137https://doi.org/10.1146/annurev-physiol-022516-034038Google Scholar
- 31.The BK channel-NS1619 agonist complex reveals molecular insights on allosteric activation gatingbioRxiv :2025.03.27.645783https://doi.org/10.1101/2025.03.27.645783Google Scholar
- 32.Central cavity dehydration as a gating mechanism of potassium channelsNature Communications 14:2178https://doi.org/10.1038/s41467-023-37531-8Google Scholar
- 33.Dose-Independent ADME Properties and Tentative Identification of Metabolites of α-Mangostin from Garcinia mangostana in Mice by Automated Microsampling and UPLC-MS/MS MethodsPLOS One 10:e0131587https://doi.org/10.1371/journal.pone.0131587Google Scholar
- 34.The beta subunit of the high-conductance calcium-activated potassium channel contributes to the high-affinity receptor for charybdotoxinProceedings of the National Academy of Sciences of the United States of America 94:2853–2858https://doi.org/10.1073/pnas.94.7.2853Google Scholar
- 35.Lack of effect of long-term supplementation with beta carotene on the incidence of malignant neoplasms and cardiovascular diseaseThe New England Journal of Medicine 334:1145–1149https://doi.org/10.1056/NEJM199605023341801Google Scholar
- 36.Calcium signaling in smooth muscleCold Spring Harbor Perspectives in Biology 3:a004549https://doi.org/10.1101/cshperspect.a004549Google Scholar
- 37.Structural basis for gating the high-conductance Ca(2+)-activated K(+) channelNature 541:52–57https://doi.org/10.1038/nature20775Google Scholar
- 38.Coupling between voltage sensor activation, Ca2+ binding and channel opening in large conductance (BK) potassium channelsThe Journal of General Physiology 120:267–305https://doi.org/10.1085/jgp.20028605Google Scholar
- 39.A point mutation in the human Slo1 channel that impairs its sensitivity to omega-3 docosahexaenoic acidJournal of General Physiology 142:507–522https://doi.org/10.1085/jgp.201311061Google Scholar
- 40.Mechanism of the modulation of BK potassium channel complexes with different auxiliary subunit compositions by the omega-3 fatty acid DHAProceedings of the National Academy of Sciences 110:4822–4827https://doi.org/10.1073/pnas.1222003110Google Scholar
- 41.Modulation of BK Channels by Small Endogenous Molecules and Pharmaceutical Channel OpenersInternational Review of Neurobiology 128:193–237https://doi.org/10.1016/bs.irn.2016.03.020Google Scholar
- 42.Cancer chemopreventive activity of resveratrol, a natural product derived from grapesScience (New York, N.Y.) 275:218–220https://doi.org/10.1126/science.275.5297.218Google Scholar
- 43.Hydrophobic gating in BK channelsNature Communications 9:3408https://doi.org/10.1038/s41467-018-05970-3Google Scholar
- 44.The metabolic and molecular mechanisms of α-mangostin in cardiometabolic disorders (Review)International Journal of Molecular Medicine 50:120https://doi.org/10.3892/ijmm.2022.5176Google Scholar
- 45.Baicalein and wogonin are activators of rat TREK-2 two-pore domain K+ channelActa Physiologica (Oxford, England) 202:185–192https://doi.org/10.1111/j.1748-1716.2011.02263.xGoogle Scholar
- 46.Inhibitory effects of α-Mangostin on T cell cytokine secretion via ORAI1 calcium channel and K+ channels inhibitionPeerJ 9:e10973https://doi.org/10.7717/peerj.10973Google Scholar
- 47.Multi-target modulation of ion channels underlying the analgesic effects of α-mangostin in dorsal root ganglion neuronsPhytomedicine 115:154791https://doi.org/10.1016/j.phymed.2023.154791Google Scholar
- 48.Subunit composition of the high conductance calcium-activated potassium channel from smooth muscle, a representative of the mSlo and slowpoke family of potassium channelsThe Journal of Biological Chemistry 269:3921–3924Google Scholar
- 49.Bioavailability and Antioxidant Effects of a Xanthone-Rich Mangosteen (Garcinia mangostana) Product in HumansJournal of Agricultural and Food Chemistry 57:8788–8792https://doi.org/10.1021/jf901012fGoogle Scholar
- 50.γ-mangostin attenuates amyloid-β42-induced neuroinflammation and oxidative stress in microglia-like BV2 cells via the mitogen-activated protein kinases signaling pathwayEuropean Journal of Pharmacology 917:174744https://doi.org/10.1016/j.ejphar.2022.174744Google Scholar
- 51.Effects of the novel BK (KCa1.1) channel opener GoSlo-SR-5-130 are dependent on the presence of BKβ subunitsBritish Journal of Pharmacology 172:2544–2556https://doi.org/10.1111/bph.13085Google Scholar
- 52.Molecular Determinants of BK Channel Functional Diversity and FunctioningPhysiological Reviews 97:39–87https://doi.org/10.1152/physrev.00001.2016Google Scholar
- 53.Virus-induced senescence is a driver and therapeutic target in COVID-19Nature 599:283–289https://doi.org/10.1038/s41586-021-03995-1Google Scholar
- 54.Modulation of BK Channel Function by Auxiliary Beta and Gamma SubunitsInternational review of neurobiology 128:51–90https://doi.org/10.1016/bs.irn.2016.03.015Google Scholar
- 55.Alpha-Mangostin Ameliorates Bleomycin-Induced Pulmonary Fibrosis in Mice Partly Through Activating Adenosine 5′-Monophosphate-Activated Protein KinaseFrontiers in Pharmacology 10:1305https://doi.org/10.3389/fphar.2019.01305Google Scholar
- 56.Unique Inner Pore Properties of BK Channels Revealed by Quaternary Ammonium BlockJournal of General Physiology 124:43–57https://doi.org/10.1085/jgp.200409067Google Scholar
- 57.Gating mechanism of BK (Slo1) channels: so near, yet so farThe Journal of General Physiology 121:81–96https://doi.org/10.1085/jgp.20028721Google Scholar
- 58.α-Mangostin: A Xanthone Derivative in Mangosteen with Potent Anti-Cancer PropertiesBiomolecules 14:1382https://doi.org/10.3390/biom14111382Google Scholar
- 59.Arachidonic acid effect on the allosteric gating mechanism of BK (Slo1) channels associated with the β1 subunitBiochimica et Biophysica Acta (BBA) - Biomembranes 1863:183550https://doi.org/10.1016/j.bbamem.2021.183550Google Scholar
- 60.An activator of calcium-dependent potassium channels isolated from a medicinal herbBiochemistry 32:6128–6133https://doi.org/10.1021/bi00075a002Google Scholar
- 61.Functional role of the beta subunit of high conductance calcium-activated potassium channelsNeuron 14:645–650https://doi.org/10.1016/0896-6273(95)90321-6Google Scholar
- 62.AutoDock4 and AutoDockTools4: Automated Docking with Selective Receptor FlexibilityJournal of computational chemistry 30:2785–2791https://doi.org/10.1002/jcc.21256Google Scholar
- 63.BK Channel Modulators: A Comprehensive OverviewCurrent Medicinal Chemistry 15:1126–1146https://doi.org/10.2174/092986708784221412Google Scholar
- 64.PubChem Compound Summary for CID 5281650, Mangostinhttps://pubchem.ncbi.nlm.nih.gov/compound/Mangostin
- 65.Small Molecule NS11021 Promotes BK Channel Activation by Increasing Inner Pore HydrationJournal of Chemical Information and Modeling 64:7616–7625https://doi.org/10.1021/acs.jcim.4c01012Google Scholar
- 66.Differential Effects of β1 and β2 Subunits on BK Channel ActivityThe Journal of General Physiology 125:395–411https://doi.org/10.1085/jgp.200409236Google Scholar
- 67.Relaxation of coronary arteries by electro-mechanical decoupling or adrenergic stimulationPflugers Archiv: European Journal of Physiology 337:107–117https://doi.org/10.1007/BF00587834Google Scholar
- 68.Ion channel molecular complexes in vascular smooth muscleFrontiers in Physiology 13:999369https://doi.org/10.3389/fphys.2022.999369Google Scholar
- 69.Pharmacokinetic characterization of mangosteen (Garcinia mangostana) fruit extract standardized to α-mangostin in C57BL/6 miceNutrition Research 34:336–345https://doi.org/10.1016/j.nutres.2014.03.002Google Scholar
- 70.The pore structure and gating mechanism of K2P channelsThe EMBO Journal 30:3607–19https://doi.org/10.1038/emboj.2011.268Google Scholar
- 71.Relationship between pore occupancy and gating in BK potassium channelsThe Journal of General Physiology 127:557–576https://doi.org/10.1085/jgp.200509482Google Scholar
- 72.Single Dose Oral Pharmacokinetic Profile of α-Mangostin in MiceCurrent Drug Targets 13:1698–1704https://doi.org/10.2174/138945012804545524Google Scholar
- 73.Flavonoids as Modulators of Potassium ChannelsInternational Journal of Molecular Sciences 24:1311https://doi.org/10.3390/ijms24021311Google Scholar
- 74.Molecular mechanism of BK channel activation by the smooth muscle relaxant NS11021Journal of General Physiology 152:e201912506https://doi.org/10.1085/jgp.201912506Google Scholar
- 75.Molecular mechanism of BK channel activation by the smooth muscle relaxant NS11021The Journal of General Physiology 152:e201912506https://doi.org/10.1085/jgp.201912506Google Scholar
- 76.Structure–Activity Relationships of a Novel Group of Large-Conductance Ca2+-Activated K+ (BK) Channel Modulators: The GoSlo-SR FamilyChemMedChem 7:1763–1769https://doi.org/10.1002/cmdc.201200321Google Scholar
- 77.Cytosolic Calcium Oscillations in Smooth Muscle CellsPhysiology 15:50–55https://doi.org/10.1152/physiologyonline.2000.15.1.50Google Scholar
- 78.A pharmacological master key mechanism that unlocks the selectivity filter gate in K + channelsScience 363:875–880https://doi.org/10.1126/science.aav0569Google Scholar
- 79.The PyMOL Molecular Graphics SystemSchrodinger LLC 1.8
- 80.Structural and Functional Coupling of Calcium-Activated BK Channels and Calcium-Permeable Channels Within Nanodomain Signaling ComplexesFrontiers in Physiology 12https://doi.org/10.3389/fphys.2021.796540Google Scholar
- 81.A gating lever and molecular logic gate that couple voltage and calcium sensor activation to opening in BK potassium channelsScience Advances 8:eabq5772https://doi.org/10.1126/sciadv.abq5772Google Scholar
- 82.Cryo-EM structure of the open high-conductance Ca2+-activated K+ channelNature 541:46–51https://doi.org/10.1038/nature20608Google Scholar
- 83.In vivo antibacterial activity of Garcinia mangostana pericarp extract against methicillin-resistant Staphylococcus aureus in a mouse superficial skin infection modelPharmaceutical Biology 54:2606–2615https://doi.org/10.3109/13880209.2016.1172321Google Scholar
- 84.Mechanisms of vasorelaxation to gamma-mangostin in the rat aortaJournal of the Medical Association of Thailand = Chotmaihet Thangphaet 95:S63–68Google Scholar
- 85.Smooth Muscle Ion Channels and Regulation of Vascular Tone in Resistance Arteries and ArteriolesIn: Comprehensive Physiology John Wiley & Sons, Ltd pp. 485–581https://doi.org/10.1002/cphy.c160011Google Scholar
- 86.Acute activation of Maxi-K channels (hSlo) by estradiol binding to the beta subunitScience (New York, N.Y.) 285:1929–1931https://doi.org/10.1126/science.285.5435.1929Google Scholar
- 87.Antihypertensive and vasorelaxing activities of synthetic xanthone derivativesBioorganic & Medicinal Chemistry 10:567–572https://doi.org/10.1016/S0968-0896(01)00315-7Google Scholar
- 88.Molecular mechanisms underlying the effect of the novel BK channel opener GoSlo: Involvement of the S4/S5 linker and the S6 segmentProceedings of the National Academy of Sciences 112:2064–2069https://doi.org/10.1073/pnas.1400555112Google Scholar
- 89.The BK potassium channel in the vascular smooth muscle and kidney: α- and β-subunitsKidney International 78:963–974https://doi.org/10.1038/ki.2010.325Google Scholar
- 90.Intersubunit Coupling in the Pore of BK ChannelsJournal of Biological Chemistry 284:23353–23363https://doi.org/10.1074/jbc.M109.027789Google Scholar
- 91.Association between circulating vitamin E and ten common cancers: evidence from large-scale Mendelian randomization analysis and a longitudinal cohort studyBMC Medicine 20:168https://doi.org/10.1186/s12916-022-02366-5Google Scholar
- 92.α-Mangostin reduces hypertension in spontaneously hypertensive rats and inhibits EMT and fibrosis in Ang II-induced HK-2 cellsInternational Journal of Medical Sciences 21:1681–1688https://doi.org/10.7150/ijms.94236Google Scholar
- 93.An allosteric modulator activates BK channels by perturbing coupling between Ca2+ binding and pore openingNature Communications 13:6784https://doi.org/10.1038/s41467-022-34359-6Google Scholar
- 94.Cysteine scanning and modification reveal major differences between BK channels and Kv channels in the inner pore regionProceedings of the National Academy of Sciences of the United States of America 108:12161–12166https://doi.org/10.1073/pnas.1104150108Google Scholar
- 95.Spontaneous Transient Outward Currents Arise from Microdomains Where BK Channels Are Exposed to a Mean Ca2+ Concentration on the Order of 10 μM during a Ca2+ SparkThe Journal of General Physiology 120:15–27https://doi.org/10.1085/jgp.20028571Google Scholar
Article and author information
Author information
Version history
- Preprint posted:
- Sent for peer review:
- Reviewed Preprint version 1:
Cite all versions
You can cite all versions using the DOI https://doi.org/10.7554/eLife.109479. This DOI represents all versions, and will always resolve to the latest one.
Copyright
© 2025, Cordeiro et al.
This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.
Metrics
- views
- 0
- downloads
- 0
- citations
- 0
Views, downloads and citations are aggregated across all versions of this paper published by eLife.