Korean J Physiol Pharmacol.  2020 Nov;24(6):503-516. 10.4196/kjpp.2020.24.6.503.

The agonistic action of URO-K10 on Kv7.4 and 7.5 channels is attenuated by co-expression of KCNE4 ancillary subunit

Affiliations
  • 1Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
  • 2Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Korea
  • 3Department of Urology, Samsung Medical Center, Seoul 06351, Korea

Abstract

KCNQ family constitutes slowly-activating potassium channels among voltage-gated potassium channel superfamily. Recent studies suggested that KCNQ4 and 5 channels are abundantly expressed in smooth muscle cells, especially in lower urinary tract including corpus cavernosum and that both channels can exert membrane stabilizing effect in the tissues. In this article, we examined the electrophysiological characteristics of overexpressed KCNQ4, 5 channels in HEK293 cells with recently developed KCNQ-specific agonist. With submicromolar EC50 , the drug not only increased the open probability of KCNQ4 channel but also increased slope conductance of the channel. The overall effect of the drug in whole-cell configuration was to increase maximal whole-cell conductance, to prolongate the activation process, and left-shift of the activation curve. The agonistic action of the drug, however, was highly attenuated by the co-expression of one of the βancillary subunits of KCNQ family, KCNE4. Strong in vitro interactions between KCNQ4, 5 and KCNE4 were found through Foster Resonance Energy Transfer and co-immunoprecipitation. Although the expression levels of both KCNQ4 and KCNE4 are high in mesenteric arterial smooth muscle cells, we found that 1 μM of the agonist was sufficient to almost completely relax phenylephrine-induced contraction of the muscle strip. Significant expression of KCNQ4 and KCNE4 in corpus cavernosum together with high tonic contractility of the tissue grants highly promising relaxational effect of the KCNQspecific agonist in the tissue.

Keyword

KCNE4; KCNQ4; KCNQ5; Vasorelaxation

Figure

  • Fig. 1 Chemical nature, reference, purchase information of the agonist/antagonists used in the study. (1) URO-K10 was synthesized by Sundia MediTech Company (Sanghai, China) based on reaction scheme suggested by Seefeld et al. [24]. (2) ML-213 was purchased from Tocris Bioscience (Bristol, UK). (3) XE-991 was purchased from Sigma Aldrich (St. Louis, MO, USA).

  • Fig. 2 Electrophysiologic characteristics of overexpressed Kv7.4 and Kv7.5 channels in HEK293 cells. (A) Voltage clamp current traces of control (black) and Kv7.4 with URO-K10 (blue) are shown. (B) Corresponding I–V curve is plotted with steady-state currents measured from –100 mV to +100 mV at 10 mV step intervals. At +100 mV, Kv7.4-expressing cells showed 6.08 ± 1.60 nA whole-cell current in the presence of 1 μM URO-K10. (C) Boltzmann function-fitted conductance curves with URO-K10 show a leftward shift compared to the control (half-maximal voltage of –59.13 ± 6.33 mV). (D) Activation processes are slower with URO-K10 (blue), and (E) activation time constant values increased with the presence of the drug. (F) Voltage clamp traces of control (black) and Kv7.5 with URO-K10 (blue) are shown. (G) I–V curve of Kv7.5 expressing cells is shown. At +100 mV, Kv7.5-expressing cells showed 5.26 ± 2.19 nA whole-cell current in the presence of 1 μM URO-K10. (H) Boltzmann function-fitted steady-state conductance curve of Kv7.5 expressing cells is shown (half-maximal voltage of –74.93 ± 8.64 mV). (I, J) Activation time constant values of Kv7.5-expressing cells in 1 μM URO-K10 are plotted against voltage. In (H) and (J), both regression of conductance-voltage data to Boltzmann function and regression of current-time data to single exponential function were deliberately disregarded in Kv7.5-expressing cells without 1μM URO-K10 (control) (see manuscript for detailed description).

  • Fig. 3 Dose-dependent action of URO-K10 on Kv7.4 and Kv7.5. (A) Kv7.4 current traces are obtained at +50 mV with URO-K10 concentrations ranging from 1 pM to 10 μM. At each concentration, drug-affected current traces at +50 mV was subtracted from drug-free current traces at the same cell and at the same membrane potential (Idiff). (B) Dose-response curve in a logarithmic scale shows EC50 value of 210.8 nM for Kv7.4. (C, D) Dose-dependent action of URO-K10 on Kv7.5 is also shown in parallel. Dose-response curve in a logarithmic scale shows EC50 value of 142.0 nM for Kv7.5. In both (A) and (C), black inset lines indicate corresponding voltage-clamp step.

  • Fig. 4 URO-K10 increases both open probability and slope conductance of KCNQ4 channel. (A) Current traces from cell-attached single channel recordings at given commanding potentials. Traces in blue lines indicate pipette solution with URO-K10 (1 μM) while traces in black lines indicate control. At each commanding potential, dotted lines and full lines indicate closed states (C) and open states (O), respectively. (B) Open probabilities of KCNQ4 channel in 30 sec recording interval. In both traces, –160 mV of commanding potential was applied. URO-K10 (1 μM) significantly increased mean open probability of KCNQ4 channel (0.15 vs. 0.46). (C) Slope conductances calculated from unitary current amplitude at given commanding potential. 1 μM URO-K10 significantly increased slope conductance of KCNQ4 channel (4.77 pS vs. 18.85 pS).

  • Fig. 5 Kv7.4 and Kv7.5 have a molecular interaction with ancillary subunit KCNE4. (A) Fluorescence imaging of KCNQ4-ECFP, KCNQ5-ECFP and KCNE4-EYFP. Both KCNQ4-ECFP and KCNQ5-ECFP showed strong cyan fluorescence in plasma membrane and inside the cell. KCNE4-EYFP, however, showed discrete, puncta-like YFP signal inside the cell while signal in plasma membrane was highly limited. (B) Fluorescence imaging of KCNQ4-ECFP+KCNE4-EYFP and KCNQ5-ECFP+KCNE4-EYFP. When co-expressed with KCNQ4 or KCNQ5, prominent localization of YFP signal in plasma membrane was observable. High effective FRET efficiency (EEFF, see Methods) between KCNQ4-ECFP and KCNE4-EYFP (11.3% ± 1.5%, n = 7), and between KCNQ5-ECFP and KCNE4-EYFP (12.5% ± 1.6%, n = 7) were also observable, suggesting strong in vitro interaction between each α subunit and KCNE4. (C) Co-immunoprecipitation blot shows interaction with KCNE4 at a molecular level. KCNQ4-EGFP band (104 kDa) and KCNQ5-EGFP band (130 kDa) is observed when pulled down with the anti-Flag antibody. Conversely, KCNE4-Flag (23 kDa) is observed when pulled down with anti-GFP.

  • Fig. 6 Electrophysiology of KCNQ4/KCNE4 and KCNQ5/KCNE4 complex. (A) KCNE4 suppresses the activity of KCNQ4, as shown in the whole-cell potassium current traces. Whole-cell potassium current in KCNQ4/KCNE4 cells is 1.66 ± 0.72 nA at +100 mV. (B, C) I–V and conductance (G–V) curves are shown for control (KCNQ4/E4, black) and with 1 μM URO-K10 (blue). Half-maximal voltage in the presence of the drug is –67.35 ± 5.50 mV. (D–F) Electrophysiological characteristics of KCNQ5/KCNE4 are also shown in parallel. In 1 μM URO-K10, whole cell potassium current in KCNQ5/KCNE4 was 2.51 ± 1.29 nA at +100 mV and the half-maximal voltage was –72.73 ± 7.09 mV. (G) Maximum steady-state conductances of KCNQ4/E4 and KCNQ5/E4 are compared. KCNE4 suppresses both agonist-activated and intrinsic current of KCNQ4. KCNE4 suppresses the agonist-activated current of KCNQ5, but its effects on intrinsic currents are not shown due to the small current size indistinguishable from the background HEK293 cell current. (H) Half maximal voltages of KCNQ4/E4 and KCNQ5/E4 are compared. KCNE4 exerts a negligible effect on the voltage-dependency of KCNQ4 and KCNQ5 channels. **p < 0.05.

  • Fig. 7 Effect of URO-K10 onto mesenteric arterial vasorelaxation. (A) Isometric pressure myograph shows almost complete relaxation of phenylephrine-induced contraction upon administration of 1 μM URO-K10 (blue) (11.42% ± 2.06%, n = 4). (B) This relaxation effect is abolished in 80 mM potassium extracellular concentration (86.88% ± 3.05%, n = 6). (C) Bar graph summarizes percentages of contraction with and without 1 μM URO-K10 in 5 mM or 80 mM potassium. (D) Various concentrations of URO-K10 (from 0.1 to 1 μM) was applied to measure EC50 in a tissue-specific scale. (E) Percentages of contraction is plotted against concentration at a logarithmic scale. EC50 is estimated to be 521.0 nM. ***p < 0.001.


Reference

1. Hille B. 2001. Ion channels of excitable membranes. 3rd ed. Sinauer Associates;Sunderland (MA):
2. Barhanin J, Lesage F, Guillemare E, Fink M, Lazdunski M, Romey G. 1996; K(V)LQT1 and lsK (minK) proteins associate to form the I(Ks) cardiac potassium current. Nature. 384:78–80. DOI: 10.1038/384078a0. PMID: 8900282.
3. Biervert C, Schroeder BC, Kubisch C, Berkovic SF, Propping P, Jentsch TJ, Steinlein OK. 1998; A potassium channel mutation in neonatal human epilepsy. Science. 279:403–406. DOI: 10.1126/science.279.5349.403. PMID: 9430594.
Article
4. Charlier C, Singh NA, Ryan SG, Lewis TB, Reus BE, Leach RJ, Leppert M. 1998; A pore mutation in a novel KQT-like potassium channel gene in an idiopathic epilepsy family. Nat Genet. 18:53–55. DOI: 10.1038/ng0198-53. PMID: 9425900.
Article
5. Delmas P, Brown DA. 2005; Pathways modulating neural KCNQ/M (Kv7) potassium channels. Nat Rev Neurosci. 6:850–862. DOI: 10.1038/nrn1785. PMID: 16261179.
Article
6. Nelson MT, Quayle JM. 1995; Physiological roles and properties of potassium channels in arterial smooth muscle. Am J Physiol. 268(4 Pt 1):C799–C822. DOI: 10.1152/ajpcell.1995.268.4.C799. PMID: 7733230.
Article
7. Jepps TA, Olesen SP, Greenwood IA. 2013; One man's side effect is another man's therapeutic opportunity: targeting Kv7 channels in smooth muscle disorders. Br J Pharmacol. 168:19–27. DOI: 10.1111/j.1476-5381.2012.02133.x. PMID: 22880633. PMCID: PMC3569999.
Article
8. Stott JB, Jepps TA, Greenwood IA. 2014; K(V)7 potassium channels: a new therapeutic target in smooth muscle disorders. Drug Discov Today. 19:413–424. DOI: 10.1016/j.drudis.2013.12.003. PMID: 24333708.
Article
9. Haick JM, Byron KL. 2016; Novel treatment strategies for smooth muscle disorders: targeting Kv7 potassium channels. Pharmacol Ther. 165:14–25. DOI: 10.1016/j.pharmthera.2016.05.002. PMID: 27179745.
Article
10. Petkov GV. 2011; Role of potassium ion channels in detrusor smooth muscle function and dysfunction. Nat Rev Urol. 9:30–40. DOI: 10.1038/nrurol.2011.194. PMID: 22158596. PMCID: PMC3759241.
Article
11. Cellek S, Cameron NE, Cotter MA, Fry CH, Ilo D. 2014; Microvascular dysfunction and efficacy of PDE5 inhibitors in BPH-LUTS. Nat Rev Urol. 11:231–241. DOI: 10.1038/nrurol.2014.53. PMID: 24619381.
Article
12. Yafi FA, Jenkins L, Albersen M, Corona G, Isidori AM, Goldfarb S, Maggi M, Nelson CJ, Parish S, Salonia A, Tan R, Mulhall JP, Hellstrom WJ. 2016; Erectile dysfunction. Nat Rev Dis Primers. 2:16003. DOI: 10.1038/nrdp.2016.3. PMID: 27188339. PMCID: PMC5027992.
Article
13. Mónica FZ, Antunes E. 2018; Stimulators and activators of soluble guanylate cyclase for urogenital disorders. Nat Rev Urol. 15:42–54. DOI: 10.1038/nrurol.2017.181. PMID: 29133940.
Article
14. Lee JH, Chae MR, Kang SJ, Sung HH, Han DH, So I, Park JK, Lee SW. 2020; Characterization and functional roles of KCNQ-encoded voltage-gated potassium (Kv7) channels in human corpus cavernosum smooth muscle. Pflugers Arch. 472:89–102. DOI: 10.1007/s00424-019-02343-7. PMID: 31919767.
Article
15. Abbott GW, Sesti F, Splawski I, Buck ME, Lehmann MH, Timothy KW, Keating MT, Goldstein SA. 1999; MiRP1 forms IKr potassium channels with HERG and is associated with cardiac arrhythmia. Cell. 97:175–187. DOI: 10.1016/S0092-8674(00)80728-X. PMID: 10219239.
Article
16. McCrossan ZA, Abbott GW. 2004; The MinK-related peptides. Neuropharmacology. 47:787–821. DOI: 10.1016/j.neuropharm.2004.06.018. PMID: 15527815.
Article
17. Jepps TA, Carr G, Lundegaard PR, Olesen SP, Greenwood IA. 2015; Fundamental role for the KCNE4 ancillary subunit in Kv7.4 regulation of arterial tone. J Physiol. 593:5325–5340. DOI: 10.1113/JP271286. PMID: 26503181. PMCID: PMC4704525.
Article
18. Abbott GW, Jepps TA. 2016; Kcne4 deletion sex-dependently alters vascular reactivity. J Vasc Res. 53:138–148. DOI: 10.1159/000449060. PMID: 27710966. PMCID: PMC5166573.
19. Yeung SY, Pucovský V, Moffatt JD, Saldanha L, Schwake M, Ohya S, Greenwood IA. 2007; Molecular expression and pharmacological identification of a role for K(v)7 channels in murine vascular reactivity. Br J Pharmacol. 151:758–770. DOI: 10.1038/sj.bjp.0707284. PMID: 17519950.
Article
20. Panaghie G, Tai KK, Abbott GW. 2006; Interaction of KCNE subunits with the KCNQ1 K+ channel pore. J Physiol. 570(Pt 3):455–467. DOI: 10.1113/jphysiol.2005.100644. PMID: 16308347.
21. Strutz-Seebohm N, Seebohm G, Fedorenko O, Baltaev R, Engel J, Knirsch M, Lang F. 2006; Functional coassembly of KCNQ4 with KCNE-beta- subunits in Xenopus oocytes. Cell Physiol Biochem. 18:57–66. DOI: 10.1159/000095158. PMID: 16914890.
22. Roura-Ferrer M, Etxebarria A, Solé L, Oliveras A, Comes N, Villarroel A, Felipe A. 2009; Functional implications of KCNE subunit expression for the Kv7.5 (KCNQ5) channel. Cell Physiol Biochem. 24:325–334. DOI: 10.1159/000257425. PMID: 19910673.
Article
23. Sun J, MacKinnon R. 2020; Structural basis of human KCNQ1 modulation and gating. Cell. 180:340–347.e9. DOI: 10.1016/j.cell.2019.12.003. PMID: 31883792. PMCID: PMC7083075.
Article
24. Seefeld MA, Lin H, Holenz J, Downie D, Donovan B, Fu T, Pasikanti K, Zhen W, Cato M, Chaudhary KW, Brady P, Bakshi T, Morrow D, Rajagopal S, Samanta SK, Madhyastha N, Kuppusamy BM, Dougherty RW, Bhamidipati R, Mohd Z, et al. 2018; Novel KV7 ion channel openers for the treatment of epilepsy and implications for detrusor tissue contraction. Bioorg Med Chem Lett. 28:3793–3797. DOI: 10.1016/j.bmcl.2018.09.036. PMID: 30327146.
Article
25. Ng FL, Davis AJ, Jepps TA, Harhun MI, Yeung SY, Wan A, Reddy M, Melville D, Nardi A, Khong TK, Greenwood IA. 2011; Expression and function of the K+ channel KCNQ genes in human arteries. Br J Pharmacol. 162:42–53. DOI: 10.1111/j.1476-5381.2010.01027.x. PMID: 20840535.
Article
26. Mackie AR, Brueggemann LI, Henderson KK, Shiels AJ, Cribbs LL, Scrogin KE, Byron KL. 2008; Vascular KCNQ potassium channels as novel targets for the control of mesenteric artery constriction by vasopressin, based on studies in single cells, pressurized arteries, and in vivo measurements of mesenteric vascular resistance. J Pharmacol Exp Ther. 325:475–483. DOI: 10.1124/jpet.107.135764. PMID: 18272810. PMCID: PMC2597077.
Article
27. Hodgkin AL, Huxley AF. 1952; A quantitative description of membrane current and its application to conduction and excitation in nerve. J Physiol. 117:500–544. DOI: 10.1113/jphysiol.1952.sp004764. PMID: 12991237. PMCID: PMC1392413.
Article
28. Myeong J, Kwak M, Hong C, Jeon JH, So I. 2014; Identification of a membrane-targeting domain of the transient receptor potential canonical (TRPC)4 channel unrelated to its formation of a tetrameric structure. J Biol Chem. 289:34990–35002. DOI: 10.1074/jbc.M114.584649. PMID: 25349210. PMCID: PMC4263895.
Article
29. Erickson MG, Alseikhan BA, Peterson BZ, Yue DT. 2001; Preassociation of calmodulin with voltage-gated Ca(2+) channels revealed by FRET in single living cells. Neuron. 31:973–985. DOI: 10.1016/S0896-6273(01)00438-X. PMID: 11580897.
Article
30. Epe B, Steinhäuser KG, Woolley P. 1983; Theory of measurement of Förster-type energy transfer in macromolecules. Proc Natl Acad Sci U S A. 80:2579–2583. DOI: 10.1073/pnas.80.9.2579. PMID: 16593305. PMCID: PMC393869.
31. Patterson G, Day RN, Piston D. 2001; Fluorescent protein spectra. J Cell Sci. 114(Pt 5):837–838. PMID: 11181166.
Article
32. Hoshi T, Zagotta WN, Aldrich RW. 1991; Two types of inactivation in Shaker K+ channels: effects of alterations in the carboxy-terminal region. Neuron. 7:547–556. DOI: 10.1016/0896-6273(91)90367-9. PMID: 1931050.
Article
33. Brueggemann LI, Haick JM, Cribbs LL, Byron KL. 2014; Differential activation of vascular smooth muscle Kv7.4, Kv7.5, and Kv7.4/7.5 channels by ML213 and ICA-069673. Mol Pharmacol. 86:330–341. DOI: 10.1124/mol.114.093799. PMID: 24944189. PMCID: PMC4152906.
Article
34. Sun J, MacKinnon R. 2017; Cryo-EM structure of a KCNQ1/CaM complex reveals insights into congenital long QT syndrome. Cell. 169:1042–1050.e9. DOI: 10.1016/j.cell.2017.05.019. PMID: 28575668. PMCID: PMC5562354.
Article
35. Rodriguez-Menchaca AA, Adney SK, Tang QY, Meng XY, Rosenhouse-Dantsker A, Cui M, Logothetis DE. 2012; PIP2 controls voltage-sensor movement and pore opening of Kv channels through the S4-S5 linker. Proc Natl Acad Sci U S A. 109:E2399–E2408. DOI: 10.1073/pnas.1207901109. PMID: 22891352. PMCID: PMC3437867.
Article
36. Zaydman MA, Cui J. 2014; PIP2 regulation of KCNQ channels: biophysical and molecular mechanisms for lipid modulation of voltage-dependent gating. Front Physiol. 5:195. DOI: 10.3389/fphys.2014.00195. PMID: 24904429. PMCID: PMC4034418.
Article
37. Suh BC, Inoue T, Meyer T, Hille B. 2006; Rapid chemically induced changes of PtdIns(4,5)P2 gate KCNQ ion channels. Science. 314:1454–1457. DOI: 10.1126/science.1131163. PMID: 16990515. PMCID: PMC3579521.
Article
38. Li Y, Gamper N, Hilgemann DW, Shapiro MS. 2005; Regulation of Kv7 (KCNQ) K+ channel open probability by phosphatidylinositol 4,5-bisphosphate. J Neurosci. 25:9825–9835. DOI: 10.1523/JNEUROSCI.2597-05.2005. PMID: 16251430. PMCID: PMC6725574.
Article
39. Lerche C, Scherer CR, Seebohm G, Derst C, Wei AD, Busch AE, Steinmeyer K. 2000; Molecular cloning and functional expression of KCNQ5, a potassium channel subunit that may contribute to neuronal M-current diversity. J Biol Chem. 275:22395–22400. DOI: 10.1074/jbc.M002378200. PMID: 10787416.
Article
40. Li Y, Gamper N, Shapiro MS. 2004; Single-channel analysis of KCNQ K+ channels reveals the mechanism of augmentation by a cysteine-modifying reagent. J Neurosci. 24:5079–5090. DOI: 10.1523/JNEUROSCI.0882-04.2004. PMID: 15175377. PMCID: PMC6729199.
Article
Full Text Links
  • KJPP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr