<<

View metadata, citation and similar papers at core.ac.uk brought to you by CORE

provided by Elsevier - Publisher Connector

Biochimica et Biophysica Acta 1757 (2006) 715–720 http://www.elsevier.com/locate/bba

Mitochondrial potassium channels: From to function ⁎ Adam Szewczyk a, , Jolanta Skalska a, Marta Głąb a, Bogusz Kulawiak a, Dominika Malińska a, Izabela Koszela-Piotrowska a, Wolfram S. Kunz b

a Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, 3 Pasteur st., 02-093 Warsaw, Poland b Department of Epileptology, University Bonn Medical Center, Sigmund-Freud-Str. 25, D-53105 Bonn, Germany Received 15 December 2005; received in revised form 19 April 2006; accepted 2 May 2006 Available online 12 May 2006

Abstract

Mitochondrial potassium channels, such as ATP-regulated or large conductance Ca2+-activated and voltage gated channels were implicated in cytoprotective phenomenon in different tissues. Basic effects of these channels activity include changes in mitochondrial matrix volume, mitochondrial respiration and membrane potential, and generation of reactive oxygen species. In this paper, we describe the pharmacological properties of mitochondrial potassium channels and their modulation by channel inhibitors and openers. We also discuss potential side effects of these substances. © 2006 Elsevier B.V. All rights reserved.

Keywords: Mitochondria; Potassium channels; Potassium channel openers; Sulfonylurea;

1. Introduction drial metabolism primarily due to the regulation of matrix volume [7]. The basic pharmacological properties of mitochondrial Mitochondria play a fundamental role in energy generation potassium channels such as ATP-regulated potassium (mitoKATP) 2+ within the cell. Apart from this canonical role, mitochondria are channel, large conductance Ca -activated potassium (mitoBKCa) involved in other complex processes such as apoptosis. Mitochon- channel and voltage dependent potassium (mitoKv1.3) channel drial membrane permeabilization induces the release of cytochrome were found to be similar to some of the potassium channels present c, Smac/DIABLO, and AIF, starting various apoptosis pathways. in the plasma membrane of various cell types [8]. Recently, an essential position of potassium transport through mi- Rational application of different pharmacological substances tochondrial inner membrane was identified to trigger cytoprotection (channel inhibitors or potassium channel openers) helps in ex- [1–3]. This transport is strictly dependent, and accor- plaining the functional role of mitochondrial potassium channels dingly, resembles plasma membrane ion channels activity. Po- within the cell. In this paper, we describe the properties of mito- tassium selective ion channels were found to be present in inner chondrial potassium channels, their modulation by channel in- mitochondrial membrane [4–6]. Potassium ions control mitochon- hibitors and activators (potassium channel openers). Finally, we also discuss side effects of these substances and their potential contribution to cytoprotective effects.

Abbreviations: ANT, mitochondrial adenine nucleotide translocase; BKCa 2. Mitochondrial ATP-regulated K+ channel channel, large conductance Ca2+-activated potassium channel; BLM, black lipid

membrane technique; ChTx, ; IbTx, iberiotoxin; KATP channel, ATP-regulated potassium channel; mitoBKCa channel, mitochondrial large The mitoKATP channel was initially described in isolated liver conductance calcium-activated potassium channel; mitoKATP channel, mito- mitochondria [9]. Subsequently, it was also identified in heart [10], chondrial ATP-regulated potassium channel; mitoKv1.3 channel, mitochondrial brain [11,12], renal [13], skeletal muscle [14] and human T-lym- voltage gated potassium channel; NS1619, 1,3-dihydro-1-[2-hydroxy-5-(tri- phocyte mitochondria [15]. The activity of this channel is modu- fluoromethyl)phenyl]-5-(trifluoromethyl)-2H-benzimidazole-2-one; SMP, sub- mitochondrial particles lated by various nucleotides [10] (see Fig. 1). Recently, it was ⁎ Corresponding author. Tel.: +48 22 5892269; fax: +48 22 8225342. shown that ATP effects on the mitoKATP channel can be modulated 2+ E-mail address: [email protected] (A. Szewczyk). by Ca [15].

0005-2728/$ - see front matter © 2006 Elsevier B.V. All rights reserved. doi:10.1016/j.bbabio.2006.05.002 716 A. Szewczyk et al. / Biochimica et Biophysica Acta 1757 (2006) 715–720

acid has some mitochondrial specificity as mitoKATP . An important contribution to understanding the pharmacolog- ical properties of the mitoKATP channel comes from the use of the techniques allowing single channel recording i.e. patch clamp or black lipid membranes [9,15,23–25]. The observed K+ channel activity is in fact a mitoKATP channel if the following phar- macological properties are present in the recorded potassium channel:

(1) The channel has to be blocked by ATP/Mg [9,10,23,26], (2) The ATP/Mg-inhibited channel should be activated by the potassium diazoxide [26,27], (3) The channel should be blocked by 5-hydroxydecanoic acid (5-HD), a substance known to be a blocker of the mito- chondrial channel [23,24,26], (4) The channel should be blocked by [9,10,26], (5) The plasma membrane cardiac KATP channel blocker HMR1098 should be without effect on the channel activity [23,28],

Some of the substances acting on mitoKATP show additional effect on isolated mitochondria. Diazoxide is known to inhibit mitochondrial succinate dehydrogenase [29] and induce uncou- pling of mitochondria [30,31]. Similar effects were observed with [30]. Glibenclamide is also able to uncouple mitochondria at a high concentration [32] or to Fig. 1. Physiological regulators of the mitochondrial potassium channels: ATP- induce mitochondrial permeability transition [33].Recently,it 2+ regulated potassium (mitoKATP) channel, large conductance Ca -activated was shown that 5-HD is rapidly converted to 5-HD-CoA by potassium (mitoBKCa) channel and voltage dependent potassium (mitoKv1.3) mitochondrial fatty acyl-CoA synthetase and acts as a weak channel. ROS, reactive oxygen species; NO, nitric oxide. substrate or inhibitor of respiration in the employed conditions [34,35]. Hence, in some reports the existence of the mitoKATP channel in inner mitochondrial membrane is questioned [19,35– The molecular identity of the mitoKATP channel is presently 37]. It is proposed that the pharmacological preconditioning by unknown. Immunological studies identify both pore forming Kir mitoKATP channel modulators may be related to partial inhibition 6.1 and Kir6.2 subunits in brain mitochondria [16]. It has also been of respiratory chain complexes [35,38]. hypothesized that a complex of five proteins (including succinate These side effects may contribute to the overall effects of dehydrogenase) in the mitochondrial inner membrane is capable of substances used in experimental models with active metabolism, transporting K+ with characteristics similar to those of the mito- for example in experiments on isolated mitochondria or cells (see KATP channel [17]. Recently, adenine nucleotide translocase was Table 1). For example, application of 5-HD, as the KATP channel implicated in ATP-sensitive K+ transport into mitochondria [18– blocker, may be questioned in experimental models with active 20]. Interestingly, the involvement of adenine nucleotide translo- acyl-CoA synthase [35]. Additionally, the channel opener – dia- + case in K traffic was also suggested before description of the KATP zoxide – has a complex action on mitochondrial membrane po- channel activity in inner mitochondrial membrane [20]. tential [39], ROS production [40] and mitochondrial permeability Similarly, molecular properties of the mitochondrial sulfonyl- transition pore [41]. urea (mitoSUR) are also not clear. The use of the sulfo- nylurea derivative [125I]glibenclamide leads to labeling of a 28 3. Mitochondrial large conductance calcium-activated kDa protein in heart mitochondria [21]. A 64 kDa protein was potassium channel labeled in brain mitochondria with the use of the fluorescent probe BODIPY-glibenclamide [11]. A putative mitochondrial large conductance Ca2+-activated Activity of this channel is modulated by known as potas- potassium channel (mitoBKCa channel) was originally described sium channel openers [22]. All of the potassium channel openers using patch-clamp technique in human glioma cells LN229 [42]. applied on mitochondria were previously used to regulate plasma This channel, with a conductance of 295 pS, was stimulated by membrane ATP-regulated potassium channel or large conductance Ca2+ and blocked by charybdotoxin. Later the presence of a calcium-activated potassium channel. Despite this, potassium channel with properties similar to the surface membrane calcium- channel openers such as diazoxide and BMS191095, are consi- activated K+ channel (stimulated by the potassium channel opener dered mitoKATP-selective openers. Similarly, 5-hydroxydecanoic NS1619 and blocked by charybdotoxin, iberiotoxin and paxilline) A. Szewczyk et al. / Biochimica et Biophysica Acta 1757 (2006) 715–720 717

Table 1

Pharmacological regulators of mitochondrial potassium channels: mitochondrial ATP-regulated potassium (mitoKATP) channel, large conductance calcium-activated potassium (mitoBKCa) channel and voltage gated potassium (mitoKv1.3) channel. BLM—black lipid membranes technique Postulated Compound Experimental Applied Comments [References] channel target [inhibitor/activator] setup concentration or dose mitoKATP Glibenclamide Patch-clamp/ 10–100 μM Studies performed on liver [9] and heart mitochondria [23,25] [inhibitor] BLM Proteoliposomes 0.3–1 μMK+ flux with reconstituted partially purified channel [27] flux Isolated 0.3–50 μM Studies performed on rat liver mitochondria [27] mitochondria and skeletal muscle mitochondria [14] Tissue 50 μM [52] 5-hydroxydecanoic acid Patch-clamp/ 10 μM–1mM [15,23,24,53] [inhibitor] BLM Proteoliposomes 200 μM [54] flux Isolated 40 μ M–1 mM Studies performed on mitochondria isolated from heart [55,56], mitochondria skeletal muscle [14] liver [26], kidney [13] and brain [46] Cells/tissue 10 μM–1 mM Studies performed on cardiomiocytes [57–59], vascular smooth muscle cells [60], endothelial cells [61] and neuronal cultures [62]. quinine [inhibitor] BLM 3–100 μM Additionally 86Rb+ flux and [3H]glibenclamode binding experiments were performed [53] N-ethylmaleimide BLM 2 mM Reconstitution of inner mitochondrial membrane from beef [23] [inhibitor] Diazoxide [activator] Proteoliposomes 0.3–3 μMK+ flux with reconstituted partially purified channel [27] flux Isolated 10–100 μM [14] mitochondria BLM 10–50 μM Reconstitution of inner mitochondrial membrane from beef [23], [53] and rat heart mitochondria [24] were performed Cells/tissue 3.5 mg/kg [63] [activator] Proteoliposomes 0.3–10 μMK+ flux with reconstituted partially purified channel, flux two developmental cromakalim analogues EMD60480 and EMD57970 were also applied [27] Pinacidil [activator] Isolated 50 μM [30] mitochondria BMS191095 [activator] Isolated 10–30 μM Studies performed on heart and brain mitochondria [46,64,73] mitochondria Cells 10–40 μM Studies were performed with primary rat cortical neuronal cultures [65] Tissue 0.4–10 mg/kg [66–69] 6 μM [70,71], RP66471 [activator] Isolated 100 μM Studies performed with liver mitochondria [72] mitochondria [activator] Tissue 1.75 mg/kg [63] Isolated 10–100 μM Studies performed with rat skeletal muscle mitochondria [14] mitochondria P1075 [activator] Tissue 100 μM [52] Proteoliposomes 0.1–1 μM [52] flux Isoflurane [activator] BLM 0.8 mM Reconstitution of inner membrane from rat heart mitochondria [24] were performed mitoBKCa Charybdotoxin (ChTx) Patch-clamp 1–200 nM Mitoplasts from glioma cell line and cardiac tissue were used [42,43] [inhibitor] Mitochondrial 100 nM The surface of cardiac myocytes was permeabilized flux with the use of saponin. ChTx slowed K+ uptake (measured with fluorescent probe PBFI) by approximately 20-fold [43] Iberiotoxin (IbTx) Mitochondrial 100 nM The surface of cardiac myocytes was permeabilized [inhibitor] flux with the use of saponin [43] Paxilline [inhibitor] 2 μM Paxilline was also used to block the protective effect of NS1619 in the experiments of the ischemic damage experiments [43] NS1619 [activator] Mitochondrial 30 μM [43] flux (continued on next page) 718 A. Szewczyk et al. / Biochimica et Biophysica Acta 1757 (2006) 715–720

Table 1 (continued) Postulated Compound Experimental Applied Comments [References] channel target [inhibitor/activator] setup concentration or dose mitoBKCa Cells 30 μM [44] mitoKv1.3 Margatoxin (MgTx) Patch-clamp 2 nM Studies were performed on T lymphocytes mitochondria [51] [inhibitor] Isolated 10 nM A relatively high concentration of toxin was used to mitochondria promote a rapid diffusion to the inner membrane and to achieve complete inhibition [51]

was observed in patch-clamp recordings from mitoplasts of mitoBKCa channel in cardiac mitochondria [48].Additionally,it guinea pig ventricular cells [43]. Immunoblots of cardiac mito- was suggested that the β1 subunit of BKCa channel is present in chondria with antibodies against the C terminal part of BKCa inner mitochondrial membrane and interacts with cytochrome c channel identified a 55 kDa protein as putative channel which oxidase subunit I [48]. + may contribute to the cardioprotective effect of K influx into It has also been reported that two BKCa channel openers, NS004 mitochondria. Very recently, it was reported that cardiac mito and NS1619, inhibit mitochondrial function in human glioma cells BKCa activation by NS1619 (measured by flavoproteins oxida- due to inhibition of the complex I of mitochondrial respiratory tion) is amplified by 8-bromoadenosine-3′,5′-cyclic monophos- chain [49]. Recently, a channel opener NS1619 was shown to phate and forskolin [44]. This suggests that opening of mitoBKCa inhibit the function of isolated cardiac mitochondria in similar is modulated by cAMP-dependent protein kinase. manner [50]. It should be mentioned that the mitoBKCa channel may offer a novel link between cellular/mitochondrial calcium signaling and 4. Voltage regulated potassium channel in inner mitochondrial mitochondrial membrane potential-dependent reactions. Altered membrane intramitochondrial calcium levels directly affect the potassium permeability of the mitochondrial inner membrane thus modula- Recently, a margatoxin-sensitive voltage gated Kv1.3 channel ting the membrane potential. This type of interaction can mo- was identified in T lymphocytes mitochondria [51] with the use of dulate the efficiency of oxidative phosphorylation in a calcium- patch-clamp, electron microscopy and immunological studies. dependent manner. Interestingly, it was shown that Kv1.3 is present both in the plas- The pharmacological, biophysical and molecular character- ma and mitochondrial membranes, despite lacking the N-terminal istics of this channel appear to be similar to the following pro- mitochondrial targeting sequence. Mitochondrial Kv1.3 channel perties of the well known plasma membrane large conductance my represent an important factor in the apoptotic signal trans- potassium channel (BKCa-channel, slo 1) [45]: duction [51].

(i) The mitochondrial channel is potassium selective and ac- 5. Final remarks tivated by calcium ions. (ii) The channel is inhibited by iberiotoxin and charybdotoxin Until presently, three different potassium channels have been at nanomolar concentrations. reported to contribute to the potassium permeability of mito- (iii) The channel is activated by NS1619 at low micromolar chondria—the ATP-regulated potassium channel (mitoKATP concentrations. channel), the large conductance Ca2+-activated potassium channel (mitoBKCa channel) and the voltage gated Kv1.3 chan- Very likely, the mitochondrial channel has its charybdotoxin/ nel. Although the precise molecular identity of these channels iberiotoxin binding site close to the cytosolic compartment since remains to be elucidated, their control of mitochondrial potassium those compounds as peptides cannot easily enter the mitochon- transport has two important implications: drial matrix space. Consequently, the calcium binding site of the Small alterations in the potassium permeability contribute to mitoBKCa channel is close to the matrix compartment (cf. orien- physiological regulation of the mitochondrial matrix volume. tation of the plasma membrane BKCa channel [45]). A larger increase in the potassium permeability of the mito- Similar to the mitochondrial ATP-regulated potassium chan- chondrial inner membrane affects the mitochondrial membrane nel, this novel potassium channel is expected to affect mito- potential (mild uncoupling) with potential beneficial effects on chondrial metabolism due to regulation of matrix volume [7].In cell survival under pathological conditions. addition to this classical physiological effect of mitochondrial In this review, various classes of substances that interact with potassium transport, a pivotal role of mitochondrial potassium mitochondrial potassium channels have been presented. They play channels has been implicated in cardio- and neuroprotection an important role in identification and characterization of mito- [1,2,46]. It is therefore reasonable to expect a possible cyto- chondrial channels in different tissues. In light of the ubiquitous protective effect of mitoBKCa channel activation, probably in the presence of mitochondrial identified potassium channels, novel presence of superoxide radicals [47]. Recently, it was shown that opportunities for novel development have been emerged. cardioprotective effects of estradiol include the activation of To reach this stage, however, the molecular identification of A. Szewczyk et al. / Biochimica et Biophysica Acta 1757 (2006) 715–720 719

[19] T. Brustovetsky, N. Shalbuyeva, N. Brustovetsky, Lack of manifestations mitochondrial potassium channels (especially mitoKATP channel) requires significant progress. Moreover, drugs specific to the new of diazoxide/5-hydroxydecanoate-sensitive KATP channel in rat brain nonsynaptosomal mitochondria, J. Physiol. 568 (2005) 47–59. mitochondrial channels (lacking cross-reactivity with plasma [20] A. Panov, S. Filippova, V. Lyakhovich, Adenine nucleotide translocase as membrane channels or other present in mitochondria) site of regulation by ADP of the rat liver mitochondria permeability to H+ will have to be synthesized. and K+ ions, Arch. Biochem. Biophys. 199 (1980) 420–426. [21] A. Szewczyk, Intracellular targets for antidiabetic sulfonylureas and – Acknowledgments potassium channel openers, Biochem. Pharmacol. 54 (1997) 961 965. [22] G. Edwards, A.H. Weston, Structure–activity relationships of K+ channel openers, Trends Pharmacol. Sci. 11 (1990) 417–422. This study was supported by the Ministry of Education and [23] D.X. Zhang, Y.F. Chen, W.B. Campbell, A.P. Zou, G.J. Gross, P.L. Li, Science grants No. 3P04A05025 and PBZ-MIN-001/P05/11 (to Characteristics and superoxide-induced activation of reconstituted myo- AS) and the BMBF grant No. 01GZ0308 (to WSK). cardial mitochondrial ATP-sensitive potassium channels, Circ. Res. 89 (2001) 1177–1183. [24] Y. Nakae, W.M. Kwok, Z.J. Bosnjak, M.T. Jiang, Isoflurane activates rat References mitochondrial ATP-sensitive K+ channels reconstituted in lipid bilayers, Am. J. Physiol. 284 (2003) H1865–H1871. [1] B. O'Rourke, Evidence for mitochondrial K+ channels and their role in [25] P. Bednarczyk, K. Dolowy, A. Szewczyk, Matrix Mg2+ regulates cardioprotection, Circ. Res. 94 (2004) 420–432. mitochondrial ATP-dependent potassium channel from heart, FEBS Lett. [2] M.P. Mattson, D. Liu, Mitochondrial potassium channels and uncoupling 579 (2005) 1625–1632. proteins in synaptic plasticity and neuronal cell death, Biochem. Biophys. [26] M. Jaburek, V. Yarov-Yarovoy, P. Paucek, K.D. Garlid, State-dependent Res. Commun. 304 (2003) 539–549. inhibition of the mitochondrial KATP channel by glyburide and 5-hydro- [3] D.W. Busija, Z. Lacza, N. Rajapakse, K. Shimiz, B. Kis, F. Bari, F. Domoki, T. xydecanoate, J. Biol. Chem. 273 (1998) 13578–13582. Horiguchi, Targeting mitochondrial ATP-sensitive potassium channels—A [27] K.D. Garlid, P. Paucek, V. Yarov-Yarovoy, X. Sun, P.A. Schindler, The novel approach to neuroprotection, Brain Res. Brain Res. Rev. 46 (2004) mitochondrial KATP channel as a receptor for potassium channel openers, 282–294. J. Biol. Chem. 271 (1996) 8796–8799. [4] A. Szewczyk, The intracellular potassium and chloride channels: properties, [28] T. Sato, N. Sasaki, J. Seharaseyon, B. O'Rourke, E. Marban, Selective pharmacology and function, Mol. Membr. Biol. 15 (1998) 49–58. pharmacological agents implicate mitochondrial but not sarcolemmal [5] P. Bernardi, Mitochondrial transport of cations: channels, exchangers, and KATP channels in ischemic cardioprotection, Circulation 101 (2000) permeability transition,, Physiol. Rev. 79 (1999) 1127–1155. 2418–2423. [6] A. Szewczyk, L. Wojtczak, Mitochondria as a pharmacological target, [29] T. Grimmsmann, I. Rustenbeck, Direct effects of diazoxide on mitochon- Pharmacol. Rev. 54 (2002) 101–127. dria in pancreatic B-cells and on isolated liver mitochondria, Br. J. [7] A.P. Halestrap, Regulation of mitochondrial metabolism through changes Pharmacol. 123 (1998) 781–788. in matrix volume, Biochem. Soc. Trans. 22 (1994) 522–529. [30] A.J. Kowaltowski, S. Seetharaman, P. Paucek, K.D. Garlid, Bioenergetic [8] A. Szewczyk, E. Marban, Mitochondria: a new target for potassium channel consequences of opening the ATP-sensitive K+ channel of heart openers? Trends Pharmacol. Sci. 20 (1999) 157–161. mitochondria, Am. J. Physiol. 280 (2001) 649–657. [9] I. Inoue, H. Nagase, K. Kishi, T. Higuti, ATP-sensitive K+ channel in the [31] E.L. Holmuhamedov, A. Jahangir, A. Oberlin, A. Komarov, M. Colombini, mitochondrial inner membrane, Nature 352 (1991) 244–247. A. Terzic, Potassium channel openers are uncoupling protonophores: [10] P.Paucek, G. Mironova, F. Mahdi, A.D. Beavis, G. Woldegiorgis, K.D. Garlid, implication in cardioprotection, FEBS Lett. 568 (2004) 167–170. Reconstitution and partial purification of the glibenclamide-sensitive, ATP- [32] A. Szewczyk, A. Czyz, M.J. Nalecz, ATP-regulated potassium channel dependent K+ channel from rat liver and beef heart mitochondria, J. Biol. blocker, glibenclamide, uncouples mitochondria, Pol. J. Pharmacol. 49 Chem. 267 (1992) 26062–26069. (1997) 49–52. [11] R. Bajgar, S. Seetharaman, A.J. Kowaltowski, K.D. Garlid, P. Paucek, [33] J. Skalska, G. Debska, W.S. Kunz, A. Szewczyk, Antidiabetic sulfonylur- Identification and properties of a novel intracellular (mitochondrial) ATP- eas activate mitochondria permeability transition in rat skeletal muscle, Br. sensitive potassium channel in brain, J. Biol. Chem. 276 (2001) 33369–33374. J. Pharmacol. (2005) 1–7. [12] G. Debska, R. May, A. Kicinska, A. Szewczyk, C.E. Elger, W.S. Kunz, [34] K.H. Lim, S.A. Javadov, M. Das, S.J. Clarke, M.S. Suleiman, A.P. Halestrap, Potassium channel openers depolarize hippocampal mitochondria, Brain The effects of ischaemic preconditioning, diazoxide and 5-hydroxydecanoate Res. 892 (2001) 42–50. on rat heart mitochondrial volume and respiration, J. Physiol. 545 (2002) [13] D.V. Cancherini, L.G. Trabuco, N.A. Reboucas, A.J. Kowaltowski, ATP- 961–974. sensitive K+ channels in renal mitochondria, Am. J. Physiol., Renal Fluid [35] P.J. Hanley, M. Mickel, M. Lõffler, U. Brandt, J. Daut, KATP channel- Electrolyte Physiol. 285 (2003) F1291–F1296. independent targets of diazoxide and 5-hydroxydecanoate in the heart, [14] G. Debska, A. Kicinska, J. Skalska, A. Szewczyk, R. May, C.E. Elger, W.S. J. Physiol. 542.3 (2002) 735–741. Kunz, Opening of potassium channels modulates mitochondrial function in [36] M. Das, J.E. Parker, A.P.Halestrap, Matrix volume measurements challenge the

rat skeletal muscle, Biochim. Biophys. Acta 1556 (2002) 97–105. existence of diazoxide/glibenclamide-sensitive KATP channels in rat mitochon- [15] Y.A. Dahlem, T.F.W. Horn, L. Buntinas, T. Gonoi, G. Wolf, D. Siemen, The dria, J. Physiol. 547.3 (2003) 893–902. human mitochondrial KATP channel is modulated by calcium and nitric [37] S. Ovide-Bordeaux, R. Ventura-Clapier, V. Veksler, Do modulators of the oxide: a patch-clamp approach, Biochim. Biophys. Acta 1656 (2004) 46–56. mitochondrial KATP channel change the function of mitochondria in situ? [16] Z. Lacza, J.A. Snipes, B. Kis, C. Szabo, G. Grover, D.W.Busija, Investigation J. Biol. Chem. 275 (2000) 37291–37295. of the subunit composition and the pharmacology of the mitochondrial ATP- [38] B. Kis, N.C. Rajapakse, J.A. Snipes, K. Nagy, T. Horiguchi, D.W. Busija, dependent K+ channel in the brain, Brain Res. 994 (2003) 27–36. Diazoxide induces delayed pre-conditioning in cultured rat cortical [17] H. Ardehali, Z. Chen, Y. Ko, R. Mejia-Alvarez, E. Marban, Multiprotein neurons, J. Neurochem. 87 (2003) 969–980.

complex containing succinate dehydrogenase confers mitochondrial ATP- [39] C.L. Lawrence, B. Billups, G.C. Rodrigo, N.B. Standen, The KATP channel sensitive K+ channel activity, Proc. Natl. Acad. Sci. U. S. A. 101 (2004) opener diazoxide protects cardiac myocytes during metabolic inhibition 11880–11885. without causing mitochondrial depolarization of flavoprotein oxidation, [18] D.M. Kopustinskiene, A. Toleikis, N.E. Saris, Adenine nucleotide translo- Br. J. Pharmacol. 134 (2001) 535–542. case mediates the K(ATP)-channel-openers-induced proton and potassium [40] H. Katoh, N. Nishigaki, H. Hayashi, Diazoxide opens the mitochondria flux to the mitochondrial matrix, J. Bioenerg. Biomembranes 35 (2003) permeability transition pore and alters Ca2+ transients in rat ventricular 141–148. myocytes, Circulation 105 (2002) 2666–2671. 720 A. Szewczyk et al. / Biochimica et Biophysica Acta 1757 (2006) 715–720

[41] K. Nagy, B. Kis, N.C. Rajapakse, F. Bari, D.W. Busija, Diazoxide [59] M. Matsunga, M. Saotome, H. Satoh, H. Katoh, H. Tereda, H. Hayashi, preconditioning protects against cell Heath by attenuation of oxidative Different actions of cardioprotective agents on mitochondria Ca2+ regulation in stress upon glutamate stimulation, J. Neurosci. Res. 76 (2004) 697–704. Ca2+ pardox-induced Ca2+ overload, Circ. J. 69 (2005) 1132–1140. [42] D. Siemen, C. Loupatatzis, J. Borecky, E. Gulbins, F. Lang, Ca2+-activated [60] S. Kimura, G.X. Zhang, A. Nishiyama, T. Shokoji, L. Yao, Y.Y. Fan, M. K+ channel of the BK-type in the inner mitochondrial membrane of a human Rahman, Y. Abe, Mitochondrial-derived reactive oxygen species and glioma cell line, Biochem. Biophys. Res. Commun. 257 (1999) 549–554. vascular MAP kineses: comparisong of angiotensin II and diazoxide, [43] W. Xu, Y. Liu, S. Wang, T. McDonald, J.E. Van Eyk, A. Sidor, B. O’Rourke, Hypertension 45 (2005) 1132–1140. Cytoprotective role of Ca2+-activated K+ channels in the cardiac inner [61] M.J. de Klaver, M.G. Buckingham, G.F. Rich, Isolurane pretreatment mitochondrial membrane, Science 298 (2002) 1029–1033. Has immediate and delayed protective effects against cytokine-induced [44] T. Sato, T. Saito, N. Saegusa, H. Nakaya, Mitochondrial Ca2+-activated K+ injury in endothelial and vascular muscle cells, Anesthesiology 99 (2003) channels in cardiac myocytes: a mechanism of the cardioprotective effect 896–903. and modulation by protein kinase A, Circulation 111 (2005) 198–203. [62] T. Teshima, M. Akao, W.A. Baumgartner, E. Marban, Nicorandil prevents [45] S.N. Wu, Large-conductance Ca2+-activated K+ channels: physiological oxidative stress-induced apoptosis in neurons by activating mitochondrial role and pharmacology, Curr. Med. Chem. 10 (2003) 649–661. ATP-sensitive potassium channels, Brain Res. 990 (2003) 45–50. [46] D.W. Busija, P. Katakam, N.C. Rajapakse, B. Kis, G. Grover, F. Domoki, F. [63] Y. Dairaku, T. Miura, N. Harada, M. Kimura, T. Okamura, H. Iwamoto, R. Bari, Effects of ATP-sensitive potassium channel activators diazoxide and Kametani, M. Yamada, Y. Ikeda, M. Iwatate, S. Kawamura, M. Matsuzaki,

BMS-191095 on membrane potential and reactive oxygen species production Effect of ischemic preconditioning and mitochondria KATP channel in isolated piglet mitochondria, Brain Res. Bull. 66 (2005) 85–90. openers on chronic left ventricular remodeling in the ischemic–reperfused [47] D.F. Stowe, M. Aldakkak, A.K. Camara, M.L. Riess, A. Heinen, S.G. rat heart, Circ. J. 66 (2002) 411–415. Varadarajan, M.T. Jiang, Cardiac mitochondrial preconditioning by big [64] Z. Lacza, J.A. Snipes, A.W. Miller, C. Szabo, G. Grover, D.W. Busija, Heart Ca2+ sensitive K+ channel opening requires superoxide radical generation, mitochondria contain functional ATP-dependent K+ channels, J. Mol. Cell. Am. J. Physiol. 290 (2005) H434–H440. Cardiol. 35 (2003) 1339–1347. [48] S. Ohya, Y. Kuwata, K. Sakamoto, K. Muraki, Y. Imaizumi, Cardiopro- [65] B. Kis, K. Nagy, J.A. Scipes, N.C. Rapajakse, T. Horiguchi, G.J. Grover, 2+ tective effects of estradiol include the activation of large-conductance Ca - D.W. Busija, The mitochondrial KATP channel opener BMS-191095 activated K+ channels in cardiac mitochondria, Am. J. Physiol, Heart Circ. induces neuronal preconditioning, NeuroReport 15 (2004) 345–349. Physiol. 289 (2005) 1635–1642. [66] J. Neckar, O. Szarszoi, L. Koten, F. Papusek, B. Ostadal, G.J. Grover,

[49] G. Debska, A. Kicinska, J. Dobrucki, B. Dworakowska, E. Nurowska, J. F. Kolar, Effects of mitochondria KATP modulators on cardioprotection Skalska, K. Dolowy, A. Szewczyk, Large-conductance K+ channel openers induced by chronic hihh altitude hypoxia in rats, Cardiovasc. Res. 55 NS1619 and NS004 as inhibitors of mitochondrial function in glioma cells, (2002) 567–575. Biochem. Pharmacol. 65 (2003) 1827–1834. [67] G.J. Grover, A.J. D’Alonzo, R.B. Darbenzio, C.S. Parham, T.A. Hess, M.S.

[50] A. Kicinska, A. Szewczyk, Large conductance potassium cation channel Bathala, In vitro characterization of the mitochondrial selective KATP opener opener NS1619 inhibits cardiac mitochondria respiratory chain, Toxicol. (3R)-trans-4-((4-chlorophenyl)-N-(1H-imidazol-2-ylmethyl)dimethyl-2H- Mech. Methods 14 (2004) 59–61. 1-benzopyran-6-carbonitril monohydrochloride (BMS-191095: cardiopro- [51] I. Szabo, J. Bock, A. Jekle, M. Soddemann, C. Adams, F. Lang, M. Zoratti, E. tective, hemodynamic, and electrophysiological effects, J. Pharmacol. Exp. Gulbins, A novel potassium channel in lymphocyte mitochondria, J. Biol. Ther. 303 (2002) 132–140. Chem. 280 (2005) 12790–12798. [68] G.J. Grover, D.E. Burkett, C.S. Parham, R.J. Scales, K.K. Sadanaga,

[52] O. Oldenburg, X.-M. Yang, T. Krieg, K.D. Garlid, M.V. Cohen, G.J. Protective effect of mitochondrial KATP activation in an isolated gracilis Grover, J.M. Downey, P1075 opens mitochondrial KATP channels and model of ischemia and reperfusion in dogs, J. Cardiovasc. Pharmacol. 47 generates reactive oxygen species resulting in cardioprotection of rabbit (2003) 790–792. hearts, J. Mol. Cell. Cardiol. 35 (2003) 1035–1042. [69] M. Moses, P.D. Addison, P.C. Neligan, H. Ashrafpour, N. Huang, M. Zair, A.

[53] P. Bednarczyk, A. Kicinska, V. Kominkova, K. Ondrias, K. Dolowy, A. Rassuli, C.R. Forrest, G.J. Grover, C.Y. Pang, Mitochondrial KATP channels Szewczyk, Quinine inhibits mitochondrial ATP-regulated potassium in hindlimb remote ischemic preconditioning of skeletal muscle against channel from bovine heart, J. Membr. Biol. 199 (2004) 63–72. infraction, Am. J. Physiol, Heart Circ. Physiol. 288 (2005) H559–H567. [54] G.D. Mironova, A.E. Negoda, B.S. Marinov, P. Paucek, A.D. Costa, S.M. [70] G.J. Grover, A.J. D’Alonzo, K.D. Garlid, R. Bajgar, N.J. Lodge, P.G. Grigoriev, Y.Y. Skarga, K.D. Garlid, Functional distinctions between the Sleph, R.B. Darbenzio, T.A. Hess, M.A. Smith, P. Paucek, K.S. Atawal, + mitochondrial ATP-dependent K channel (mitoKATP) and its inward Pharmacological characterization of BMS-191095, a mitochondrial KATP rectifier subunit (mitoKIR), J. Biol. Chem. 279 (2004) 32562–32568. opener with no peripheral vasodilator or cardiac action potential shortening [55] P. Korge, H.M. Honda, J.N. Weiss, Protection of cardiac mitochondria by activity, J. Pharmacol. Exp. Ther. 303 (2001) 132–140. diazoxide and protein kinase C: implications for ischemic preconditioning, [71] P.S. Fischbach, A. White, T.D. Barret, B.R. Lucchesi, Risk of ventricular Proc. Natl. Acad. Sci. U. S. A. 99 (2002) 3312–3317. proarrythmia with selective opening of the myocardial sarcolemmal versus [56] C. Ozcan, M. Bienengraeber, P.P. Dzeja, A. Terzic, Potassium channel mitochondrial ATP-gated potassium channel, J. Pharmacol. Exp. Ther. 309 openers protect cardiac mitochondrial by attenuating oxidant stress at (2004) 554–559. reaxygenation, Am. J. Physiol, Heart Circ. Physiol. 282 (2002) H531–H539. [72] A. Szewczyk, G. Wojcik, M.J. Nalecz, Potassium channel opener, RP [57] M. Ichinose, H. Yonemochi, T. Sato, T. Saikawa, Diazoxide triggers 66471, induces membrane depolarization of rat liver mitochondria, cardioprotection against apoptosis induced by oxidative stress, Am. J. Biochem. Biophys. Res. Commun. 207 (1995) 126–132. Physiol, Heart Circ. Physiol. 284 (2003) H2235–H2241. [73] Z. Lacza, J.A. Snipes, B. Kis, C. Szabo, G. Grover, D.W.Busija, Investigation [58] T. Sato, Y. Li, T. Saito, H. Nakaya, opens mitochondrial K(ATP) of the subunit composition and pharmacology of the mitochondrial ATP- channels and cenfers cardioprotection, Br. J. Pharmacol. 141 (2004) 360–366. dependent K+ channel in the brain, Brain Res. 994 (2003) 27–36.