Download ATP-sensitive potassium channels: metabolic sensing and

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts
no text concepts found
Transcript
J Appl Physiol 103: 1888–1893, 2007;
doi:10.1152/japplphysiol.00747.2007.
Invited Review
HIGHLIGHTED TOPIC
Perspectives in Innate and Acquired Cardioprotection
ATP-sensitive potassium channels: metabolic sensing and cardioprotection
L. V. Zingman,1,2 A. E. Alekseev,2 D. M. Hodgson-Zingman,1 and A. Terzic2
1
University of Iowa, Carver College of Medicine, Iowa City, Iowa; and 2Mayo Clinic College of Medicine,
Rochester, Minnesota
homeostasis; energetics; heart failure; energetic signaling
bodily homeostasis, facilitating delivery of oxygen and nutrients, while assisting with
metabolic end-product removal. The cardiovascular system
operates under a variety of demands, ranging from conditions
of rest to extreme stress, the later requiring significant increase
in the amount of blood that needs to be pumped. Even a minor
mismatch between cardiac workload and organism needs can
precipitate a fatal outcome (9, 20, 44). Heart muscle itself is a
high-energy, demanding organ with ⬃2% of ATP content
consumed with every beat, primarily supporting excitationcontraction coupling and force generation. Without proper
adjustment of ATP production to match workload, energetic
resources would be rapidly consumed (⬃15 s to 1 min, depends on heart rate), underscoring the energy dependence of
the working myocardium (9, 44).
How the heart muscle matches the rates of ATP production
with utilization is an area of active investigation. Traditional
models of feedback adjustment of ATP hydrolysis and synthesis, based simply on cytosolic levels of ADP and inorganic
phosphate, are reevaluated, as no tight correlation between
cardiac workload and changes in bulk adenine nucleotide or
inorganic phosphate levels is readily demonstrable in vivo or
THE HEART FUNCTIONS TO MAINTAIN
Address for reprint requests and other correspondence: L. Zingman, Univ. of
Iowa, Dept. of Internal Medicine, 285 Newton Rd., CBRB2296, Iowa City, IA
52242 (e-mail: [email protected]).
1888
in vitro (4, 9, 20, 44, 59). This creates an apparent paradox
where, at maintained ATP concentrations, ATPases with a high
affinity toward ATP would function virtually in an independent
mode from energy stores. The explanation may lie in metabolic
compartmentation, metabolic “channeling”, cooperative kinetics, or parallel regulation of ATP-producing and -consuming
cellular systems (4, 9, 20, 31, 37, 44, 46, 53, 59). In this regard,
metabolic sensors, exemplified by the ATP-sensitive potassium
(KATP) channel, serve a critical role in the orchestration of cell
well-being (4, 33, 47, 58, 59, 69, 76, 78).
Formed as heteromultimers of inwardly-rectifying potassium K⫹ channel (Kir) 6.2 and ATP-binding cassette (ABC)
sulfonylurea receptor (SUR) 2A proteins, myocardial KATP
channels are unique channel-enzyme complexes that provide
linkage between cellular energetics and membrane excitability
(2– 4, 14, 26, 28 –30, 36, 43, 47, 54, 55, 58, 64, 70 –72, 75, 78).
KATP channels are coupled with phosphotransfer reactions that,
in cells in which the mobility of metabolites between intracellular microdomains is otherwise limited, integrate energetic
signaling systems (1, 4, 10, 11, 13, 15, 18, 19, 25, 59, 75).
Coupling of phosphotransfer pathways with KATP channels
permits a high-fidelity translation of energy fluxes into channel
gating, adjusting membrane excitability to match demand (1, 4,
13, 25, 33, 76). KATP channel-dependent modulation of cardiac
action potential duration, in accord with the cellular energetic
state, allows synchronized energy consumption and produc-
8750-7587/07 $8.00 Copyright © 2007 the American Physiological Society
http://www. jap.org
Downloaded from http://jap.physiology.org/ by 10.220.32.247 on May 12, 2017
Zingman LV, Alekseev AE, Hodgson-Zingman DM, Terzic A. ATP-sensitive
potassium channels: metabolic sensing and cardioprotection. J Appl Physiol 103:
1888 –1893, 2007. doi:10.1152/japplphysiol.00747.2007.—The cardiovascular system operates under a wide scale of demands, ranging from conditions of rest to
extreme stress. How the heart muscle matches rates of ATP production with
utilization is an area of active investigation. ATP-sensitive potassium (KATP)
channels serve a critical role in the orchestration of myocardial energetic wellbeing. KATP channel heteromultimers consist of inwardly-rectifying K⫹ channel
6.2 and ATP-binding cassette sulfonylurea receptor 2A that translates local ATP/
ADP levels, set by ATPases and phosphotransfer reactions, to the channel pore
function. In cells in which the mobility of metabolites between intracellular
microdomains is limited, coupling of phosphotransfer pathways with KATP channels permits a high-fidelity transduction of nucleotide fluxes into changes in
membrane excitability, matching energy demands with metabolic resources. This
KATP channel-dependent optimization of cardiac action potential duration preserves
cellular energy balance at varying workloads. Mutations of KATP channels result in
disruption of the nucleotide signaling network and generate a stress-vulnerable
phenotype with excessive susceptibility to injury, development of cardiomyopathy,
and arrhythmia. Solving the mechanisms underlying the integration of KATP
channels into the cellular energy network will advance the understanding of
endogenous cardioprotection and the development of strategies for the management
of cardiovascular injury and disease progression.
Invited Review
KATP CHANNELS: METABOLIC SENSING AND CARDIOPROTECTION
Fig. 1. Schematic presentation of the ATP-sensitive potassium (KATP) channel’s role in the balance of cardiac ATP production and utilization. KATP
channels couple cardiac energetic load (set by heart rate arterial pressure and
action potential duration) and energy production through control of membrane
excitability. The homeostatic role of the KATP channel function is particularly
critical in conditions elevating cardiac workload: hypertension (HTN) and
tachycardia (THC).
J Appl Physiol • VOL
pore closure or negative channel gating (75). Recruitment of a
posthydrolytic MgADP-bound state promotes opening of ATPinhibited KATP channels or positive channel gating (75). Elevated levels of intracellular MgADP, as occurs under metabolic stress, effectively decelerate ATPase cycling, increasing
the probability of the posthydrolytic conformation associated
with channel opening. NBD2 ATPase catalysis is characterized
by a relatively long lifetime of the NBD2䡠ADP intermediate
state compared with shorter lifetimes in other intermediates,
NBD2䡠ATP and NBD2䡠ADP䡠Pi (10, 11, 75). Thus the probability that NBD2 will adopt the ADP-bound state through the
ATPase cycle will be higher than the probability that this state
will form through simple competitive binding in the absence of
ATPase activity (11, 59). An additional conformational rearrangement that transmits the information provided by catalytic
intermediates of SUR to the channel pore may rely on the
dimerization of the two NBDs, as recently established for a
number of ABC proteins (4, 5, 24, 27, 61). In the KATP channel
complex, cooperative interaction, rather than separate contributions of each of the NBDs in SUR2A, is critical for coupling
NBD2 ATP intermediates and the functional state of the KATP
channel pore (40 – 42, 68, 77).
KATP channels operate in a compartmentalized milieu with
diffusional restrictions. Intracellular bulk nucleotide levels have
apparently only limited direct effect on KATP channels (59). The
channel agonist, MgADP, even at saturating levels (⬎100 ␮M),
shifts the range for ATP-induced KATP channel inhibition to an
IC50 from ⬃30 to ⬃300 ␮M, which is below actual bulk intracellular ATP levels (6 –10 mM) and, therefore, cannot per se
explain promotion of channel activity (59). However, several
observations support the notion that energy consumption is
compartmentalized in cardiac cells and that the free diffusional
space within the cellular microenvironment is limited, thus
restricting metabolite mobility (4, 31, 37, 59). Thereby, in the
presence of local ATP-consuming systems, submembrane nucleotide concentrations will be distinct from average cytosolic
levels (59). This would provide a possible explanation for the
activity of KATP channels measured even in the absence of
detectable gross changes in cytosolic ATP/ADP levels. Although the existence of diffusional barriers may explain
KATP channel regulation by local nucleotide levels, their
function as cellular energetic sensors, securing beneficial
channel activity in accord with cellular energetic demand,
requires a network mechanism that would shunt diffusional
barriers, allowing cellular energetic signal transfer (18, 19,
46, 59).
KATP channels: components of the cellular energetic network. Nucleotide exchange between the SUR2A ATPase and
intracellular metabolic pathways provides a mechanism coupling KATP channel function with cellular energetics (10, 11,
75). Cardiomyocytes possess well-defined phosphotransfer
systems that facilitate energetic signaling between sites of ATP
production and ATP utilization or sensing (18, 19, 46, 50, 59).
Structural and functional interactions of KATP channel proteins
with the prototypic phosphotransfer enzymes, adenylate kinase
(AK; 2ADP 7 ATP ⫹ AMP) and creatine kinase (CK; ADP ⫹
CrP 7 ATP ⫹ Cr), are assured by the distribution of these
enzymes in distinct cellular compartments, including membranes, and their physical association with KATP channel subunits (1, 13, 18, 19). The high rate of the major phosphotransfer
reaction, catalyzed by CK, scavenges ATPase products and
103 • NOVEMBER 2007 •
www.jap.org
Downloaded from http://jap.physiology.org/ by 10.220.32.247 on May 12, 2017
tion, preserving cellular energy balance at different workloads
(4, 76) (Fig. 1).
Cardiac KATP channels: Kir6.2/SUR2A complexes. Abundant in metabolically active tissues, KATP channels were originally discovered in the heart sarcolemma (48). Tetramers of
Kir6.2 comprise the pore of cardiac KATP channels (2, 14, 28,
36, 67). Yet pore-forming Kir6.2 subunits cannot readily traffic
to the plasma membrane alone, without the regulatory SUR
module, due to a COOH-terminal RKR endoplasmic reticulum
retention signal (56, 57, 74). Truncated Kir6.2, lacking retention signals, can reach the plasma membrane-generating channels that only partially reconstitute properties of intact KATP
channels (67). Both the NH2- and COOH-termini of Kir6.2
apparently contribute to recognition of ATP, the major inhibitory channel ligand, with two Kir6.2 subunits implicated in the
coordination of one ATP molecule (6, 63, 66). Mutations that
impact the ATP responsiveness of Kir6.2 alter cardiac mechanical properties at rest and produce a poor myocardial response
to ischemic and metabolic challenge (51), while in noncardiac
tissues they are associated with disease states characterized by
deficient metabolic coupling (21).
Assembly of Kir6.2 with SUR2A enhances ATP-induced
channel inhibition. Furthermore, fundamental KATP channel
properties, including stimulation by MgADP and potassium
channel openers, as well as inhibition by sulfonylurea drugs
that are absent in truncated Kir6.2 channels, are rescued by
coexpression of Kir6.2 with the SUR protein (4, 7, 12, 16,
28 –30, 43, 45, 50, 52, 60, 64). SUR2A, the regulatory subunit
of the cardiac KATP channel, incorporates two bundles of six
hydrophobic transmembrane-spanning domains (TMD) that
are fused to hydrophilic nucleotide binding domains (NBD),
also known as the ABCs (2, 4, 5, 8, 35, 40 – 42, 75). By virtue
of structure and sequence homology, SUR belongs to the
ABCC subfamily of ABC proteins (http://www.gene.ucl.ac.uk/
nomenclature/genefamily/abc.html) that usually exist as
dimers with two ABC modules and two TMDs (TMD-ABCTMD-ABC). SUR (TMD0-TMD-ABC-TMD-ABC) (4), in addition to its unique property of association with a distinct
protein subunit, Kir6.2, also contains an additional bundle of
five TMDs (TMD0), proposed to anchor SUR to the Kir6.2
channel pore (8). NBD2 of SUR, in the presence of Mg2⫹,
hydrolyses ATP, while NBD1 is primarily responsible for
nucleotide binding (10, 11, 22, 40 – 42, 68, 75, 77). The
intrinsic SUR-mediated catalysis is involved in the regulation
of Kir6.2 pore gating (10, 11, 75) (Fig. 2). The SUR ATPase
trapped in the prehydrolytic MgATP-bound state translates into
1889
Invited Review
1890
KATP CHANNELS: METABOLIC SENSING AND CARDIOPROTECTION
facilitates disengagement of posthydrolytic conformations,
promoting SUR ATPase cycling (10, 11, 59, 75). As a consequence of metabolic stress, a drop of CK flux compromises
ADP removal from ATPase sites and suspends the ATPase
cycle in a MgADP-bound state, producing channel opening, a
regulation absent in knockout mice lacking the major CK gene
M-CK (1, 10, 75). Analogously, AMP, the substrate for AK,
antagonizes ATP-induced KATP channel closure, an effect lost
in cardiomyocytes from mice lacking the primary AK1 isoform
(13). Thus phosphotransfer reactions are identified regulators
of KATP channel activity, providing a mechanism for coupling
channel behavior with the cellular energetic state.
J Appl Physiol • VOL
KATP channels and cardioprotection. Due to integration
with the cellular energetic network, KATP channels adjust
cardiomyocyte membrane excitability and produce shortening of the cardiac action potential duration, limiting intracellular calcium loading under conditions of increased demand and high-cardiac performance. KATP channel-dependent action potential shortening secures electrical stability
and diastolic interval prolongation, preserving a systolic/
diastolic calcium differential that is optimal for cardiac
relaxation and suppression of triggered arrhythmia secondary to calcium-dependent afterdepolarizations (23, 25, 32–
34, 38, 49, 58, 73, 76).
103 • NOVEMBER 2007 •
www.jap.org
Downloaded from http://jap.physiology.org/ by 10.220.32.247 on May 12, 2017
Fig. 2. Mutations in the ABCC9-encoded regulatory channel subunit sulfonylurea receptor (SUR) 2A disrupt KATP channel integration with cellular energetic
network. A: Lineweaver-Burk plot for nucleotide binding domain (NBD) 2 ATPase activities measured by spectrophotometry revealed a Vmax ⫽ 9.98 ⫾ 0.34
min⫺1 and Km ⫽ 0.11 ⫾ 0.02 mM, determined at 0 ADP plus creatine kinase (CK; 0.01 U/ml) and creatine phosphate (CrP; 5 mM). B: Lineweaver-Burk plot
for ADP-dependent inhibition of the NBD2 ATPase revealed KADP ⫽ 8.6 ⫾ 0.3 ␮M (5). C: CK (⫹ CrP) promotes ATP hydrolysis in a fusion NBD2-maltose
binding protein (MBP) constructs. Generation of [32P]Pi by purified constructs from [␥-32P]ATP was analyzed by thin-layer chromatography, followed by
autoradiography (10). D: rate-limiting steps in the SUR2A ATPase cycle are as follows: for wild-type (WT)-ADP dissociation (k4), for constructs repeating
SUR2A human mutations associated with dilated cardiomyopathy/Fs1524-Pi dissociation (k3), and for A1513T-abnormally slow k4. In addition, A1513T displays
the lowest k04 rate constant defining ADP association (11). E: ADP-scavenging by CK accelerates the ATPase in WT, but not in NBD2 mutants. F-G: Fs1524
and A1513T SUR2A mutants produced KATP channel phenotype characterized by reduced ATP sensitivity and blunted response to ADP relative to KATP channel
activity at zero nucleotide levels. [Adapted from Refs. 11 and 75.]
Invited Review
KATP CHANNELS: METABOLIC SENSING AND CARDIOPROTECTION
J Appl Physiol • VOL
missense and frame-shift mutations were mapped to domains
bordering the catalytic ATPase pocket within SUR2A. Mutant
SUR2A proteins reduced the intrinsic channel ATPase activity,
altering reaction kinetics and translating into a dysfunctional
channel phenotype with impaired metabolic signal decoding
and processing capabilities. These data implicate a link between mutations in the cardioprotective KATP channel and
susceptibility to myopathic disease and electrical instability.
Dilated cardiomyopathy with ventricular tachycardia due to
ABCC9 KATP channel mutations has been designated CMD10
(Online Mendelian Inheritance in Man no. 608569) to distinguish this entity from other etiologies causing this heterogeneous condition. The ultimate phenotype in the Kir6.2 knockout, under pressure overload, is development of heart failure
with cardiac chamber dilation, a phenotype similar to that
observed in patients with mutations in KATP channel genes (34,
73). Even in the absence of genetic deficit in KATP channel
proteins, cardiomyocytes in heart failure undergo extensive
remodeling, including diminished mitochondrial respiration,
suppressed CK flux, decreased energy storage, and cytoskeletal
disruption, although bulk cytosolic ATP levels are preserved
(4, 11, 25, 33). These changes impact metabolic signal generation, transduction, and decoding by KATP channels. KATP
channels from failing hearts display an improper response to
ATP and creatine kinase, a reduced recognition of metabolic
distress, and do not provide for appropriate action potential
shortening upon exposure to hypoxia (25). Consequently, such
hearts are excessively vulnerable to calcium loading and myocyte necrosis under stress that can be largely ameliorated by
pharmacological restoration of KATP channel opening with KATP
channel-opening drugs, such as nicorandil and pinacidil (25).
More recently, a KATP channel mutation conferring risk for
adrenergic atrial fibrillation originating from vein of Marshall
was identified (49). The vein of Marshall, a remnant of the left
superior vena cava rich in sympathetic fibers, is a recognized
source for adrenergic atrial fibrillation. Genetic investigation
uncovered a missense mutation (T1547I) also in the ABCC9
gene, encoding the SUR2A regulatory subunit of cardiac KATP
channels. Structural modeling of mutant KATP channels predicted,
and patch-clamp electrophysiology demonstrated, compromised
function with defective stress responsiveness. Targeted knockout
of the KATP channel verified the pathogenic link between channel
dysfunction and predisposition to adrenergic atrial fibrillation. In
this first report of genetic susceptibility to adrenergic vein of
Marshall atrial fibrillation, radiofrequency ablation was curative,
disrupting the gene-environment substrate for arrhythmia conferred by KATP channelopathy (49).
Thus cardiac KATP channels through tight integration with
cellular metabolic signaling pathways respond with high fidelity to perturbations in cellular energetics. This highly responsive integrated system adapts metabolic expenditures to available resources, resulting in an effective feedback that maintains
cellular well-being under a range of physiological and pathological conditions. Further understanding of mechanisms responsible for KATP channel regulation and KATP channelopathy-associated disease susceptibility provides novel insights
into endogenous cardioprotection and the development of strategies for the management of cardiovascular injury and disease
progression.
103 • NOVEMBER 2007 •
www.jap.org
Downloaded from http://jap.physiology.org/ by 10.220.32.247 on May 12, 2017
KATP channel function is recognized as vital for adaptation
to physiological and pathological stress. Hearts genetically
deprived of KATP channels lack stress-induced cardiac action
potential shortening upon adrenergic stress (76). Indeed, loss of
this regulator of membrane electrical stability in the Kir6.2knockout predisposes to cytosolic calcium overload associated
with development of progressive contractile dysfunction and
death under acute stress (76). On autopsy, contraction bands,
pathognomonic of cytosolic calcium loading, are visible
throughout the myocardium of the Kir6.2-knockout, but not
wild-type, in response to ␤-adrenergic stress (76). Similarly,
under chronic stress associated with physical exertion, KATP
channel knockout compromises the benefit of exercise training,
resulting in cardiac functional and structural deficits (32). Thus
cardiac KATP channels, harnessing the ability to recognize
alterations in the metabolic state of the cell and translate this
information into changes in membrane excitability, provide a
link necessary for the maintenance of myocardial well-being
under stress-induced energy-demanding augmentation in performance.
In pathological conditions, KATP channels are recognized to
also serve a vital cardioprotective role. Genetic disruption of
KATP channels results in poor recovery following coronary
hypoperfusion and compromises the protective benefits of
ischemic preconditioning (23, 47, 58, 62), while overexpression of channel subunits generates a phenotype resistant to
ischemia (17). Analogous to ischemic preconditioning, pharmacological activation of the channel through use of KATP
channel openers displays cardioprotective outcome in hearts
subject to ischemic challenge (23, 33, 58, 62). This pharmacological preconditioning is abolished in the absence of Kir6.2containing KATP channels (23, 33, 58, 62). In fact, Kir6.2
knockout also negates protection afforded by ischemic preconditioning on myocardial energy generation, transfer, and utilization.
Total ATP turnover, a global parameter of myocardial energetic
dynamics, fails to increase in the ischemic-preconditioned KATP
channel knockout, as opposed to wild type, correlating with the
failure of preconditioned hearts lacking KATP channels to functionally recover (23). As discussed above, the maintenance of
energetic stability in the myocardium requires synchronized
ATP generation and utilization accomplished through energetic
phosphotransfer relay systems transferring energy from sites of
production to sites of utilization and promptly removing end
products from sites of utilization. In this regard, in wild-type
but not Kir6.2-knockout ischemic hearts, preconditioning preserves CK phosphotransfer, the major energy transfer pathway
in the heart (23, 33). Disruption of KATP channel function
forms the basis for reduction in energetic production and/or
consumption that contributes to contractile dysfunction. These
findings may provide a mechanistic basis for the potentially
deleterious outcomes associated with use of KATP channel
blockers, sulfonylurea medication, in the setting of myocardial
infarction (33).
Defects in the function of KATP channel proteins themselves,
disruption of intracellular metabolic networks, and/or disturbed
communication between channel proteins and the energetic
network can all contribute to susceptibility toward cardiac
disease and poor outcome (11, 49). In humans, mutations in the
ABCC9-encoded regulatory channel subunit SUR2A have been
demonstrated in a subset of patients with dilated cardiomyopathy and ventricular arrhythmia (11) (Fig. 2). The identified
1891
Invited Review
1892
KATP CHANNELS: METABOLIC SENSING AND CARDIOPROTECTION
REFERENCES
J Appl Physiol • VOL
103 • NOVEMBER 2007 •
www.jap.org
Downloaded from http://jap.physiology.org/ by 10.220.32.247 on May 12, 2017
1. Abraham MR, Selivanov VA, Hodgson DM, Pucar D, Zingman LV,
Wieringa B, Dzeja PP, Alekseev AE, Terzic A. Coupling of cell
energetics with membrane metabolic sensing. Integrative signaling
through creatine kinase phosphotransfer disrupted by M-CK gene knockout. J Biol Chem 277: 24427–24434, 2002.
2. Aguilar-Bryan L, Nichols CG, Wechsler SW, Clement JP 4th, Boyd
AE 3rd, Gonzalez G, Herrera-Sosa H, Nguy K, Bryan J, Nelson DA.
Cloning of the beta cell high-affinity sulfonylurea receptor: a regulator of
insulin secretion. Science 268: 423– 426, 1995.
3. Alekseev AE, Brady PA, Terzic A. Ligand-insensitive state of cardiac
ATP-sensitive K⫹ channels. Basis for channel opening. J Gen Physiol
111: 381–394, 1998.
4. Alekseev AE, Hodgson DM, Karger AB, Park S, Zingman LV, Terzic
A. ATP-sensitive K⫹ channel/enzyme multimer: metabolic gating in the
heart. J Mol Cell Cardiol 38: 895–905, 2005.
5. Altenberg GA. Structure of multidrug-resistance proteins of the ATPbinding cassette (ABC) superfamily. Curr Med Chem Anticancer Agents
4: 53– 62, 2004.
6. Antcliff JF, Haider S, Proks P, Sansom MS, Ashcroft FM. Functional
analysis of a structural model of the ATP-binding site of the K(ATP)
channel Kir6.2 subunit. EMBO J 24: 229 –239, 2005.
7. Ashcroft FM, Gribble FM. New windows on the mechanism of action of
K(ATP) channel openers. Trends Pharmacol Sci 21: 439 – 445, 2000.
8. Babenko AP, Bryan J. SUR domains that associate with and gate KATP
pores define a novel gatekeeper. J Biol Chem 278: 41577– 41580, 2003.
9. Balaban RS. Cardiac energy metabolism homeostasis: role of cytosolic
calcium. J Mol Cell Cardiol 38: 895–905, 2005.
10. Bienengraeber M, Alekseev AE, Abraham MR, Carrasco AJ, Moreau
C, Vivaudou M, Dzeja PP, Terzic A. ATPase activity of the sulfonylurea
receptor: a catalytic function for the KATP channel complex. FASEB J 14:
1943–1952, 2000.
11. Bienengraeber M, Olson TM, Selivanov VA, Kathmann EC,
O’Coclain F, Gao F, Karger AB, Ballew JD, Hodgson DM, Zingman
LV, Pang YP, Alekseev AE, Terzic A. ABCC9 mutations identified in
human dilated cardiomyopathy disrupt catalytic KATP channel gating. Nat
Genet 36: 382–387, 2004.
12. Campbell JD, Sansom MS, Ashcroft FM. Potassium channel regulation.
EMBO Rep 4: 1038 –1042, 2003.
13. Carrasco AJ, Dzeja PP, Alekseev AE, Pucar D, Zingman LV, Abraham MR, Hodgson DM, Bienengraeber M, Puceat M, Janssen E,
Weringa B, Terzic A. Adenylate kinase phosphotransfer communicates
cellular energetic signals to ATP-sensitive potassium channels. Proc Natl
Acad Sci USA 98: 7623–7628, 2001.
14. Clement JP 4th, Kunjilwar K, Gonzalez G, Schwanstecher M, Panten
U, Aguilar-Bryan L, Bryan J. Association and stoichiometry of K(ATP)
channel subunits. Neuron 18: 827– 838, 1997.
15. Crawford RM, Ranki JH, Botting CH, Budas GR, Jovanovic A.
Creatine kinase is physically associated with the cardiac ATP-sensitive
K⫹ channel in vivo. FASEB J 16: 102–104, 2002.
16. Drain P, Geng X, Li L. Concerted gating mechanism underlying KATP
channel inhibition by ATP. Biophys J 86: 2101–2112, 2004.
17. Du Q, Jovanoviæc S, Clelland A, Sukhodub A, Budas G, Phelan K,
Murray-Tait V, Malone L, Jovanoviæc A. Overexpression of SUR2A
generates a cardiac phenotype resistant to ischemia. FASEB J 20: 1131–
1141, 2006.
18. Dzeja PP, Terzic A. Phosphotransfer reactions in the regulation of
ATP-sensitive K⫹ channels. FASEB J 12: 523–529, 1998.
19. Dzeja PP, Redfield MM, Burnett JC, Terzic A. Failing energetics in
failing hearts. Curr Cardiol Rep 2: 212–217, 2000.
20. Gibbs CL. Cardiac energetics: sense and nonsense. Clin Exp Pharmacol
Physiol 30: 598 – 603, 2003.
21. Gloyn AL, Pearson ER, Antcliff JF, Proks P, Bruining GJ, Slingerland AS, Howard N, Srinivasan S, Silva JM, Molnes J, Edghill EL,
Frayling TM, Temple IK, Mackay D, Shield JP, Sumnik Z, van Rhijn
A, Wales JK, Clark P, Gorman S, Aisenberg J, Ellard S, Njolstard
PR, Ashcroft FM, Hattersley AT. Activating mutations in the gene
encoding the ATP-sensitive potassium-channel subunit Kir6.2 and permanent neonatal diabetes. N Engl J Med 350: 1838 –1849, 2004.
22. Gribble FM, Tucker SJ, Ashcroft FM. The essential role of the Walker
A motifs of SUR1 in K-ATP channel activation by Mg-ADP and diazoxide. EMBO J 16: 1145–1152, 1997.
23. Gumina RJ, Pucar D, Bast P, Hodgson DM, Kurtz CE, Dzeja PP, Miki
T, Seino S, Terzic A. Knockout of Kir6.2 negates ischemic preconditioning-induced protection of myocardial energetics. Am J Physiol Heart Circ
Physiol 284: H2106 –H2113, 2003.
24. Higgins CF, Linton KJ. The ATP switch model for ABC transporters.
Nat Struct Mol Biol 11: 918 –926, 2004.
25. Hodgson DM, Zingman LV, Kane GC, Perez-Terzic C, Bienengraeber
M, Ozcan C, Gumina RJ, Pucar D, O’Cochlain F, Mann DL, Alekseev
AE, Terzic A. Cellular remodeling in heart failure disrupts KATP channeldependent stress tolerance. EMBO J 22: 1732–1742, 2003.
26. Hu K, Huang CS, Jan YN, Jan LY. ATP-sensitive potassium channel
traffic regulation by adenosine and protein kinase C. Neuron 38: 417– 432,
2003.
27. Hung LW, Wang IX, Nikaido K, Liu PQ, Ames GFL, Kim SH. Crystal
structure of the ATP-binding subunit of an ABC transporter. Nature 396:
703–707, 1998.
28. Inagaki N, Gonoi T, Clement JP, Namba N, Inazawa J, Gonzales G,
Aguilar-Bryan L, Seino S, Bryan J. Reconstitution of I KATP: an inward
rectifier subunit plus the sulfonylurea receptor. Science 270: 1166 –1170,
1995.
29. Inagaki N, Gonoi T, Clement JP, Wang CZ, Aguilar-Bryan L, Bryan
J, Seino S. A family of sulfonylurea receptors determines the pharmacological properties of ATP-sensitive K channels. Neuron 16: 1011–1017,
1996.
30. John SA, Weiss JN, Ribalet B. Regulation of cloned ATP-sensitive K⫹
channels by adenine nucleotides and sulfonylureas: interactions between
SUR1 and positively charged domains on Kir6.2. J Gen Physiol 118:
391– 405, 2001.
31. Kaasik A, Veksler V, Boehm E, Novotova M, Minajeva A, VenturaClapier R. Energetic crosstalk between organelles: architectural integration of energy production and utilization. Circ Res 89: 153–159, 2001.
32. Kane GC, Behfar A, Yamada S, Perez-Terzic C, O’Cochlain F, Reyes
S, Dzeja PP, Miki T, Seino S, Terzic A. ATP-sensitive K⫹ channel
knockout compromises the metabolic benefit of exercise training, resulting
in cardiac deficits. Diabetes 53: S169 –S175, 2004.
33. Kane GC, Liu XK, Yamada S, Olson TM, Terzic A. Cardiac KATP
channels in health and disease. J Mol Cell Cardiol 38: 937–943, 2005.
34. Kane GC, Behfar A, Dyer RB, O’Cochlain DF, Liu XK, Hodgson DM,
Reyes S, Miki T, Seino S, Terzic A. KCNJ11 gene knockout of the Kir6.2
KATP channel causes maladaptive remodeling and heart failure in hypertension. Hum Mol Genet 15: 2285–2297, 2006.
35. Kerr ID. Structure and association of ATP-binding cassette transporter
nucleotide-binding domains. Biochim Biophys Acta 1561: 47– 64, 2002.
36. Lederer WJ, Nichols CG. Nucleotide modulation of the activity of rat
heart ATP-sensitive K⫹ channels in isolated membrane patches. J Physiol
419: 193–211, 1989.
37. Lederer WJ, Niggli E, Hadley RW. Sodium-calcium exchange in excitable cells: fuzzy space. Science 248: 283, 1990.
38. Liu XK, Yamada S, Kane GC, Alekseev AE, Hodgson DM,
O’Cochlain F, Jahangir A, Miki T, Seino S, Terzic A. Genetic disruption of Kir6.2, the pore-forming subunit of ATP-sensitive K⫹ channel,
predisposes to catecholamine-induced ventricular dysrhythmia. Diabetes
53: S165–S168, 2004.
39. Lorenz E, Terzic A. Physical association between recombinant cardiac
ATP-sensitive K⫹ channel subunits Kir6.2 and SUR2A. J Mol Cell
Cardiol 31: 425– 434, 1999.
40. Matsuo M, Dabrowski M, Ueda K, Ashcroft FM. Mutations in the
linker domain of NBD2 of SUR inhibit transduction but not nucleotide
binding. EMBO J 21: 4250 – 4258, 2002.
41. Matsuo M, Kioka N, Amachi T, Ueda K. ATP binding properties of the
nucleotide-binding folds of SUR1. J Biol Chem 274: 37479 –37482, 1999.
42. Matsuo M, Tanabe K, Kioka N, Amachi T, Ueda K. Different binding
properties and affinities for ATP and ADP among sulfonylurea receptor
subtypes, SUR1, SUR2A, and SUR2B. J Biol Chem 275: 28757–28763,
2000.
43. Matsushita K, Kinoshita K, Matsuoka T, Fujita A, Fujikado T, Tano
Y, Nakamura H, Kurachi Y. Intramolecular interaction of SUR2 subtypes for intracellular ADP-induced differential control of K(ATP) channels. Circ Res 90: 554 –561, 2002.
44. Mootha VK, Arai AE, Balaban RS. Maximum oxidative phosphorylation capacity of the mammalian heart. Am J Physiol Heart Circ Physiol
272: H769 –H775, 1997.
Invited Review
KATP CHANNELS: METABOLIC SENSING AND CARDIOPROTECTION
J Appl Physiol • VOL
62.
63.
64.
66.
67.
68.
69.
70.
71.
72.
73.
74.
75.
76.
77.
78.
drives formation of a nucleotide sandwich dimmer. Mol Cell 10: 139 –149,
2002.
Suzuki M, Sasaki N, Miki T, Sakamoto N, Ohmoto-Sekine Y, Tamagawa M, Seino S, Marban E, Nakaya H. Role of sarcolemmal K(ATP)
channels in cardioprotection against ischemia/reperfusion injury in mice.
J Clin Invest 109: 509 –516, 2002.
Tanabe K, Tucker SJ, Matsuo M, Proks P, Ashcroft FM, Seino S.
Direct photoaffinity labeling of the Kir6.2 subunit of the ATP-sensitive
K⫹ channel by 8-azido-ATP. J Biol Chem 274: 3931–3933, 1999.
Terzic A, Jahangir A, Kurachi Y. Cardiac ATP-sensitive K⫹ channels:
regulation by intracellular nucleotides and K⫹ channel-opening drugs.
Am J Physiol Cell Physiol 269: C525–C545, 1995.
Trapp S, Haider S, Jones P, Sansom MS, Ashcroft FM. Identification
of residues contributing to the ATP binding site of Kir6.2. EMBO J 22:
2903–2912, 2003.
Tucker SJ, Gribble FM, Zhao C, Trapp S, Ashcroft FM. Truncation of
Kir6.2 produces ATP-sensitive K⫹ channels in the absence of the sulphonylurea receptor. Nature 387: 179 –183, 1997.
Ueda K, Komine J, Matsuo M, Seino S, Amachi T. Cooperative binding
of ATP and MgADP in the sulfonylurea receptor is modulated by glibenclamide. Proc Natl Acad Sci USA 96: 1268 –1272, 1999.
Vivaudou M, Forestier C. Modification by protons of frog skeletal
muscle KATP channels: effects on ion conduction and nucleotide inhibition. J Physiol 486: 629 – 645, 1995.
Weiss JN, Lamp ST. Glycolysis preferentially inhibits ATP-sensitive K⫹
channels in isolated guinea pig cardiac myocytes. Science 238: 67– 69,
1987.
Weiss JN, Venkatesh N, Lamp ST. ATP-sensitive K⫹ channels and
cellular K⫹ loss in hypoxic and ischaemic mammalian ventricle. J Physiol
447: 649 – 673, 1992.
Weiss JN, Venkatesh N. Metabolic regulation of cardiac ATP-sensitive
K⫹ channels. Cardiovasc Drugs Ther 7: 499 –505, 1993.
Yamada S, Kane GC, Behfar A, Liu XK, Dyer RB, Faustino RS, Miki
T, Seino S, Terzic A. Protection conferred by myocardial ATP-sensitive
K⫹ channels in pressure overload-induced congestive heart failure revealed in KCNJ11 Kir6.2-null mutant. J Physiol 577: 1053–1065, 2006.
Zerangue N, Schwappach B, Jan YN, Jan LY. A new ER trafficking
signal regulates the subunit stoichiometry of plasma membrane KATP
channels. Neuron 22: 537–548, 1999.
Zingman LV, Alekseev AE, Bienengraeber M, Hodgson DM,
Karger AB, Dzeja PP, Terzic A. Signaling in channel/enzyme multimers: ATPase transitions in SUR module gate ATP-sensitive K⫹ conductance. Neuron 31: 233–245, 2001.
Zingman LV, Hodgson DM, Bast PH, Kane GC, Perez-Terzic C,
Gumina RJ, Pucar D, Bienengraeber M, Dzeja PP, Miki T, Seino S,
Alekseev AE, Terzic A. Kir6.2 is required for adaptation to stress. Proc
Natl Acad Sci USA 99: 13278 –13283, 2002.
Zingman LV, Hodgson DM, Bienengraeber M, Karger AB, Kathmann EC, Alekseev AE, Terzic A. Tandem function of nucleotide
binding domains confers competence to sulfonylurea receptor in gating
ATP-sensitive K⫹ channels. J Biol Chem 277: 14206 –14210, 2002.
Zingman LV, Hodgson DM, Alekseev AE, Terzic A. Stress without
distress: homeostatic role for KATP channels. Mol Psychiatry 8: 253–254,
2003.
103 • NOVEMBER 2007 •
www.jap.org
Downloaded from http://jap.physiology.org/ by 10.220.32.247 on May 12, 2017
45. Moreau C, Jacquet H, Prost AL, D’hahan N, Vivaudou M. The
molecular basis of the specificity of action of K(ATP) channel openers.
EMBO J 19: 6644 – 6651, 2000.
46. Nascimben L, Ingwall JS, Pauletto P, Friedrich J, Gwathmey JK,
Saks V, Pessina AC, Allen PD. Creatine kinase system in failing and
non-failing human myocardium. Circulation 94: 1894 –1901, 1996.
47. Nichols CG, Lederer WJ. Adenosine triphosphate-sensitive potassium
channels in the cardiovascular system. Am J Physiol Heart Circ Physiol
261: H1675–H1686, 1991.
48. Noma A. ATP-regulated K⫹ channels in cardiac muscle. Nature 305:
147–148, 1983.
49. Olson TM, Alekseev AE, Moreau C, Liu XK, Zingman LV, Miki T,
Seino S, Asirvatham SJ, Jahangir A, Terzic A. KATP channel mutation
confers risk for vein of Marshall adrenergic atrial fibrillation. Nat Clin
Pract Cardiovasc Med 4: 110 –116, 2007.
50. O’Rourke B, Ramza BM, Marban E. Oscillations of membrane current
and excitability driven by metabolic oscillations in heart cells. Science
265: 962–966, 1994.
51. Rajashree R, Koster JC, Markova KP, Nichols CG, Hofmann PA.
Contractility and ischemic response of hearts from transgenic mice with
altered sarcolemmal K(ATP) channels. Am J Physiol Heart Circ Physiol
283: H584 –H590, 2002.
52. Reimann F, Gribble FM, Ashcroft FM. Differential response of K(ATP)
channels containing SUR2A or SUR2B subunits to nucleotides and pinacidil. Mol Pharmacol 58: 1318 –1325, 2000.
53. Saks VA, Aliev MK. Is there the creatine kinase equilibrium in working
heart cells? Biochem Biophys Res Commun 227: 360 –367, 1996.
54. Sasaki N, Sato T, Marban E, O’Rourke B. ATP consumption by
uncoupled mitochondria activates sarcolemmal K(ATP) channels in cardiac myocytes. Am J Physiol Heart Circ Physiol 280: H1882–H1888,
2001.
55. Satoh E, Yamada M, Kondo C, Repunte VP, Horio Y, Iijima T,
Kurachi Y. Intracellular nucleotide-mediated gating of SUR/Kir6.0 complex potassium channels expressed in a mammalian cell line and its
modification by pinacidil. J Physiol 11: 663– 674, 1998.
56. Schwanstecher M, Sieverding C, Dorschner H, Gross I, AguilarBryan L, Schwanstecher C, Bryan J. Potassium channel openers require
ATP to bind to and act through sulfonylurea receptors. EMBO J 17:
5529 –5535, 1998.
57. Schwappach B, Zerangue N, Jan YN, Jan LY. Molecular basis for
K(ATP) assembly: transmembrane interactions mediate association of a
K⫹ channel with an ABC transporter. Neuron 26: 155–167, 2000.
58. Seino S, Miki T. Physiological and pathophysiological roles of ATPsensitive K⫹ channels. Prog Biophys Mol Biol 81: 133–176, 2003.
59. Selivanov V, Alekseev AE, Hodgson DM, Dzeja PP, Terzic A. Nucleotide-gated KATP channels integrated with creatine and adenylate kinases: amplification, tuning and sensing of energetic signals in the compartmentalized cellular environment. Mol Cell Biochem 256: 243–256,
2004.
60. Shyng SL, Ferrigni T, Nichols CG. Regulation of KATP channel activity
by diazoxide and MgADP: distinct functions of the two nucleotide binding
folds of the sulfonylurea receptor. J Gen Physiol 110: 643– 654, 1997.
61. Smith PC, Karpowich N, Millen L, Moody JE, Rosen J, Thomas PJ,
Hunt JF. ATP binding to the motor domain from an ABC transporter
1893