Sie sind auf Seite 1von 9

REVIEW Circ J 2009; 73: 1184 – 1192

GSK-3β, a Therapeutic Target for Cardiomyocyte Protection

Tetsuji Miura, MD; Takayuki Miki, MD

Glycogen synthase kinase-3β(GSK-3β) is a multifunctional Ser/Thr kinase that plays important roles in necrosis
and apoptosis of cardiomyocytes. A major mechanism of cell necrosis is the opening of the mitochondrial per-
meability transition pore (mPTP), which consists of multiple protein subunits, including adenine nucleotide
translocase (ANT). The threshold for mPTP opening is elevated by phosphorylation of GSK-3β at Ser9, which
reduces activity of this kinase. How inactivation of GSK-3βsuppresses mPTP opening has not been fully under-
stood, but evidence to date suggests that preservation of hexokinase-II in the mPTP complex, inhibition of
cyclophilin-D-ANT binding, inhibition of p53 and inhibition of ANT into the mitochondria are contributory.
GSK-3βphosphorylation is a step to which multiple protective signaling pathways converge, and thus GSK-3β
phosphorylation is crucial in cardioprotection of a variety of interventions against ischemia/reperfusion injury.
Apoptosis of cardiomyocytes by pressure overload or ischemia/reperfusion is also suppressed by inactivation of
GSK-3β, in which reduced phosphorylation of p53, heat shock factor-1 and myeloid cell leukemia sequence-1 and
inhibition of Bax translocation might be involved. Considering predominant roles of GSK-3β in cardiomyocyte
death, manipulation of this protein kinase is a promising strategy for myocardial protection in coronary artery
disease and heart failure.   (Circ J 2009; 73: 1184 – 1192)
Key Words: Apoptosis; Heart failure; Myocardial infarction; Preconditioning; Signal transduction

G
lycogen synthase kinase-3β (GSK-3β) is a constitu- Mechanisms of Cardiomyocyte Necrosis
tively active 47-kDa Ser/Thr protein kinase that
was purified more than 2 decades ago as a kinase During Ischemia/Reperfusion
that reduces glycogen synthase activity. However, GSK-3β When myocardial blood perfusion is interrupted, for
is now known as a multifunctional kinase having more than example by occluding the coronary artery, ischemic car-
40 substrates, playing roles not only in glycogen metabolism diomyocytes suffer from several critical intracellular events
but also cell proliferation, growth and death.1–3 To properly that can lead to cell necrosis and/or prime the cells to reper-
execute its functions, GSK-3βhas multiple regulatory mech- fusion-induced necrosis. First, intracellular level of high-
anisms including phosphorylation at Ser and Tyr residues, energy phosphate is reduced due to oxygen deficiency.
complex formation with scaffold proteins, priming of sub- Although ischemic cardiomyocytes cease contraction within
strates and intracellular translocation. Recently, pathophysi- a few minutes after the onset of ischemia, adenosine triphos-
ological roles of GSK-3β have also received attention phate (ATP) consumption continues during ischemia mainly
because it is involved in common and serious diseases such by mitochondrial ATPase for maintenance of mitochondrial
as Alzheimer’s disease, bipolar mood disorder, cancer and membrane potential.6,7 Anaerobic glycolysis supplies ATP
ischemia/reperfusion injury.1,3 In the cardiovascular system, during the early period of ischemia, but it is ultimately halted
GSK-3β has major roles in glucose metabolism,4 cardio- by inhibition of glyceraldehyde phosphate dehydrogenase
myocyte hypertrophy5 and cell death. The roles of GSK-3β due to accumulated H+ and NADH.7,8 Second, intracellular
in hypertrophy have recently been reviewed by Sugden et Na+ accumulates due to Na+ influx via Na+–H+ exchangers
al5, so therefore we have focused on the roles of this protein and Na+ channels and due to reduced Na+ efflux via the
kinase in myocyte death, particularly that after ischemia/ Na+–K+ pump.9–11 This Na+ overload predisposes ischemic
reperfusion. We first briefly overview mechanisms of cardiomyocytes to Ca2+ influx by the Na+–Ca2+ exchanger.
ischemia/reperfusion injury and then discuss the contribu- Third, Ca2+ influx via the Na+–Ca2+ exchanger, reduced Ca2+
tion of GSK-3β to cardiomyocyte death and manipulation uptake into the sarcoplasmic reticulum and reduced Ca2+
of GSK-3βfor myocardial protection. efflux via the sarcolemmal Ca2+ pump induce Ca2+ overload
in ischemic cardiomyocytes.9–12 However, elevation of intra-
cellular Ca2+ is modest during ischemia because acidosis
inhibits the Na+–Ca2+ exchanger and cytosolic Ca2+ is taken
up by the mitochondria as long as its membrane potential is
(Received April 22, 2009; revised manuscript received May 13, 2009; maintained by use of ATP.9–11,13
accepted May 14, 2009; released online June 9, 2009) Severely ischemic cardiomyocytes ultimately ‘starve to
Division of Cardiology, Second Department of Internal Medicine, death’, although sarcolemmal damage by detergent actions
Sapporo Medical University School of Medicine, Sapporo, Japan of accumulated long-chain Acyl-CoA and by Ca2+-activated
Mailing address:  Tetsuji Miura, MD, Division of Cardiology, Second proteases and phospholipases might also be involved in
Department of Internal Medicine, Sapporo Medical University School
of Medicine, South-1 West-16, Chuo-ku, Sapporo 060-8543, Japan.
ischemia-induced necrosis.7 Nevertheless, reperfusion is nec-
E-mail: miura@sapmed.ac.jp essary to salvage ischemic cardiomyocytes from necrosis.
All rights are reserved to the Japanese Circulation Society. For permis- However, reperfusion per se can trigger lethal mechanisms.
sions, please e-mail: cj@j-circ.or.jp Reperfusion washes out H+ in the extracellular space, which

Circulation Journal Vol.73, July 2009


GSK-3βin Cardioprotection 1185

Figure 1.   Models of mitochondrial permeability tran-


sition pore (PTP). (A) A classic model. OMM, outer
mitochondrial membrane; IMM, inner mitochondrial
membrane; IMS, intermembrane space; HK, hexokinase;
PBR, peripheral benzodiazepine receptor; CK, creatine
kinase; CypD, cyclophilin-D; VDAC, voltage-dependent
anion channel; ANT, adenine nucleotide translocator.
The pore consists of VDAC and ANT. In response to
cellular stress, CypD in the matrix binds to ANT, which
sensitizes the pore to opening stimuli such as Ca2+.
(B) A new model. Components of the pore itself (PTP)
are not unidentified but are known to be regulated by
CypD, ANT and other elements depicted in the figure.
From Juhaszova et al28 with permission.

retracts inhibition of the reverse mode operation of the apoptosis is involved in cardiomyocyte death after ischemia/
Na+–Ca2+ exchanger, resulting in massive Ca2+ influx into reperfusion. Ischemia/reperfusion induces activation of
cardiomyocytes.9–11 The abrupt increase in intracellular Ca2+ caspase-3, -8, -9 and translocation of Bax and Bcl-2 in the
level induces activation of calpain, a Ca2+-activated protease, myocardium.18,19 Myocardial infarct size (ie, size of necrotic
which compromises Na+–K+ pump function10 and Ca2+ tissue mass) was reduced to 37–78% of controls by caspase
overload of the mitochondria.6,7,9,11 At the same time as Ca2+ inhibitors20,21 or genetic deletion of Fas,22 and this protec-
overload, cardiomyocytes abruptly develop cell edema after tion was associated with reduction in TUNEL-positive cells
reperfusion as a result of osmotic loading during ischemia. in reperfused regions. Furthermore, recent studies have
ATP production restituted by re-supply of oxygen in Ca2+- shown that apoptotic process is converted to necrotic process
overloaded and swelling myocytes with fragile sarcolemma when essential co-factors for apoptosis are lacking.23–25
induces hypercontraction and sarcolemmal rupture. Involve- ATP is one of such co-factors, and switching from apoptosis
ment of this mechanical factor in cardiomyocyte necrosis is to necrosis by inhibition of mitochondrial ATP generation
supported by findings that suppression of myocardial con- has been shown in non-cardiac cells.23,24 Taken together,
traction during the early period of reperfusion by the use of these findings are consistent with the notion that apoptotic
butanedione monoxime significantly reduced infarct size.14,15 cell death triggered by intrinsic mechanisms and/or by
However, a major mechanism of lethal reperfusion injury is Fas-Fas ligand system is ultimately converted to necrotic
likely to be opening of the mitochondrial permeability tran- cell death in the myocardium during ischemia/reperfusion.
sition pore (mPTP) at the time of reperfusion, which will be However, this possibility remains to be critically tested.
discussed in detail below.
mPTP and Reperfusion Injury
Apoptosis of Cardiomyocytes After
The mPTP is a non-selective large conductance channel in
Ischemia/Reperfusion the mitochondrial inner membrane, which is physiologically
Most, if not all, of dead cardiomyocytes after ischemia/ closed. Opening of mPTPs is involved in cell death induced
reperfusion show morphology of necrosis under examina- by a variety of causes (for example, ischemia/reperfusion,
tions with macro- and micro-histochemistry and electron alcohol, endotoxin, anti-cancer agents).26 Although the
microscopy. However, contribution of apoptosis to myocyte molecular structure of the mPTP has not yet been clarified,
loss after ischemia/reperfusion is rather inconclusive.16 A several proteins have been suggested to be subunits of this
major problem in this issue is limitations of methods used for multi-protein complex.26–29 In a classic model of mPTP, the
detecting apoptosis. Terminal deoxynucleoidyl tranferease- pore is formed by adenine nucleotide translocase (ANT) in
mediated dUTP in-situ nick end labeling (TUNEL) and the inner membrane and voltage-dependent anion channel
detection of non-random fragmentation of DNA (DNA lad- (VDAC) (Figure 1A). In this model, binding of cyclophilin-
dering) are somewhat non-specific, and TUNEL-positive D (CypD), a matrix protein, to ANT increases sensitivity of
cardiomyocytes after ischemia/reperfusion in situ actually ANT to Ca2+, a trigger of mPTP opening. A crucial role of
showed necrotic morphology under electron microscopy.17 CypD in mPTP opening has been indicated by 3 indepen-
However, there are some observations suggesting that dent studies demonstrating that deletion of a gene coding

Circulation Journal Vol.73, July 2009


1186 MIURA T et al.

Figure 2.   Interaction of phospho-GSK-3βwith mitochondrial permeability transition pore subunit proteins in rat hearts.
Isolated perfused rat hearts were subjected to 25-min ischemia and reperfusion with or without IPC and EPO infusion
before ischemia. Tissue samples were taken for IP and IB at 5 min after reperfusion. (A) IPC + EPO increased level of
ANT in phospho-GSK-3β immunoprecipitates by 50%, whereas phospho-GSK-3β-VDAC interaction was not detected.
(B) Level of CypD in ANT immunoprecipitates was reduced by IPC + EPO to 40% of the control level, though ANT-
VDAC binding was unchanged. ANT, adenine nucleotide translocase; CypD, cyclophilin-D; EPO, erythropoietin; GSK,
glycogen synthase kinase; IB, immunoblotting; IP, immunoprecipitation; VDAC, voltage-dependent anion channel.
*P<0.05 vs Control. From Nishihara et al35 with permission.

mitochondrial CypD (Ppif) markedly increased resistance TNF-α plus cyclohexamide was not different from that of
of cells to mPTP opening and necrosis.30–32 However, recent normal control cells.32 There is no clear explanation for
studies using ANT- and VADAC-deficient mice indicate the contradictory observations, but it may reflect complex
that ANT and VDAC are not indispensable for mPTP cross-talks of redundant signaling pathways of apoptosis
opening.33,34 Based on these findings, a new model of mPTP and necrosis.24
was recently proposed by Sollott’s group (Figure 1B).28 In
this new model, ANT and VDAC are regulatory factors of
an unknown channel protein. GSK-3β and mPTP
In addition to Ca2+, reactive oxygen species (ROS), accu- Johaszova et al first reported that GSK-3β activity is a
mulated inorganic phosphate and depletion of ATP promote determinant of the threshold for mPTP opening in car-
opening of mPTPs.35,36 Although the relative importance of diomyocytes.47 They showed that the threshold for ROS-
each factor is not clear, all of these mPTP opening stimuli induced opening of mPTPs is elevated by inhibitory phos-
are induced in cardiomyocytes subjected to long-sustained phorylation of GSK-3β, pharmacological inhibitors of
ischemia and reperfusion. Studies using radio-labeled 2- GSK-3β and knockdown of GSK-3β protein expression
deoxyglucose as a tracer of opened mPTPs indicated that using siRNA. Recently, Gomez et al examined the role of
mPTPs open within 5–10 min after reperfusion but not GSK-3βin mPTP regulation by using the amount of loading
during ischemia.37 This finding is consistent with the notion Ca2+ required to induce irreversible Ca2+ release in isolated
that ROS production and Ca2+ influx upon reperfusion are mitochondria as an index of the threshold for mPTP
triggers of mPTP opening in the reperfused myocardium. opening.48 Postconditioning significantly elevated the thresh-
Because molecules smaller than 1.5 kDa pass through old of mPTP opening in cardiac mitochondria from wild-
mPTPs, irreversible mPTP opening abolishes mitochondrial type mice, but such a protective effect was not detected in
membrane potential, disabling the mitochondria to produce mitochondria from transgenic mice expressing GSK-S9A.
ATP. Without ATP generation, reperfused cardiomyocytes These findings indicate that GSK-3β activity promotes
cannot recover Na+ and Ca2+ homeostasis and thus cannot mPTP opening in response to ROS and/or Ca2+ overloading.
maintain cell viability. Results of experiments using phar- How inactivation of GSK-3β (or its phosphorylation at
macological inhibitors of mPTP and CypD knockout mice Ser9) increases the threshold for mPTP opening remains
generally support the notion that opening of mPTPs is unclear. However, several mechanisms have been suggested
responsible for cardiomyocyte necrosis after ischemia/reper- to date. First, hexokinase II (HK-II) might be preserved
fusion.32–34,38–40 in the mPTP complex by inactivation of GSK-3β.28,49,50
In contrast to its role in cell necrosis, the role of mPTP in Although experiments were conducted by using non-cardiac
apoptosis is still controversial. Earlier studies using non- cells, Pastorino et al showed that activation of GSK-3β
cardiac cells have shown that mPTP opening induces Bax induces release of HK-II from the mitochondria via phos-
translocation to the mitochondria, leading to apoptosis.41,42 phorylation of VDAC, which enhanced susceptibility of the
Furthermore, inhibition of mPTP by pharmacological inhib- cells to necrosis.49 A role of HK-II as a stabilizer of mPTP
itors (cyclosporine A, bongkrekic acid, NIM811) suppressed was also indicated by finding that the inhibitory effect of
apoptosis induced by H2O2 in vitro43–45 and by ischemia/ leukemia inhibitory factor on mPTP opening was sig-
reperfusion in vivo.46 However, susceptibility of CypD nificantly attenuated by dissociation of HK-II from the
deleted cells to apoptosis induced by etoposide, saturosporin, mitochondria by the use of glucose-6-phosphate or HK-II

Circulation Journal Vol.73, July 2009


GSK-3βin Cardioprotection 1187

Figure 3.   Current hypothesis of GSK-3β-


mediated regulation of cell death and survival
in the heart. ANT, adenine nucleotide trans-
locase; ATP, adenosine triphosphate; CypD,
cyclophilin-D; EPO-R, erythropoietin receptor;
ERK, extracellular signal-regulated kinase;
GPCRs, G-protein-coupled receptors; HK-II,
hexokinase-II; HSF-1, heat shock factor-1;
IGF-R, IGF-1 receptor; I-R, insulin receptor;
mATPase, mitochondrial ATPase; MCL-1,
myeloid cell leukemia sequence-1; mPTP,
mitochondrial permeability transition pore;
OMP, outer membrane permeabilization; PI3K,
phosphatidylinositol 3-phosphate kinase; Pi,
inorganic phosphate; PKC, protein kinase C;
PP1, protein phosphatase-1; PP2A, protein
phosphatase-2A; VDAC, voltage-dependent
anion channel.

dissociating peptide.50 protection has been suggested in a wide variety of inter-


Second, our recent study suggests that binding of ventions against infarction. Elevation (or preservation) of
phospho-GSK-3β to ANT suppresses interaction of ANT phospho-Ser9-GSK-3βlevel on reperfusion and anti-infarct
with CypD, a trigger of mPTP opening.51 In isolated rat tolerance have been observed in rat, rabbit or mouse hearts
hearts, reperfusion after sustained ischemia induced trans- treated with ischemic preconditioning,51,55–57 ischemic post-
location of cytosolic GSK-3βto the mitochondria, where it conditioning,48,58 bradykinin,59 opioids,60,61 EPO,57,62 an ade-
formed a complex with ANT and VDAC. Phosphorlylation nosine A1/A2 receptor agonist,63 an A3 receptor agonist,64
of GSK-3βby ischemic preconditioning and erythropoietin PKC-ε activator,65 PKC-δ inhibitor,66 isoflurane,67 lipopoly-
(EPO) receptor activation was dependent on protein kinase saccharide,68 sildenafil,69 xenon70 or resveratrol.71 GSK-3β
C (PKC) and phosphatidiylinositol 3-phosphate kinase phosphorylation by these apparently unrelated interven-
(PI3K), and phospho-GSK-3β was bound to ANT but not tions is explained by the fact that GSK-3β is a substrate
to VDAC at reperfusion (Figure 2A). Interestingly, this of multiple pro-survival protein kinases, including Akt,
phopsho-GSK-3β-ANT interaction was associated with PKC-ε, extracellular signal-regulated kinase (ERK) and
reduction of CypD-ANT binding by 60% compared with protein kinase G, and GSK-3βphosphorylation is therefore
the untreated control group (Figure 2B). Because neither a step to which multiple protective signaling pathways
ANT nor CypD is a putative substrate of GSK-3β3, modifi- converge. Direct inhibition of GSK-3β by the use of struc-
cation of CypD-ANT interaction by phospho-GSK-3β is turally different pharmacological inhibitors (SB-216763,
presumably an indirect one. lithium chloride) administered before either ischemia or
Third, p53-mediated regulation of mPTP might be sup- reperfusion limits infarct size.55,57,60,72,73 It is interesting to
pressed by inhibition of GSK-3β activity. Phosphorylation note that inhibitors of GSK-3βincrease the level of phospho-
of p53 by GSK-3βenhances its functional activity and trans- GSK-3β, which is explained by GSK-3β-mediated positive
location to the nucleus and mitochondria.52 Venkatapuram regulation of protein phosphatase-1 (Figure 3).74 Neverthe-
et al showed that an inhibitor of p53, pifithrin-α, sensitized less, these findings support the notion that inactivation of
the myocardium to isoflurane-induced protection, which is GSK-3β by phosphorylation at Ser9 is a common mecha-
presumably phospho-GSK-3β-mediated.53 This beneficial nism of protection of cardiomyocytes against necrosis in
effect of pifithrin-αwas abolished by an activator of mPTP, many cardioprotective interventions.
atractyloside, indicating that a target of the p53 inhibitor We hypothesized that phospho-Ser9-GSK-3βat the time
was mPTP. of reperfusion determines the extent of reperfusion-induced
As another possible mechanism of protection by GSK-3β myocardial necrosis, because it suppresses mPTP opening.
inactivation, Das et al recently showed that inhibition of To test this possibility, different levels of GSK-3β phos-
GSK-3β suppressed ATP hydrolysis by reducing ATP phorylation were induced in rat hearts in situ by IPC, admin-
transport from the cytosol to the mitochondria.54 This effect istration of EPO, the combination of these two, or inhibitors
was associated with reduced phosphorylation of VDAC, of PKC or PI3K together with IPC-EPO combination before
suggesting modification of ATP transport through VDAC. 20 min ischemia/2 h reperfusion. Levels of phospho-GSK-
Suppression of ATP hydrolysis during ischemia would 3β at 5 min after reperfusion were tightly correlated with
prevent both ATP depletion and accumulation of inorganic infarct sizes (% of ischemic area) 2 h after reperfusion
phosphate, 2 factors promoting mPTP opening. All of the (r=0.809, P<0.05), indicating that approximately 60% of
mechanisms discussed here are not mutually exclusive and infarct size variation is explained by variation in the level
might contribute in concert to mPTP inhibition (Figure 3). of GSK-3β phosphorylation.57 This relationship between
phospho-GSK-3β and infarct size is unlikely to be non-
GSK-3β and Tolerance of Cardiomyocytes specific, because there was no correlation between infarct
size with level of phospho-Ser473-Akt or phospho-Tyr705-
to Necrosis STAT3 on reperfusion.
Involvement of GSK-3β phosphorylation in myocardial However, that role of phospho-GSK-3βin cardiomyocyte

Circulation Journal Vol.73, July 2009


1188 MIURA T et al.

protection might not be equivalent across animal species. To mimicked by pharmacological inhibitors of GSK-3β69,94–98
.
our surprise, protective effects of ischemic preconditioning Transgenic mice that cardio-specifically express dominant-
and postconditioning were not lost in GSK-3α/β knock-in negative GSK-3β showed significantly fewer apoptotic
mice in which Ser21 of GSK-3α and Ser9 of GSK-3β were cardiomyocytes after pressure overloading by aortic con-
changed to Ala.75 Furthermore, pharmacological inhibitors striction.99 These findings indicate that GSK-3β activity
of GSK-3βfailed to limit infarct size in these knock-in mice determines the fate of cardiomyocytes exposed to apoptosis
and also in wild-type mice. In contrast, another study using inducers. However, the intracellular localization of phospho-
transgenic mice showed apparently opposite results. Gomez GSK-3βresponsible for the anti-apoptotic function and the
et al reported that the infarct size-limiting effect of post- mechanism downstream of GSK-3β phosphorylation in
conditioning was lost in mice cardiospecifically expressing cardiomyocytes remain unclear.
GSK-3β-S9A and that SB216763 could mimic postcondi- Recently, we tested the hypothesis that translocation of
tioning in wild-type mice.48 The reasons for this apparent phosphorylated GSK-3β to the mitochondria is important
discrepancy in results of the 2 mouse studies remains for inhibition of oxidant stress-induced apoptosis of car-
unclear, although differences in intracellular distribution of diomyocytes.100 Apoptosis of H9c2 cells was induced by
mutant GSK-3β, function of extra-cardiac GSK-3β and hydrogen peroxide or hypoxia/reoxygenation. EPO and
function of GSK-3αare possibilities. In addition, Skyschally insulin-like growth factor-1 significantly suppressed apop-
et al recently reported that the infarct size-limiting effect of tosis of H9c2 cells, which was associated with activation of
ischemic postconditioning was maintained in pigs in which Akt and phosphorylation of GSK-3β at Ser9. Furthermore,
phosphorylation of Akt, ERK and GSK-3β was inhibited H9c2 cells transfected with S9A were insensitive to EPO-
by inhibitors of PI3K and ERK.76 Collectively, the negative induced protection, and siRNA knockdown of GSK-3βcould
results in mouse and pig studies suggest that role of phospho- mimic the effect of the EPO receptor activation on H2O2-
GSK-3β as a determinant of myocyte tolerance to reperfu- induced apoptosis. However, translocation of GSK-3βto the
sion-induced necrosis may be species dependent. mitochondria after EPO receptor activation was not detected
by tracing EGFP-tagged GSK-3βor by co-immunostaining
Role of GSK-3β in Apoptosis of GSK-3βand mitochondria. Instead, these methods showed
that GSK-3β was phosphorylated within the mitochondria
of Cardiomyocytes after EPO receptor activation in a PI3K-dependent manner.
It is known that GSK-3β has 2 opposite roles in apop- No significant changes were detected for p53, MCL-1 or
totic death of non-cardiac cells depending on the trigger of Bcl-2 after H2O2 challenge or EPO treatments in our proto-
apoptosis. GSK-3β activity restrains pro-apoptotic signal- cols, but translocation of Bax to mitochondria in response
ing from death receptors (ie, TNF-R1, Fas, DR4, DR5).77–80 to oxidant stress was suppressed. These results suggest that
Inhibition of GSK-3βin the cells with activated death recep- phosphorylation of GSK-3β pre-existing in the mitochon-
tors enhanced activation of caspases-8 and -3, Bid cleavage dria by Akt affords protection from oxidant stress-induced
and apoptosis, indicating that GSK-3βsuppresses pro-apop- apoptosis, possibly by suppressing Bax translocation, in car-
totic signals upstream of caspase activation.78–80 Thus, inac- diomyocytes. Phosphorylation of GSK-3βin the mitochon-
tivation of GSK-3β under the condition of stimulation of dria100 is possibly achieved by translocation of Akt to the
death receptors promotes apoptosis. mitochondria.50,62
In contrast, apoptosis induced by intrinsic mechanisms
is facilitated by active GSK-3β. Pro-apoptotic functions of Do We Need a Novel Therapy for Cardiomyocyte
GSK-3β have been shown in apoptosis by withdrawal of
growth factors,81–83 DNA damage,52,84 mitochondrial toxins,85 Protection in the PCI Era?
ischemia,86,87 oxidant stress88 and other conditions that trigger Introduction of reperfusion therapy and its continuous
apoptosis via the mitochondrial pathway. Phosphorylation refinement have improved myocardial salvage in patients
of 4 GSK-3β substrates (p53, heat shock factor-1 [HSF-1], with acute myocardial infarction (AMI) and their progno-
myeloid cell leukemia sequence-1 [MCL-1], Bax) is involved sis. Our recent analysis of data on infarct size and ischemic
in the pro-apoptotic functions. Phosphorylation of p53 in the zone size in the literature indicates that current reperfusion
nucleus leads to p53-mediated transcription of pro-apoptotic therapy salvages more than 50% of the ischemic myocar-
genes52,89 phosphorylation of HSF-1 inhibits its function as dium in approximately half of the patients with AMI.101
a survival-promoting transcription factor,90,91 phosphory- However, there is wide variation of the effect of reperfu-
lation of MCL-1 induces ubiquitinylation and subsequent sion therapy, and the infarct size is larger than 75% of the
degradation of this anti-apoptotic Bcl-2 protein,92,93 and phos- ischemic zone despite successful coronary reperfusion in
phorylation of Bax promotes its localization in the mito- 25% of AMI patients.101 Patients with infarct size larger than
chondria and apoptosis.81 Contribution of the 4 GSK-3βsub- 20% of the left ventricle at the acute phase of AMI were
strates to apoptosis appears different depending on cell types, also approximately 25% of the total patients. Interestingly,
experimental conditions and triggers of apoptosis. the variation of infarct size cannot be explained by symptom-
Although the role of GSK-3βin apoptosis of cardiomyo- to-treatment time alone. Collectively, it is clear that a novel
cytes has not been fully clarified, evidence to date supports and potent therapy is necessary for at least approximately
its significant contribution to apoptosis induced by ischemia/ 25% of AMI patients in order to improve their clinical out-
reperfusion,69,94–96 hypoxia/re-oxygenation,97β-adrenoceptor comes.
activation98 and pressure overload.99 Apoptosis by these Recently, 2 randomized clinical studies, AMISTAD II102
insults was suppressed by overexpression of the adreno- and J-WIND103 showed that administration of adenosine
medullin gene95 or kallikrein gene96 or treatment with and that of human atrial natriuretic peptide, respectively,
statins,97 all of which induced phosphorylation of GSK-3β. significantly reduced infarct size in AMI patients. However,
Furthermore, this protection from apoptosis was inhibited the protection afforded by these pharmacological agents was
by dominant-negative Akt or active GSK-3β mutant and not associated with improvement of prognosis or suppres-

Circulation Journal Vol.73, July 2009


GSK-3βin Cardioprotection 1189

sion of ventricular remodeling after AMI. Possible reasons    2. Cohen P, Goedert M. GSK3 inhibitors: Development and therapeutic
for the apparent discrepancy between infarct size limitation potential. Nat Rev Drug Discov 2004; 3: 479 – 487.
and lack of clinical benefit were extensively discussed in    3. Jope RS, Johnson GV. The glamour and gloom of glycogen syn-
thase kinase-3. Trends Biochem Sci 2004; 29: 95 – 102.
our recent review.101 Nevertheless, it is notable that ‘signifi-    4. Mora A, Sakamoto K, McManus EJ, Alessi DR. Role of the PDK1-
cant’ limitation of infarct size might not always reduce mor- PKB-GSK3 pathway in regulating glycogen synthase and glucose
tality after AMI or its surrogate markers such as ventricular uptake in the heart. FEBS Lett 2005; 579: 3632 – 3638.
dimension.    5. Sugden PH, Fuller SJ, Weiss SC, Clerk A. Glycogen synthase kinase
3 (GSK3) in the heart: A point of integration in hypertrophic signal-
Myocardial necrosis is a major problem not only in coro- ling and a therapeutic target? A critical analysis. Br J Pharmacol
nary artery disease but also in heart failure. Necrosis of car- 2008; 153(Suppl 1): S137 – S153.
diomyocytes, in addition to their apoptosis, has been observed    6. Di Lisa F, Menabò R, Canton M, Petronilli V. The role of mito-
in animal models of heart failure.104 and in end-stage heart chondria in the salvage and the injury of the ischemic myocardium.
Biochim Biophys Acta 1998; 1366: 69 – 78.
failure of patients with dilated cardiomyopathy and aortic    7. Jennings RB, Reimer KA. The cell biology of acute myocardial
stenosis.105,106 Recent studies have shown that plasma tropo- ischemia. Annu Rev Med 1991; 42: 225 – 246.
nin concentration increases during acute severe heart failure    8. Opie LH, Sack MN. Metabolic plasticity and the promotion of
and during exacerbation of chronic heart failure, and their cardiac protection in ischemia and ischemic preconditioning. J Mol
Cell Cardiol 2002; 34: 1077 – 1089.
prognosis correlates with the concentration of troponin.107–112    9. Piper HM, Abdallah Y, Schäfer C. The first minutes of reperfusion:
However, no current therapy directly targets myocardial A window of opportunity for cardioprotection. Cardiovasc Res
necrosis in failing hearts. 2004; 61: 365 – 371.
  10. Inserte J, Gacrcia-Dorado D, Hernando V, Barba I, Soler-Soler J.
Ischemic preconditioning prevents calpain-mediated impairment of
GSK-3β Inhibitors for Clinical Application Na+/K+-ATPase activity during early reperfusion. Cardiovasc Res
2006; 70: 364 – 373.
Lithium chloride is the only GSK-3βinhibitor in clinical   11. Varadarajan SG, An J, Novalija E, Smart SC, Stowe DF. Changes in
use for bipolar mood disorders at the present time.2,3 Find- [Na(+)](i), compartmental [Ca(2+)], and NADH with dysfunction
ings in experiments discussed in the present article indicate after global ischemia in intact hearts. Am J Physiol Heart Circ
Physiol 2001; 280: H280 – H293.
that GSK-3β inhibitors are promising agents for use in   12. Tanno M, Miura T. Protecting ischemic hearts by modulation of SR
adjunctive therapy with PCI for AMI patients. Side effects calcium handling. Cardiovasc Res 2007; 75: 453 – 454.
of the agent for this use could be minimal, because a single   13. Inserte J, Garcia-Dorado D, Ruiz-Meana M, Padilla F, Barrabés JA,
dose administration of a GSK-3β inhibitor would be suffi- Pina P, et al. Effect of inhibition of Na(+)/Ca(2+) exchanger at the
time of myocardial reperfusion on hypercontracture and cell death.
cient to inhibit mPTP opening at the time of reperfusion. Cardiovasc Res 2002; 55: 739 – 748.
However, chronic use of a GSK-3β inhibitor for heart fail-   14. Schlack W, Uebing A, Schäfer M, Bier F, Schäfer S, Piper HM, et al.
ure, for example, could be problematic because its effects Regional contractile blockade at the onset of reperfusion reduces
on ventricular hypertrophy and tumor growth might result infarct size in the dog heart. Pflugers Arch 1994; 428: 134 – 141.
  15. Sebbag L, Verbinski SG, Reimer KA, Jennings RB. Protection of
in adverse outcomes.2,3 Actually such serious side effects ischemic myocardium in dogs using intracoronary 2,3-butanedione
have not been reported for chronic use of lithium chloride. monoxime (BDM). J Mol Cell Cardiol 2003; 35: 165 – 176.
However, more specific and potent inhibitors of GSK-3β   16. Takemura G, Fujiwara H. Morphological aspects of apoptosis in
could have different side effects. heart diseases. J Cell Mol Med 2006; 10: 56 – 75.
Although pre-clinical studies except for one using   17. Ohno M, Takemura G, Ohno A, Misao J, Hayakawa Y, Minatoguchi
S, et al. “Apoptotic” myocytes in infarct area in rabbit hearts may be
mice55,57,60,72,73,75 have shown that inhibitors of GSK-3β oncotic myocytes with DNA fragmentation: Analysis by immuno-
could limit infarct size in animal models of AMI, efficacy gold electron microscopy combined with in situ nick end-labeling.
of the GSK-3βinhibitors has not yet been assessed in human Circulation 1998; 98: 1422 – 1430.
cardiac tissues. However, protection of the human heart was   18. Zhao ZQ, Vinten-Johansen J. Myocardial apoptosis and ischemic
preconditioning. Cardiovasc Res 2002; 55: 438 – 455.
reportedly achieved by an inhibitor of mPTP, cyclosporine   19. Mani K. Programmed cell death in cardiac myocytes: Strategies to
A,110 ischemic preconditioning,113–115 and postconditioning.116 maximize post-ischemic salvage. Heart Fail Rev 2008; 13: 193 – 
If the roles of GSK-3βin mPTP opening, ischemic precon- 209.
ditioning and postconditioning are similar between human,   20. Yaoita H, Ogawa K, Maehara K, Maruyama Y. Attenuation of isch-
emia/reperfusion injury in rats by a caspase inhibitor. Circulation
rat and rabbit hearts, we could expect that GSK-3β inhibi- 1998; 97: 276 – 281.
tors afford significant protection in human hearts as well.   21. Holly TA, Drincic A, Byun Y, Nakamura S, Harris K, Klocke FJ, et
However, a recent preliminary study by Yang et al suggests al. Caspase inhibition reduces myocyte death induced by myocar-
that the extent of myocardial protection by inhibition of dial ischemia and reperfusion in vivo. J Mol Cell Cardiol 1999; 31:
reperfusion injury is much less in the primate than in the rat 1709 – 1715.
  22. Lee P, Sata M, Lefer DJ, Factor SM, Walsh K, Kitsis RN. Fas
and rabbit because ischemic injury is more predominant as a pathway is a critical mediator of cardiac myocyte death and MI
mechanism of myocardial necrosis in this species.117 Never- during ischemia-reperfusion in vivo. Am J Physiol Heart Circ
theless, roles of GSK-3β in the human myocardium and Physiol 2003; 284: H456 – H463.
efficient strategies to directly or indirectly inhibit GSK-3β   23. Leist M, Single B, Naumann H, Fava E, Simon B, Kuhnle S, et al.
Inhibition of mitochondrial ATP generation by nitric oxide switches
for protection of cardiomyocytes warrant further investiga- apoptosis to necrosis. Exp Cell Res 1999; 249: 396 – 401.
tion.   24. Single B, Leist M, Nicotera P. Differential effects of Bcl-2 on cell
death triggered under ATP-depleting conditions. Exp Cell Res 2001;
Acknowledgements 262: 8 – 16.
  25. Leist M, Jaattela M. Four deaths and a funeral: From caspases to
The scientific work of the authors is supported by Grants-in-Aid for Sci- alternative mechanisms. Nature Rev Mol Cell Biol 2001; 2: 1 – 10.
entific Research from the Japanese Society for the Promotion of Science.   26. Rasola A, Bernardi P. The mitochondrial permeability transition
pore and its involvement in cell death and in disease pathogenesis.
References Apoptosis 2007; 12: 815 – 833.
  27. Baines CP. The mitochondrial permeability transition pore and isch-
   1. Jope RS, Yuskaitis CJ, Beurel E. Glycogen synthase kinase-3 emia-reperfusion injury. Basic Res Cardiol 2009; 104: 181 – 188.
(GSK3): Inflammation, diseases, and therapeutics. Neurochem Res   28. Juhaszova M, Wang S, Zorov DB, Nuss HB, Gleichmann M, Mattson
2007; 32: 577 – 595. MP, et al. The identity and regulation of the mitochondrial permea-

Circulation Journal Vol.73, July 2009


1190 MIURA T et al.

bility transition pore: Where the known meets the unknown. Ann N 10554.
Y Acad Sci 2008; 1123: 197 – 212.   50. Miyamoto S, Murphy AN, Brown JH. Akt mediates mitochondrial
  29. Halestrap AP, Clarke SJ, Javadov SA. Mitochondrial permeability protection in cardiomyocytes through phosphorylation of mitochon-
transition pore opening during myocardial reperfusion-a target for drial hexokinase-II. Cell Death Differ 2008; 15: 521 – 529.
cardioprotection. Cardiovasc Res 2004; 61: 372 – 385.   51. Nishihara M, Miura T, Miki T, Tanno M, Yano T, Naitoh K, et al.
  30. Basso E, Fante L, Fowlkes J, Petronilli V, Forte MA, Bernard P. Modulation of the mitochondrial permeability transition pore com-
Properties of the permeability transition pore in mitochondria devoid plex in GSK-3β-mediated myocardial protection. J Mol Cell Cardiol
of cyclophilin D. J Biol Chem 2005; 280: 18558 – 18561. 2007; 43: 564 – 570.
  31. Baines CP, Kaiser RA, Purcell NH, Blair NS, Osinska H, Hambleton   52. Watcharasit P, Bijur GN, Song L, Zhu J, Chen X, Jope RS. Glycogen
MA, et al. Loss of cyclophilin D reveals a critical role for mitochon- synthase kinase-3β (GSK3β) binds to and promotes the actions of
drial permeability transition in cell death. Nature 2005; 434: 658 –  p53. J Biol Chem 2003; 278: 48872 – 48879.
662.   53. Venkatapuram S, Wang C, Krolikowski JG, Weihrauch D, Kersten
  32. Nakagawa T, Shimizu S, Watanabe T, Yamaguchi O, Otsu K, JR, Warltier DC, et al. Inhibition of apoptotic protein p53 lowers the
Yamagata H, et al. Cyclophilin D-dependent mitochondrial permea- threshold of isoflurane-induced cardioprotection during early reper-
bility transition regulates some necrotic but not apoptotic cell death. fusion in rabbits. Anesth Analg 2006; 103: 1400 – 1405.
Nature 2005; 434: 652 – 658.   54. Das S, Wong R, Rajapakse N, Murphy E, Steenbergen C. Glycogen
  33. Kokoszka JE, Waymire KG, Levy SE, Sligh JE, Cai J, Jones DP, et synthase kinase 3 inhibition slows mitochondrial adenine nucleotide
al. The ADP/ATP translocator is not essential for the mitochondrial transport and regulates voltage-dependent anion channel phosphory-
permeability transition pore. Nature 2004; 427: 461 – 465. lation. Circ Res 2008; 103: 983 – 991.
  34. Baines CP, Kaiser RA, Sheiko T, Craigen WJ, Molkentin JD.   55. Tong H, Imahashi K, Steenbergen C, Murphy E. Phosphorylation of
Voltage-dependent anion channels are dispensable for mitochondrial- glycogen synthase kinase-3beta during preconditioning through a
dependent cell death. Nat Cell Biol 2007; 9: 550 – 555. phosphatidylinositol-3-kinase-dependent pathway is cardioprotec-
  35. Kowaltowski AJ, Castilho RF, Grijalba MT, Bechara EJ, Vercesi tive. Circ Res 2002; 90: 377 – 379.
AE. Effect of inorganic phosphate concentration on the nature of   56. Budas GR, Sukhodub A, Alessi DR, Jovanović A. 3’Phosphoinosit-
inner mitochondrial membrane alterations mediated by Ca2+ ions: A ide-dependent kinase-1 is essential for ischemic preconditioning of
proposed model for phosphate-stimulated lipid peroxidation. J Biol the myocardium. FASEB J 2006; 20: 2556 – 2558.
Chem 1996; 271: 2929 – 2934.   57. Nishihara M, Miura T, Miki T, Sakamoto J, Tanno M, Kobayashi H,
  36. Crompton M. Mitochondrial intermembrane junctional complexes et al. Erythropoietin affords additional cardioprotection to precondi-
and their role in cell death. J Physiol 2000; 529(Pt 1): 11 – 21. tioned hearts by enhanced phosphorylation of glycogen synthase
  37. Griffiths EJ, Halestrap AP. Mitochondrial non-specific pores remain kinase-3 beta. Am J Physiol Heart Circ Physiol 2006; 291: H748 – 
closed during cardiac ischaemia, but open upon reperfusion. Biochem H755.
J 1995; 307: 93 – 98.   58. Yin Z, Gao H, Wang H, Li L, Di C, Luan R, et al. Ischemic post-
  38. Clarke SJ, McStay GP, Halestrap AP. Sanglifehrin A acts as a potent conditioning protects both adult and aged Sprague Dawley rat hearts
inhibitor of the mitochondrial permeability transition and reperfu- from ischemia/reperfusion injury through the PI3-K/Akt and GSK-
sion injury of the heart by binding to cyclophilin-D at a different site 3beta pathway. Clin Exp Pharmacol Physiol 2009 (in press).
from cyclosporin A. J Biochem 2002; 277: 34793 – 34799.   59. Park SS, Zhao H, Mueller RA, Xu Z. Bradykinin prevents reperfu-
  39. Khaspekov L, Friberg H, Halestrap A, Viktorov I, Wieloch T. sion injury by targeting mitochondrial permeability transition pore
Cyclosporin A and its nonimmunosuppressive analogue N-Me- through glycogen synthase kinase 3beta. J Mol Cell Cardiol 2006;
Val-4-cyclosporin A mitigate glucose/oxygen deprivation-induced 40: 708 – 716.
damage to rat cultured hippocampal neurons. Eur J Neurosci 1999;   60. Gross ER, Hsu AK, Gross GJ. Opioid-induced cardioprotection
9: 3194 – 3198. occurs via glycogen synthase kinase beta inhibition during reperfu-
  40. Li Y, Johnson N, Capano M, Edwards M, Crompton M. Cyclophilin- sion in intact rat hearts. Circ Res 2004; 94: 960 – 966.
D promotes the mitochondrial permeability transition but has oppo-   61. Gross ER, Hsu AK, Gross GJ. The JAK/STAT pathway is essential
site effects on apoptosis and necrosis. Biochem J 2004; 383: 101 –  for opioid-induced cardioprotection: JAK2 as a mediator of STAT3,
109. Akt, and GSK-3 beta. Am J Physiol Heart Circ Physiol 2006; 291:
  41. De George F, Lartigue L, Bauer MK, Schubert A, Grimm S, Hanson H827 – H834.
GT, et al. The permeability transition pore signals apoptosis by   62. Kobayashi H, Miura T, Ishida H, Miki T, Tanno M, Yano T, et al.
directing Bax translocation and multimerization. FASEB J 2002; 16: Limitation of infarct size by erythropoietin is associated with trans-
607 – 609. location of Akt to the mitochondria after reperfusion. Clin Exp
  42. Precht TA, Phelps RA, Liseman DA, Butts BD, Le SS, Laessig TA, Pharmacol Physiol 2008; 35: 812 – 819.
et al. The permeability transition pore triggers Bax translocation to   63. Förster K, Paul I, Solenkova N, Staudt A, Cohen MV, Downey JM,
mitochondria during neuronal apoptosis. Cell Death Diff 2005; 12: et al. NECA at reperfusion limits infarction and inhibits formation
255 – 265. of the mitochondrial permeability transition pore by activating p70S6
  43. Fatokun AA, Stone TW, Smith RA. Cell death in rat cerebellar kinase. Basic Res Cardiol 2006; 101: 319 – 326.
granule neurons induced by hydrogen peroxide in vitro: Mechanisms   64. Park SS, Zhao H, Jang Y, Mueller RA, Xu Z. N6-(3-iodobenzyl)-
and protection by adenosine receptor ligands. Brain Res 2007; 1132: adenosine-5’-N-methylcarboxamide confers cardioprotection at
193 – 202. reperfusion by inhibiting mitochondrial permeability transition pore
  44. Takeyama N, Miki S, Hirakawa A, Tanaka T. Role of the mitochon- opening via glycogen synthase kinase 3 beta. J Pharmacol Exp Ther
drial permeability transition and cytochrome C release in hydrogen 2006; 318: 124 – 131.
peroxide-induced apoptosis. Exp Cell Res 2002; 274: 16 – 24.   65. Kostyak JC, Hunter JC, Korzick DH. Acute PKCdelta inhibition
  45. Takuma K, Phuagphong P, Lee E, Mori K, Baba A, Matsuda T. limits ischaemia-reperfusion injury in the aged rat heart: Role of
Anti-apoptotic effect of cGMP in cultured astrocytes: Inhibition by GSK-3beta. Cardiovasc Res 2006; 70: 325 – 334.
cGMP-dependent protein kinase of mitochondrial permeable transi-   66. Korzick DH, Kostyak JC, Hunter JC, Saupe KW. Local delivery of
tion pore. J Biol Chem 2001; 276: 48093 – 48099. PKCepsilon-activating peptide mimics ischemic preconditioning in
  46. Argaud L, Gateau-Roesch O, Muntean D, Chalabreysse L, Loufouat aged hearts through GSK-3beta but not F1-ATPase inactivation.
J, Robert D, et al. Specific inhibition of the mitochondrial permeabil- Am J Physiol Heart Circ Physiol 2007; 293: H2056 – H2063.
ity transition prevents lethal reperfusion injury. J Mol Cell Cardiol   67. Feng J, Lucchinetti E, Ahuja P, Pasch T, Perriard JC, Zaugg M.
2005; 38: 367 – 374. Isoflurane postconditioning prevents opening of the mitochondrial
  47. Juhaszova M, Zorov DB, Kim SH, Pepe S, Fu Q, Fishbein KW, et permeability transition pore through inhibition of glycogen synthase
al. Glycogen synthase kinase-3βmediates convergence of protection kinase 3beta. Anesthesiology 2005; 103: 987 – 995.
signaling to inhibit the mitochondrial permeability transition pore.   68. Ha T, Hua F, Liu X, Ma J, McMullen JR, Shioi T, et al. Lipopolysac-
J Clin Invest 2004; 113: 1535 – 1549. charide-induced myocardial protection against ischaemia/reperfusion
  48. Gomez L, Paillard M, Thibault H, Derumeaux G, Ovize M. Inhibi- injury is mediated through a PI3K/Akt-dependent mechanism. Car-
tion of GSK3βby postconditioning is required to prevent opening of diovasc Res 2008; 78: 546 – 553.
the mitochondrial permeability transition pore during reperfusion.   69. Das A, Xi L, Kukreja RC. Protein kinase G-dependent cardiopro-
Circulation 2008; 117: 2761 – 2768. tective mechanism of phosphodiesterase-5 inhibition involves phos-
  49. Pastorino JG, Hoek JB, Shulga N. Activation of glycogen synthase phorylation of ERK and GSK3beta. J Biol Chem 2008; 283:
kinase 3beta disrupts the binding of hexokinase II to mitochondria 29572 – 295785.
by phosphorylating voltage-dependent anion channel and potentiates   70. Mio Y, Shim YH, Richards E, Bosnjak ZJ, Pagel PS, Bienengraeber
chemotherapy-induced cytotoxicity. Cancer Res 2005; 65: 10545 –  M. Xenon preconditioning: The role of prosurvival signaling, mito-

Circulation Journal Vol.73, July 2009


GSK-3βin Cardioprotection 1191

chondrial permeability transition and bioenergetics in rats. Anesth to protection from apoptosis and anchorage-independent growth.
Analg 2009; 108: 858 – 866. Oncogene 2005; 24: 6564 – 6573.
  71. Xi J, Wang H, Mueller RA, Norfleet EA, Xu Z. Mechanism for res-   92. Maurer U, Charvet C, Wagman AS, Dejardin E, Green DR. Glycogen
veratrol-induced cardioprotection against reperfusion injury involves synthase kinase-3 regulates mitochondrial outer membrane permea-
glycogen synthase kinase 3beta and mitochondrial permeability bilization and apoptosis by destabilization of MCL-1. Mol Cell 2006;
transition pore. Eur J Pharmacol 2009; 604: 111 – 116. 21: 749 – 760.
  72. Obame FN, Plin-Mercier C, Assaly R, Zini R, Dubois-Randé JL,   93. Zhao Y, Altman BJ, Coloff JL, Herman CE, Jacobs SR, Wieman HL,
Berdeaux A, et al. Cardioprotective effect of morphine and a blocker et al. Glycogen synthase kinase 3alpha and 3beta mediate a glucose-
of glycogen synthase kinase 3 beta, SB216763 [3-(2,4-dichlorophe- sensitive antiapoptotic signaling pathway to stabilize Mcl-1. Mol
nyl)-4(1-methyl-1H-indol-3-yl)-1H-pyrrole-2,5-dione], via inhibition Cell Biol 2007; 27: 4328 – 4339.
of the mitochondrial permeability transition pore. J Pharmacol Exp   94. Gao HK, Yin Z, Zhou N, Feng XY, Gao F, Wang HC. Glycogen
Ther 2008; 326: 252 – 258. synthase kinase 3 inhibition protects the heart from acute ischemia-
  73. Mozaffari MS, Schaffer SW. Effect of pressure overload on cardio- reperfusion injury via inhibition of inflammation and apoptosis.
protection of mitochondrial KATP channels and GSK-3beta: Interac- J Cardiovasc Pharmacol 2008; 52: 286 – 292.
tion with the MPT pore. Am J Hypertens 2008; 21: 570 – 575.   95. Yin H, Chao L, Chao J. Adrenomedullin protects against myocardial
  74. Zhang F, Phiel CJ, Spece L, Gurvich N, Klein PS. Inhibitory phos- apoptosis after ischemia/reperfusion through activation of Akt-GSK
phorylation of glycogen synthase kinase-3 (GSK-3) in response to signaling. Hypertension 2004; 43: 109 – 116.
lithium: Evidence for autoregulation of GSK-3. J Biol Chem 2003;   96. Yin H, Chao L, Chao J. Kallikrein/kinin protects against myocardial
278: 33067 – 33077. apoptosis after ischemia/reperfusion via Akt-glycogen synthase
  75. Nishino Y, Webb IG, Davidson SM, Ahmed AI, Clark JE, Jacquet S, kinase-3 and Akt-Bad: 14-3-3 signaling pathways. J Biol Chem 2005;
et al. Glycogen synthase kinase-3 inactivation is not required for 280: 8022 – 8030.
ischemic preconditioning or postconditioning in the mouse. Circ Res   97. Bergmann MW, Rechner C, Freund C, Baurand A, El Jamali A,
2008; 103: 307 – 314. Dietz R. Statins inhibit reoxygenation-induced cardiomyocyte apop-
  76. Skyschally A, van Caster P, Boengler K, Gres P, Musiolik J, tosis: Role for glycogen synthase kinase 3beta and transcription
Schilawa D, et al. Ischemic postconditioning in pigs: No causal role factor beta-catenin. J Mol Cell Cardiol 2004; 37: 681 – 690.
for RISK activation. Circ Res 2009; 104: 15 – 18.   98. Menon B, Johnson JN, Ross RS, Singh M, Singh K. Glycogen syn-
  77. Rottmann S, Wang Y, Nasoff M, Deveraux QL, Quon KC. A TRAIL thase kinase-3beta plays a pro-apoptotic role in beta-adrenergic
receptor-dependent synthetic lethal relationship between MYC acti- receptor-stimulated apoptosis in adult rat ventricular myocytes: Role
vation and GSK3β/FBW7 loss of function. Proc Natl Acad Sci USA of beta1 integrins. J Mol Cell Cardiol 2007; 42: 653 – 661.
2005; 102: 15195 – 15200.   99. Hirotani S, Zhai P, Tomita H, Galeotti J, Marquez JP, Gao S, et al.
  78. Song L, Zhou T, Jope RS. Lithium facilitates apoptotic signaling Inhibition of glycogen synthase kinase 3beta during heart failure is
induced by activation of the Fas death domain-containing receptor. protective. Circ Res 2007; 101: 1164 – 1174.
BMC Neurosci 2004; 5: 20 – 27. 100. Ohori K, Miura T, Tanno M, Miki T, Sato T, Ishikawa S, et al. Ser9
  79. Liao X, Zhang L, Thrasher JB, Du J, Li B. Glycogen synthase kinase- phosphorylation of mitochondrial GSK-3beta is a primary mecha-
3β suppression eliminates tumor necrosis factor-related apoptosis- nism of cardiomyocyte protection by erythropoietin against oxidant-
inducing ligand resistance in prostate cancer. Mol Cancer Ther 2003; induced apoptosis. Am J Physiol Heart Circ Physiol 2008; 295:
2: 1215 – 1222. H2079 – H2086.
  80. Schwabe RF, Brenner DA. Role of glycogen synthase kinase-3 in 101. Miura T, Miki T. Limitation of myocardial infarct size in the clinical
TNF-α-induced NF-κB activation and apoptosis in hepatocytes. Am setting: Current status and challenges in translating animal experi-
J Physiol Gastrointest Liver Physiol 2002; 283: G204 – G211. ments into clinical therapy. Basic Res Cardiol 2008; 103: 501 – 513.
  81. Linseman DA, Butts BD, Precht TA, Phelps RA, Le SS, Laessig TA, 102. Kloner RA, Forman MB, Gibbons RJ, Ross AM, Alexander RW,
et al. Glycogen synthase kinase-3βphosphorylates Bax and promotes Stone GW. Impact of time to therapy and reperfusion modality on the
its mitochondrial localization during neuronal apoptosis. J Neurosci efficacy of adenosine in acute myocardial infarction: The AMISTAD-
2004; 24: 9993 – 10002. 2 trial. Eur Heart J 2006; 27: 2400 – 2405.
  82. Cross DA, Culbert AA, Chalmers KA, Facci L, Skaper SD, Reith 103. Kitakaze M, Asakura M, Kim J, Shintani Y, Asanuma H, Hamasaki
AD. Selective small-molecule inhibitors of glycogen synthase kinase- T, et al. Human atrial natriuretic peptide and nicorandil as adjuncts
3 activity protect primary neurones from death. J Neurochem 2001; to reperfusion treatment for acute myocardial infarction (J-WIND):
77: 94 – 102. Two randomised trials. Lancet 2007; 370: 1483 – 1493.
  83. Sinha D, Wang Z, Ruchalski KL, Levine JS, Krishnan S, Lieberthal 104. Nakayama H, Chen X, Baines CP, Klevitsky R, Zhang X, Zhang H,
W, et al. Lithium activates the Wnt and phos-phatidylinositol 3- et al. Ca2+- and mitochondrial-dependent cardiomyocyte necrosis as
kinase Akt signaling pathways to promote cell survival in the absence a primary mediator of heart failure. J Clin Invest 2007; 117: 2431 – 
of soluble survival factors. Am J Physiol Renal Physiol 2005; 288: 2444.
F703 – F713. 105. Hein S, Arnon E, Kostin S, Schönburg M, Elsässer A, Polyakova V,
  84. Tan J, Geng L, Yazlovitskaya EM, Hallahan DE. Protein kinase B/ et al. Progression from compensated hypertrophy to failure in the
Akt-dependent phosphorylation of glycogen synthase kinase-3β in pressure-overloaded human heart: Structural deterioration and com-
irradiated vascular endothelium. Cancer Res 2006; 66: 2320 – 2327. pensatory mechanisms. Circulation 2003; 107: 984 – 991.
  85. King TD, Bijur GN, Jope RS. Caspase-3 activation induced by inhi- 106. Kostin S, Pool L, Elsässer A, Hein S, Drexler HC, Arnon E, et al.
bition of mitochondrial complex I is facilitated by glycogen synthase Myocytes die by multiple mechanisms in failing human hearts. Circ
kinase-3β and attenuated by lithium. Brain Res 2001; 919: 106 –  Res 2003; 92: 715 – 724.
114. 107. Ilva T, Lassus J, Siirilä-Waris K, Melin J, Peuhkurinen K, Pulkki K,
  86. Kaga S, Zhan L, Altaf E, Maulik N. Glycogen synthase-3β/β-catenin et al. Clinical significance of cardiac troponins I and T in acute heart
promotes angiogenic and anti-apoptotic signaling through the induc- failure. Eur J Heart Fail 2008; 10: 772 – 779.
tion of VEGF, Bcl-2 and survivin expression in rat ischemic pre- 108. Latini R, Masson S, Anand IS, Missov E, Carlson M, Vago T, et al.
conditioned myocardium. J Mol Cell Cardiol 2005; 40: 138 – 147. Prognostic value of very low plasma concentrations of troponin T
  87. Brywe KG, Mallard C, Gustavsson M, Hedtjarn M, Leverin AL, in patients with stable chronic heart failure. Circulation 2007; 116:
Wang X, et al. IGF-I neuroprotection in the immature brain after 1242 – 1249.
hypoxia-ischemia, involvement of Akt and GSK3β? Eur J Neurosci 109. Kuwabara Y, Sato Y, Miyamoto T, Taniguchi R, Matsuoka T, Isoda
2005; 21: 1489 – 1502. K, et al. Persistently increased serum concentrations of cardiac tropo-
  88. King TD, Jope RS. Inhibition of GSK3 protects cells from intrinsic nin in patients with acutely decompensated heart failure are predic-
but not extrinsic oxidative stress. Neuroreport 2005; 16: 597 – 601. tive of adverse outcomes. Circ J 2007; 71: 1047 – 1051.
  89. Beurel E, Kornprobst M, Blivet-Van Eggelpoel MJ, Ruiz-Ruiz C, 110. Nishio Y, Sato Y, Taniguchi R, Shizuta S, Doi T, Morimoto T, et
Cadoret A, Capeau J, et al. GSK3β inhibition by lithium confers al. Cardiac troponin T vs other biochemical markers in patients with
resistance to chemotherapy-induced apoptosis through the repression congestive heart failure. Circ J 2007; 71: 631 – 635.
of CD95 (Fas/APO-1) expression. Exp Cell Res 2004; 300: 354 –  111. Setsuta K, Seino Y, Kitahara Y, Arau M, Ohbayashi T, Takano T, et
364. al. Elevated levels of both cardiomyocyte membrane and myofibril
  90. Bijur GN, Jope RS. Opposing actions of phosphatidylinositol 3- damage markers predict adverse outcomes in patients with chronic
kinase and glycogen synthase kinase-3βin the regulation of HSF-1 heart failure. Circ J 2008; 72: 569 – 574.
activity. J Neurochem 2000; 75: 2401 – 2408. 112. Peacock WF 4th, De Marco T, Fonarow GC, Diercks D, Wynne J,
  91. Khaleque MA, Bharti A, Sawyer D, Gong J, Benjamin IJ, Stevenson Apple FS, et al. Cardiac troponin and outcome in acute heart failure.
MA, et al. Induction of heat shock proteins by heregulinβ1 leads N Engl J Med 2008; 358: 2117 – 2126.

Circulation Journal Vol.73, July 2009


1192 MIURA T et al.

113. Piot C, Croisille P, Staat P, Thibault H, Rioufol G, Mewton N, et al. 102: 1487 – 1488.
Effect of cyclosporine on reperfusion injury in acute myocardial 116. Staat P, Rioufol G, Piot C, Cottin Y, Cung TT, L’Huillier I, et al.
infarction. N Engl J Med 2008; 359: 473 – 481. Postconditioning the human heart. Circulation 2005; 112: 2143 – 
114. Rezkalla SH, Kloner RA. Preconditioning in humans. Heart Fail Rev 2148.
2007; 12: 201 – 206. 117. Yang XM, Liu Y, Liu Y, Tandon N, Kambayashi J, Downey JM, et
115. Takagi H, Manabe H, Kawai N, Goto SN, Umemoto T. Review and al. Preconditioning in cynomolgus monkey heart protects against
meta-analysis of randomized controlled clinical trials of remote isch- both ischemic and reperfusion injury. Circulation 2008; 118(Suppl):
emic preconditioning in cardiovascular surgery. Am J Cardiol 2008; S401 – S402.

Circulation Journal Vol.73, July 2009

Das könnte Ihnen auch gefallen