ArticlePDF AvailableLiterature Review

Diabetic Inhibition of Preconditioning- and Postconditioning-Mediated Myocardial Protection against Ischemia/Reperfusion Injury

Authors:

Abstract and Figures

Ischemic preconditioning (IPC) or postconditioning (Ipost) is proved to efficiently prevent ischemia/reperfusion injuries. Mortality of diabetic patients with acute myocardial infarction was found to be 2-6 folds higher than that of non-diabetic patients with same myocardial infarction, which may be in part due to diabetic inhibition of IPC- and Ipost-mediated protective mechanisms. Both IPC- and Ipost-mediated myocardial protection is predominantly mediated by stimulating PI3K/Akt and associated GSK-3β pathway while diabetes-mediated pathogenic effects are found to be mediated by inhibiting PI3K/Akt and associated GSK-3β pathway. Therefore, this review briefly introduced the general features of IPC- and Ipost-mediated myocardial protection and the general pathogenic effects of diabetes on the myocardium. We have collected experimental evidence that indicates the diabetic inhibition of IPC- and Ipost-mediated myocardial protection. Increasing evidence implies that diabetic inhibition of IPC- and Ipost-mediated myocardial protection may be mediated by inhibiting PI3K/Akt and associated GSK-3β pathway. Therefore any strategy to activate PI3K/Akt and associated GSK-3β pathway to release the diabetic inhibition of both IPC and Ipost-mediated myocardial protection may provide the protective effect against ischemia/reperfusion injuries.
Content may be subject to copyright.
Hindawi Publishing Corporation
Experimental Diabetes Research
Volume 2012, Article ID 198048, 9 pages
doi:10.1155/2012/198048
Review A rticle
Diabet ic Inhibition of Preconditioning- and
Postconditioning-Mediated Myocardial Protect ion against
Ischemia/Reperfusion Injury
Xia Yin,
1, 2
Yang Z h e n g ,
1
Xujie Zhai,
3
Xin Zhao,
1
and Lu Cai
1, 2
1
The Cardiovascular Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
2
KCHRI, The Depart ment of Pediatrics, University of Louisville, Louisville, KY 40202, USA
3
Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
Correspondence should be addressed to Yang Zheng, zhengyang@jlu.edu.cn
Received 6 May 2011; Accepted 31 May 2011
Academic Editor: Yingmei Zhang
Copyright © 2012 Xia Yin et al. This is an op en access article distributed under the Creative Commons Attribution License, which
permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Ischemic preconditioning (IPC) or postconditioning (Ipost) is proved to eciently prevent ischemia/reperfusion injuries. Mortal-
ity of diabetic patients with acute myocardial infarction was found to be 2–6 folds higher than that of non-diabetic patients with
same myocardial infarction, which may be in part due to diabetic inhibition of IPC- and Ipost-mediated protective mechanisms.
Both IPC- and Ipost-mediated myocardial protection is predominantly mediated by stimulating PI3K/Akt and associated GSK-3β
pathway while diabetes-mediated pathogenic eects are found to be mediated by inhibiting PI3K/Akt and associated GSK-3β
pathway. Therefore, this review briefly introduced the general features of IPC- and Ipost-mediated myocardial protection and the
general pathogenic eects of diabetes on the myocardium. We have collected experimental evidence that indicates the diabetic
inhibition of IPC- and Ipost-mediated myocardial protection. Increasing evidence implies that diabetic inhibition of IPC- and
Ipost-mediated myocardial protection may be mediated by inhibiting PI3K/Akt and associated GSK-3β pathway. Therefore any
strategy to activate PI3K/Akt and associated GSK-3β pathway to release the diabetic inhibition of both IPC and Ipost-mediated
myocardial protection may provide the protective eect against ischemia/reperfusion injuries.
1. Introduction
Acute myocardial infarction (AMI) is a worldwide problem
that threatens the humans health both in the developed
and developing countries. AMI is often induced by the
complete thrombotic occlusion of coronary arteries at the
site of a ruptured atherosclerotic plaque. Prompt reperfusion
is a definitive treatment to salvage ischemic myocardium
from inevitable death. Experimental and clinical investi-
gations suggest that although reperfusion can salvage the
ischemic myocardium, it can also induce side eect, called
as ischemia/reperfusion injuries. It is appreciated now that
lethal myocardial injury caused by ischemia/reperfusion
accounts for up to 50% of the final infarct size of a myocardial
infarct [1].
Myocardial ischemia/reperfusion injury is a complex
pathophysiological event, resulting in serious acute and
chronic myocardial damage. It is characterized by a cas-
cade of acutely initiated local inflammatory responses,
metabolic disorder, and cell death, leading to myocardial
ultrastructural changes and remodeling and subsequently
myocardial systolic and diastolic dysfunction [24]. Myocar-
dial ischemia/reperfusion injury also induces ventricular
arrhythmias, resulting in circulation collapse and sudden
death [5, 6].
Nu merous studies have demonstr a ted that inflammation
following ischemia/reperfusion injury exacerbates myocar-
dial injury [4, 7]. In addition to inflammation, profound
alterations in myocardial metabolism, such as the disarr ange-
ment of glycolysis and fatty acid oxidation, also significantly
2 Experimental Diabetes Research
impact on the cell integrity and functional recovery of the
myocardium [8]. Evidence from previous studies suggests
that reactive oxygen or nitrogen species (ROS or RNS),
including superoxide radicals, hydrogen peroxide, hydroxyl
radicals, singlet oxygen, nitric oxide, and peroxynitrite
play major contribution to myocardial ischemia/reperfusion
injury [9, 10]. These ROS and/or RNSs, which are formed
within the ischemic myocardial cells and in the first few
moments of reperfusion, are known to be cytotoxic to
surrounding cells. In addition, it is also widely accepted
that apoptotic cell death is involved in the development of
ischemic myocardial damage [11]. Therefore, how to protect
the ischemic myocardium from reperfusion injury is the
key issue for cardiologist and cardiovascular physicians. This
review briefly overviews the status of ischemic precondition-
ing (IPC) and ischemic postconditioning (Ipost) with an
emphasis of the diabetic eects on the myocardial protection
of IPC and Ipost as well as possible mechanisms.
2. Ischemic Preconditioning, Postconditioning,
and Their Myocardial Protective Mechanisms
2.1. Ischemic Preconditioning and Its Myocardial Protection.
Mur ry et al. (1986) first found the potent myocardial pro-
tection by preconditioning the ischemic myocardium when
they gave transient and repeat ischemia and reperfusion
before the occlusion of the coronary artery in dog heart
[12]. They found that multiple brief ischemic episodes
actually protected the myocardium from a subsequent
sustained ischemic insult. They called this prote ctive eect
as IPC (Figure 1). IPC is a well-described adaptive response
by which brief exposure to ischemia/reperfusion before
sustained ischemia markedly enhances the ability of the
myocardium to withstand a subsequent ischemic insult [13].
The protection of IPC is displayed as the reduction of
ischemia/reperfusion-induced infarct size, arrhythmia, and
the improvement of contractile and diastolic function of the
myocardial muscle. Consequently, many studies indicated
that IPC was an endogenous protection for AMI, by induc-
ing phosphatidylinositol 3-kinase (PI3K), protein kinase C
(PKC) and JAK/STAT pathways [12, 1417]. Among these,
the activation of PI3K/protein kinase B (Akt) pathway was
found to play an important role in protecting myocardial
ischemia/reperfusion injury [15, 16, 18]. The PI3K/Akt
pathway aects cell survival by a variety of substrates,
including apoptotic proteins, endothelial nitric oxide syn-
thase (eNOS), and PKC [19, 20 ]. More recent interest
has focused on glycogen synthase kinase-3β (GSK-3β)as
a distal kinase, phosphorylated (and hence inactivated) by
other kinases, including Akt and p42/p44 MAPK/ERK [21,
22]. GSK-3β is a multifunctional Ser/Thr kinase that plays
important roles in necrosis and apoptosis of cardiomyocytes.
GSK-3 activity has been associated with many cell processes,
including the regulation of multiple transcription factors,
the Wnt pathway, nuclear factor κB, endoplasmic reticulum
stress, embryogenesis, apoptosis and cell survival, cell cycle
progression, cell migration, and so on [23, 24].
Preconditioning Occlusion
Reperfusion
Occlusion
Postconditioning
Reperfusion
Figure 1: The illustration of IPC and Ipost. IPC means that
transient and repeat ischemia and reperfusions were given before
the occlusion of the coronary artery. Ipost means that transient and
repeat ischemia and reperfusions were given after the occlusion and
before the reperfusion of coronary artery.
IPC produces myocardial protection by phosphorylating
and consequently inactivating GSK-3β [21]. Howe ver, since
ischemic event is unpredictable and IPC is also invasive,
myocardial protection by IPC is dicult to be used in clinics.
In this review, we do not introduce the detail status of
IPC myocardial protection and possible mechanisms since
these issues have been extensively discussed in a few recent
excellent reviews [2528].
2.2. Ischemic Postconditioning and Its Myocardial Protection.
The Ipost came into notice of Zhao et al. (2003) when
they moved the transient and repeat ischemia/reperfusions to
after the occlusion and before the reperfusion, as illustrated
in Figure 1 [2]. Subsequently, a lot of researchers reported
the same protective eects [29, 30]. They found that cycles
of brief reperfusion and ischemia performed immediately
at the onset of reperfusion following a prolonged ischemic
insult markedly limited reperfusion injury. Like IPC, the
Ipost is also a powerful approach to protect the ischemic
myocardium from reperfusion-induced damage [3133]. In
clinics, with the development of percutaneous coronary
intervention emerged as an exciting innovative treatment
strategy, it makes Ipost possible to intervene AMI. A recent
analysis of data on infarct size and ischemic zone size
indicates that current reperfusion therapy salvages more than
50% of the ischemic myocardium in approximately half of
the patients with AMI [34].
It has been supported by several studies that Ipost
protected the myocardium against the detrimental eects
of lethal myocardial reperfusion injury by limiting oxida-
tive stress, reducing calcium accumulation, maintaining
endothelial function, and reducing inflammation [3537].
Subsequent studies have identified a number of signaling
pathways which are activated by Ipost, and involve in the
myocardial protection of Ipost. Among these pathways,
reperfusion injury salvage kinase (RISK) pathway was the
first sig naling cascade to be linked to Ipost [21], which
showed that Ipost was capable of recruiting prosurvival
signal cascades including PI3K/Akt, PKC, GSK-3β,eNOS,
and guanylyl-cyclase, as disclosed for the mechanisms of IPC
myocardial protection (see the above discussion).
The discovery of IPC and Ipost, including pharmaco-
logical preconditioning and postconditioning, as the two
major forms of endogenously protective mechanisms in the
Experimental Diabetes Research 3
myocardium have encouraged us to explore new ways to
protect the myocardium from ischemia/reperfusion and have
enriched our knowledge of the molecular basis of injury and
survival during ischemia/reperfusion [13]. In both IPC- and
Ipost-mediated myocardial protections, PI3K/Akt activation
is considered as an initial step that induces phosphory-
lation of downstream kinases to inhibit the several pro-
apoptotic factors and mitochondrial permeability transition
pore (mPTP)’s opening at reperfusion, as illustrated in
Figure 2 [23 , 3844]. One of the downstream targets of
the RISK pathway is GSK-3β that plays important roles
in necrosis and apoptosis of cardiomyocytes [23]. GSK-3β
links to the regulation of a variety of cellular functions
including glycogen metabolism, gene expression, and cellular
survival. Experimental studies have demonstrated that the
phosphorylation or inactivation of GSK-3β confers myocar-
dial protective eects through its potential mitochondrial
eects that include the inhibition of mPTP’s opening and
the control of mitochondrial a denine nucleotide transport
through the outer mitochondrial membrane [3537]. The
mPTP is a nonselective large conductance channel in the
mitochondrial inner membrane, which is physiologically
closed. The mPTP remains closed during ischemia but
opens at the onset of reperfusion [45], and modulation
of the mPTP opening at early reperfusion can protect the
myocardium from reperfusion injury [46, 47 ].
Opening of mPTPs is involved in cell death induced by
a variety of causes, including ischemia/reperfusion, alcohol,
endotoxin, and anticancer agents [48]. In addition to
Ca
2+
, ROS and/or RNS-caused accumulation of inorganic
phosphate and depletion of ATP all can open mPTPs [49,
50]. It is also clear that all of these mPTP opening stimuli
are induced in cardiomyocy tes subjected to long-sustained
ischemia/reperfusion. Ipost significantly elevated the thresh-
old of mPTP’s opening in myocardial mitochondria [23].
The inhibition of mPTP’s opening plays a critical end eector
for the myocardial protective eects of Ipost. Juhaszova et
al. [21] first reported that GSK-3β activity is a determinant
of the threshold for mPTP’s opening in cardiomyocytes.
Therefore, GSK-3β plays a critical role in IPC- and Ipost-
mediated myocardial protect ion.
So far, there were two studies that have examined the
role of GSK-3β as an obligatory mediator of Ipost using
transgenic mice and showed dierent results. Gomez et al.
[35] found that mice containing a mutant form of GSK-
3β (which cannot be phosphorylated and inhibited) were
resistant to the myocardial infarct-limiting eects of Ipost
in situ, suggesting that GSK-3β inactivation is required
for Ipost’s myocardial protection. Contrast to the study of
Gomez et al., Nishino et al. [51]havereportedthatmicewith
amutantformofbothGSK-3β and GSK-3α in which the
Akt phosphorylation sites were changed, thereby rendering
them to resistant to inactivation, were still amenable to the
myocardial infarct-limiting eects of both IPC and Ipost.
This study suggests that GSK-3β and GSK-3α inactivation
are not necessary for myocardial protection in these settings.
Therefore, the exact role of GSK-3β in the setting of Ipost
remains further investigation, par ticularly under dierent
conditions.
Preconditioning Postconditioning
Growth factors
G-protein-coupled receptor
PDK1
JAK2
PI3K
STAT3
Akt
p-Akt
GSK-3β
DM
mPTP’s
opening
Cell death
p-GSK-3β
Figure 2: Major signaling pathways of IPC- and Ipost-mediated
protection against cardiac cell death. Myocardial protection of
IPC and Ipost were proposed to be mediated by stimulation of
the prosurvival signaling pathway—PI3K/Akt pathway to inhibit
the GSK-3β activation either via PI3K pathway or JAK2/STAT3
pathway. Diabetes (DM) can inhibit the activation of STAT3 or Akt
to consequently activate GSK-3β that in turn induces mitochondrial
cell death that is the critical cellular event for ischemia/reperfusion-
induced myocardial infarction.
3. Diabetic Inhibition of Ischemic
Preconditioning- and Postconditioning-
Mediated Myocardial Protection against
Ischemia/Reperfusion Injury
Epidemiological data show that diabetes is a major risk
for cardiovascular morbidity and mortality [52, 53]. Coro-
nary artery diseases leading to myocardial infarction and
myocardium failure are one of the major chronic complica-
tions of diabetes, accounting for >75% of hospitalizations in
diabetic patients. The mortality rate of diabetic patients after
AMI is 2–6 folds higher than that of nondiabetic patients
[54, 55]. Increased mortality or increased myocardial injury
following AMI in diabetes is thought probably because of the
high prevalence of other risk factors, that is, hypertension,
hyperlipidemia, and advanced coronary artery diseases [56].
The poor prognosis may b e also in part because of an
increase in the myocardial injury in response to ischemia and
reperfusion [57].
It is well known that insulin regulates metabolism in the
myocardium by modulating glucose transport, glycolysis,
glycogen synthesis, lipid metabolism, protein synthesis,
4 Experimental Diabetes Research
growth, contractility, and apoptosis in cardiomyocytes [58,
59]. Myocardial insulin resistance develops in animal models
of both type 1 and type 2 diabetes [59]. These insulin-
stimulated eects have been shown to be reduced in the
myocardium and cardiomyocytes of diabetic rats [60], which
may be the main reason for the increase in myocardial
injury in response to ischemia and reperfusion in diabetic
subjects.
In normal physiological status, insulin can regulate the
metabolism of glucose through PI3K/Akt pathway. Insulin
binds to its receptor and phosphorylates insulin receptor’s
substrates (IRS) such as IRS protein 1–4, Shc, Grb-2 asso-
ciated binder-1, and APS adapter protein. These substrates
have the SH2 structural domain and can provide the orienta-
tion sites for other signaling protein molecules, including the
downstream signaling molecules of PI3K [61, 62]. Activated
PI3K can phosphorylate the PI’s substrates specifically to
produce PIP2 and PIP3. PIP1 and PIP2 can translocate the
PI3K-dependent kinase (PDK1) and Akt from the cytoplasm
to plasma membrane. Under these conditions, Akt can be
phosphorylated at Thr308 and Ser473, and the activated Akt
then phosphorylates GSK-3β. The phosphorylation of GSK-
3β inactivates its ac tivity, which will release its inhibition
of the synthesis of glycogen, as shown in Figure 2.The
activity of GSK-3β is two-fold higher in diabetes than that of
nondiabetes. Hyperglycemia and hyperinsulinemia can both
activate the GSK-3β [43, 44, 63]. The activated GSK-3β can
inhibit the myocardial transduction of insulin signaling and
the utilization of glucose through the phosphorylation of
IRS-1.
We have recently reported for the first time that the
activation of GSK-3β played the pivotal role in diabetes-
induced energy disarrangement a nd consequently patholog-
ical remodeling in the myocardium [63]. This study suggests
that the activation of GSK-3β plays an important role in the
development of diabetic cardiomyopathy.
Diabetes is an independent risk factor for ischemic
myocardium disease; therefore, whether diabetes could
decrease the IPC and/or Ipost protection against ischemia/
reperfusion-induced myocardial damage has been ques-
tioned. Tosaki et al. found that IPC did not aord protection
against ischemic damage in diabetic subjects [76]. Other
studies also showed that STZ-induced diabetes signifi-
cantly aggravated myocardial ischemia/reperfusion injury
and blunted the protective eects of IPC [77, 78]. However,
whether diabetes abrogates IPC- or Ipost-mediated myocar-
dial protection depends on IPC times or the periods of dia-
betes. For instance, Tsang et al. [15] discovered that in nor-
mal Wistar rats, one, two, and three cycles of IPC significantly
reduced infarct size induced by ischemia/reperfusion; how-
ever, in diabetic Goto-Kakizaki (GK) rats, only three cycles
of IPC reduced infarct size induced by ischemia/reperfusion,
compared with GK control hearts. Both one and two cycles
ofIPCfailedtoaord reductive eect on the infarct,
suggesting that the diabetic heart has a high threshed to IPC
stimulus-induced myocardial protection. In addition, Shi-
Ting et al. [79] also showed that mice with diabetes for 4
weeks showed a tolerance to ischemia/reperfusion-induced
damage as compared to normal rats; IPC of these diabetic
mice remained aording partial myocardial protection. In
contrast, mice with diabetes for 8 weeks showed a low
tolerance to ischemia/reperfusion damage as compared to
normal mice, and the IPC-induced myocardial protection
was not evident. These findings suggest that shor t-term dia-
betes makes the myocardium more tolerant, like an adaptive
response, but long-term diabetes makes the myocardium
more susceptible to ischemia/reperfusion-induced damage,
like a decompensated response.
Recently, Przyklenk et al. [80] have assessed the con-
sequences of a major risk factor—diabetes on the infarct-
sparing eect of stuttered reflow using type 1 and type 2 dia-
betic mouse models. They gave the isolated bu
er-perfused
myocardium for 30 min ischemia, and the myocardium
received either standard reperfusion or three to six 10s cycles
of stuttered reflow as Ipost. They found that Ipost-reduced
infarct size via upregulation of extracellular signal-regulated
kinase 1/2 (ERK1/2) in nor moglycemic mice, but diabetic
myocardium was refractory to Ipost-induced cardioprotec-
tion. They also found that in the type-1 diabetic model,
Ipost’s protective eects were reversed by the restoration
of normoglycemia. Therefore, this study provided strong
evidence for a profound, but potentially reversible, defect
induced by diabetes in the myocardial protection of Ipost.
In a study by Drenger et al. [81], however, the protective
eects of Ipost were found to be inhibited in the diabetes rats,
and the diabetic inhibition of Ipost’s myocardial protection
was not relieved by insulin-induced normoglycemia. The
discrepancy between these two studies may be also due to
hyperglycemic times; as the hyperglycemic time increases,
the inhibited protective function of Ipost by diabetes may
become irreversible.
As the myocardial protection of IPC and Ipost is medi-
ated by a number of signaling pathways, the blunted myocar-
dial protection mediated by IPC in diabetes may be related to
the impairment in myocardial protective signaling pathways
such as the PI3K/Akt pathway, as illustrated in Figure 2 [2,
15, 77]. Since signal tr ansducer and activator of transcription
(STAT) 3-mediated signaling pathway has been found to
play an important role in the cardiac protection induced
by IPC [14]andIpost[69]. Downregulation of STAT3
was found to be a causative of abolishment of the cardiac
protection mediated by IPC [17]andIpost[8183 ]under
several conditions. Therefore, STAT3 downregulation may
be one of the mechanisms for diabetic inhibition of Ipost-
mediated cardiac protection, as discussed by Drenger et al.
[81]. Reportedly erythropoietin (EPO) has an IPC-like eect
to show myocardial protection against ischemia/reperfusion-
induced damage [73, 74]. However, Ghaboura et al. have
shown the attenuation of EPO-mediated myocardial protec-
tion under diabetic condition [43].
4. Diabetic Activation of GSK-3β Plays a Critical
Role in Diminishing IPC- and Ipost-Mediated
Myocardial Protective Function
In the above sections, we mentioned that there are several
signaling pathways that may involve in the myocardial
Experimental Diabetes Research 5
Table 1: Potential candidates that may have protective eect against ischemia reperfusion injury related with Akt/GSK-3β pathway.
Potential candidates Target of signaling pathway Reference
Lithium chloride GSK-3β inhibitor [44]
Indirubin-3 monooxime GSK-3β inhibitor [44]
SB216763 GSK-3β inhibitor [44]
Zinc Inactivation of GSK-3β directly or indirectly [42, 6467]
Adenosine Activation/translocation of PKC, PI3K, and MAPK [68]
Endogenous opioids JAK-STAT pathway and then inactivation of GSK-3β [6972]
Erythropoietin Activate Akt and inhibit GSK-3β [43, 73, 74]
Sevoflurane Phosphor ylates Akt and then GSK-3 β [75]
Except for the GSK-3β inhibitors, most of other potential candidates may exert their protective eect against ischemia reperfusion injury through activation
of Akt and then inactivation of GSK-3β.
protection mediated by IPC or Ipost. As shown in Figure 2,
however, inactivation of GSK-3β has been considered as the
pivotal step for both IPC and Ipost’s myocardial protec-
tion. Furthermore, studies have demonstrated that diabetes-
induced activation of GSK-3β and impairment of RISK
play critical roles in diabetes-induced myocardial oxidative
damage and remodeling [43, 63]; other studies also reported
that the activity of GSK-3β is twice in diabetic patients
compared to that of nondiabetic patients [84]. Therefore,
whether diabetic activation of GSK-3β blunts IPC and Ipost’s
myocardial protection really needs to be investigated [83, 85,
86].
To date, studies have demonstrated a decreased protective
eectofIPConAMIindiabeticsubjects[43, 44, 83, 85
87]. It is clear that IPC produces myocardial protection
by phosphorylation of GSK-3β that inhibited the opening
of mPTP, but the activity of GSK-3β was found to be
elevated during diabetes [21, 23, 35, 63]. Yadav et al.
[44] investigated the role of GSK-3β in attenuating the
cardioprotective eect of IPC using a ty pe-1diabetic rat
model. They found that IPC had protective eect on
normal rat myocardium, but this cardioprotective eect
of IPC was significantly attenuated in diabetic rat. At the
same time, they found that GSK-3β inhibitors, including
lithium chloride, indirubin-3 monooxime, and SB216763,
significantly reduced the myocardial damage and decreased
infarct size in diabetic rat myocardium. This study suggests
that diabetes-induced attenuation of myocardial protection
mediated by IPC involves in the activation of GSK-3β.In
addition, Ghaboura et al. [43] also demonstrated that the
attenuation of EPO-mediated myocardial protection from
ischemia/reperfusion under diabetic condition was related
to the decrease in EPO-stimulated GSK-3β phosphorylation.
The administration of GSK-3β inhibitor SB216763 protected
the hearts from ischemia/reperfusion-induced damage in
control and diabetic g roups [43]. Therefore, the inhibition
of IPC myocardial protection in the diabetes is most likely
related to the activation of GSK-3β [43, 44].
Because Ipost and IPC share some common signal
transduction cascades proposed above (Figure 2
), which
include the activation of survival protein kinase pathways
[13]. In the study by Drenger et al. [81], they demonstrated
that diabetes can impair the protective eect of Ipost on
myocardial damage or infarction through inhibition of STAT
3-mediated PI3K/Akt pathways. Up to now, there remains
no proof to indicate that diabetes can inhibit the protective
eect of Ipost on the myocardium; therefore, it remains to be
further explored.
5. Is It Possible to Prevent
the Diabeti c Inhibition of IPC or
Ipost Myocardial Protection against
Ischemia/Reperfusion Injury?
We have demonstrated that diabetes-induced myocardial
oxidative damage and inflammation mainly due to the
activation of GSK-3β. When we inactivated GSK-3β activ-
ity with its inactivator in diabetic mice, diabetes-induced
myocardial damage were almost completely prevented [63].
In addition, we have discussed above that inactivation
of GSK-3β w ith its specific inactivators can also directly
aord the myocardial protection in diabetic animals treated
with GSK-3β inactivators [43, 44]. Therefore, any reagents
that can inactivate GSK-3β may have the potential to be
applied for the prevention of diabetic inhibition of IPC-
and/or Ipost-mediated myocardial protection. Except for the
consideration of GSK-3β inhibitors as discussed above and
also listed in the Table 1, the following reagents (Table 1)may
also have such potential.
Zinc (Zn) is an interesting candidate because Zn is an
important trace element found in most body tissues as
bivalent cations and has essential roles in human health.
Zn has also an insulin-like function that was found also
to be related to its inactivation of GSK-3β [88]. We have
demonstrated that Zn supplementation to diabetic mice
could significantly prevent the development of myocardial
oxidative damage, remodeling, and dysfunction in these
diabetic mice [ 64]. Although we did not explore whether
the myocardial protection by Zn supplementation in these
diabetic mice is mediated by the inactivation of GSK-3β
by supplied Zn, other studies have reported that Zn also
inactivated GSK-3β in several conditions. In the experiment
from Chanoit et al. [42], for instance, they found that the
treatment of myocardial H9c2 cells with ZnCl
2
(10 μM) for
20 min significantly enhanced GSK-3β phosphorylation at
6 Experimental Diabetes Research
Ser9, indicating that exogenous Zn can inactivate GSK-3β in
H9c2 ce lls. Other experiments [41] also demonstrated that
Zn also increased mitochondrial GSK-3β phosphorylation.
This may indicate an involvement of the mitochondria in the
action of Zn.
Zn applied at reperfusion period reduced cell death in
the cells subjected to ischemia/reperfusion, which confirmed
thatZnmayactasaninactivatorofGSK-3β to provide a
myocardial protection at reperfusion [41, 42, 89]. Besides
the direct inactivation of GSK-3β, Zn was also reported to
stimulate Akt phosphorylation by inhibiting Akt negative
regulators, including phosphatase and tensin homologue on
chromosome 10 (PTEN) and protein tyrosine phosphatase
1B (PTP1B) [6567]. Inactivation of PTEN and/or PTP1B
may also contribute to Zns inac tivation of GSK-3β via Akt
activation [41]. Therefore, Zn may inhibit GSK-3β by direct
and indirect mechanisms to protect the myocardium from
diabetic activation of GSK-3β-mediated pathogenic eects.
In addition to Zn protective eects, other substrates
are also reported to exert their protective eect, as IPC
and Ipost, on ischemia/reperfusion-induced cardiac damage.
For instance, adenosine leads to the activation and/or
translocation of PKC, PI3K, and mitogen-activated protein
kinase (MAPK) and, subsequently, aords IPC- or Ipost-
like myocardial protection at the level of mitochondrial
targets [68]. Endogenous opioids have also been documented
to be involved in protective eects of Ipost [69, 70].
The administration of EPO at the time of reperfusion
aorded a beneficial eect on Ipost myocardial prote ction
in rabbits [90]andmice[73]. EPO administration just
prior to reperfusion has reduced infarct size in isolated rat
and dog hearts, and even in canine hearts. Furthermore,
EPO administration even 5 min after the reperfusion has
also provided protective responses [74, 91]. Lamont et al.
reported that both melatonin and resveratrol, as found in
red wine, protected the myocardium in an experimental
model from myocardial infarction via the survivor activating
factor enhancement pathway [92]. Fang et al. demonstrate
that sevoflurane administered immediately during early
reperfusion prevented the myocardial infarction [75].
Although all these substances can aord myocardial
protective eects on ischemia/reperfusion in the models
without diabetes, whether these substances can modify
diabetic individuals to maintain the myocardial protection
of IPC and Ipost remains to be explored in the future
studies.
6. Conclusion
Epidemiological data show that diabetes is a major risk
for cardiovascular diseases and the mortality of diabetic
patients with acute myocardial infarction is 2–6 folds higher
than that of nondiabetic patients with the same myocardial
infarction. The poor prognosis may be at least in part because
of diabetic inhibition of IPC- and Ipost-mediated pro-
tective mechanisms against ischemia/reperfusion injuries.
Emerging evidence indicates that both IPC- and Ipost-
mediated myocardial protection pre dominantly be mediated
by stimulating PI3K/Akt and associated GSK-3β pathway
while diabetes-mediated pathogenic eects are found to be
mediated by inhibiting PI3K/Akt and associated GSK-3β
pathway. Therefore, diabetic inhibition of IPC- and Ipost-
mediated myocardial protection may be mediated by the
activation of GSK-3β pathway, which suggests a possibility
that we may activate PI3K/Akt indirectly to inactivate GSK-
3β pathway or use GSK-3β inactivator directly to inactive
GSK-3β pathway to preserve IPC- and/or Ipost-mediated
myocardial protection under diabetic conditions. Although
there is not enough experimental and epidemiological
evidence to support our assumption, it was worthy to be
explored in the future studies.
Acknowledgment
The studies cited from the authors laboratories in the article
were supported in part by Basic Science Award from ADA
(01-11-BS-17 to LC), and a 50101 project from the First
Hospital of Jilin University (To LC).
References
[1] D. M. Yellon and D. J. Hausenloy, “Myocardial reperfusion
injury, The New England Journal of Medicine, vol. 357, no. 11,
pp. 1074–1135, 2007.
[2] Z. Q. Zhao, J. S. Corvera, M. E. Halkos et al., “Inhibition of
myocardial injury by ischemic postconditioning during reper-
fusion: comparison with ischemic preconditioning, American
Journal of Physiology, vol. 285, no. 2, pp. H579–H588, 2003.
[3]K.Matsumura,R.W.Jeremy,J.Schaper,andL.C.Becker,
“Progression of myocardial necrosis during reperfusion of
ischemic myocardium, Circulation, vol. 97, no. 8, pp. 795–
804, 1998.
[4] N. G. Frangogiannis, C. W. Smith, and M. L. Entman, The
inflammatory response in myocardial infarction, Cardiovas-
cular Research, vol. 53, no. 1, pp. 31–47, 2002.
[5] F.F.Gao,S.Y.Hao,Z.Q.Huangetal.,“Cardiacelectrophys-
iological and antiarrhythmic eects of N-n-butyl haloperidol
iodide, Cellular Physiolog y and Biochemistry,vol.25,no.4-5,
pp. 433–442, 2010.
[6] A.L.Moens,M.J.Claeys,J.P.Timmermans,andC.J.Vrints,
“Myocardial ischemia/reperfusion-injury, a clinical view on a
complex pathophysiological process, International Journal of
Cardiology, vol. 100, no. 2, pp. 179–190, 2005.
[7] J. E. Jordan, Z. Q. Zhao, and J. Vinten-Johansen, “The role
of neutrophils in myocardial ischemia-reperfusion injury,
Cardiovascular Research, vol. 43, no. 4, pp. 860–878, 1999.
[8] N. Sambandam and G. D. Lopaschuk, AMP-activated protein
kinase (AMPK) control of fatty acid and glucose metabolism
in the ischemic heart, Progress in Lipid Research, vol. 42, no.
3, pp. 238–256, 2003.
[9] J. L. Park and B. R. Lucchesi, “Mechanisms of myocardial
reperfusion injury, Annals of Thoracic Surgery,vol.68,no.5,
pp. 1905–1912, 1999.
[10] G. M. C. Rosano, M. Fini, G. Caminiti, and G. Barbaro,
“Cardiac metabolism in myocardial ischemia, Current Phar-
maceutical Design, vol. 14, no. 25, pp. 2551–2562, 2008.
[11] J. W. Ding, X. H. Tong, J. Yang et al., Activated protein C pro-
tects myocardium via activation of anti-apoptotic pathways of
survival in ischemia-reperfused rat heart, Journal of Korean
Medical Science, vol. 25, no. 11, pp. 1609–1615, 2010.
Experimental Diabetes Research 7
[12] C. E. Murry, R. B. Jennings, and K. A. Reimer, “Precondi-
tioning with ischemia: a delay of lethal cell injury in ischemic
myocardium, Circulation, vol. 74, no. 5, pp. 1124–1136, 1986.
[13] P. Ferdinandy, R. Schulz, and G. F. Baxter, “Interaction of car-
diovascular risk factors with myocardial ischemia/reperfusion
injury, preconditioning, and postconditioning, Pharmacolog-
ical Reviews, vol. 59, no. 4, pp. 418–458, 2007.
[14] E. R. Gross, A. K. Hsu, and G. J. Gross, “The JAK/STAT
pathway is essential for opioid-induced cardioprotection:
JAK2 as a mediator of STAT3, Akt, and GSK-3β,” American
Journal of Physiology, vol. 291, no. 2, pp. H827–H834, 2006.
[15] A. Tsang, D. J. Hausenloy, M. M. Mocanu, R. D. Carr, and D.
M. Yellon, “Preconditioning the diabetic heart: the importance
of Akt phosphorylation, Diabetes, vol. 54, no. 8, pp. 2360–
2364, 2005.
[16] D. J. Hausenloy and D. M. Yellon, “Survival kinases in
ischemic preconditioning and postconditioning, Cardiovas-
cular Research, vol. 70, no. 2, pp. 240–253, 2006.
[17] K. Boengler, A. Buechert, Y. Heinen et al., “Cardioprotection
by ischemic postconditioning is lost in aged and STAT3-
deficient mice, Circulation Research, vol. 102, no. 1, pp. 131–
135, 2008.
[18] S. V. Penumathsa, M. Thirunavukkarasu, S. M. Samuel et
al., “Niacin bound chromium treatment induces myocardial
Glut-4 translocation and caveolar interaction via Akt, AMPK
and eNOS phosphorylation in streptozotocin induced diabetic
rats after ischemia-reperfusion injury, Biochimica et Biophys-
ica Acta, vol. 1792, no. 1, pp. 39–48, 2009.
[19] F. Gao, E. Gao, T. L. Yue et al., “Nitric oxide mediates
the antiapoptotic eect of insulin in myocardial ischemia-
reperfusion: the roles of PI3-kinase, Akt, and endothelial nitric
oxide synthase phosphorylation, Circulation, vol. 105, no. 12,
pp. 1497–1502, 2002.
[20] D. J. Hausenloy and D. M. Yellon, “New directions for
protecting the heart against ischaemia-reperfusion injury:
targeting the Reperfusion Injury Salvage Kinase (RISK)-
pathway, Cardiovascular Research, vol. 61, no. 3, pp. 448–460,
2004.
[21] M. Juhaszova, D. B. Zorov, S. H. Kim et al., “Glycogen synthase
kinase-3β mediates convergence of protection signalling to
inhibit the mitochondrial permeability transition pore, Jour-
nal of Clinical Investigation, vol. 113, no. 11, pp. 1535–1549,
2004.
[22] H. Tong, K. Imahashi, C. Steenbergen, and E. Murphy,
“Phosphor ylation of glycogen synthase kinase-3β during
preconditioning through a phosphatidylinositol-3-kinase—
dependent pathway is cardioprotective, Circulation Research,
vol. 90, no. 4, pp. 377–379, 2002.
[23] T. Miura and T. Miki, “GSK-3β, a therapeutic target for
cardiomyocyte protection, Circulation Journal, vol. 73, no. 7,
pp. 1184–1192, 2009.
[24] M. Juhaszova, D. B. Zorov, Y. Yaniv, H. B. Nuss, S. Wang,
and S. J. Sollott, “Role of glycogen synthase kinase-3β in
cardioprotection, Circulation Research, vol. 104, no. 11, pp.
1240–1252, 2009.
[25] X. Yang, M. V. Cohen, and J. M. Downe y, “Mechanism of
cardioprotection by early ischemic preconditioning, Cardio-
vascular Drugs and Therapy, vol. 24, no. 3, pp. 225–234, 2010.
[26] T. Miura, T. Miki, and T. Yano, “Role of the gap junction in
ischemic preconditioning in the hear t, American Journal of
Physiology, vol. 298, no. 4, pp. H1115–H1125, 2010.
[27] C. Penna, D. Mancardi, R. Rastaldo, and P. Pagliaro, “Cardio-
protection: a radical view. Free radicals in pre and postcon-
ditioning, Biochimica et Biophysica Acta, vol. 1787, no. 7, pp.
781–793, 2009.
[28] M. Das and D. K. Das, “Molecular mechanism of precondi-
tioning, IUBMB Life, vol. 60, no. 4, pp. 199–203, 2008.
[29] D. M. Yellon and L. H. Opie, “Postconditioning for protection
of the infarcting heart, The Lancet, vol. 367, no. 9509, pp. 456–
458, 2006.
[30] H. Kin, Z. Q. Zhao, H. Y. Sun et al., “Postconditioning atten-
uates myocardial ischemia-reperfusion injury by inhibiting
events in the early minutes of reperfusion, Cardiovascular
Research, vol. 62, no. 1, pp. 74–85, 2004.
[31] S. Philipp, X. M. Yang, L. Cui, A. M. Davis, J. M. Downey,
and M. V. Cohen, “Postconditioning protects rabbit hearts
through a protein kinase C-adenosine A2b receptor cascade,
Cardiovascular Research, vol. 70, no. 2, pp. 308–314, 2006.
[32] C. Penna, S. Cappello, D. Mancardi et al., “Post-conditioning
reduces infarct size in the isolated rat heart: role of coronary
flow and pressure and the nitric oxide/cGMP pathway, Basic
Research in Cardiology, vol. 101, no. 2, pp. 168–179, 2006.
[33] L. Argaud, O. Gateau-Roesch, L. Augeul et al., “Increased
mitochondrial calcium coexists with decreased reperfusion
injur y in postconditioned (but not preconditioned) hearts,
American Journal of Physiology, vol. 294, no. 1, pp. H386–
H391, 2008.
[34] T. Miura and T. Miki, “Limitation of myocardial infarct size in
the clinical setting: current status and challenges in translating
animal experiments into clinical therapy, Basic Research in
Cardiology, vol. 103, no. 6, pp. 501–513, 2008.
[35] L. Gomez, M. Paillard, H. Thibault, G. Derumeaux, and M.
Ovize, “Inhibition of GSK3β by postconditioning is required
to prevent opening of the mitochondrial permeability transi-
tion pore during reperfusion, Circulation, vol. 117, no. 21, pp.
2761–2768, 2008.
[36] A. Tsang, D. J. Hausenloy, M. M. Mocanu, and D. M. Yellon,
“Postconditioning: a form of “modified reperfusion protects
the myocardium by activating the phosphatidylinositol 3-
kinase-Akt pathway, Circulation Research,vol.95,no.3,pp.
230–232, 2004.
[37]S.A.Javador,S.Clarke,M.Das,E.J.Griths, K. H. H.
Lim, and A. P. Halestrap, “Ischaemic preconditioning inhibits
opening of mitochondrial permeability transition pores in the
reperfused rat heart, Journal of Physiology, vol. 549, no. 2, pp.
513–524, 2003.
[38] M. S. Mozaari, J. Y. Liu, and S. W. Schaer, Eect of pressure
overload on cardioprotection via PI3K-Akt: comparison of
postconditioning, insulin, and pressure unloading, American
Journal of Hypertension, vol. 23, no. 6, pp. 668–674, 2010.
[39] J. C. Bopassa, R. Ferrera, O. Gateau-Roesch, E. Couture-
Lepetit, and M. Ovize, “PI 3-kinase regulates the mitochon-
drial transition pore in controlled reperfusion and postcondi-
tioning, Cardiovascular Research, vol. 69, no. 1, pp. 178–185,
2006.
[40] E. Murphy and C. Steenbergen, “Preconditioning: the mito-
chondrial connection, Annual Review of Physiology, vol. 69,
pp. 51–67, 2007.
[41] S. Lee, G. Chanoit, R. McIntosh, D. A. Zvara, and Z. Xu,
“Molecular mechanism underlying Akt activation in zinc-
induced cardioprotection, American Journal of Physiology,
vol. 297, no. 2, pp. H569–H575, 2009.
8 Experimental Diabetes Research
[42] G. Chanoit, S. Lee, J. Xi et al., “Exogenous zinc protects cardiac
cells from reperfusion injury by targeting mitochondrial
permeability transition pore through inactivation of glycogen
synthase kinase-3β,” American Journal of Physiology, vol. 295,
no. 3, pp. H1227–H1233, 2008.
[43] N. Ghaboura, S. Tamareille, P.-H. Ducluzeau et al., “Diabetes
mellitus abrogates erythropoietin-induced cardioprotection
against ischemic-reperfusion injury by alteration of the
RISK/GSK-3β signaling, Basic Research in Cardiology, vol.
106, no. 1, pp. 147–162, 2011.
[44] H. N. Yadav, M. Singh, and P. L. Sharma, “Involvement
of GSK-3β in attenuation of the cardioprotective eect of
ischemic preconditioning in diabetic rat heart, Molecular and
Cellular Biochemistry, vol. 343, no. 1-2, pp. 75–81, 2010.
[45] E. J. Griths and A. P. Halestrap, “Mitochondrial non-specific
pores remain closed during cardiac ischaemia, but open upon
reperfusion, Biochemical Journal, vol. 307, no. 1, pp. 93–98,
1995.
[46] L. Argaud, O. Gateau-Roesch, D. Muntean et al., “Specific
inhibition of t he mitochondrial permeabilit y transition pre-
vents lethal reperfusion injury, Journal of Molecular and
Cellular Cardiology, vol. 38, no. 2, pp. 367–374, 2005.
[47] L. Argaud, O. Gateau-Roesch, O. Raisky, J. Loufouat, D.
Robert, and M. Ovize, “Postconditioning inhibits mitochon-
drial permeability transition, Circulation, vol. 111, no. 2, pp.
194–197, 2005.
[48] A. Rasola and P. Bernardi, The mitochondrial permeability
transition pore and its involvement in cell death and in disease
pathogenesis, Apoptosis, vol. 12, no. 5, pp. 815–833, 2007.
[49]A.J.Kowaltowski,R.F.Castilho,M.T.Grijalba,E.J.H.
Bechara, and A. E. Vercesi, “Eect of inorganic phosphate
concentration on the nature of inner mitochondrial mem-
brane alterations mediated by Ca
2+
ions: a proposed model for
phosphate-stimulated lipid peroxidation, Journal of Biolog ical
Chemistry, vol. 271, no. 6, pp. 2929–2934, 1996.
[50] M. Crompton, “Mitochondrial intermembrane junctional
complexes and their role in cell death, Journal of Physiology,
vol. 529, no. 1, pp. 11–21, 2000.
[51] Y. Nishino, I. G. Webb, S. M. Davidson et al., “Glycogen
synthase kinase-3 inactivation is not required for ischemic
preconditioning or postconditioning in the mouse, Circula-
tion Research, vol. 103, no. 3, pp. 307–314, 2008.
[52] S. M. Donahoe, G. C. Stewart, C. H. McCabe et al., “Diabetes
and mortality following acute coronary syndromes, Journal of
the American Medical Association, vol. 298, no. 7, pp. 765–775,
2007.
[53] S. Boudina and E. D. Abel, “Diabetic cardiomyopathy revis-
ited, Circulation, vol. 115, no. 25, pp. 3213–3223, 2007.
[54] S. M. Haner, S. Lehto, T. R
¨
onnemaa, K. Py
¨
or
¨
al
¨
a, and M.
Laakso, “Mortality from coronary heart disease in subjects
with type 2 diabetes and in nondiabetic subjects w ith and
without prior myocardial infarction, The New England Jour-
nal of Medicine, vol. 339, no. 4, pp. 229–234, 1998.
[55] A. Juutilainen, S. Lehto, T. R
¨
onnemaa, K. Py
¨
or
¨
al
¨
a, and
M. Laakso, “Type 2 diabetes as a coronary heart disease
equivalent”: an 18-year prospective population-based study in
Finnish subjects, Diabetes Care, vol. 28, no. 12, pp. 2901–
2907, 2005.
[56] G. Zuanetti, R. Latini, A. P. Maggioni, L. Santoro, and M.
G. Franzosi, “Influence of diabetes on mortality in acute
myocardial infarction: data from the GISSI-2 study, Journal
of the American College of Cardiology, vol. 22, no. 7, pp. 1788–
1794, 1993.
[57] J. Herlitz, B. W. Karlson, J. Lindqvist, and M. Sj
¨
olin, “Rate and
mode of death during five years of follow-up among patients
with acute chest pain with and without a history of diabetes
mellitus, Diabetic Medicine, vol. 15, no. 4, pp. 308–314, 1998.
[58] E. D. Abel, “Insulin signaling in heart muscle: lessons from
genetically engineered mouse models, Current Hypertension
Reports, vol. 6, no. 6, pp. 416–423, 2004.
[59] E. D. Abel, “Myocardial insulin resistance and cardiac compli-
cations of diabetes, Current Drug Targets: Immune, Endocrine
and Metabolic Disorders, vol. 5, no. 2, pp. 219–226, 2005.
[60] L. Laviola, G. Belsanti, A. M. Davalli et al., “Eects of strepto-
zocin diabetes and diabetes treatment by islet transplantation
on in vivo insulin signaling in rat h eart, Diabetes, vol. 50, no.
12, pp. 2709–2720, 2001.
[61] J. Ali Raza and A. Movahed, “Current concepts of cardiovas-
cular diseases in diabetes mellitus, International Journal of
Cardiology, vol. 89, no. 2-3, pp. 123–134, 2003.
[62] C. Yu, Y. Chen, G. W. Cline et al., “Mechanism by which fatty
acids inhibit insulin activation of insulin receptor substrate-
1 (IRS-1)-associated phosphatidylinositol 3-kinase activity in
muscle, Journal of Biological Chemistry, vol. 277, no. 52, pp.
50230–50236, 2002.
[63] Y. Wang, W. Feng, W. Xue et al., “Inactivation of GSK-3β
by
metallothionein prevents diabetes-related changes in cardiac
energy metabolism, inflammation, nitrosative damage, and
remodeling, Diabetes, vol. 58, no. 6, pp. 1391–1402, 2009.
[64] J. Wang, Y. Song, L. Elsherif et al., “Cardiac metallothionein
induction plays the major role in the prevention of diabetic
cardiomyopathy by zinc supplementation, Circulation, vol.
113, no. 4, pp. 544–554, 2006.
[65] A. Barthel, E. A. Ostrakhovitch, P. L. Walter, A. Kampk
¨
otter,
and L. O. Klotz, “Stimulation of phosphoinositide 3-
kinase/Akt signaling by copper and zinc ions: mechanisms and
consequences, Archives of Biochemistry and Biophysics, vol.
463, no. 2, pp. 175–182, 2007.
[66] M. M. Mocanu and D. M. Yellon, “PTEN, the Achilles heel
of myocardial ischaemia/reperfusion injury?” British Jour n al
of Pharmacology, vol. 150, no. 7, pp. 833–838, 2007.
[67] W. Wu, X. Wang, W. Zhang et al., “Zinc-induced PTEN
protein degradation through the proteasome pathway in
human airway epithelial cells, Journal of Biological Chemistry,
vol. 278, no. 30, pp. 28258–28263, 2003.
[68] J. N. Peart and J. P. Headrick, Adenosinergic cardioprotection:
multiple receptors, multiple pathways, Pharmacology and
Therapeutics, vol. 114, no. 2, pp. 208–221, 2007.
[69] L. You, L. Li, Q. Xu, J. Ren, and F. Zhang, “Postconditioning
reduces infarct size and cardiac myocyte apoptosis via the
opioid receptor and JAK-STAT signaling pathway, Molecular
Biology Reports, pp. 1–7, 2010.
[70] A. J. Zatta, H. Kin, D. Yoshishige et al., “Evidence that
cardioprotection by postconditioning involves preservation
of myocardial opioid content and selective opioid receptor
activation, American Journal of Physiology, vol. 294, no. 3, pp.
H1444–H1451, 2008.
[71] E. R. Gross, A. K. Hsu, and G. J. Gross, Opioid-induced car-
dioprotection occurs via glycogen synthase kinase β inhibition
during reperfusion in intact rat hearts, Circulation Research,
vol. 94, no. 7, pp. 960–966, 2004.
[72] M. Nishihara, T. Miura, T. Miki et al., “Erythropoietin
aords additional cardioprotection to preconditioned hearts
by enhanced phosphorylation of glycogen synthase kinase-
3β,” American Journal of Physiology, vol. 291, no. 2, pp. H748–
H755, 2006.
Experimental Diabetes Research 9
[73] S. Namiuchi, Y. Kagaya, J. Ohta et al., “High serum erythropoi-
etin level is associated with smaller infarct size in patients with
acute myocardial infarction who undergo successful primary
percutaneous coronary intervention, Journal of the American
College of Cardiology, vol. 45, no. 9, pp. 1406–1412, 2005.
[74] A. J. Bullard, P. Govewalla, and D. M. Yellon, “Er ythropoietin
protects the myocardium against reperfusion injury in vitro
and in vivo, Basic Research in Cardiology, vol. 100, no. 5, pp.
397–403, 2005.
[75] N. X. Fang, Y. T. Yao, C. X. Shi, and L. H. Li, Attenuation of
ischemia-reperfusion injury by sevoflurane postconditioning
involves protein kinase B and glycogen synthase kinase 3 beta
activation in isolated rat hearts, Molecular Biology Reports,
vol. 37, no. 8, pp. 3763–3769, 2010.
[76] A. Tosaki, D. T. Engelman, R. M. Engelman, and D. K. Das,
“The evolution of diabetic response to ischemia/reperfusion
and preconditioning in isolated working rat hearts, Cardio-
vascular Research, vol. 31, no. 4, pp. 526–536, 1996.
[77] E. R. Gross, A. K. Hsu, and G. J. Gross, “Diabetes abolishes
morphine-induced cardioprotection via multiple pathways
upstream of glycogen synthase kinase-3β,” Diabetes, vol. 56,
no. 1, pp. 127–136, 2007.
[78] H. Hotta, T. Miura, T. Miki et al., “Short communication:
angiotensin II type 1 receptor-mediated upregulation of
calcineurin activity underlies impairment of cardioprotective
signaling in diabetic hearts, Circulation Research, vol. 106, no.
1, pp. 129–132, 2010.
[79] W. Shi-Ting, X. Mang-Hua, C. Wen-Ting, G. Feng-Hou, and
G. Zhu-Ying, “Study on tolerance to ischemia-reperfusion
injur y and protection of ischemic preconditioning of type
1 diabetes rat heart, Biomedicine and Pharmacotherapy.In
press.
[80] K. Przyklenk, M. Maynard, D. L. Greiner, and P. Whittaker,
“Cardioprotection with postconditioning: loss of ecacy in
murine models of type-2 and type-1 diabetes, Antioxidants
and Redox Signaling, vol. 14, no. 5, pp. 781–790, 2011.
[81] B. Drenger, I. A. Ostrovsky, M. Barak, Y. Nechemia-Arbely,
E. Ziv, and J. H. Axelrod, “Diabetes blockade of sevoflu-
rane postconditioning is not restored by insulin in the rat
heart: phosphorylated signal transducer and activator of
transcription 3- and phosphatidylinositol 3-kinase-mediated
inhibition, Anesthesiology, vol. 114, no. 6, pp. 1364–1372,
2011.
[82] C. Zhuo, Y. Wang, X. Wang, Y. Wang, and Y. Chen, “Car-
dioprotection by ischemic postconditioning is abolished in
depressed rats: role of Akt and signal transducer and activator
of transcription-3, Molecular and Cellular Biochemistry , vol.
346, no. 1-2, pp. 39–47, 2011.
[83] R. Huhn, A. Heinen, N. C. Weber, M. W. Hollmann, W.
Schlack, and B. Preckel, “Hyperglycaemia blocks sevoflurane-
induced postconditioning in the rat heart in vivo: cardio-
protection can be restored by blocking the mitochondrial
permeability transition pore, British Journal of Anaesthesia,
vol. 100, no. 4, pp. 465–471, 2008.
[84] Y. Balaraman, A. R. Limaye, A. I. Levey, and S. Srini-
vasan, “Glyco gen synthase kinase 3β and Alzheimer’s disease:
pathophysiological and therapeutic significance, Cellular and
Molecular Life Sciences, vol. 63, no. 11, pp. 1226–1235, 2006.
[85]J.Raphael,Y.Gozal,N.Navot,andZ.Zuo,“Hyperglycemia
inhibits anesthetic-induced postconditioning in the rabbit
heart via modulation of phosphatidylinositol-3-kinase/Akt
and endothelial nitric oxide synthase signaling, Journal of
Cardiovascular Pharmacology, vol. 55, no. 4, pp. 348–357,
2010.
[86] K. Kupai, C. Csonka, V. Fekete et al., “Cholesterol diet-
induced hyperlipidemia impairs the cardioprotective eect of
postconditioning: role of peroxynitrite, American Journal of
Physiology, vol. 297, no. 5, pp. H1729–H1735, 2009.
[87] H. Su, X. Sun, H. Ma et al., Acute hyperglycemia exacerbates
myocardial ischemia/reperfusion injury and blunts cardiopro-
tective eect of GIK,
American Journal of Physiology, vol. 293,
no. 3, pp. E629–E635, 2007.
[88] R. Ilouz, O. Kaidanovich, D. Gurwitz, and H. Eldar-
Finkelman, “Inhibition of glycogen synthase kinase-3β by
bivalent zinc ions: insight into the insulin-mimetic action of
zinc, Biochemical and Biophysical Research Communications,
vol. 295, no. 1, pp. 102–106, 2002.
[89] K. Viswanath, S. Bodiga, V. Balogun, A. Zhang, and V. L.
Bodiga, Cardioprotective eect of zinc requires ErbB2 and
Akt during hypoxia/reoxygenation, BioMetals,vol.24,no.1,
pp. 171–180, 2010.
[90] C. Penna, D. Mancardi, R. Rastaldo, G. Losano, and P. Pagliaro,
“Intermittent activation of bradykinin B2 receptors and mito-
chondrial KATP channels trigger cardiac postconditioning
through redox signaling, Cardiovascular Research, vol. 75, no.
1, pp. 168–177, 2007.
[91] C. J. Parsa, A. Matsumoto, J. Kim et al., A novel protective
eect of erythropoietin in the infarcted heart, Journal of
Clinical Investigation, vol. 112, no. 7, pp. 999–1007, 2003.
[92] K. T. Lamont, S. Somers, L. Lacerda, L. H. Opie, and S.
Lecour, “Is red wine a SAFE sip away from cardioprotection?
Mechanisms involved in resveratrol- and melatonin-induced
cardioprotection, Journal of Pineal Research, vol. 50, no. 4, pp.
374–380, 2011.
... GSK-3β plays an important role in necrosis and apoptosis of cardiomyocytes as one of the downstream targets of PI3K/Akt pathway (Miura and Miki, 2009). GSK-3β activity is a determinant of the threshold for mPTP's opening in cardiomyocytes (Yin et al., 2012). Its phosphorylation or inactivation can inhibit the opening of mPTP, which is associated with the upregulation of apoptotic cascade response process and oxidative stress (Yin et al., 2012). ...
... GSK-3β activity is a determinant of the threshold for mPTP's opening in cardiomyocytes (Yin et al., 2012). Its phosphorylation or inactivation can inhibit the opening of mPTP, which is associated with the upregulation of apoptotic cascade response process and oxidative stress (Yin et al., 2012). In the diabetic state, GSK-3β is activated and its activity is 2-fold higher than in non-diabetic state (Yin et al., 2012). ...
... Its phosphorylation or inactivation can inhibit the opening of mPTP, which is associated with the upregulation of apoptotic cascade response process and oxidative stress (Yin et al., 2012). In the diabetic state, GSK-3β is activated and its activity is 2-fold higher than in non-diabetic state (Yin et al., 2012). DEX preconditioning could promote the phosphorylation of GSK-3β, inhibit mPTP opening, maintain mitochondrial function, block apoptotic cascade initiation, and enhance myocardial antioxidant defense (Kip et al., 2015;Guo et al., 2020). ...
Article
Full-text available
Mechanisms contributing to the pathogenesis of myocardial ischemia-reperfusion (I/R) injury are complex and multifactorial. Many strategies have been developed to ameliorate myocardial I/R injuries based on these mechanisms. However, the cardioprotective effects of these strategies appear to diminish in diabetic states. Diabetes weakens myocardial responses to therapies by disrupting intracellular signaling pathways which may be responsible for enhancing cellular resistance to damage. Intriguingly, it was found that Dexmedetomidine (DEX), a potent and selective α2-adrenergic agonist, appears to have the property to reverse diabetes-related inhibition of most intervention-mediated myocardial protection and exert a protective effect. Several mechanisms were revealed to be involved in DEX’s protection in diabetic rodent myocardial I/R models, including PI3K/Akt and associated GSK-3β pathway stimulation, endoplasmic reticulum stress (ERS) alleviation, and apoptosis inhibition. In addition, DEX could attenuate diabetic myocardial I/R injury by up-regulating autophagy, reducing ROS production, and inhibiting the inflammatory response through HMGB1 pathways. The regulation of autonomic nervous function also appeared to be involved in the protective mechanisms of DEX. In the present review, the evidence and underlying mechanisms of DEX in ameliorating myocardial I/R injury in diabetes are summarized, and the potential of DEX for the treatment/prevention of myocardial I/R injury in diabetic patients is discussed.
... These cardioprotective results are supported by the other studies. [10,35,36] However, the cardioprotective effect of IPOC was significantly modulated in the diabetic rat, our results supported earlier published studies. [10,35] In our present study, perfusion of ANP significantly restored the attenuated effect of IPOC in diabetic rat myocardium in the terms of decreasing the myocardial infarct size, CK-MB and LDH. ...
... [10,35,36] However, the cardioprotective effect of IPOC was significantly modulated in the diabetic rat, our results supported earlier published studies. [10,35] In our present study, perfusion of ANP significantly restored the attenuated effect of IPOC in diabetic rat myocardium in the terms of decreasing the myocardial infarct size, CK-MB and LDH. IPOC induces the cardioprotection by activating the PI3K/Akt pathway and endothelial nitric oxide (eNOS). ...
Article
Full-text available
Background: Diabetes decreased cardioprotective potential of ischemic postconditioning (IPOC), atrial natriuretic peptide (ANP) induced the cardioprotection against ischemic-reperfusion (I/R) injury. The present study has been designed to investigate the role of ANP induced postconditioning in diabetic rat heart. Methods: Isolated Langendorff perfused normal and diabetic rat hearts were stabilized for 10 min proceed by global ischemia further followed by four cycles of IPOC, each cycle comprised 5 min of reperfusion and 5 min of ischemia at onset of 120 min of reperfusion. Perfusion of ANP (0.1μM/l) with Krebs–Henseleit Buffer solution in isolated diabetic rat heart for four-cycle of IPOC significantly decreased I/R-induced myocardial infarct size and release of CK-MB and lactate dehydrogenase (LDH) level in coronary effluent. Results: Four cycles of IPOC-induced cardioprotection noted by decreased in infarct size and also in release of LDH and CK-MB in normal rat heart. However, IPOC-induced cardioprotection was completely attenuated in isolated heart obtained from diabetic rat. Perfusion of ANP (0.1μM/L) significantly restored the attenuated cardioprotection in diabetic rat heart, which was completely blocked by perfusion of L-NAME (100μM/L), an eNOS inhibitor. Conclusion: So that, ANP restored cardioprotective affect in diabetic rat heart, which was completely abolished by the perfusion of L-NAME (100μM/L), an eNOS inhibitor.
... [33] A mass of H 2 O 2 and hydroxyl radicals is produced during ischemic injury. [40,41] However, a series of antioxidants can control the levels of reactive oxygen species and scavenge free radicals. [42] In addition, activation of Nrf2-ARE signaling is a major mechanism underlying the anti-oxidative and cytoprotective processes. ...
... Another routes of imparting function revealed that generation of nitric oxide (NO) and opening of mitochondrial ATP-sensitive potassium channel (mito-K ATP ) mediate cardioprotection (Selzner et al., 2012). The cardio-protective effect of IPTC is weakened in different physiological and disease conditions like hyperhomocysteinia (Rohilla et al., 2009), hyperlipidemia (Zhao et al., 2009), estrogen inadequacy (Couvreur et al., 2009) and diabetes mellitus (Yin et al., 2012). ...
Article
Full-text available
Abstract Ischemic postconditioning (IPTC) brings cardioprotection endogenously, Atrial natriuretic peptide (ANP) produces the same effect. It happens due to down expression of endothelial nitric oxide synthase (eNOS). Thus, experimental protocol associating IPTC has been formulated to find the role of ANP in the cardioprotection of heart in OVX rats. For this experiment, heart was isolated from OVX rat and held tightly on Langendorff’s apparatus in a manner that ischemia of 30 min and reperfusion of 120 min were also given. Simultaneously, IPTC with four cycles of 5 min ischemia and 5 min reperfusion of each was applied. Parameters like size of myocardial infarct, levels of lactate dehydrogenase (LDH) and release of creatine kinase- MB (CK-MB) in coronary effluent were noted after each stage of experiment for ensuring the extent of myocardial injury. Some significant changes were also seen in the histopathology of cardiovascular tissues. The cardio-protection has been made by four cycles of IPTC. It was confirmed by decline in the size of myocardial infarct. It diminishes the release of LDH and CK-MB in heart of OVX rat. Thus, IPTC induces cardio-protection in the isolated heart from OVX rat. Perfusion of ANP associating with IPTC favors the cardioprotection which is further confirmed by rise in the NO release and heart rate. The level of myocardial damage changes using IPTC, IPTC+OVX, IPTC+OVX+ANP, IPTC+ OVX+ANP+L-NAME and other groups were observed significantly and were found to be less than those in I/R control group. Thus, it is recommended that ANP involving IPTC restores attenuated cardio-protection in OVX rat heart. Therefore, Post-conditioning is useful in various clinical aspects.
... Therefore, the threshold for cardioprotection provided by activity of the PI3K/Akt pathway is elevated in diabetic myocardium. DM inhibits preconditioning and postconditioning, affecting PI3K/Akt on many levels [122,242]. For example, activation of STAT3 is strongly reduced in STZ-induced diabetic rats, which results in the inhibition of PI3K/Akt activity and decreased cardioprotection [243]. ...
Article
Full-text available
Despite the significant decline in mortality, cardiovascular diseases are still the leading cause of death worldwide. Among them, myocardial infarction (MI) seems to be the most important. A further decline in the death rate may be achieved by the introduction of molecularly targeted drugs. It seems that the components of the PI3K/Akt signaling pathway are good candidates for this. The PI3K/Akt pathway plays a key role in the regulation of the growth and survival of cells, such as cardiomyocytes. In addition, it has been shown that the activation of the PI3K/Akt pathway results in the alleviation of the negative post-infarct changes in the myocardium and is impaired in the state of diabetes. In this article, the role of this pathway was described in each step of ischemia and subsequent left ventricular remodeling. In addition, we point out the most promising substances which need more investigation before introduction into clinical practice. Moreover, we present the impact of diabetes and widely used cardiac and antidiabetic drugs on the PI3K/Akt pathway and discuss the molecular mechanism of its effects on myocardial ischemia and left ventricular remodeling.
... I/R is characterized by a limited blood supply to the organs and subsequent tissue damage as a result of recovery. Many studies have shown that inflammation associated with I/R injury can exacerbate myocardial damage [96], which may cause a condition experimentally similar to IH. On the basis of these studies [97], we suspected that IH had a direct effect on myokine secretion and conducted the following experiments. ...
Article
Full-text available
Sleep apnea syndrome (SAS) is a prevalent disorder characterized by recurrent apnea or hypoxia episodes leading to intermittent hypoxia (IH) and arousals during sleep. Currently, the relationship between SAS and metabolic diseases is being actively analyzed, and SAS is considered to be an independent risk factor for the development and progression of insulin resistance/type 2 diabetes (T2DM). Accumulating evidence suggests that the short cycles of decreased oxygen saturation and rapid reoxygenation, a typical feature of SAS, contribute to the development of glucose intolerance and insulin resistance. In addition to IH, several pathological conditions may also contribute to insulin resistance, including sympathetic nervous system hyperactivity, oxidative stress, vascular endothelial dysfunction, and the activation of inflammatory cytokines. However, the detailed mechanism by which IH induces insulin resistance in SAS patients has not been fully revealed. We have previously reported that IH stress may exacerbate insulin resistance/T2DM, especially in hepatocytes, adipocytes, and skeletal muscle cells, by causing abnormal cytokine expression/secretion from each cell. Adipose tissues, skeletal muscle, and the liver are the main endocrine organs producing hepatokines, adipokines, and myokines, respectively. In this review, we focus on the effect of IH on hepatokine, adipokine, and myokine expression.
Article
Myocardial ischemia-reperfusion (IR) in diabetes can cause severe myocardial damages. In this study, resveratrol (RES) nanoparticles were used in diabetic myocardial IR rat model injury to assess its effect on mitochondria function. Rat models were assigned into sham group, IR group, IR+RES group, IR+RES+mir-NC group, and IR+RES+miR-20b-5p inhibitor group. Myocardial infarction area was measured by TTC in 5 rats from each group, and ultrasound was used to detect left ventricular end-systolic internal diameters (LVIDs) and end-diastolic internal diameters (LVIDd), along with analysis of cardiomyopathy by HE staining. miR-20b-5p and Stromal interaction molecule 2 (STIM2) expressions, cardiomyocyte proliferation, apoptosis, cell viability, mitochondrial function, and relationship between miR-20b-5p and STIM2 were also analyzed. Resveratrol (RES) nanoparticles were prepared successfully. Myocardial infarct size, LVIDd and LVIDs of rats in IR+RES group decreased (vs. IR group), but were higher than sham group. miR-20b-5p expression also increased in the IR+RES group (vs. IR group), and the above indicators were decreased by the miR-20b-5p inhibitor (vs. IR+RES group, P <0.05). The myocardial changes in rats from the IR+RES+miR-20b-5p antagomir group were smaller (vs. IR group), while STIM2 expression was lower than in the IR group after using the RES nanoparticles ( P < 0.05). RES nanoparticles can thus enhance mitochondrial function and cell viability of cardiomyocytes, increasing cell proliferation rate and decreasing apoptosis rate (vs. IR group, P <0.05). After using the RES nanoparticles to interfere with myocardial IR in the diabetic rats, they were found to inhibit STIM2 and improve mitochondria by regulating miR-20b-5p signaling pathway.
Article
The public health issue of glucolipid metabolic disorders (GLMD) has grown significantly, posing a grave threat to human wellness. Its prevalence is rising yearly and tends to affect younger people. Metaflammation is an important mechanism regulating body metabolism. Through a complicated multi-organ crosstalk network involving numerous signaling pathways such as NLRP3/caspase-1/IL-1, NF-B, p38 MAPK, IL-6/STAT3, and PI3K/AKT, it influences systemic metabolic regulation. Numerous inflammatory mediators are essential for preserving metabolic balance, but more research is needed to determine how they contribute to the co-morbidities of numerous metabolic diseases. Whether controlling the inflammatory response can influence the progression of GLMD determines the therapeutic strategy for such diseases. This review thoroughly examines the role of metaflammation in GLMD and combs the research progress of related therapeutic approaches, including inflammatory factor-targeting drugs, traditional Chinese medicine (TCM), and exercise therapy. Multiple metabolic diseases, including diabetes, non-alcoholic fatty liver disease (NAFLD), cardiovascular disease, and others, respond therapeutically to anti-inflammatory therapy on the whole. Moreover, we emphasize the value and open question of anti-inflammatory-based means for treating GLMD.
Article
Background: Myocardial insulin resistance is a hallmark of diabetic cardiac injury. However, the underlying molecular mechanisms remain unclear. Recent studies demonstrate that the diabetic heart is resistant to other cardioprotective interventions, including adiponectin and preconditioning. The "universal" resistance to multiple therapeutic interventions suggests impairment of the requisite molecule(s) involved in broad prosurvival signaling cascades. Cav (Caveolin) is a scaffolding protein coordinating transmembrane signaling transduction. However, the role of Cav3 in diabetic impairment of cardiac protective signaling and diabetic ischemic heart failure is unknown. Methods: Wild-type and gene-manipulated mice were fed a normal diet or high-fat diet for 2 to 12 weeks and subjected to myocardial ischemia and reperfusion. Insulin cardioprotection was determined. Results: Compared with the normal diet group, the cardioprotective effect of insulin was significantly blunted as early as 4 weeks of high-fat diet feeding (prediabetes), a time point where expression levels of insulin-signaling molecules remained unchanged. However, Cav3/insulin receptor-β complex formation was significantly reduced. Among multiple posttranslational modifications altering protein/protein interaction, Cav3 (not insulin receptor-β) tyrosine nitration is prominent in the prediabetic heart. Treatment of cardiomyocytes with 5-amino-3-(4-morpholinyl)-1,2,3-oxadiazolium chloride reduced the signalsome complex and blocked insulin transmembrane signaling. Mass spectrometry identified Tyr73 as the Cav3 nitration site. Phenylalanine substitution of Tyr73 (Cav3Y73F) abolished 5-amino-3-(4-morpholinyl)-1,2,3-oxadiazolium chloride-induced Cav3 nitration, restored Cav3/insulin receptor-β complex, and rescued insulin transmembrane signaling. It is most important that adeno-associated virus 9-mediated cardiomyocyte-specific Cav3Y73F reexpression blocked high-fat diet-induced Cav3 nitration, preserved Cav3 signalsome integrity, restored transmembrane signaling, and rescued insulin-protective action against ischemic heart failure. Last, diabetic nitrative modification of Cav3 at Tyr73 also reduced Cav3/AdipoR1 complex formation and blocked adiponectin cardioprotective signaling. Conclusion: Nitration of Cav3 at Tyr73 and resultant signal complex dissociation results in cardiac insulin/adiponectin resistance in the prediabetic heart, contributing to ischemic heart failure progression. Early interventions preserving Cav3-centered signalsome integrity is an effective novel strategy against diabetic exacerbation of ischemic heart failure.
Article
All eukaryotes consist of kinases with a serine/threonine residue called glycogen synthase kinase 3 (GSK-3) which mediates cellular functions by causing phosphorylation of glycogen synthase and regulating glucose metabolism. It establishes disease mechanisms through cell signalling and different transcription factors. Glycogen synthase kinase-3β (GSK-3β) has pharmacological role in cardiac fibrosis, hyperlipidaemia, hyperglycaemia, hyperhomocysteinemia and in case of myocardial reperfusion injury and estrogen deficiency on the heart. The lead compounds were discovered from natural products possessing GSK-3β inhibitory activity. New signalling pathways involving mitochondrion have been investigated for ischemic preconditioning. GSK-3β may bind with mitochondrial protein and mediate mitochondrion function by binding with PI3K-Akt, PGC-1α, HK-II, PKCε subunits of mPTP. The present study explores the structural functionalities of GSK-3β and their contributory role in cardiac disorders and various other diseases. Therefore, GSK-3β is believed to be an imperative target for the discovery and development of newer drugs.
Article
Full-text available
The purpose of this study was to determine whether exogenous zinc prevents cardiac reperfusion injury by targeting the mitochondrial permeability transition pore (mPTP) via glycogen synthase kinase-3beta (GSK-3beta). The treatment of cardiac H9c2 cells with ZnCl2 (10 microM) in the presence of zinc ionophore pyrithione for 20 min significantly enhanced GSK-3beta phosphorylation at Ser9, indicating that exogenous zinc can inactivate GSK-3beta in H9c2 cells. The effect of zinc on GSK-3beta activity was blocked by the phosphatidylinositol 3-kinase (PI3K) inhibitor LY-294002 but not by the mammalian target of rapamycin (mTOR) inhibitor rapamycin or the PKC inhibitor chelerythrine, implying that PI3K but not mTOR or PKC accounts for the action of zinc. In support of this interpretation, zinc induced a significant increase in Akt but not mTOR phosphorylation. Further experiments found that zinc also increased mitochondrial GSK-3beta phosphorylation. This may indicate an involvement of the mitochondria in the action of zinc. The effect of zinc on mitochondrial GSK-3beta phosphorylation was not altered by the mitochondrial ATP-sensitive K+ channel blocker 5-hydroxydecanoic acid. Zinc applied at reperfusion reduced cell death in cells subjected to simulated ischemia/reperfusion, indicating that zinc can prevent reperfusion injury. However, zinc was not able to exert protection in cells transfected with the constitutively active GSK-3beta (GSK-3beta-S9A-HA) mutant, suggesting that zinc prevents reperfusion injury by inactivating GSK-3beta. Cells transfected with the catalytically inactive GSK-3beta (GSK-3beta-KM-HA) also revealed a significant decrease in cell death, strongly supporting the essential role of GSK-3beta inactivation in cardioprotection. Moreover, zinc prevented oxidant-induced mPTP opening through the inhibition of GSK-3beta. Taken together, these data suggest that zinc prevents reperfusion injury by modulating the mPTP opening through the inactivation of GSK-3beta. The PI3K/Akt signaling pathway is responsible for the inactivation of GSK-3beta by zinc.
Article
Full-text available
The possibility of restoring sevoflurane postconditioning (sevo-postC) cardioprotection in diabetic animals is uncertain. We hypothesized that attenuation of myocardial injury by sevo-postC might be hindered by inhibition of signal transducer and activator of transcription (STAT) 3-regulated activity of phosphatidylinositol 3-kinase (PI3K) in diabetic animals. To determine whether postC cardioprotection can be restored by normoglycemia, we treated rats with insulin. Diabetic or nondiabetic rats were randomly subjected to 30-min ischemia/reperfusion, with ischemic postC or sevo-postC, with and without mitochondrial adenosine triphosphate-dependent potassium channel blocker 5-hydroxy decanoate sodium and PI3K antagonist wortmannin. The infarct area, phosphorylated STAT3, and apoptosis were examined. Studies were repeated after insulin treatment. Ischemic postC and sevo-postC significantly reduced infarct size by 50% in the nondiabetic rats (P < 0.002), a phenomenon completely reversed by 5-hydroxy decanoate sodium and wortmannin. Diabetes mellitus blocked the protective effect of postC, and insulin treatment to achieve normoglycemia did not restore cardioprotection. Phosphorylated STAT3 nuclear retention was significantly increased after ischemia-reperfusion and was further enhanced in response to ischemic postC (P < 0.05) but was significantly reduced in diabetic rats (by 43%; P < 0.01). The effective reduction in infarct size and apoptosis in the nondiabetic rat heart by postC was completely abrogated in diabetic rats. This inhibition is not relieved by insulin-induced normoglycemia. The PI3K pathway and mitochondrial adenosine triphosphate-dependent potassium channel activation are involved in the mechanism of postC. In diabetic rats, STAT3 activation was strongly reduced, as was postC cardioprotection, suggesting that the inability of insulin to restore postC may be attributed to diabetes-induced STAT3-mediated inhibition of PI3K signaling.
Article
Full-text available
Activated protein C (APC) is known to be beneficial on ischemia reperfusion injury in myocardium. However, the protection mechanism of APC is not fully understood. The purpose of this study was to investigate the effects and possible mechanisms of APC on myocardial ischemic damage. Artificially ventilated anaesthetized Sprague-Dawley rats were subjected to a 30 min of left anterior descending coronary artery occlusion followed by 2 hr of reperfusion. Rats were randomly divided into four groups; Sham, I/R, APC preconditioning and postconditioning group. Myocardial infarct size, apoptosis index, the phosphorylation of ERK1/2, Bcl-2, Bax and cytochrome c genes and proteins were assessed. In APC-administrated rat hearts, regardless of the timing of administration, infarct size was consistently reduced compared to ischemia/reperfusion (I/R) rats. APC improved the expression of ERK1/2 and anti-apoptotic protein Bcl-2 which were significantly reduced in the I/R rats. APC reduced the expression of pro-apoptotic genes, Bax and cytochrome c. These findings suggest that APC produces cardioprotective effect by preserving the expression of proteins and genes involved in anti-apoptotic pathways, regardless of the timing of administration.
Article
Background— Recent evidence from cultured endothelial cell studies suggests that phosphorylation of endothelial nitric oxide synthase (eNOS) through the PI3-kinase– Akt pathway increases NO production. This study was designed to elucidate the signaling pathway involved in the antiapoptotic effect of insulin in vivo and to test the hypothesis that phosphorylation of eNOS by insulin may participate in the cardioprotective effect of insulin after myocardial ischemia and reperfusion. Methods and Results— Male Sprague-Dawley rats were subjected to 30 minutes of myocardial ischemia and 4 hours of reperfusion. Rats were randomized to receive vehicle, insulin, insulin plus wortmannin, or insulin plus L-NAME. Treatment with insulin resulted in 2.6-fold and 4.3-fold increases in Akt and eNOS phosphorylation and a significant increase in NO production in ischemic/reperfused myocardial tissue. Phosphorylation of Akt and eNOS and increase of NO production by insulin were completely blocked by wortmannin, a PI3-kinase inhibitor. Pretreatment with L-NAME, a nonselective NOS inhibitor, had no effect on Akt and eNOS phosphorylation but significantly reduced NO production. Moreover, treatment with insulin markedly reduced myocardial apoptotic death ( P <0.01 versus vehicle). Pretreatment with wortmannin abolished the antiapoptotic effect of insulin. Most importantly, pretreatment with L-NAME also significantly reduced the antiapoptotic effect of insulin ( P <0.01 versus insulin). Conclusions— These results demonstrated that in vivo administration of insulin activated Akt through the PI3-kinase–dependent mechanism and reduced postischemic myocardial apoptotic death. Phosphorylation of eNOS and the concurrent increase of NO production contribute significantly to the antiapoptotic effect of insulin.
Article
Postconditioning (PostC) maneuvers allow post-ischemic accumulation of autacoids, which trigger protection. We tested if PostC-triggering includes bradykinin (BK) B2 receptor activation and its downstream pathway. Isolated rat hearts underwent 30 min ischemia and 120 min reperfusion. Infarct size was evaluated using nitro-blue tetrazolium staining. In Control hearts infarct size was 61+/-5% of risk area. PostC (5 cycles of 10 s reperfusion/ischemia) reduced infarct size to 22+/-4% (p<0.01). PostC protection was abolished by B2 BK receptor-antagonists (HOE140 or WIN64338), nitric oxide synthase-inhibitor (L-nitro-arginine-methylester), protein kinase G (PKG)-blocker (8-bromoguanosine-3',5'-cyclic-monophosphorothioate), and mitochondrial K(ATP) (mK(ATP))-blocker (5-hydroxydecanoate) each given for 3 min only. Since 3 min of BK-infusion (100 nM) did not reproduce PostC protection, protocols with Intermittent-BK infusion were used to mimic PostC: a) 5 cycles of 10 s oxygenated-no-BK/oxygenated+BK buffer; b) 5 cycles of 10 s oxygenated-no-BK/hypoxic+BK buffer. Both protocols with Intermittent-BK attenuated infarct size (36+/-5% and 38+/-4%, respectively; p<0.05 vs Control and NS vs PostC for both; NS vs each other). Intermittent-BK protection was abolished by the same antagonists used to prevent PostC protection. Intermittence of re-oxygenation only (5 cycles of 10 s oxygenated/hypoxic buffer) did not reproduce PostC. Yet, cardioprotection was triggered by intermittent mK(ATP) activation with diazoxide, but not by intermittent reactive oxygen species (ROS) generation with purine/xanthine oxidase. ROS scavengers (N-acetyl-L-cysteine or 2-mercaptopropionylglycine), given for 3 min only, abolished PostC-, Intermittent BK-and diazoxide-induced protection. Intermittent targeting of specific cellular sites (i.e. BK B2 receptors and mK(ATP) channels) during early reperfusion triggers PostC protection via ROS signaling. Since neither intermittent oxygenation nor exogenous ROS generators can trigger protection, it is likely that intermittent autacoid accumulation and ROS compartmentalization may play a pivotal role in PostC-triggering.
Article
Reperfusion of ischemic myocardium is necessary to salvage tissue from eventual death. However, reperfusion after even brief periods of ischemia is associated with pathologic changes that represent either an acceleration of processes initiated during ischemia per se, or new pathophysiological changes that were initiated after reperfusion. This 'reperfusion injury' shares many characteristics with inflammatory responses in the myocardium. Neutrophils feature prominently in this inflammatory component of postischemic injury. Ischemia-reperfusion prompts a release of oxygen free radicals, cytokines and other proinflammatory mediators that activate both the neutrophils and the coronary vascular endothelium. Activation of these cell types promotes the expression of adhesion molecules on both the neutrophils and endothelium, which recruits neutrophils to the surface of the endothelium and initiate a specific cascade of cell-cell interactions, leading first to adherence of neutrophils to the vascular endothelium, followed later by transendothelial migration and direct interaction with myocytes. This specific series of events is a prerequisite to the phenotypic expression of reperfusion injury, including endothelial dysfunction, microvascular collapse and blood flow defects, myocardial infarction and apoptosis. Pharmacologic therapy can target the various components in this critical series of events. Effective targets for these pharmacologic agents include: (a) inhibiting the release or accumulation of proinflammatory mediators, (b) altering neutrophil or endothelial cell activation and (c) attenuating adhesion molecule expression on endothelium, neutrophils and myocytes. Monoclonal antibodies to adhesion molecules (P-selectin, L-selectin, CD11, CD18), complement fragments and receptors attenuate neutrophil-mediated injury (vascular injury, infarction), but clinical application may encounter limitations due to antigen-antibody reactions with the peptides. Humanized antibodies and non-peptide agents, such as oligosaccharide analogs to sialyl Lewis, may prove effective in this regard. Both nitric oxide and adenosine exhibit broad spectrum effects against neutrophil-mediated events and, therefore, can intervene at several critical points in the ischemic-reperfusion response, and may offer greater benefit than agents that interdict at a single point in the cascade. The understanding of the molecular processes regulating actions of neutrophils in ischemic-reperfusion injury may be applicable to other clinical situations, such as trauma, shock and organ or tissue (i.e. vascular conduits) transplantation.
Article
The occurrence of myocyte necrosis during reperfusion of ischemic myocardium is controversial. This study measured myocardial 2-deoxyglucose uptake, correlated with histology, to determine whether loss of viability occurred during reperfusion of ischemic myocardium. In 12 anesthetized dogs, the left anterior descending coronary artery was occluded for 90 minutes before 4 hours reperfusion. Myocardial blood flow was measured by microspheres and the tracers 14C-2-deoxyglucose and 18F-2-deoxyglucose were injected intravenously after 5 and 180 minutes of reperfusion, respectively. After 240 minutes, the heart was stained with thioflavin-S (size of no-reflow zone) and triphenyl-tetrazolium chloride (TTC, extent of necrosis). Samples from normal, salvaged, and necrotic myocardium were counted for 14C- and 18F-deoxyglucose and microspheres. With the use of a three-compartment model of 2-deoxyglucose uptake, the rate constant k3 for phosphorylation of 14C- and 18F-2-deoxyglucose was calculated for each sample. Viability was defined as k3> or = 0.125 min(-1) (predictive accuracy 88% versus electron microscopy and 97% versus TTC). Among 58 samples from no-reflow regions, 97% were nonviable after 5 minutes of reperfusion (k3=0.096 +/- 0.027 min[-1]). Among 164 samples from salvaged myocardium, 95% were viable after both 5 and 180 minutes of reperfusion (k3=0.170 +/- 0.056 min[-1] P<.01 versus no-reflow). Among 179 samples from infarcted myocardium, mean k3 after 5 minutes of reperfusion was 0.184 +/- 0.070 min(-1) and 65% of samples were viable, but after 180 minutes of reperfusion mean k3 had decreased to 0.077 +/- 0.032 min(-1) (P<.0001) and 98% of samples were nonviable. A large proportion of samples from infarcted myocardium are viable at the end of the ischemic period but lose viability during the first hours of reperfusion.
Article
Epidemiological studies suggest that regular moderate consumption of red wine confers cardioprotection but the mechanisms involved in this effect remain unclear. Recent studies demonstrate the presence of melatonin in wine. We propose that melatonin, at a concentration found in red wine, confers cardioprotection against ischemia-reperfusion injury. Furthermore, we investigated whether both melatonin and resveratrol protect via the activation of the newly discovered survivor activating factor enhancement (SAFE) prosurvival signaling pathway that involves the activation of tumor necrosis factor alpha (TNFα) and the signal transducer and activator of transcription 3 (STAT3). Isolated perfused male mouse (wild type, TNFα receptor 2 knockout mice, and cardiomyocyte-specific STAT3-deficient mice) or rat hearts (Wistars) were subjected to ischemia-reperfusion. Resveratrol (2.3 mg/L) or melatonin (75 ng/L) was perfused for 15 min with a 10-min washout period prior to an ischemia-reperfusion insult. Infarct size was measured at the end of the protocol, and Western blot analysis was performed to evaluate STAT3 activation prior to the ischemic insult. Both resveratrol and melatonin, at concentrations found in red wine, significantly reduced infarct size compared with control hearts in wild-type mouse hearts (25 ± 3% and 25 ± 3% respectively versus control 69 ± 3%, P < 0.001) but failed to protect in TNF receptor 2 knockout or STAT3-deficient mice. Furthermore, perfusion with either melatonin or resveratrol increased STAT3 phosphorylation prior to ischemia by 79% and 50%, respectively (P < 0.001 versus control). Our data demonstrate that both melatonin and resveratrol, as found in red wine, protect the heart in an experimental model of myocardial infarction via the SAFE pathway.
Article
Recent studies reported cardioprotective effects of erythropoietin (EPO) against ischemia-reperfusion (I/R) injury through activation of the reperfusion injury salvage kinase (RISK) pathway. As RISK has been reported to be impaired in diabetes and insulin resistance syndrome, we examined whether EPO-induced cardioprotection was maintained in rat models of type 1 diabetes and insulin resistance syndrome. Isolated hearts were obtained from three rat cohorts: healthy controls, streptozotocin (STZ)-induced diabetes, and high-fat diet (HFD)-induced insulin resistance syndrome. All hearts underwent 25 min ischemia and 30 min or 120 min reperfusion. They were assigned to receive either no intervention or a single dose of EPO at the onset of reperfusion. In hearts from healthy controls, EPO decreased infarct size (14.36 ± 0.60 and 36.22 ± 4.20% of left ventricle in EPO-treated and untreated hearts, respectively, p < 0.05) and increased phosphorylated forms of Akt, ERK1/2, and their downstream target GSK-3β. In hearts from STZ-induced diabetic rats, EPO did not decrease infarct size (32.05 ± 2.38 and 31.88 ± 1.87% in EPO-treated and untreated diabetic rat hearts, respectively, NS) nor did it increase phosphorylation of Akt, ERK1/2, and GSK-3β. In contrast, in hearts from HFD-induced insulin resistance rats, EPO decreased infarct size (18.66 ± 1.99 and 34.62 ± 3.41% in EPO-treated and untreated HFD rat hearts, respectively, p < 0.05) and increased phosphorylation of Akt, ERK1/2, and GSK-3β. Administration of GSK-3β inhibitor SB216763 was cardioprotective in healthy and diabetic hearts. STZ-induced diabetes abolished EPO-induced cardioprotection against I/R injury through a disruption of upstream signaling of GSK-3β. In conclusion, direct inhibition of GSK-3β may provide an alternative strategy to protect diabetic hearts against I/R injury.
Article
This study is to investigate changes of tolerance to ischemia-reperfusion (IR) injury and protection of ischemic preconditioning (IPC) of type1 diabetes rat heart. Type1 diabetes rat model was made by single injection of streptozotocin and divided into 4wk (D-4w) group and 8wk (D-8w) group randomly. The two groups were further assigned to six teams: D-4w-con, D-4w-IR, D-4w-IPC, D-8w-con, D-8w-IR and D-8w-IPC team. Normal rats were correspondingly divided into N-4w-con, N-4w-IR, N-4w-IPC, N-8w-con, N-8w-IR and N-8w-IPC team. Heart function, myocardial infarct size, release of lactate dehydrogenase (LDH) and creatine kinase (CK) were detected. And injury increment rate, injury rate and protection rate were calculated. Ultrastructure of cardiomyocyte was observed by electron microscope. The myocardial injury increment rate, infarct sizes in D-con group were all significantly higher than those in N-con rat group, and the degree of injury in D-8w rat hearts was exacerbated compared to D-4w rat hearts. The myocardial injury rate and the injury of cardiomyocyte ultrastructure in D-4w-IR team were lower compared with N-4w-IR group. Compared with D-4w-IR team, IPC decreased infarct sizes in D-4w-IPC group, but its protection rate was lower than that in N-4w-IPC team. Infarct sizes in D-8w-IPC team had no significance with those in D-8w-IR team. The fundament cardiac function of type1 diabetes rat heart was weakened. Diabetes self could induce heart injury, which could aggravate along with time. The tolerance of 4w type1 diabetes rat to IR injury is stronger but the protection of IPC was weaker compared to normal rat. The protection of IPC disappears in D-8w group.