ArticlePDF AvailableLiterature Review

Hyperglycemia in stroke and possible treatments

Authors:

Abstract and Figures

Hyperglycemia affects approximately one-third of acute ischemic stroke patients and is associated with poor clinical outcomes. In experimental and clinical stroke studies, hyperglycemia has been shown to be detrimental to the penumbral tissue for several reasons. First, hyperglycemia exacerbates both calcium imbalance and the accumulation of reactive oxygen species (ROS) in neurons, leading to increased apoptosis. Second, hyperglycemia fuels anaerobic energy production, causing lactic acidosis, which further stresses neurons in the penumbral regions. Third, hyperglycemia decreases blood perfusion after ischemic stroke by lowering the availability of nitric oxide (NO), which is a crucial mediator of vasodilation. Lastly, hyperglycemia intensifies the inflammatory response after stroke, causing edema, and hemorrhage through disruption of the blood brain barrier and degradation of white matter, which leads to a worsening of functional outcomes. Many neuroprotective treatments addressing hyperglycemia in stroke have been implemented in the past decade. Early clinical use of insulin provided mixed results due to insufficiently controlled glucose levels and heterogeneity of patient population. Recently, however, the latest Stroke Hyperglycemia Insulin Network Effort trial has addressed the shortcomings of insulin therapy. While glucagon-like protein-1 administration, hyperbaric oxygen preconditioning, and ethanol therapy appear promising, these treatments remain in their infancy and more research is needed to better understand the mechanisms underlying hyperglycemia-induced injuries. Elucidation of these mechanistic pathways could lead to the development of rational treatments that reduce hyperglycemia-associated injuries and improve functional outcomes for ischemic stroke patients.
Content may be subject to copyright.
Review
Hyperglycemia in stroke and possible
treatments
William A. Li
1
, Shannon Moore-Langston
1
, Tia Chakraborty
1
, Jose A. Rafols
2
,
Alana C. Conti
1,3
, Yuchuan Ding
1
1
Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA,
2
Department
of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI, USA,
3
Research Service,
John D. Dingell VA Medical Center, Detroit, MI, USA
Hyperglycemia affects approximately one-third of acute ischemic stroke patients and is associated with
poor clinical outcomes. In experimental and clinical stroke studies, hyperglycemia has been shown to be
detrimental to the penumbral tissue for several reasons. First, hyperglycemia exacerbates both calcium
imbalance and the accumulation of reactive oxygen species (ROS) in neurons, leading to increased
apoptosis. Second, hyperglycemia fuels anaerobic energy production, causing lactic acidosis, which
further stresses neurons in the penumbral regions. Third, hyperglycemia decreases blood perfusion after
ischemic stroke by lowering the availability of nitric oxide (NO), which is a crucial mediator of vasodilation.
Lastly, hyperglycemia intensifies the inflammatory response after stroke, causing edema, and hemorrhage
through disruption of the blood brain barrier and degradation of white matter, which leads to a worsening of
functional outcomes. Many neuroprotective treatments addressing hyperglycemia in stroke have been
implemented in the past decade. Early clinical use of insulin provided mixed results due to insufficiently
controlled glucose levels and heterogeneity of patient population. Recently, however, the latest Stroke
Hyperglycemia Insulin Network Effort trial has addressed the shortcomings of insulin therapy. While
glucagon-like protein-1 administration, hyperbaric oxygen preconditioning, and ethanol therapy appear
promising, these treatments remain in their infancy and more research is needed to better understand the
mechanisms underlying hyperglycemia-induced injuries. Elucidation of these mechanistic pathways could
lead to the development of rational treatments that reduce hyperglycemia-associated injuries and improve
functional outcomes for ischemic stroke patients.
Keywords: Stroke, Hyperglycemia, Reactive oxygen species, Metabolism, Blood–brain barrier, Ethanol, Insulin, NADPH oxidase
Introduction
Stroke is a major cause of mortality and morbidity in
America, with an annual combined direct and
indirect cost exceeding 68 billion dollars.
1
In recent
years, accumulating literature supports the notion
that persisting hyperglycemia leads to an unfavorable
prognosis in stroke patients. While diabetes mellitus
is a well-known risk factor contributing to poor
prognosis after ischemic strokes, hyperglycemia with-
out pre-existing diabetes mellitus is also linked to
increased mortality and morbidity after stroke. In
fact, epidemiologic meta-analysis of 30-day post-
stroke mortality revealed a greater than three-fold
increase for non-diabetic hyperglycemic patients,
compared with a two-fold increase for hyperglycemic
patients with diabetes mellitus.
2
Elevated blood glucose, defined as levels greater
than 6.0 mmol/l (108 mg/dl), has been observed in
over one-third of all ischemic stroke patients.
Furthermore, evidence in experimental animal stroke
models supports the observation that hyperglycemia
not only exacerbates stroke related injury but that it
also adversely affects overall functional outcome.
3,4
In this review, the mechanisms by which hyperglyce-
mia exacerbates negative outcomes in ischemic stroke
and several potential treatments are discussed.
Mechanisms of Hyperglycemia-Mediated
Damage in Ischemic Stroke
Following experimental ischemic stroke, hyperglyce-
mic animals generally tend to have higher incidence
of brain edema, cell death, hemorrhagic transforma-
tion, and larger infarct volume.
3,5
Possible mechan-
isms include intracellular acidification, reactive
oxygen species (ROS) accumulation, blood-brain
barrier (BBB) disruption, inflammatory response
induction, and axonal degradation.
6–9
Correspondence to: Y. C. Ding, Department of Neurological Surgery,
Wayne State University School of Medicine, 550 E Canfield, Detroit, MI
48201, USA. Email: yding@med.wayne.edu
ßW. S. Maney & Son Ltd 2013
DOI 10.1179/1743132813Y.0000000209 Neurological Research 2013 VOL.35 NO.5 479
Figure 1 Elevated cell apoptosis due to calcium imbalance. Excess glucose causes calcium imbalance and exacerbates
cytochrome c leakage into the cytosol. Cytochrome cthen interacts with dATP to activate downstream apoptogenic proteins
that ultimately trigger apoptosis.
Figure 2 Elevated cell stress due to lactate accumulation. Ischemic stroke causes ATP depletion and a decrease in blood
perfusion. To compensate for decreased ATP, anaerobic metabolism of glucose produces lactic acid as a by-product. Owing to
poor blood perfusion, lactic acid accumulates and exacerbates neuronal apoptosis.
Li et al. Hyperglycemia in stroke and possible treatments
480 Neurological Research 2013 VOL.35 NO.5
Hyperglycemia-mediated metabolic alteration
Metabolic pathways play a crucial role in regulating
ischemic pathogenesis. Ischemic stroke in the nor-
moglycemic brain obstructs the supply of substrates
such as glucose and oxygen to the brain, thereby
slowing the synthesis of ATP.
10
As oxygen is an
irreplaceable substrate of mitochondrial respiration,
disruption in oxygen delivery leads to an immediate
mismatch between ATP production and consump-
tion. In an oxygen deprivation (OGD) brain slice
culture model, we previously reported
11
that the ADP
to ATP ratio may increase by as much as 72% after
8 hours of hypoxia. In vivo investigations reported
similar results.
12–14
Another metabolic issue related
to ATP metabolism after stroke relates to calcium
homeostasis. Considerable evidence suggests that N-
methyl-D-aspartate (NMDA) receptor hyperactiva-
tion during and after ischemic insult triggers a massive
intracellular calcium influx. Without a sufficient
energy supply, ATPase-dependent ion transport sys-
tems become dysfunctional and are unable to
maintain intracellular ion homeostasis, resulting in
a massive accumulation of intracellular and mito-
chondrial calcium.
15,16
These pathophysiological
events then trigger several deleterious developments
in the cell, including cellular swelling, organelle dege-
neration, loss of membrane integrity, and eventual
cell death.
17
The increase in intracellular and mitochondrial
calcium that occurs during hyperglycemia may indir-
ectly trigger apoptosis in the stroke brain via an
interaction with the electron transport chain compo-
nent cytochrome c. Elevated intracellular calcium
levels could lead to mitochondrial depolarization and
loss of cytochrome cinto the cytosol.
18
Once in the
cytosol, cytochrome cinteracts with procaspase-9,
apoptotic protease-activating factor-1, and dATP to
form an activated complex.
19
This complex then
activates caspase-3, the irreversible trigger of apopto-
sis. Caspase-3 in turn proteolyzes an inhibitor of
caspase-activated DNase. In apoptosis, the degrada-
tion of this inhibitor results in the translocation of
caspase-activated DNase from the cytoplasm to the
nucleus, where it catalyses internucleosomal DNA
degradation and subsequent cell apoptosis.
20
Hyperglycemia has been shown to amplify intra-
cellular calcium imbalance after ischemic stroke by
upregulating neuronal NMDA receptors and exacer-
bating the aforementioned apoptotic pathway.
Li et al.
21
demonstrated that hyperglycemia increases
extracellular glutamate concentration following fore-
brain ischemia, this excessive glutamate stimulating
Figure 3 Hyperglycemia-mediated increases in ROS gen-
eration. Glucose fuels the production of NADPH, which
donates an electron to oxygen and generates superoxides
via NADPH oxidase. Hyperglycemia also exacerbates cal-
cium and proton imbalance, causing the mitochondrial
membrane to hyperpolarize and increase ROS leakage into
the cytosol.
Figure 4 Hyperglycemia-induced vasoconstriction by
decreased nitric oxide (NO) production and increased NO
elimination. Hyperglycemia amplifies the production of ROS,
whichinteractswithNOtoproduce peroxynitrite and
depletes NO bioavailability. Hyperglycemia also decreases
the activity of eNOS through the PI3K/AKT pathway to
diminish NO production.
Li et al. Hyperglycemiainstrokeandpossibletreatments
Neurological Research 2013 VOL.35 NO.5 481
NMDA receptors which further increases influx of
calcium.
22
Elevated intracellular calcium then pro-
motes the release of cytochrome cinto the cytoplasm,
thus upregulating caspase-3 activity and subsequent
DNA fragmentation (Fig. 1). Indeed, the role of
hyperglycemia and calcium in apoptotic cell death
after stroke has been further elaborated in two more
studies.
23,24
In summary, the above studies demon-
strate that the compounding deleterious effect of
hyperglycemia in stroke-related injuries may be
caused in part by glutamate release, upregulated
intracellular calcium levels, and increased cytoplas-
mic cytochrome c, all of which are known to induce
neuronal death.
Hyperglycemia-mediated lactate accumulation
and acidosis in the penumbral region
Intracellular acidification as a result of persistent
glycolysis increases proton and lactate levels, which
stresses the penumbral tissue, making it more suscep-
tible to pro-apoptotic signals. After ischemic stroke,
cerebral blood perfusion rates decrease to less than
20% of the normal rate in the penumbral region.
25
The
availability of oxygen is even more limited in the
ischemic core. Glucose is metabolized to pyruvate via
glycolysis, which anaerobically undergoes fermenta-
tion to produce lactic acid, causing a decrease in
intracellular pH.
26
In the penumbral tissue, lactate accumulates at a
level many times its normal concentration. This shift
to anaerobic metabolism in cells causes diminished
mitochondrial ATP production which creates condi-
tions that lead to tissue necrosis. Using an animal
middle cerebral artery occlusion (MCAO) model,
Parsons et el.
3
demonstrated acute hyperglycemia to
be associated with higher acute and subacute lactate
production. Although the exact mechanism is not
well elucidated, it is conceivable that high intracel-
lular glucose levels may drive the metabolic process
forward as a result of increased pyruvate availability
for lactic acid formation (Fig. 2). However, several
studies have highlighted the fact that hyperglycemia
can be beneficial to the energy starved brain in
specific situations.
27,28
For example, hyperglycemia
may be beneficial in situations where lactate can be
quickly flushed away by uninterrupted blood flow. By
contrast, blood flow in ischemia is stagnant, allowing
lactate to accumulate. The combination of stagnant
blood and lactate build-up may decrease penumbral
salvage, increase final infarct volume, and ultimately
worsen functional outcome.
3
Hyperglycemia-mediated oxidative stress and
ROS accumulation in penumbral neurons
Normally mitochondria produce ATP with remark-
able efficiency through the reduction of oxygen to
water by cytochrome coxidase. Under normal
conditions, only approximately 1–2% of the oxygen
reduced by mitochondria becomes ROS, such as
superoxide (O{
2).
29
The two known locations in the
Figure 5 Hyperglycemia-mediated increase in leukocyte transmigration. Hyperglycemia upregulates the PKC pathway, which
in turn increases calpain expression. Calpain in turn upregulates the expression of ICAM-1 on endothelial cell surface.
Alternatively, hyperglycemia also increases the translocation of NF-kappaB into the nucleus by upregulating IkappaB
proteosomal degradation. Activated NF-kappaB then translocates to the nucleus and binds DNA at specific kappaB sites in
promoter regions to activate the transcription of ICAM-1 and VCAM-1.
Li et al. Hyperglycemia in stroke and possible treatments
482 Neurological Research 2013 VOL.35 NO.5
oxidative phosphorylation chain at which superoxides
are produced are the nicotinamide adenine dinucleo-
tide dehydrogenase and ubiquinone–cytochrome bc1
regions.
30
Oxidative stress has a profound effect on
stroke pathogenesis due to the high susceptibility of
the brain to ROS-induced injuries. The brain has high
concentrations of unsaturated lipids, low levels of
antioxidants, high levels of oxygen consumption, and
high levels of iron that acts as an oxidizing agent under
pathological conditions.
31–33
All of these conditions
favor lipid peroxidation.
There are two known pathways by which ROS may
be produced following ischemia, one as a consequence
of mitochondrial membrane hyperpolarization and the
other through the use of the nicotinamide adenine
dinucleotide phosphate oxidase (NADPH oxidase)
pathway.
34
Both are upregulated by hyperglycemia.
The accumulation of intracellular calcium and protons
during ischemia onset causes hyperpolarization of the
mitochondrial membrane.
35,36
The rate of superoxide
production increases exponentially as the mitochon-
drial membrane potential increases above 140 mV.
37,38
Using an oxygen and glucose deprivation model,
Abramov et al. showed ROS production to be elevated
1.6-fold during the initial onset of hypoxia and 2.6-
fold during reoxygenation.
The NADPH oxidase pathway is an alternative
mechanism for ROS production. NADPH oxidase is a
multi-subunit enzyme that catalyzes oxygen reduction
by using NADPH as the electron donor. The five
subunits of NADPH oxidase consist of p47phox,
p67phox, p40phox, p22phox, and the catalytic subunit
gp91phox.
39
In normal brains, p47phox, p67phox, and
p40phox exist in the cytosol, while p22phox and
gp91phox are attached to the cell membrane.
However, upon stimulation, such as during ischemia,
p47phox is phosphorylated and the cytosolic subunits
form a complex that translocates to the cell membrane,
where it associates with p22phox and gp91phox to
assemble the active oxidase.
40
High intracellular glucose levels could exacerbate
reperfusion-induced injuries by upregulating super-
oxide production. Thus, Suh et al.
41
demonstrated
that glucose is central to ROS generation after OGD
and reoxygenation, and that it acts independent of
lactic acidosis in neurons. Glucose sustains the hexose
monophosphate shunt, allowing it to regenerate
NADPH, which serves as a substrate for ROS
production (Fig. 3).
42
Superoxide production is
significantly downregulated in the absence of glucose
as well as when the hexose monophosphate shunt is
inhibited after OGD.
43
Furthermore, the increase in
superoxide production during hyperglycemia is abol-
ished in p47 knockout mice and after NADPH
oxidase inhibition with apocynin.
42,44
Additionally,
2-deoxyglucose, which slows glucose transport and
impairs glucose utilization via hexokinase, may
reverse the accumulation of superoxide effectuated
as a result of hyperglycemia.
45
Hyperglycemia-mediated BBB disruption and
perfusion reduction
The BBB is formed by endothelial cells, tight junction
proteins, pericytes, and astrocytes. The BBB plays a
critical role both in maintaining brain homeostasis and
restricting blood-borne substances from entering the
parenchyma. Hyperglycemia is a risk factor for aug-
mented BBB injury during ischemia. Hyperglycemia-
induced cerebral acidosis and superoxide production
are generally considered the mechanisms of enhanced
brain injury, particularly as pertains to neurons.
46,47
However, recent reports suggest that damage to brain
endothelial cells and the BBB has a profound impact
on overall functional outcome and may even exceed
the impact on neuronal damage.
First, hyperglycemia has been shown to disrupt the
stability of the BBB by indirectly upregulating the
phosphorylation of occludin fragments, leading to
their ubiquitination and subsequent BBB permeabil-
ity. Short-term hyperglycemia enhances de novo
synthesis of diacylglycerol (DAG) in capillary en-
dothelial cells from glycolytic intermediates.
48
DAG
is then responsible for activating protein kinase C-
beta II, one of nine PKC family proteins altered by
DAG during hyperglycemia.
49
Activated PKC-beta
II then phosphorylates occludin fragments at the
Ser490 position, which leads to the ubiquitination
and redistribution of tight junction proteins in
endothelial cells.
50
The end result of these molecular
changes is increased BBB permeability. This mechan-
ism occurs within minutes of reperfusion, which
concurs with the time course observed in in vivo
models.
51
Together, the evidence from human and
animal studies supports the notion that hyperglyce-
mia worsens cerebrovascular reperfusion injuries
through the PKC-beta II pathway.
Second, hyperglycemia has been shown to signifi-
cantly reduce blood flow in the penumbral region. In
animal studies, induction of hyperglycemia by acute
glucose injection caused changes in cerebral blood
flow after either 2 or 4 hours of occlusion and
2 hours of reperfusion. Cerebral blood flow in the
ischemic hemisphere of hyperglycemic animals was
approximately 40% of that in the contralateral non-
ischemic hemisphere, compared to 100% in the
normoglycemic controls.
52
Additionally, induction
of acute hyperglycemia by glucose infusion decreased
penumbral blood flow to 60% of pre-ischemic values
in the hyperglycemic group, compared to 89% in the
normoglycemic group.
53
Another mechanism for the
reduced rate of perfusion in hyperglycemia has been
attributed to decreased bioavailability of nitric oxide
(NO), which is a potent vasodilator and inhibitor of
Li et al. Hyperglycemiainstrokeandpossibletreatments
Neurological Research 2013 VOL.35 NO.5 483
platelet aggregation.
54,55
Although this mechanism is
not well delineated, it has been suggested that
hyperglycemia could upregulate the dephosphoryla-
tion of endothelial nitric oxide synthase (eNOS) at
the Ser1177 position and its phosphorylation at the
Thr495 position, both of which decrease eNOS
activity.
55,56
Indeed, hyperglycemia has been shown
to downregulate eNOS activity by upregulating the
hexosamine and PKC pathways, thereby downregu-
lating the phosphoinositide 3-kinase/protein kinase B
(PI3K/AKT) pathway.
57,58
This process inhibits
vasodilation, thus decreasing blood flow to the
ischemic regions of the brain (Fig. 4).
Finally, ROS are known to interact with NO to
produce the highly reactive peroxynitrite radical
which decreases the availability of NO and further
impairs the blood vessels’ ability to dilate.
56
Even
mild acute hyperglycemia (y150 mg/dl) significantly
reduces blood flow to the penumbral region and
causes red blood cells to be trapped within areas of
the ischemic vasculature. The end result of these
hyperglycemic sequelae is the diminishing or absence
of oxygen delivery to a portion of the penumbra.
59
These oxygen deprived areas exhibited marked
increases in BBB permeability after reperfusion.
59
In summary, vascular injury appears to be an
important factor associated with hyperglycemia-
induced derangements and the consequences of vascu-
lar damage may significantly limit the benefit derived
from available protective neuronal treatments. As later
mentioned in this Review, more effective treatments
may emerge from a better understanding of hypergly-
cemia’s effects on the cerebrovasculature after ischemia.
Hyperglycemia-mediated inflammatory response
The brain’s inflammatory responses to ischemia are
characterized by the rapid activation of resident cells.
Beginning two hours after stroke onset, microglia are
activated via CD14 receptors and subsequently by
toll-like receptor 4.
60–62
Activated microglia undergo
morphological transformation into phagocytes and
act as cytotoxin-producing factories releasing proin-
flammatory cytokines such as tumor-necrosis factor-
alpha (TNF-alpha), IL-1, and IL-6 in addition to NO
and ROS.
63,64
Similar to microglia, astrocytes are
also activated during the acute phase to produce pro-
inflammatory cytokines, chemokines, and inducible
nitric oxide synthase.
65
Elevated levels of the afore-
mentioned molecules initiate the second step of the
post-ischemic inflammatory response, which includes
circulating inflammatory cell chemotaxis and trans-
migration into the injured brain.
66–68
Lymphocytes,
normally excluded from the neuronal compartment
under quiescent conditions, can be visualized by
immunohistochemistry in the same compartment in
the post-ischemic brain. Recent work suggests that
components of the immune system are not only
intimately involved in the ischemic cascade, but that
they interact with tight junction proteins and the basal
lamina to disrupt the BBB and degrade white matter to
cause parenchymal injuries.
69–72
Hyperglycemia has
been shown to have a tremendous effect on the
inflammatory pathway post ischemia, elevating several
transcription pathways to increase leukocyte migra-
tion to the brain and their ability to cross into the
injured tissue.
Elevated intracellular glucose results in an increase
in nuclear factor-kappaB (NF-kappaB) and a de-
crease in IkappaB expressions in endothelial cells.
73,74
Under quiescent conditions, NF-kappaB is bound to
IkappaB in the cytosol. However during IkappaB
degradation, NF-kappaB translocates into the nucleus
and stimulates the expression of pro-inflammatory
cytokines such as TNF-alpha, interleukin (IL)-1 and
IL-6 and chemokines, such as monocyte chemoattrac-
tant protein.
74–77
These cytokines and chemokines
attract mononuclear cells to the site of injury and
upregulate endothelial cell surface receptors to facil-
itate mononuclear cell attachment and their transmi-
gration into the parenchyma.
78,79
Indeed, Morigi
et al.
80
found that maintaining a blood glucose level
of 174 mg/dl for 24 hours significantly increased levels
of NF-kappaB in endothelial cells and adhesion
molecules including intercellular cell adhesion mole-
cule-1 (ICAM-1) and vascular cell adhesion molecule-
1. The inhibition of NF-kappaB with pyrrolidine
dithiocarbamate and N-tosyl-L-phenylalanine chloro-
methyl ketone reversed hyperglycemia-induced
increases in NF-kappaB expression and subsequent
mononuclear cell adhesion to endothelial cells.
80
The second major pathway in hyperglycemia media-
tion of the immune response was elucidated by
Smolock et al.,
81
who demonstrated that elevated
glucose levels activated the PKC pathway. In turn,
PKC upregulated its downstream molecule calpain in a
calcium-dependent manner.
81
This upregulation of
calpain ultimately resulted in the overexpression of
ICAM-1 on the surface of endothelial cells.
81
However,
there is controversy over whether elevated glucose
concentrations alone cause the increase in ICAM-1
expression. Supporting the direct role of hyperglyce-
mia, Kado et al.
82
demonstrated that incubating
endothelial cells with 30 mM of glucose for 24 hours
significantly increased ICAM-1 levels (Fig. 5). Several
other studies have confirmed the above results.
80,83
In contrast, studies conducted by Rasmussen
et al.
84
suggested that the induction of cellular
adhesion molecules in human endothelial cells could
be associated with the upregulation of pro-inflamma-
tory cytokines. Esposito et al.
85
further demonstrated
that acute hyperglycemia could induce an increase in
the concentrations of plasma IL-6 and TNF-alpha.
Li et al. Hyperglycemia in stroke and possible treatments
484 Neurological Research 2013 VOL.35 NO.5
This last finding has been substantiated by in vitro
studies.
86–89
Taken together, the above observations
suggest multiple mechanisms by which hyperglycemia
orchestrates pro-inflammatory phenotypes.
Inflammatory mechanisms intrinsic to the brain
and its vasculature are crucial mediators of increased
BBB permeability during ischemia. Hyperglycemia
may upregulate activator protein-1, which then
stimulates the transcription of matrix metalloprotei-
nases -2 (MMP-2) and -9 (MMP-9) in leukocytes.
90
Furthermore, hyperglycemia promotes the recruit-
ment of blood-borne leukocytes, mainly neutrophils,
which further increase the levels of MMP-9.
91
Expression of MMP-2 and MMP-9 has been inver-
sely correlated with BBB tight junction protein
(zonula occludens-1) expression and basal laminin
levels, both of which are integral to the integrity of
the BBB.
92
Decreases in the latter two protein
concentrations are thought to affect the integrity of
the BBB and increase its permeability, resulting in
edema and hemorrhagic transformation.
The source of MMP-9 is still a matter of debate.
Gasche et al.
93
found active MMP-9 to be colocalized
with endothelial cells and astrocytic processes. In
contrast, our initial results show OGD significantly
increased the expression of MMP-9 by resident micro-
glial cells in the brain. Recent studies using chimeric mice
with GFP bone marrow cells provided the opportunity
to distinguish brain versus blood macrophages and their
contribution to vascular injury after ischemic stroke.
Although it is generally thought that both resident and
blood-derived macrophages play significant roles in
the inflammatory pathway, resident microglial cells
predominate.
94
This conclusion is supported by our
studies using an in vitro OGD-reoxygenation brain
slice culture method in which blood-borne macro-
phages are eliminated from the preparation.
Compelling evidence suggests that hyperglycemia-
induced MMP-9 overexpression may also enhance
white matter degeneration following ischemic stroke,
leading to poorer outcomes.
95
Patients with inade-
quately regulated serum glucose levels are more
susceptible to white matter lesions.
96
Similarly, dis-
ruption of the BBB as a result of ischemic onset
exposes the brain parenchyma to blood-borne macro-
phages and has been associated with white matter
lesions.
97,98
Asahi et al.
99
showed that MMP-9 knock-
out mice have reduced myelin degradation after
ischemic stroke. Given the critical role of white matter
cells in ischemic outcomes, further investigation is
needed to better understand the effects of the
inflammatory response on white matter degeneration.
Treatments
Despite the ongoing effort to better understand
the mechanisms underlying hyperglycemia-enhanced
ischemic injuries, the translation of knowledge gath-
ered from experimental studies into effective treat-
ments has been uniformly disappointing. While tissue
plasminogen activator (tPA) administration after
stroke is considered the gold standard in re-establish-
ing blood flow to the brain, co-administration of
neuroprotective agents that interrupt pathways of
ischemic cell death could improve stroke outcomes. A
therapy that would both salvage ischemic tissue and
protect the brain from hyperglycemia-induced injuries
post-ischemia could benefit millions, yet this medical
need remains unmet.
Insulin therapy
There is an emerging body of evidence supporting the
beneficial effects of normalizing serum glucose levels
within 48 hours of ischemic stroke onset.
100
In animal
models, insulin therapy has been shown to reduce
infarct volume and enhance neuroprotection during
acute focal ischemia.
101
However, the translation of
these findings from the laboratory to the bedside has
yielded mixed results.
102,103
Evidence supports the notion that hyperglyce-
mia can have a significantly deleterious effect on the
neurovasculature. In rodent MCAO models, insulin
exerts a neuroprotective effect on the brain vasculature
through the upregulation of NO via the PI3K/AKT
pathway.
104
The activation of the PI3K/AKT path-
way negatively modulates genes that promote vascular
permeability and inflammation, thereby protecting
vascular integrity.
105
eNOS, a molecule downstream
of AKT, is a known catalyst of NO. As previously
discussed, NO has a crucial role in regulating vascular
tone and integrity. Generation of NO could potentially
increase vasodilation and improve blood flow to the
penumbra, thus diminishing platelet aggregation.
106
In
addition to increasing NO synthesis, insulin therapy
also decreases ROS accumulation by downregulating
NADPH oxidase activity. This change in NADPH
oxidase increases the bioavailability of NO.
73
In addition to lowering glucose levels, insulin may
also have a direct neuroprotective effect on the CNS
parenchyma by reducing inflammation.
107
In patients
who received intensive insulin therapy (target glucose
level between 80 and 110 mg/dl), the activity of the
transcription regulator NF-kappaB and its down-
stream targets, ICAM-1 and E-selectin, were signifi-
cantly reduced.
107
As ICAM-1 and E-selectin are
cardinal mediators of blood macrophage transmigra-
tion across the endothelium after stroke onset,
decreased expression of both receptors produces an
oppositional effect to that of hyperglycemia on the
vascular endothelium.
However, a broad-spectrum reduction in the
inflammatory response may paradoxically limit recov-
ery since immune cells and inflammation also play a
Li et al. Hyperglycemiainstrokeandpossibletreatments
Neurological Research 2013 VOL.35 NO.5 485
crucial role in tissue repair. Although there is evidence
to support the beneficial effects of the anti-inflamma-
tory response during the acute phase in ameliorating
tissue damage, counteracting this response may
actually worsen long-term outcomes by reducing
overall tissue repair and reorganization.
108–113
More
work in the area of the brain’s immune response after
injury would likely lead to effective novel therapies.
Early insulin based clinical trails have failed to
show significant results. The Glucose Insulin in
Stroke Trial (GIST) conducted in the UK reported
no evidence of benefit from insulin therapy.
102
There
are several possible explanations for this result. First,
the difference in mean glucose levels between the
control group and the experimental group was only
10 mg/dl after insulin treatment.
114
As a result, there
were no significant differences in clinical outcomes
as measured by mortality rate and neurological
deficits.
102
Second, the optimal range of serum
glucose was not well defined and a heterogeneous
group of patients was accepted into the study.
Nonetheless, the GIST study helped establish a 24-
hour glucose-insulin infusion safety parameter com-
prised of maintaining glucose levels between 72 and
126 mg/dl in order to avoid the significant risk of
hypoglycemia or mortality in excess of 40 weeks.
Currently, the Stroke Hyperglycemia Insulin Net-
work Effort study, a phase III single-blinded ran-
domized control trial, is looking for a correlation
between normalized serum glucose levels and a
reduction in thrombolysis-induced intracranial hemor-
rhage associated with hyperglycemia.
115
A stratified
result from this trial has the potential to alter future
treatment decisions for hyperglycemic stroke patients.
Furthermore, the Continuous Glucose Monitoring
and Insulin Pump Therapy in Diabetic Gastroparesis
clinical trial, currently conducted at the University of
Michigan, uses continuous glucose monitoring and an
insulin pump to precisely maintain the patient’s
glucose levels. The validated end point has the
potential to reduce the resulting complications of
hypoglycemia and would allow physicians to maintain
patients’ glucose at a more consistent level.
Insulin-like growth factor 1
Insulin-like growth factor I (IGF-1) is structurally
similar to insulin and thus has many analogous
functions. In animal models, it was found that IGF-1,
when injected into the ventricles, reduced ischemic
damage.
116
IGF-1 displays strong anti-apoptotic cell
survival properties and is a potent neuroprotective
agent when administered after stroke induction.
117
Cheng et al.
117
showed neuroprotective properties of
IGF-1 are mediated by the activation of PI3K/AKT
as well as FKHRL, both of which improve cell
survival. While both insulin and IGF-1 can directly
increase neuronal survival by interacting with the
IGF-1 receptor, IGF-1 binds to the receptor with
much higher affinity.
118
Thus there is reason to
believe that IGF-1 treatment may be effective in
ameliorating the negative effects of hyperglycemia
during stroke onset.
Glucagon-like peptide-1 therapy
Glucagon-like peptide-1 (GLP-1) and artificial GLP-
1 receptor agonists such as exendin-4 (Ex-4) are well-
established diabetes medications.
119
In recent years,
several studies have shown GLP-1 receptor (GLP-
1R) stimulation to ameliorate the adverse effects of
ischemic stroke.
119,120
GLP-1 and Ex-4 have a wide
range of physiological actions in glucose metabolism,
including the stimulation of insulin secretion, inhibi-
tion of glucagon release, and reduction of appetite
and food intake.
120,121
GLP-1R is expressed through-
out the brain, and GLP-1 and Ex-4 are able to cross
the BBB due to high lipophilicity.
122
GLP-1 admin-
istration elicits the combined outcomes of decreased
intracerebral glucose content and increased cerebral
metabolic rate.
123
Models of transient MCAO stroke
in rodent have shown that treatment with Ex-4
provides marked benefits on locomotor activity and
infarct volume that are absent in GLP-1R knock out
mice.
124
GLP-1R stimulation has also been shown to reduce
the synthesis of pro-inflammatory cytokine IL-1beta
in activated astrocytes via stimulation of adenylate
cyclase and the accumulation of cAMP.
125
The
mechanism by which GLP-1R stimulation drives
neuroprotection is unclear, and future research is
necessary to investigate the potential of GLP-1 as a
treatment.
Hyperbaric oxygen preconditioning
Accumulating evidence indicates that hyperbaric
oxygen therapy (HBO) may have beneficial effects in
stroke. Qin et al.
126
demonstrated that HBO precon-
ditioning decreased hyperglycemia-induced hemorrha-
gic transformation. While the exact mechanism by
which HBO limits hemorrhagic transformation is not
well-defined, recent reports suggest that HBO pre-
conditioning may ameliorate hyperglycemia-induced
injuries by reducing BBB disruption. Veltkamp
et al.
127
showed that HBO therapy reduces basal
lamina degradation and MMP-9 levels, which are
associated with hemorrhagic transformation.
HBO preconditioning also protects against brain
ischemia–reperfusion injury by upregulating antiox-
idant species.
128,129
Evidence suggests that several
antioxidant genes may act synergistically to remove
ROS through sequential enzymatic reactions. It is
believed that nuclear factor erythroid 2 mediates the
expression of these genes. The relevant enzymes
include heme oxygenase-1, glutathione S-transferase,
Li et al. Hyperglycemia in stroke and possible treatments
486 Neurological Research 2013 VOL.35 NO.5
and NADPH quinone oxidoreductase.
130
Soejima
et al.
128
found that HBO preconditioning significantly
attenuated transient MCAO-induced neurological
deficits, infarct volume, and BBB permeability in
hyperglycemic rodents.
Hypothermia
Hypothermia has long been suspected to have
neuroprotective effects when administered immedi-
ately after stroke and has been a topic of intense
research. In temporary ischemic stroke models,
hypothermia has been shown to reduce infarct size
and improve functional outcome.
131
The physiological
effects of hypothermia are multifaceted. Hypothermia
decreases inflammation,
132–134
neuronal apoptosis,
135
mitochondrial dysfunction,
136
and excitotoxicity.
137
Mild hypothermia (33C) significantly reduces cyto-
chrome ctranslocation into the cytoplasm and Bax
and Bcl-2 levels.
135
Furthermore, hypothermia re-
stores elevated calcium levels after stroke outcome and
reduces calcium dependent degradative pathways
leading to cell death.
138
Ethanol
The efficacy of ethanol therapy as a treatment for
hyperglycemia-induced brain injury post-ischemic
stroke has yet to be tested. However, there is evidence
to suggest that a moderate concentration of etha-
nol treatment may be neuroprotective and can re-
duce hyperglycemia-associated injuries. Previously
we showed that acute ethanol administration re-
duces both infarct lesion volume and hemorrhagic
transformation.
139
One of the pathways by which
ethanol exerts its neuroprotective effects may be
through a reduction of ROS. Thus, in a MCAO
model, we found that a moderate ethanol treatment
(1.5 g/kg) administered immediately upon reperfu-
sion significantly reduced NADPH oxidase and
gp91(phox) in the brain.
140
Furthermore, acute ethanol treatment reduces
disruption of the BBB caused by reperfusion. In the
same model, we showed that ischemic rodents gained
neuroprotection as exhibited by decreased expression
of aquaporins 4 and 9, two important mediators of
cerebral edema post-ischemic stroke, when adminis-
tered moderate concentrations of ethanol immedi-
ately following reperfusion.
92
In addition, using an in
vitro oxygen and glucose deprivation and reoxygena-
tion model, we found that ethanol administration
at a moderate concentration of 30 mM significantly
reduced the levels of MMP-2 and MMP-9 and
increased basal lamina and tight-junction zonula
occludins-1 levels.
92
These effects likely helped to
stabilize the BBB after stroke onset.
Ethanol-derived neuroprotection may constitute one
method of combating hyperglycemiainduced post-
ischemic injuries. Evidence that ethanol treatment
interferes with pathways of ischemic hemorrhagic
transformation, neuronal cell death, and increased
blood vessel permeability shows promise. However,
further studies are needed to determine whether
ethanol’s effect on these post-ischemic injuries could
be clinically significant.
Conclusion
Acutely elevated glucose levels are associated with
poorer outcomes in ischemic stroke patients.
141
Substantial evidence from experimental work and
clinical studies indicate that hyperglycemia further
complicates the metabolic state and mitochondrial
function in the ischemic penumbra, increases both
infarct volume and hemorrhagic transformation, and
worsens outcomes even after thrombolytic therapy.
Using in vivo (e.g. MCAO models) and in vitro (e.g.
OGD) models, investigators have shown hyperglyce-
mia to exacerbate intracellular calcium imbalance
and the accumulation of ROS in endothelial cells.
These intracellular changes are thought to cause
detrimental effects in the penumbral region and the
neurovascular system. In addition, the activation of
both microglia and macrophages in the neuronal
compartment after ischemic stroke aggravate the
extent of injury by disrupting the BBB and causing
degeneration of the white matter.
Although experimental studies have elucidated
several mechanisms by which hyperglycemia influ-
ences the survival of ischemic penumbral tissue,
studies bridging the gap between clinical stroke and
experimental models have been limited. Recently,
MRI data have shown a strong correlation between
loss of penumbral tissue and elevated blood glucose.
3
Although several treatments have been proposed
to ameliorate hyperglycemia-induced injury after
ischemic stroke, their translation from the laboratory
to the bedside has been disappointing thus far. While
insulin therapy to normalize glucose levels has shown
promise in animal models, clinical application of this
strategy in the GIST trial failed to show significant
improvement. Furthermore, the GIST trial revealed
the difficulty with managing patient’s glucose levels in
a clinical setting. Lowering the patient’s glucose levels
using insulin has the potential to induce hypoglyce-
mia, causing more harm than good. Anti-inflamma-
tory therapies have shown paradoxical outcomes; as a
result, non-specific anti-inflammatory therapy has yet
to fulfill its promise. Research is needed to delineate
specific inflammatory pathways and their effects on
ischemia-induced injuries. A better understanding of
the inflammatory pathways involved in ischemic
stroke will allow physicians to inhibit destructive
pathways while promoting those that are neuro-
restorative. Finally, though in its infancy, ethanol ther-
apy is promising, as it exerts significant depressive
Li et al. Hyperglycemiainstrokeandpossibletreatments
Neurological Research 2013 VOL.35 NO.5 487
effect on glucose metabolism and influence on the
neurovascular system. Ethanol therapy is one of the
few treatments that impedes both the metabolic
pathway and the inflammatory pathway to protect
the penumbral tissue. However, more research is
necessary to establish ethanol-derived neuroprotection
in hyperglycemic conditions.
The effect of hyperglycemia on ischemic stroke
outcome is profound and may significantly affect
treatment outcome. Accumulating data suggests
diabetic rodents, when treated with tPA, exhibit
negative side effects including increased BBB perme-
ability and an orders of magnitude increase in
intracerebral hemorrhage volumes compared to
non-hyperglycemic rodents treated with tPA.
142,143
A recent study published by Chen et al. showed that
bone marrow stromal cell treatment of stroke in
diabetic rats precipitated adverse effects, including
increased BBB leakage, cerebral artery neointimal
formation, arteriosclerosis, and worse functional
outcome.
144
Traditional methods known to improve
functional outcome after ischemic stroke have shown
adverse effects when used in hyperglycemic animal
models.
As the diabetic population continues to expand in
both developed and developing countries, the effect
of hyperglycemia post-ischemic stroke should be
investigated. Successful therapeutic intervention for
the large population facing the social, financial, and
health consequences of diabetes and ischemic stroke
will depend on understanding the interplay between
glucose and different systems of the brain, including
the cerebrovascular system, the inflammatory system,
and the metabolic system.
References
1 Rosamond W, Flegal K, Furie K, Go A, Greenlund K, Haase
N, et al. Heart disease and stroke statistics 2008 update: a
report from the American Heart Association Statistics
Committee and Stroke Statistics Subcommittee. Circulation.
2008;117:e25–46.
2 Capes SE, Hunt D, Malmberg K, Pathak P, Gerstein HC.
Stress hyperglycemia and prognosis of stroke in nondiabetic
and diabetic patients: a systematic overview. Stroke.
2001;32:2426–32.
3 Parsons MW, Barber PA, Desmond PM, Baird TA, Darby
DG, Byrnes G, et al. Acute hyperglycemia adversely affects
stroke outcome: a magnetic resonance imaging and spectro-
scopy study. Ann Neurol. 2002;52:20–8.
4 Martini SR, Kent TA. Hyperglycemia in acute ischemic
stroke: a vascular perspective. J Cereb Blood Flow Metab.
2007;27:435–51.
5 Baird TA, Parsons MW, Phanh T, Butcher KS, Desmond PM,
Tress BM, et al. Persistent poststroke hyperglycemia is
independently associated with infarct expansion and worse
clinical outcome. Stroke. 2003;34:2208–14.
6 Ginsberg MD. Hyperglycemia and stroke outcome: vindica-
tion of the ischemic penumbra. Ann Neurol. 2002;52:5–6.
7 Kent TA, Soukup VM, Fabian RH. Heterogeneity affecting
outcome from acute stroke therapy: making reperfusion
worse. Stroke. 2001;32:2318–27.
8 Suarez JI. Does hyperglycemia contribute to secondary injury
in subarachnoid hemorrhage? Stroke. 2006;37:8–9.
9 Song EC, Chu K, Jeong SW, Jung KH, Kim SH, Kim M, et al.
Hyperglycemia exacerbates brain edema and perihematomal
cell death after intracerebral hemorrhage. Stroke.
2003;34:2215–20.
10 Silver IA, Deas J, Erecinska M. Ion homeostasis in brain cells:
differences in intracellular ion responses to energy limitation
between cultured neurons and glial cells. Neuroscience.
1997;78:589–601.
11 Yuan Y, Peng C, Li K, Hussain M, Sikharam C, Guthikonda
M, et al. Ethanol reduces expression of apoptotic proteins
after hypoxia/reoxygenation in a brain slice model. Neurol
Res. 2012;34:373–8.
12 Rosenblum WI. ATP-sensitive potassium channels in the
cerebral circulation. Stroke. 2003;34:1547–52.
13 Klaidman L, Morales M, Kem S, Yang J, Chang ML, Adams
JD, Jr. Nicotinamide offers multiple protective mechanisms in
stroke as a precursor for NAD
z
, as a PARP inhibitor and by
partial restoration of mitochondrial function. Pharmacology.
2003;69:150–7.
14 Yang GY, Chen SF, Kinouchi H, Chan PH, Weinstein PR.
Edema, cation content, and ATPase activity after middle
cerebral artery occlusion in rats. Stroke. 1992;23:1331–6.
15 Hansen AJ, Nedergaard M. Brain ion homeostasis in cerebral
ischemia. Neurochem Pathol. 1988;9:195–209.
16 Hossmann KA. Glutamate-mediated injury in focal cerebral
ischemia: the excitotoxin hypothesis revised. Brain Pathol.
1994;4:23–36.
17 Lipton P. Ischemic cell death in brain neurons. Physiol Rev.
1999;79:1431–568.
18 Luetjens CM, Bui NT, Sengpiel B, Munstermann G, Poppe
M, Krohn AJ, et al. Delayed mitochondrial dysfunction in
excitotoxic neuron death: cytochrome crelease and a
secondary increase in superoxide production. J Neurosci.
2000;20:5715–23.
19JiangX,WangX.Cytochromecpromotes caspase-9
activation by inducing nucleotide binding to APAF-1. J Biol
Chem. 2000;275:31199–203.
20 Krantic S, Mechawar N, Reix S, Quirion R. Apoptosis-
inducing factor: a matter of neuron life and death. Prog
Neurobiol. 2007;81:179–96.
21 Li PA, Shuaib A, Miyashita H, He QP, Siesjo BK, Warner
DS. Hyperglycemia enhances extracellular glutamate accumu-
lation in rats subjected to forebrain ischemia. Stroke.
2000;31:183–92.
22 Araki N, Greenberg JH, Sladky JT, Uematsu D, Karp A,
Reivich M. The effect of hyperglycemia on intracellular
calcium in stroke. J Cereb Blood Flow Metab. 1992;12:469–76.
23 Li PA, Rasquinha I, He QP, Siesjo BK, Csiszar K, Boyd CD,
et al. Hyperglycemia enhances DNA fragmentation after
transient cerebral ischemia. J Cereb Blood Flow Metabolism.
2001;21:568–76.
24 Li P, He QP, Ouyang YB, Liu CL, Hu BR, Siesjo BK. Early
release of cytochrome cand activation of caspase-3 in
hyperglycemic rats subjected to transient forebrain ischemia.
Brain Res. 2001;896:69–76.
25 Belayev L, Zhao W, Busto R, Ginsberg MD. Transient middle
cerebral artery occlusion by intraluminal suture: I. Three-
dimensional autoradiographic imageanalysis of local cerebral
glucose metabolism–blood flow interrelationships during
ischemia and early recirculation. J Cereb Blood Flow
Metabolism. 1997;17:1266–80.
26 Folbergrova J, Zhao Q, Katsura K, Siesjo BK. N-tert-butyl-
alpha-phenylnitrone improves recovery of brain energy state
in rats following transient focal ischemia. Proc Natl Acad Sci
USA. 1995;92:5057–61.
27 Goldberg MP, Choi DW. Combined oxygen and glucose
deprivation in cortical cell culture: calcium-dependent and
calcium-independent mechanisms of neuronal injury. J
Neurosci. 1993;13:3510–24.
28 Pringle AK, Iannotti F, Wilde GJ, Chad JE, Seeley PJ,
Sundstrom LE. Neuroprotection by both NMDA and non-
nmda receptor antagonists in in vitro ischemia. Brain Res.
1997;755:36–46.
29 Turrens JF, Boveris A. Generation of superoxide anion by the
NADH dehydrogenase of bovine heart mitochondria.
Biochem J. 1980;191:421–7.
30 Turrens JF, Alexandre A, Lehninger AL. Ubisemiquinone is
the electron donor for superoxide formation by complex III of
heart mitochondria. Arch Biochem Biophys. 1985;237:408–14.
31 Saeed SA, Shad KF, Saleem T, Javed F, Khan MU. Some new
prospects in the understanding of the molecular basis of the
pathogenesis of stroke. Exp Brain Res 2007;182:1–10.
Li et al. Hyperglycemia in stroke and possible treatments
488 Neurological Research 2013 VOL.35 NO.5
32 Hewett SJ, Uliasz TF, Vidwans AS, Hewett JA.
Cyclooxygenase-2 contributes to N-methyl-D-aspartate-
mediated neuronal cell death in primary cortical cell culture.
J Pharmacol Exp Therap. 2000;293:417–25.
33 Sugawara T, Fujimura M, Noshita N, Kim GW, Saito A,
Hayashi T, et al. Neuronal death/survival signaling pathways
in cerebral ischemia. NeuroRx. 2004;1:17–25.
34 Miller AA, Dusting GJ, Roulston CL, Sobey CG. NADPH-
oxidase activity is elevated in penumbral and non-ischemic
cerebral arteries following stroke. Brain Res. 2006;1111:111–6.
35 Piantadosi CA, Zhang J. Mitochondrial generation of reactive
oxygen species after brain ischemia in the rat. Stroke.
1996;27:327–31; discussion 332.
36 Choi K, Kim J, Kim GW, Choi C. Oxidative stress-induced
necrotic cell death via mitochondira-dependent burst of
reactive oxygen species. Curr Neurovasc Res. 2009;6:213–22.
37 Starkov AA, Fiskum G. Regulation of brain mitochondrial
H
2
O
2
production by membrane potential and NAD(P)H
redox state. J Neurochem. 2003;86:1101–7.
38 Liu SS. Mitochondrial q cycle-derived superoxide and
chemiosmotic bioenergetics. Ann NY Acad Sci.
2010;1201:84–95.
39 Chabrashvili T, Tojo A, Onozato ML, Kitiyakara C, Quinn
MT, Fujita T, et al. Expression and cellular localization of
classic NADPH oxidase subunits in the spontaneously
hypertensive rat kidney. Hypertension. 2002;39:269–74.
40 Cifuentes ME, Pagano PJ. C-Src and smooth muscle Nad(P)H
oxidase: assembling a path to hypertrophy. Arterioscler
Thromb Vasc Biol. 2003;23:919–21.
41 Suh SW, Shin BS, Ma H, van Hoecke M, Brennan AM,
Yenari MA, et al. Glucose and NADPH oxidase drive
neuronal superoxide formation in stroke. Ann Neurol.
2008;64:654–63.
42 Tang XN, Cairns B, Kim JY, Yenari MA. Nadph oxidase in
stroke and cerebrovascular disease. Neurol Res. 2012;34:338–
45.
43 Suh SW, Gum ET, Hamby AM, Chan PH, Swanson RA.
Hypoglycemic neuronal death is triggered by glucose reperfu-
sion and activation of neuronal NADPH oxidase. J Clin
Invest. 2007;117:910–8.
44 Brennan AM, Suh SW, Won SJ, Narasimhan P, Kauppinen
TM, Lee H, et al. NADPH oxidase is the primary source of
superoxide induced by NMDA receptor activation. Nat
Neurosci. 2009;12:857–63.
45 Bertoni JM. Competitive inhibition of rat brain hexokinase by
2-deoxyglucose, glucosamine, and metrizamide. J Neurochem.
1981;37:1523–8.
46 Dietrich WD, Alonso O, Busto R. Moderate hyperglycemia
worsens acute blood–brain barrier injury after forebrain
ischemia in rats. Stroke. 1993;24:111–6.
47 Siemkowicz E. Brain uptake of mannitol and sucrose after
cerebral ischemia: effect of hyperglycemia. Acta Physiol
Scand. 1981;112:359–63.
48 Lee TS, Saltsman KA, Ohashi H, King GL. Activation of
protein kinase C by elevation of glucose concentration:
proposal for a mechanism in the development of diabetic
vascular complications. Proc Natl Acad Sci USA.
1989;86:5141–5.
49 Clarke H, Marano CW, Peralta Soler A, Mullin JM.
Modification of tight junction function by protein kinase C
isoforms. Adv Drug Delivery Rev. 2000;41:283–301.
50 Murakami T, Frey T, Lin C, Antonetti DA. Protein kinase
Cbeta phosphorylates occludin regulating tight junction
trafficking in vascular endothelial growth factorinduced
permeability in vivo. Diabetes. 2012;61:1573–83.
51 Geraldes P, King GL. Activation of protein kinase C isoforms
and its impact on diabetic complications. Circ Res.
2010;106:1319–31.
52 Kawai N, Keep RF, Betz AL. Hyperglycemia and the vascular
effects of cerebral ischemia. Stroke. 1997;28:149–54.
53 Venables GS, Miller SA, Gibson G, Hardy JA, Strong AJ. The
effects of hyperglycaemia on changes during reperfusion
following focal cerebral ischaemia in the cat. J Neurol
Neurosurg Psychiatry. 1985;48:663–9.
54 Yilmaz G, Granger DN. Leukocyte recruitment and ischemic
brain injury. Neuromol Med. 2010;12:193–204.
55 Du XL, Edelstein D, Dimmeler S, Ju Q, Sui C, Brownlee M.
Hyperglycemia inhibits endothelial nitric oxide synthase
activity by posttranslational modification at the AKT site. J
Clin Invest. 2001;108:1341–8.
56 Forstermann U, Munzel T. Endothelial nitric oxide synthase
in vascular disease: from marvel to menace. Circulation.
2006;113:1708–14.
57 Federici M, Menghini R, Mauriello A, Hribal ML, Ferrelli F,
Lauro D, et al. Insulindependent activation of endothelial
nitric oxide synthase is impaired by O-linked glycosylation
modification of signaling proteins in human coronary
endothelial cells. Circulation. 2002;106:466–72.
58 Fleming I, Busse R. Molecular mechanisms involved in the
regulation of the endothelial nitric oxide synthase. Am J
Physiol. 2003;284:R1–12.
59 Ennis SR, Keep RF. Effect of sustained-mild and transient-
severe hyperglycemia on ischemia-induced blood–brain barrier
opening. J Cerebral Blood Flow Metab. 2007;27:1573–82.
60 Wood PL. Microglia as a unique cellular target in the
treatment of stroke: potential neurotoxic mediators produced
by activated microglia. Neurol Res. 1995;17:242–8.
61 Saito S, Matsuura M, Tominaga K, Kirikae T, Nakano M.
Important role of membrane-associated CD14 in the induction
of IFN-beta and subsequent nitric oxide production by murine
macrophages in response to bacterial lipopolysaccharide. Eur
J Biochem. 2000;267:37–45.
62 Beschorner R, Schluesener HJ, Gozalan F, Meyermann R,
Schwab JM. Infiltrating CD14
z
monocytes and expression of
CD14 by activated parenchymal microglia/macrophages con-
tribute to the pool of CD14
z
cells in ischemic brain lesions. J
Neuroimmunol. 2002;126:107–15.
63 Ekdahl CT, Kokaia Z, Lindvall O. Brain inflammation and
adult neurogenesis: the dual role of microglia. Neuroscience.
2009;158:1021–9.
64 Lucas SM, Rothwell NJ, Gibson RM. The role of inflamma-
tion in CNS injury and disease. Br J Pharmacol.
2006;147(Suppl 1):S232–40.
65 Endoh M, Maiese K, Wagner J. Expression of the inducible
form of nitric oxide synthase by reactive astrocytes after
transient global ischemia. Brain Res. 1994;651:92–100.
66 Huang J, Upadhyay UM, Tamargo RJ. Inflammation in
stroke and focal cerebral ischemia. Surg Neurol. 2006;66:232–
45.
67 Springer TA. Traffic signals for lymphocyte recirculation and
leukocyte emigration: the multistep paradigm. Cell.
1994;76:301–14.
68 Colgan SP, Dzus AL, Parkos CA. Epithelial exposure to
hypoxia modulates neutrophil transepithelial migration. J Exp
Med. 1996;184:1003–15.
69 Emsley HC, Smith CJ, Tyrrell PJ, Hopkins SJ. Inflammation
in acute ischemic stroke and its relevance to stroke critical
care. Neurocrit Care. 2008;9:125–38.
70 Jin R, Yang G, Li G. Inflammatory mechanisms in ischemic
stroke: role of inflammatory cells. J Leuk Biol. 2010;87:779–
89.
71 Fernandez-Lopez D, Faustino J, Daneman R, Zhou L, Lee
SY, Derugin N, et al. Blood–brain barrier permeability is
increased after acute adult stroke but not neonatal stroke in
the rat. J Neurosci. 2012;32:9588–600.
72 Belayev L, Busto R, Zhao W, Ginsberg MD. Quantitative
evaluation of blood brain barrier permeability following
middle cerebral artery occlusion in rats. Brain Res.
1996;739:88–96.
73 Dhindsa S, Tripathy D, Mohanty P, Ghanim H, Syed T,
Aljada A, et al. Differential effects of glucose and alcohol on
reactive oxygen species generation and intranuclear nuclear
factor-kappaB in mononuclear cells. Metabolism. 2004;53:
330–4.
74 Barnes PJ, Karin M. Nuclear factor-kappaB: a pivotal
transcription factor in chronic inflammatory diseases. N
Engl J Med. 1997;336:1066–71.
75 Stein B, Baldwin AS, Jr. Distinct mechanisms for regulation of
the interleukin-8 gene involve synergism and cooperativity
between C/EBP and NF-kappa B. Mol Cell Biol.
1993;13:7191–8.
76 Schreck R, Rieber P, Baeuerle PA. Reactive oxygen inter-
mediates as apparently widely used messengers in the
activation of the NF-kappa B transcription factor and HIV-
1. EMBO J. 1991;10:2247–58.
77 Xie QW, Kashiwabara Y, Nathan C. Role of transcription
factor NF-kappa B/REL in induction of nitric oxide synthase.
J Biol Chem. 1994;269:4705–8.
78 Okada Y, Copeland BR, Mori E, Tung MM, Thomas WS, del
Zoppo GJ. Pselectin and intercellular adhesion molecule-1
Li et al. Hyperglycemiainstrokeandpossibletreatments
Neurological Research 2013 VOL.35 NO.5 489
expression after focal brain ischemia and reperfusion. Stroke.
1994;25:202–11.
79 Clark WM, Lauten JD, Lessov N, Woodward W, Coull BM.
Time course of ICAM-1 expression and leukocyte subset
infiltration in rat forebrain ischemia. Mol Chem Neuropathol.
1995;26:213–30.
80 Morigi M, Angioletti S, Imberti B, Donadelli R, Micheletti G,
Figliuzzi M, et a l. Leukocyte–endothelial interaction is
augmented by high glucose concentrations and hyperglycemia
in a NF-kB-dependent fashion. J Clin Invest. 1998;101:1905–
15.
81 Smolock AR, Mishra G, Eguchi K, Eguchi S, Scalia R.
Protein kinase C upregulates intercellular adhesion molecule-1
and leukocyte–endothelium interactions in hyperglycemia via
activation of endothelial expressed calpain. Arterioscler
Thromb Vasc Biol. 2011;31:289–96.
82 Kado S, Wakatsuki T, Yamamoto M, Nagata N. Expression
of intercellular adhesion molecule-1 induced by high glucose
concentrations in human aortic endothelial cells. Life Sci.
2001;68:727–37.
83 Piconi L, Quagliaro L, Da Ros R, Assaloni R, Giugliano D,
Esposito K, et al. Intermittent high glucose enhances ICAM-1,
VCAM-1, E-selectin and interleukin-6 expression in human
umbilical endothelial cells in culture: the role of poly(adpri-
bose) polymerase. J Thromb Haemost. 2004;2:1453–9.
84 Rasmussen LM, Schmitz O, Ledet T. Increased expression of
vascular cell adhesion molecule-1 (VCAM-1) in cultured
endothelial cells exposed to serum from type 1 diabetic
patients: no effects of high glucose concentrations. Scand J
Clin Lab Invest. 2002;62:485–93.
85 Esposito K, Nappo F, Marfella R, Giugliano G, Giugliano F,
Ciotola M, et al. Inflammatory cytokine concentrations are
acutely increased by hyperglycemia in humans: role of
oxidative stress. Circulation. 2002;106:2067–72.
86 Morohoshi M, Fujisawa K, Uchimura I, Numano F. Glucose-
dependent interleukin 6 and tumor necrosis factor production
by human peripheral blood monocytes in vitro. Diabetes.
1996;45:954–9.
87 Morohoshi M, Fujisawa K, Uchimura I, Numano F. The
effect of glucose and advanced glycosylation end products on
IL-6 production by human monocytes. Ann NY Acad Sci.
1995;748:562–70.
88 Pampfer S, Vanderheyden I, de Hertogh R. Increased
synthesis of tumor necrosis factor-alpha in uterine explants
from pregnant diabetic rats and in primary cultures of uterine
cells in high glucose. Diabetes. 1997;46:1214–24.
89 Coughlan MT, Oliva K, Georgiou HM, Permezel JM, Rice
GE. Glucose-induced release of tumour necrosis factor-alpha
from human placental and adipose tissues in gestational
diabetes mellitus. Diabet Med. 2001;18:921–7.
90 Kamada H, Yu F, Nito C, Chan PH. Influence of
hyperglycemia on oxidative stress and matrix metalloprotei-
nase-9 activation after focal cerebral ischemia/reperfusion in
rats: relation to blood–brain barrier dysfunction. Stroke.
2007;38:1044–9.
91 Gidday JM, Gasche YG, Copin JC, Shah AR, Perez RS,
Shapiro SD, et al. Leukocyte-derived matrix metalloprotei-
nase-9 mediates blood–brain barrier breakdown and is
proinflammatory after transient focal cerebral ischemia. Am
J Phys. 2005;289:H558–68.
92 Zeng X, Asmaro K, Ren C, Gao M, Peng C, Ding JY, et al.
Acute ethanol treatment reduces blood–brain barrier dysfunc-
tion following ischemia/reperfusion injury. Brain Res.
2012;1437:127–33.
93 Gasche Y, Copin JC, Sugawara T, Fujimura M, Chan PH.
Matrix metalloproteinase inhibition prevents oxidative stress-
associated blood–brain barrier disruption after transient focal
cerebral ischemia. J Cereb Blood Flow Metab. 2001;21:1393–
400.
94 Schilling M, Besselmann M, Leonhard C, Mueller M,
Ringelstein EB, Kiefer R. Microglial activation precedes and
predominates over macrophage infiltration in transient focal
cerebral ischemia: a study in green fluorescent protein
transgenic bone marrow chimeric mice. Exp Neurol.
2003;183:25–33.
95 Chen J, Cui X, Zacharek A, Cui Y, Roberts C, Chopp M.
White matter damage and the effect of matrix metalloprotei-
nases in type 2 diabetic mice after stroke. Stroke. 2011;42:445–
52.
96 Roriz-Filho SJ, Sa-Roriz TM, Rosset I, Camozzato AL,
Santos AC, Chaves ML, et al. (Pre)diabetes, brain aging, and
cognition. Biochim Biophys Acta. 2009;1792:432–43.
97 Nakaji K, Ihara M, Takahashi C, Itohara S, Noda M,
Takahashi R, et al. Matrix metalloproteinase-2 plays a critical
role in the pathogenesis of white matter lesions after chronic
cerebral hypoperfusion in rodents. Stroke. 2006;37:2816–23.
98 Wallin A, Sjogren M, Edman A, Blennow K, Regland B.
Symptoms, vascular risk factors and blood–brain barrier
function in relation to CT white-matter changes in dementia.
Eur Neurol. 2000;44:229–35.
99 Asahi M, Wang X, Mori T, Sumii T, Jung JC, Moskowitz
MA, et al. Effects of matrix metalloproteinase-9 gene knock-
out on the proteolysis of blood–brain barrier and white matter
components after cerebral ischemia. J Neurosci. 2001;21:7724–
32.
100 Gentile NT, Seftchick MW, Huynh T, Kruus LK, Gaughan J.
Decreased mortality by normalizing blood glucose after acute
ischemic stroke. Acad Emerg Med. 2006;13:174–80.
101 Hamilton MG, Tranmer BI, Auer RN. Insulin reduction of
cerebral infarction due to transient focal ischemia. J
Neurosurg. 1995;82:262–8.
102 Gray CS, Hildreth AJ, Sandercock PA, O’Connell JE,
Johnston DE, Cartlidge NE, et al. Glucose-potassium-insulin
infusions in the management of post-stroke hyperglycaemia:
the UK Glucose Insulin in Stroke Trial (GIST-UK). Lancet
Neurol. 2007;6:397–406.
103 Bruno A, Kent TA, Coull BM, Shankar RR, Saha C, Becker
KJ, et al. Treatment of hyperglycemia in ischemic stroke
(THIS): a randomized pilot trial. Stroke. 2008;39:384–9.
104 Hartell NA, Archer HE, Bailey CJ. Insulin-stimulated
endothelial nitric oxide release is calcium independent and
mediated via protein kinase B. Biochem Pharmacol.
2005;69:781–90.
105 Takahashi S, Mendelsohn ME. Synergistic activation of
endothelial nitric-oxide synthase (eNOS) by HSP90 and
AKT: calcium-independent eNOS activation involves forma-
tion of an HSP90–AKT–CAM-bound eNOS complex. J Biol
Chem. 2003;278:30821–7.
106 Butterworth RJ, Cluckie A, Jackson SH, Buxton-Thomas M,
Bath PM. Pathophysiological assessment of nitric oxide (given
as sodium nitroprusside) in acute ischaemic stroke.
Cerebrovasc Dis. 1998;8:158–65.
107 Dandona P, Aljada A, Mohanty P, Ghanim H, Hamouda W,
Assian E, et al. Insulin inhibits intranuclear nuclear factor
kappaB and stimulates IkappaB in mononuclear cells in obese
subjects: evidence for an anti-inflammatory effect? J Clin
Endocrinol Metab. 2001;86:3257–65.
108 Genovese T, Mazzon E, Paterniti I, Esposito E, Bramanti P,
Cuzzocrea S. Modulation of NADPH oxidase activation in
cerebral ischemia/reperfusion injury in rats. Brain Res.
2011;1372:92–102.
109 Chamorro A, Meisel A, Planas AM, Urra X, van de Beek D,
Veltkamp R. The immunology of acute stroke. Nat Rev.
2012;8:401–10.
110 Moalem G, Leibowitz-Amit R, Yoles E, Mor F, Cohen IR,
Schwartz M. Autoimmune T cells protect neurons from
secondary degeneration after central nervous system axotomy.
Nat Med. 1999;5:49–55.
111 Lewitus GM, Kipnis J, Avidan H, Ben-Nun A, Schwartz M.
Neuroprotection induced by mucosal tolerance is epitope-
dependent: conflicting effects in different strains. J
Neuroimmunol. 2006;175:31–8.
112 Takeda H, Spatz M, Ruetzler C, McCarron R, Becker K,
Hallenbeck J. Induction of mucosal tolerance to E-selectin
prevents ischemic and hemorrhagic stroke in spontaneously
hypertensive genetically stroke-prone rats. Stroke.
2002;33:2156–63.
113 Chen Y, Ruetzler C, Pandipati S, Spatz M, McCarron RM,
Becker K, et al. Mucosal tolerance to E-selectin provides cell-
mediated protection against ischemic brain injury. Proc Natl
Acad Sci USA. 2003;100:15107–12.
114 Bruno A, Liebeskind D, Hao Q, Raychev R. Diabetes
mellitus, acute hyperglycemia, and ischemic stroke. Curr
Treat Options Neurol. 2010;12:492–503.
115 Southerland AM, Johnston KC. Considering hyperglycemia
and thrombolysis in the stroke hyperglycemia insulin network
effort (SHINE) trial. Ann NY Acad Sci. 2012;1268:72–8.
116 Zhu CZ, Auer RN. Intraventricular administration of insulin
and IGF-1 in transient forebrain ischemia. J Cereb Blood
Flow Metab. 1994;14:237–42.
Li et al. Hyperglycemia in stroke and possible treatments
490 Neurological Research 2013 VOL.35 NO.5
117 Cheng HL, Steinway M, Delaney CL, Franke TF, Feldman
EL. IGF-I promotes Schwann cell motility and survival via
activation of AKT. Mol Cell Endocrinol. 2000;170:211–5.
118 Guan J, Bennet L, Gluckman PD, Gunn AJ. Insulin-like
growth factor-1 and post-ischemic brain injury. Prog
Neurobiol. 2003;70:443–62.
119 Darsalia V, Mansouri S, Ortsater H, Olverling A, Nozadze N,
Kappe C, et al. Glucagon-like peptide-1 receptor activation
reduces ischaemic brain damage following stroke in type 2
diabetic rats. Clin Sci (Lond). 2012;122:473–83.
120 Drucker DJ, Nauck MA. The incretin system: glucagon-like
peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibi-
tors in type 2 diabetes. Lancet. 2006;368:1696–705.
121 Holst JJ. On the physiology of gip and GLP-1. Horm Metab
Res. 2004;36:747–54.
122 Kastin AJ, Akerstrom V. Entry of exendin-4 into brain is
rapid but may be limited at high doses. Int J Obesity Relat
Metab Disord. 2003;27:313–8.
123 Gejl M, Egefjord L, Lerche S, Vang K, Bibby BM, Holst JJ,
et al. Glucagon-like peptide-1 decreases intracerebral glucose
content by activating hexokinase and changing glucose
clearance during hyperglycemia. J Cereb Blood Flow Metab.
2012;32:2146–52.
124 Teramoto S, Miyamoto N, Yatomi K, Tanaka Y, Oishi H,
Arai H, et al. Exendin-4, a glucagon-like peptide-1 receptor
agonist, provides neuroprotection in mice transient focal
cerebral ischemia. J Cereb Blood Flow Metab. 2011;
31:1696–705.
125 Iwai T, Ito S, Tanimitsu K, Udagawa S, Oka J. Glucagon-like
peptide-1 inhibits LPS-induced IL-1beta production in cul-
tured rat astrocytes. Neurosci Res. 2006;55:352–60.
126 Qin Z, Karabiyikoglu M, Hua Y, Silbergleit R, He Y, Keep
RF, et al. Hyperbaric oxygen-induced attenuation of hemor-
rhagic transformation after experimental focal transient
cerebral ischemia. Stroke. 2007;38:1362–7.
127 Veltkamp R, Siebing DA, Sun L, Heiland S, Bieber K, Marti
HH, et al. Hyperbaric oxygen reduces blood–brain barrier
damage and edema after transient focal cerebral ischemia.
Stroke. 2005;36:1679–83.
128 Soejima Y, Ostrowski RP, Manaenko A, Fujii M, Tang J,
Zhang JH. Hyperbaric oxygen preconditioning attenuates
hyperglycemia enhanced hemorrhagic transformation after
transient MCAO in rats. Med Gas Res. 2012;2:9.
129 Wada K, Miyazawa T, Nomura N, Tsuzuki N, Nawashiro H,
Shima K. Preferential conditions for and possible mechanisms
of induction of ischemic tolerance by repeated hyperbaric
oxygenation in gerbil hippocampus. Neurosurgery.
2001;49:160–6; discussion 166–7.
130 Godman CA, Chheda KP, Hightower LE, Perdrizet G, Shin
DG, Giardina C. Hyperbaric oxygen induces a cytoprotective
and angiogenic response in human microvascular endothelial
cells. Cell Stress Chaper. 2010;15:431–42.
131 Zausinger S, Hungerhuber E, Baethmann A, Reulen H,
Schmid-Elsaesser R. Neurological impairment in rats after
transient middle cerebral artery occlusion: a comparative
study under various treatment paradigms. Brain Res.
2000;863:94–105.
132 Dietrich WD, Chatzipanteli K, Vitarbo E, Wada K, Kinoshita
K. The role of inflammatory processes in the pathophysiology
and treatment of brain and spinal cord trauma. Acta
Neurochirurg. 2004;89:69–74.
133 Suehiro E, Fujisawa H, Akimura T, Ishihara H, Kajiwara K,
Kato S, et al. Increased matrix metalloproteinase-9 in blood in
association with activation of interleukin-6 after traumatic
brain injury: influence of hypothermic therapy. J
Neurotrauma. 2004;21:1706–11.
134 Deng H, Han HS, Cheng D, Sun GH, Yenari MA. Mild
hypothermia inhibits inflammation after experimental stroke
and brain inflammation. Stroke. 2003;34:2495–501.
135 Xu L, Yenari MA, Steinberg GK, Giffard RG. Mild
hypothermia reduces apoptosis of mouse neurons in vitro
early in the cascade. J Cereb Blood Flow Metab. 2002;22:21–8.
136 Gong P, Li CS, Hua R, Zhao H, Tang ZR, Mei X, et al. Mild
hypothermia attenuates mitochondrial oxidative stress by
protecting respiratory enzymes and upregulating MNSOD in
a pig model of cardiac arrest. PloS ONE. 2012;7:e35313.
137 Tseng EE, Brock MV, Lange MS, Troncoso JC, Blue ME,
Lowenstein CJ, et al. Glutamate excitotoxicity mediates
neuronal apoptosis after hypothermic circulatory arrest. Ann
Thorac Surg. 2010;89:440–5.
138 Phillips KF, Deshpande LS, Delorenzo RJ. Hypothermia
reduces calcium entry via the N-methyl-D-aspartate and
ryanodine receptors in cultured hippocampal neurons. Eur J
Pharmacol. 2012;698:186–92.
139 Wang F, Wang Y, Geng X, Asmaro K, Peng C, Sullivan JM,
et al. Neuroprotective effect of acute ethanol administration in
a rat with transient cerebral ischemia. Stroke. 2012;43:205–
10.
140 Kochanski R, Peng C, Higashida T, Geng X, Hu¨ttemann M,
Guthikonda M, Ding Y. Neuroprotection conferred by post-
ischemia ethanol therapy in experimental stroke: an inhibitory
effect on hyperglycolysis and NADPH oxidase activation. J
Neurochem. 2013; 125(2), doi: 10.1111/jnc.12169. Epub 2013
Jan 27.
141 Poppe AY, Majumdar SR, Jeerakathil T, Ghali W, Buchan
AM, Hill MD, et al. Admission hyperglycemia predicts a
worse outcome in stroke patients treated with intravenous
thrombolysis. Diabetes Care. 2009;32:617–22.
142 Won SJ, Tang XN, Suh SW, Yenari MA, Swanson RA.
Hyperglycemia promotes tissue plasminogen activator-
induced hemorrhage by increasing superoxide production.
Ann Neurol. 2011;70:583–90.
143 Ning R, Chopp M, Yan T, Zacharek A, Zhang C, Roberts C,
et al. Tissue plasminogen activator treatment of stroke in type-
1 diabetes rats. Neuroscience. 2012;222:326–32.
144 Chen J, Ye X, Yan T, Zhang C, Yang XP, Cui X, et al.
Adverse effects of bone marrow stromal cell treatment of
stroke in diabetic rats. Stroke. 2011;42:3551–8.
Li et al. Hyperglycemiainstrokeandpossibletreatments
Neurological Research 2013 VOL.35 NO.5 491
... При експериментални и клинични изследвания при инсулт е доказано, че хипергликемията е вредна за пенумбралната тъкан по няколко причини: на първо място тя повишава както калциевия дисбаланс, така и натрупването на реактивни кислородни видове (ROS) в невроните, което води до повишена апоптоза; захранва производството на анаеробна енергия, причинявайки лактатна ацидоза, която допълнително стресира невроните в пенумбралните области; намалява перфузията след ИМИ, понижавайки нивото на азотен оксид (NO), който е решаващ медиатор на вазодилатацията, и на последно място засилва възпалителната реакция след инсулт, причинявайки оток и кръвоизлив чрез нарушаване на КМБ и разграждане на бялото мозъчно вещество, което води до влошаване на функционалните резултати (28). ...
... Преобладаващото становище е, че инсулинът е свързан с подобрение на болничните резултати индиректно, контролирайки нивата на КГ, но все повече доказателства се натрупват за налични директни благоприятни ефекти на инсулина, независими от влиянието му върху КГ (14). Експериментални и клинични модели демонстрират, че в допълнение към хипогликемизиращия си ефект инсулинът упражнява и невропротективен ефект върху мозъчната съдова система увеличавайки синтеза на NO (20), намалява натрупването на ROS чрез понижаване на активността на NADPH оксидазата (28), както и може да има пряк невропротективен ефект върху паренхима на ЦНС чрез намаляване на възпалението (15). Все пак, предвид това, че се предполага общо противовъзпалително действие на инсулина (14), трябва да се вземе под внимание фактът, че едно широкоспектърно потискане на възпалителния отговор може парадоксално да ограничи възстановяването поради съществената роля на имунните клетки и възпалението за възстановяването на тъканите (28). ...
... Експериментални и клинични модели демонстрират, че в допълнение към хипогликемизиращия си ефект инсулинът упражнява и невропротективен ефект върху мозъчната съдова система увеличавайки синтеза на NO (20), намалява натрупването на ROS чрез понижаване на активността на NADPH оксидазата (28), както и може да има пряк невропротективен ефект върху паренхима на ЦНС чрез намаляване на възпалението (15). Все пак, предвид това, че се предполага общо противовъзпалително действие на инсулина (14), трябва да се вземе под внимание фактът, че едно широкоспектърно потискане на възпалителния отговор може парадоксално да ограничи възстановяването поради съществената роля на имунните клетки и възпалението за възстановяването на тъканите (28). Въпреки доказателствата в подкрепа на благоприятното въздействие на противовъзпалителния отговор върху тъканното увреждане по време на острата фаза, противодействието на този отговор може всъщност да влоши дългосрочните резултати, като намали цялостното възстановяване и реорганизация на тъканите (28). ...
Article
Any kind of stress to the body can cause an abrupt increase in glucose levels. This gives reason to define the concept of stress hyperglycaemia (SH). In general, it refers to a sudden increase in blood glucose (BG) from normal levels in connection with the acute condition. Stress hyperglycaemia is associated with adverse outcomes in various categories of acutely ill patients, and this influence is particularly pronounced in those who are not adapted to chronically high BG levels. In this regard, acute ischemic stroke is often accompanied by SH, which can have an impact on its course and outcome. Blood glucose, as a modifiable risk factor for stroke that can be therapeutically affected, demands that we know the impact of abnormal glucose levels, the consequences they are associated with, and the prognostic power they carry.
... The percentages of hyperglycemic stroke patients on admission range from 50% to 75%, although this percentage varies among researchers. The wide range in the incidence of hyperglycemia may also be due to the different glucose lower limits as set by different authors [13,15,21]. ...
... Hyperglycemia is also a risk factor of stroke, as diabetic patients are more likely to suffer from cerebrovascular accidents as compared to non-diabetic population. It has negative impact on brain tissues and is linked with poor outcomes (12). A previously reported included 416 stroke patients and showed significantly higher mortality in patients who were hyperglycaemic (13). ...
Article
Full-text available
Hyponatremia and hyperglycemia are common metabolic disorders which could worsen the outcome after a cerebrovascular accident (stroke). This study aimed to evaluate the impact of hyponatremia and hyperglycemia on stroke and its clinical outcome, including morbidity and mortality. This prospective cohort study included 50 stroke patients admitted in tertiary care hospitals. Stroke severity, Glasgow coma scale (GCS), conscious level, gag reflex, clinical presentations, and outcomes were recorded. Following a stroke, patients were monitored for a period of six months, during which the patient's death was recorded on the event form. Linear logistic regression model was used to determine the stroke mortality. Hyponatremia and hyperglycaemia were associated with higher odds ratio of deaths, poor functional recovery, severe disability, low mean GCS score as compared to normonatremic and normoglycemic patients (odds ratio [OR] = 4.7; 95% CI= 0.929-43.782) and hyperglycaemia (OR= 2.74; 95% CI=0.577-13.03). Stroke patients admitted with hyperglycemia or hyponatremia were associated with greater morbidity and mortality and poor functional recovery.
... Hyperglycemia is also a risk factor of stroke, as diabetic patients are more likely to suffer from cerebrovascular accidents as compared to non-diabetic population. It has negative impact on brain tissues and is linked with poor outcomes (12). A previously reported included 416 stroke patients and showed significantly higher mortality in patients who were hyperglycaemic (13). ...
... The liver plays a crucial role in maintaining energy and glucose metabolism, with stress hyperglycemia concurring in around one-third of individuals after an acute ischemic stroke (AIS). [4][5][6][7] This stress hyperglycemia stems from an inflammatory hepatic pathway that promotes hepatic gluconeogenesis and reduces insulin sensitivity, resulting from the arousal of autonomic nervous system and the hypothalamic-pituitary axis during AIS. [8][9][10] Stress-induced hyperglycemia peaks within 24 h and normalizes by the 3 rd day. ...
Article
Acute ischemic stroke (AIS) not only affects the brain but also has significant implications for peripheral organs through neuroendocrine regulation. This reciprocal relationship influences overall brain function and stroke prognosis. Recent research has highlighted the importance of poststroke liver changes in determining patient outcomes. In our previous study, we investigated the relationship between stroke and liver function. Our findings revealed that the prognostic impact of stress-induced hyperglycemia in patients undergoing acute endovascular treatment for acute large vessel occlusion is closely related to their preexisting diabetes status. We found that the liver contributes to stress hyperglycemia after AIS by increasing hepatic gluconeogenesis and decreasing hepatic insulin sensitivity. These changes are detrimental to the brain, particularly in patients without diabetes. Furthermore, we examined the role of bilirubin, a byproduct of hepatic hemoglobin metabolism, in stroke pathophysiology. Our results demonstrated that blood bilirubin levels can serve as predictors of stroke severity and may hold therapeutic potential for reducing oxidative stress-induced stroke injury in patients with mild stroke. These results underscore the potential role of the liver in the oxidative stress response following AIS, paving the way for further investigation into liver-targeted therapeutic strategies to improve stroke prognosis and patient outcomes.
... Studi kultur sel in vitro menunjukkan bahwa lingkungan hiperglikemi dapat menurunkan ekspresi gen eNOS. 17,20 Penelitian lanjutan berskala lebih besar dan lebih detail perlu untuk dilakukan akibat masih terdapatnya kekurangan-kekurangan pada penelitian ini. Mengenai penentuan skor luaran fungsional dengan menggunakan skor mRS pada penelitian ini hanya mencatatnya dari data dalam rekam medis sehingga memungkinkan adanya ketidaksesuaian data yang tertulis dengan keadaan subjek yang sesungguhnya, pada penelitian selanjutnya yang ingin menggunakan skor mRS dengan desain penelitian serupa, sebaiknya keadaan subjek penelitian dapat langsung ditanyakan dengan menghubungi pihak keluarga subjek melalui kontak pada rekam medis. ...
Article
Full-text available
ABSTRAK Hiperglikemi kerap ditemukan pada pasien stroke iskemik dan dapat disebabkan oleh respon stres akut, diabetes yang telah diketahui maupun yang belum diketahui sebelumnya, atau keadaan pre-diabetes akibat terganggunya metabolisme glukosa. Peran hiperglikemi pada stroke iskemik fase akut yang mengarah pada buruknya luaran fungsional belum diketahui secara pasti, namun beberapa studi telah menemukan bukti-bukti bahwa hiperglikemi selama iskemia otak akut dapat memperparah dan mengganggu proses perbaikan sel otak. Penelitian ini bertujuan untuk melihat hubungan antara hiperglikemi 72 jam pertama dengan luaran fungsional pada penderita stroke iskemik akut di RSUP Sanglah Denpasar. Studi ini merupakan studi tipe analitik dengan menggunakan metode potong lintang. Sampel diambil dengan teknik consecutive non-random sampling dari data rekam medis pasien stroke iskemik yang tercatat di RSUP Sanglah Denpasar yang telah memenuhi kriteria inklusi dan eksklusi. Sebanyak 52 pasien stroke iskemik akut (50% laki-laki dan umur 63,02 ± 7,15 tahun) diikutsertakan dalam penelitian ini, 26,9% diantaranya memiliki kadar gula darah yang tinggi dan 55,9% dari seluruh sampel memiliki luaran fungsional buruk. Studi ini mengungkapkan bahwa derajat gula darah fase akut yang tinggi memiliki hubungan bermakna dengan derajat luaran fungsional yang buruk pada pasien stroke iskemik yang ditunjukkan dari hasil uji analisis regresi logistik biner dengan nilai p = 0,015 dan adjusted odd ratio sebesar 14,975 (IK 95% 1,70-131,55). ABSTRACT Hyperglycemia is often found in ischemic stroke patients and can be caused by acute stress response, known or unknown diabetes, or pre-diabetes conditions due to impaired glucose metabolism. The contribution of hyperglycemia in causing poor outcomes in acute phase ischemic stroke is not yet known, but several studies have found evidence that hyperglycemia during acute brain ischemia can aggravate and disrupt the process of repairing brain cells. The objective of this study is to observe the correlation between hyperglycemia in first 72 hours with functional outcome in acute ischemic stroke patients at Sanglah Hospital Denpasar. This study is an analytic type study using a cross-sectional method. Samples were taken by consecutive non-random sampling technique from the medical record data of stroke patients that treated at Sanglah General Hospital Denpasar who already fulfill the inclusion and exclusion criteria. A total of 52 acute ischemic stroke patients (50% male and age 63.02 ± 7.15 years) were included in this study, 26.9% of them had high blood sugar levels and 55.9% of all samples had poor functional outcomes. This study revealed that the acute phase of high blood sugar degree has a significant relationship with poor functional outcome in ischemic stroke patients as shown by the results of the binary logistic regression study with p = 0.015 and adjusted odd ratio of 14.975 (95% CI 1.70-131.55).
... Hyperglycemia is found in up to 36.4% of patients with cerebral infarction, most of which are large arterial occlusions. Non-diabetic stroke patients with SIH have a worse prognosis than those with normal glucose levels and those with a history of diabetes [42]. It was reported that excessive glucose level is detrimental to brain structure and metabolism [33]. ...
Article
Full-text available
The aim of this study was to systematically evaluate the incidence of stress-induced hyperglycemia (SIH) in acute ischemic stroke (AIS). Studies that reported SIH incidence in AIS and examined risk factors for SIH and non-SIH patients were systematically searched in PubMed, Embase, Cochrane Library, and Web of Science from the inception of each database to December 2021. Article screening and data extraction were performed by two independent reviewers according to the inclusion and exclusion criteria. The quality of the included studies was assessed using the Newcastle–Ottawa Scale (NOS), and meta-analysis was performed using Stata. A total of 13 studies involving 4552 patients (977 in the SIH group and 3575 in the non-SIH group) were included. Meta-analysis showed that the incidence of SIH was 24% (95% CI: 21–27%) in the total population, 33% (14–52%) in North America, 25% (20–29%) in Europe, and 21% (12–29%) in Asia. Subgroup analysis by year of publication revealed that the pooled incidence of SIH was 27% (22–32%) in studies published before 2010 and 19% (14–24%) in those published after 2010. SIH is relatively common in AIS and poses a serious public health problem. Therefore, more emphasis should be placed on the prevention and control of SIH in AIS.
Article
Full-text available
Background Stroke represents a prominent global health issue, exhibiting the third highest incidence of disability and a significant burden on both healthcare and the economy. Stress hyperglycemia, an acute reaction of the hypothalamic-pituitary-adrenal axis and the sympathetic nervous system, leading to adverse outcomes and mortality. Several previous studies have indicated that stress hyperglycemia, as evaluated by the stress hyperglycemia ratio (SHR), significantly increases the risk of adverse outcomes and mortality in stroke patients. However, there is a lack of further investigation into the influence of dynamic changes in stress hyperglycemia on the clinical outcomes of acute ischemic stroke (AIS) patients. Consequently, we performed a meticulous analysis, considering dose-response relationships from existing studies, to ascertain the correlation between dynamic changes in stress hyperglycemia and the susceptibility to adverse outcomes in patients with AIS. Methods This investigation was prospectively registered in PROSPERO and adhered to the PRISMA guidelines. A comprehensive search was performed across English and Chinese databases. A two-sided random-effects model was employed to consolidate the odds ratios (ORs) of the highest vs. lowest categories of SHR. Restricted cubic spline (RCS) models were employed to estimate potential non-linear trends between SHR and the risk of adverse outcomes in AIS patients. Egger's test was utilized to assess publication bias. Heterogeneity was evaluated using Cochran's Q-test. The Newcastle-Ottawa Scale (NOS) tool was employed to evaluate the risk of bias of the included studies. Results The final analysis incorporated a total of thirteen studies, which were published between 2019 and 2023, encompassing a participant cohort of 184,179 individuals. The SHR exhibited a significant association with the risk of various adverse outcomes. Specifically, a higher SHR was correlated with a 2.64-fold increased risk of 3-month poor functional outcomes (OR: 2.64, 95% CI 2.05–3.41, I² = 52.3%, P < 0.001), a 3.11-fold increased risk of 3-month mortality (OR: 3.11, 95% CI 2.10–4.59, I² = 38.6%, P < 0.001), a 2.80-fold increased risk of 1-year mortality (OR: 2.80, 95% CI 1.81–4.31, I² = 88%, P < 0.001), a 3.90-fold increased risk of intracerebral hemorrhage (ICH) and 4.57-fold increased risk of symptomatic ICH (sICH) (ICH-OR: 3.90, 95% CI 1.52–10.02, I² = 84.3%, P = 0.005; sICH-OR: 4.57, 95% CI 2.05–10.10, I² = 47.3%, P < 0.001), a 1.73-fold increased risk of neurological deficits (OR: 1.73, 95 CI 1.44–2.08, I² = 0%, P < 0.001), and a 2.84-fold increased risk of stroke recurrence (OR: 2.84, 95 CI 1.48–5.45, I² = 50.3%, P = 0.002). It is noteworthy that, except for hemorrhagic transformation (HT) and stroke recurrence, the remaining adverse outcomes exhibited a “J-shaped” non-linear dose-response relationship. Conclusion In summary, our findings collectively suggest that increased exposure to elevated SHR is robustly linked to a heightened risk of adverse outcomes and mortality in individuals with AIS, exhibiting a non-linear dose-response relationship. These results underscore the significance of SHR as a predictive factor for stroke prognosis. Therefore, further investigations are warranted to explore the role of SHR in relation to adverse outcomes in stroke patients from diverse ethnic populations. Furthermore, there is a need to explore the potential benefits of stress hyperglycemia control in alleviating the physical health burdens associated with AIS. Maintaining a lower SHR level may potentially reduce the risk of adverse stroke outcomes. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier: CRD42023424852.
Article
Diabetes is known to cause cognitive impairments through various mechanisms, including oxidative stress, inflammation, and apoptosis. Humanin (HN) has been shown to have protective effects on cognitive impairments induced by factors such as Aβ, muscarinic receptor antagonists, and aging in rodents. However, the mechanisms underlying the protective effects of HN in the prefrontal cortex and hippocampus in the context of diabetes are not well understood. In this study, we aimed to investigate the potential protective role of HN on oxidative stress, inflammation, and apoptosis in mice with diabetes. We divided the mice into four groups, including a control group (treated with saline), a humanin group (treated with 4mg/kg of HN), a streptozotocin (STZ) group (diabetic control), and an STZ+Humanin group. The mice were administered HN daily for 15 days. Our results showed that in the prefrontal cortex and hippocampus of the diabetes group, oxidative stress parameters, pro-inflammatory cytokines, apoptosis and, blood glucose levels were increased, while antioxidant and anti-inflammatory cytokines were diminished compared to the control group. However, HN treatment was able to modulate these markers, including blood glucose and the markers of oxidative stress, inflammation, and apoptosis. In conclusion, our findings suggest that hyperglycemia, oxidative stress, inflammation, and apoptosis may contribute to the development of diabetes-induced cognitive impairments. By regulating these changes with HN treatment, we may be able to positively contribute to the treatment of cognitive impairments induced by diabetes. DATA AVAILABILITY: Data will be made available on request.
Chapter
Diabetes mellitus (DM) is a major risk factor for cerebrovascular disease, particularly ischemic stroke. Hyperglycemia and insulin resistance promote functional and structural changes in the cerebral vasculature and induce a prothrombotic state resulting in micro- and macrovascular injury. The clinical correlates are overt stroke and vascular cognitive impairment and dementia. Diabetic patients have a high prevalence of comorbid vascular risk factors necessitating a careful and comprehensive approach to cardiovascular risk mitigation; the choice of hypoglycemic agents should be tailored to each patient’s circumstances. Hyperglycemia is detrimental in the acute phase of stroke but clinical trials of tight glycemic control regimens have not yielded the expected benefits. An individualized approach with emphasis on avoiding wide excursions and excessive glycemic corrections with hypoglycemic events is the mainstay of glycemic control in the acute phase of stroke.KeywordsStrokeCerebrovascular diseaseVascular cognitive impairmentDementiaBrain atrophyHyperglycemia
Article
Full-text available
The promoter of the murine gene encoding inducible nitric oxide synthase (iNOS) contains an NF-kappa B site beginning 55 base pairs upstream of the TATA box, designated NF-kappa Bd. Reporter constructs containing truncated promoter regions, when transfected into macrophages, revealed that NF-kappa Bd is necessary to confer inducibility by bacterial lipopolysaccharide (LPS). Oligonucleotide probes containing NF-kappa Bd plus the downstream 9 or 47 base pairs bound proteins that rapidly appeared in the nuclei of LPS-treated macrophages. The nuclear proteins bound to both probes in an NF-kappa Bd-dependent manner, but binding was resistant to cycloheximide only for the shorter probe. The proteins binding both probes reacted with antibodies against p50 and c-rel but not RelB; those binding the shorter probe also reacted with anti-RelA (p65). Pyrrolidine dithiocarbamate, which acts as a specific inhibitor of NF-kappa B, blocked both the activation of the NF-kappa Bd-binding proteins and the production of NO in LPS-treated macrophages. Thus, activation of NF-kappa Bd/Rel is critical in the induction of iNOS by LPS. However, additional, newly synthesized proteins contribute to the NF-kappa Bd-dependent transcription factor complex on the iNOS promoter in LPS treated mouse macrophages.
Article
Full-text available
Hypothermia is a powerful neuroprotective method when induced following cardiac arrest, stroke, and traumatic brain injury. The physiological effects of hypothermia are multifaceted and therefore a better knowledge of its therapeutic targets will be central to developing innovative combination therapies to augment the protective benefits of hypothermia. Altered neuronal calcium dynamics have been implicated following stroke, status epilepticus and traumatic brain injury. This study was therefore initiated to evaluate the effect of hypothermia on various modes of calcium entry into a neuron. Here, we utilized various pharmacological agents to stimulate major routes of calcium entry in primary cultured hippocampal neurons. Fluorescent calcium indicator Fura-2AM was used to compare calcium ratio under normothermic (37°C) and hypothermic (31°C) conditions. The results of this study indicate that hypothermia preferentially reduces calcium entry through N-methyl-D-aspartate receptors and ryanodine receptors. Hypothermia, on the other hand, did not have a significant effect on calcium entry through the voltage-dependent calcium channels or the inositol tri-phosphate receptors. The ability of hypothermia to selectively affect both N-methyl-D-aspartate receptors and ryanodine receptors-mediated calcium systems makes it an attractive intervention for alleviating calcium elevations that are present following many neurological injuries.
Article
This commentary addresses the article, “Ubisemiquinone is the electron donor for superoxide formation by complex III of heart mitochondria,” Arch. Biochem. Biophys. (1985) 237:408-14. It was part of a series on articles addressing the role of ubisemiquinone in mitochondrial superoxide production.
Article
OBJECTIVE: We reported previously that repeated hyperbaric oxygenation (HBO) as pretreatment induced ischemic tolerance in the gerbil hippocampus. This study was conducted to determine the preferential conditions for induction of ischemic tolerance by HBO and the mechanism of this induction through immunohistochemical analysis of Bcl-2, Bax, and manganese superoxide dismutase expression. METHODS: Five-minute forebrain ischemia was produced in gerbils after pretreatment with 2 atmospheres absolute (ATA) HBO once every other day for one, three, or five sessions, 2 ATA hyperbaric air once every other day for five sessions, or 3 ATA HBO once daily for 10 sessions. Histological examinations were then performed. Two days after pretreatment with 2 ATA HBO once every other day for five sessions or with 3 ATA HBO once daily for 10 sessions, sections were analyzed immunohistochemically. RESULTS: Pretreatment with 2 ATA HBO once every other day for three or five sessions induced ischemic tolerance; however, pretreatment with 2 ATA HBO for one session, 2 ATA hyperbaric air once every other day for five sessions, or 3 ATA HBO once daily for 10 sessions did not. Pretreatment with 2 ATA HBO once every other day for five sessions, but not with 3 ATA HBO once daily for 10 sessions, significantly increased Bcl-2 and manganese superoxide dismutase immunoreactivity in the CA1 sector. CONCLUSION: These results suggest that protection against mitochondrial alterations after ischemia through manganese superoxide dismutase and/or Bcl-2 expression may be related to induction of ischemic tolerance by repeated HBO pretreatment.
Article
Ethanol provides neuroprotection following ischemia/reperfusion. This study assessed ethanol's effect on hyperglycolysis and NADPH oxidase (NOX) activation. Adult, male Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO) for 2 h. Three sets of experiments were conducted to determine ethanol's effect on (i) conferring neuroprotection by measuring infarct volume and neurological deficits 24 h post reperfusion; (ii) cerebral glucose metabolism and lactic acidosis by measuring brain and blood glucose concentrations and protein expression of glucose transporter 1 and 3 (GLUT1, GLUT3), phosphofructokinase (PFK), as well as lactic acidosis by measuring lactate dehydrogenase (LDH), and lactate; and (iii) nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) activation by detecting enzymatic activity and subunit expression at 3 h after reperfusion. When administered upon reperfusion, ethanol (1.5 g/kg) reduced infarct volume by 40% (p < 0.01) and neurological deficits by 48% at 24 h post reperfusion while reducing (p < 0.01) elevations in glycolytic protein expression and lactate levels during early reperfusion (3 h). Ethanol increased the reductions in cerebral glucose concentration at 3 h post reperfusion by 64% (p < 0.01) while enhancing (p < 0.01) post stroke blood glucose concentration, suggesting a reduced cellular glucose uptake and utilization. Ethanol decreased (p < 0.01) stroke-induced NOX activation by reducing enzymatic activity and gp91 phox expression by 45% and 38%, respectively. Post-ischemia ethanol treatment exerts neuroprotection through attenuation of hyperglycolysis and associated NOX activation. Because of the lack of associated hypoglycemia and selectivity toward decreasing cerebral metabolism, further investigation of ethanol's use as a post-stroke therapy, especially in the context of hyperglycemia, seems warranted.
Article
Insulin-like growth factor-1 (IGF-1) is a naturally occurring neurotrophic factor that plays an important role in promoting cell proliferation and differentiation during normal brain development and maturation. The present review examines recent evidence that endogenous IGF-1 also plays a significant role in recovery from insults such as hypoxia-ischemia and that giving additional exogenous IGF-1 can actively ameliorate damage. It is now well established that neurons and other cell types die many hours or even days after initial injury due to activation of programmed cell death pathways. IGF-1 and its binding proteins and receptors are intensely induced within damaged brain regions following brain injury, suggesting a possible a role for IGF-1 in brain recovery. Exogenous administration of IGF-1 within a few hours after brain injury is now known to be protective in both gray and white matter and leads to improved somatic function. In contrast, pre-treatment is ineffective, likely reflecting limited intracerebral penetration of IGF-1 into the uninjured brain. The neuroprotective effects of IGF-1 are mediated by IGF-1 receptors and its binding proteins and are specific to particular cellular phenotypes and brain regions. The window of opportunity for treatment with IGF-1 is limited to a few hours after normothermic brain injury, reflecting its specific actions on early, intracellular events in the apoptotic cascade. However, injury-associated mild post-hypoxic hypothermia, which delays the development of cell death, can shift and dramatically extend the window of opportunity for delayed treatment with IGF-1. Such a combined approach is likely to be essential for any clinical treatment.