Sei sulla pagina 1di 13

Review

Hyperglycemia in stroke and possible


treatments
William A. Li1, Shannon Moore-Langston1, Tia Chakraborty1, Jose A. Rafols2,
Alana C. Conti1,3, Yuchuan Ding1
1
Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA, 2Department
of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI, USA, 3Research 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 Elevated blood glucose, defined as levels greater


Stroke is a major cause of mortality and morbidity in than 6.0 mmol/l (108 mg/dl), has been observed in
America, with an annual combined direct and over one-third of all ischemic stroke patients.
indirect cost exceeding 68 billion dollars.1 In recent Furthermore, evidence in experimental animal stroke
years, accumulating literature supports the notion models supports the observation that hyperglycemia
that persisting hyperglycemia leads to an unfavorable not only exacerbates stroke related injury but that it
prognosis in stroke patients. While diabetes mellitus also adversely affects overall functional outcome.3,4
is a well-known risk factor contributing to poor In this review, the mechanisms by which hyperglyce-
prognosis after ischemic strokes, hyperglycemia with- mia exacerbates negative outcomes in ischemic stroke
out pre-existing diabetes mellitus is also linked to and several potential treatments are discussed.
increased mortality and morbidity after stroke. In
fact, epidemiologic meta-analysis of 30-day post- Mechanisms of Hyperglycemia-Mediated
stroke mortality revealed a greater than three-fold Damage in Ischemic Stroke
Following experimental ischemic stroke, hyperglyce-
increase for non-diabetic hyperglycemic patients,
mic animals generally tend to have higher incidence
compared with a two-fold increase for hyperglycemic
of brain edema, cell death, hemorrhagic transforma-
patients with diabetes mellitus.2
tion, and larger infarct volume.3,5 Possible mechan-
isms include intracellular acidification, reactive
oxygen species (ROS) accumulation, blood-brain
Correspondence to: Y. C. Ding, Department of Neurological Surgery, barrier (BBB) disruption, inflammatory response
Wayne State University School of Medicine, 550 E Canfield, Detroit, MI
48201, USA. Email: yding@med.wayne.edu induction, and axonal degradation.6–9

ß W. S. Maney & Son Ltd 2013


DOI 10.1179/1743132813Y.0000000209 Neurological Research 2013 VOL . 35 NO . 5 479
Li et al. Hyperglycemia in stroke and possible treatments

Figure 1 Elevated cell apoptosis due to calcium imbalance. Excess glucose causes calcium imbalance and exacerbates
cytochrome c leakage into the cytosol. Cytochrome c then 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.

480 Neurological Research 2013 VOL . 35 NO . 5


Li et al. Hyperglycemia in stroke and possible treatments

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.

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 Figure 4 Hyperglycemia-induced vasoconstriction by
culture model, we previously reported11 that the ADP decreased nitric oxide (NO) production and increased NO
elimination. Hyperglycemia amplifies the production of ROS,
to ATP ratio may increase by as much as 72% after
which interacts with NO to produce peroxynitrite and
8 hours of hypoxia. In vivo investigations reported depletes NO bioavailability. Hyperglycemia also decreases
similar results.12–14 Another metabolic issue related the activity of eNOS through the PI3K/AKT pathway to
to ATP metabolism after stroke relates to calcium diminish NO production.
homeostasis. Considerable evidence suggests that N-
methyl-D-aspartate (NMDA) receptor hyperactiva- loss of cytochrome c into the cytosol.18 Once in the
tion during and after ischemic insult triggers a massive cytosol, cytochrome c interacts with procaspase-9,
intracellular calcium influx. Without a sufficient apoptotic protease-activating factor-1, and dATP to
energy supply, ATPase-dependent ion transport sys- form an activated complex.19 This complex then
tems become dysfunctional and are unable to activates caspase-3, the irreversible trigger of apopto-
maintain intracellular ion homeostasis, resulting in sis. Caspase-3 in turn proteolyzes an inhibitor of
a massive accumulation of intracellular and mito- caspase-activated DNase. In apoptosis, the degrada-
chondrial calcium.15,16 These pathophysiological tion of this inhibitor results in the translocation of
events then trigger several deleterious developments caspase-activated DNase from the cytoplasm to the
in the cell, including cellular swelling, organelle dege- nucleus, where it catalyses internucleosomal DNA
neration, loss of membrane integrity, and eventual degradation and subsequent cell apoptosis.20
cell death.17 Hyperglycemia has been shown to amplify intra-
The increase in intracellular and mitochondrial cellular calcium imbalance after ischemic stroke by
calcium that occurs during hyperglycemia may indir- upregulating neuronal NMDA receptors and exacer-
ectly trigger apoptosis in the stroke brain via an bating the aforementioned apoptotic pathway.
interaction with the electron transport chain compo- Li et al.21 demonstrated that hyperglycemia increases
nent cytochrome c. Elevated intracellular calcium extracellular glutamate concentration following fore-
levels could lead to mitochondrial depolarization and brain ischemia, this excessive glutamate stimulating

Neurological Research 2013 VOL . 35 NO . 5 481


Li et al. Hyperglycemia in stroke and possible treatments

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.

NMDA receptors which further increases influx of to anaerobic metabolism in cells causes diminished
calcium.22 Elevated intracellular calcium then pro- mitochondrial ATP production which creates condi-
motes the release of cytochrome c into the cytoplasm, tions that lead to tissue necrosis. Using an animal
thus upregulating caspase-3 activity and subsequent middle cerebral artery occlusion (MCAO) model,
DNA fragmentation (Fig. 1). Indeed, the role of Parsons et el.3 demonstrated acute hyperglycemia to
hyperglycemia and calcium in apoptotic cell death be associated with higher acute and subacute lactate
after stroke has been further elaborated in two more production. Although the exact mechanism is not
studies.23,24 In summary, the above studies demon- well elucidated, it is conceivable that high intracel-
strate that the compounding deleterious effect of lular glucose levels may drive the metabolic process
hyperglycemia in stroke-related injuries may be forward as a result of increased pyruvate availability
caused in part by glutamate release, upregulated for lactic acid formation (Fig. 2). However, several
intracellular calcium levels, and increased cytoplas- studies have highlighted the fact that hyperglycemia
mic cytochrome c, all of which are known to induce can be beneficial to the energy starved brain in
neuronal death. specific situations.27,28 For example, hyperglycemia
may be beneficial in situations where lactate can be
Hyperglycemia-mediated lactate accumulation quickly flushed away by uninterrupted blood flow. By
and acidosis in the penumbral region
contrast, blood flow in ischemia is stagnant, allowing
Intracellular acidification as a result of persistent
lactate to accumulate. The combination of stagnant
glycolysis increases proton and lactate levels, which
blood and lactate build-up may decrease penumbral
stresses the penumbral tissue, making it more suscep-
salvage, increase final infarct volume, and ultimately
tible to pro-apoptotic signals. After ischemic stroke,
worsen functional outcome.3
cerebral blood perfusion rates decrease to less than
20% of the normal rate in the penumbral region.25 The Hyperglycemia-mediated oxidative stress and
availability of oxygen is even more limited in the ROS accumulation in penumbral neurons
ischemic core. Glucose is metabolized to pyruvate via Normally mitochondria produce ATP with remark-
glycolysis, which anaerobically undergoes fermenta- able efficiency through the reduction of oxygen to
tion to produce lactic acid, causing a decrease in water by cytochrome c oxidase. Under normal
intracellular pH.26 conditions, only approximately 1–2% of the oxygen
In the penumbral tissue, lactate accumulates at a reduced by mitochondria becomes ROS, such as
level many times its normal concentration. This shift superoxide (O{ 2 ).
29
The two known locations in the

482 Neurological Research 2013 VOL . 35 NO . 5


Li et al. Hyperglycemia in stroke and possible treatments

oxidative phosphorylation chain at which superoxides impairs glucose utilization via hexokinase, may
are produced are the nicotinamide adenine dinucleo- reverse the accumulation of superoxide effectuated
tide dehydrogenase and ubiquinone–cytochrome b–c1 as a result of hyperglycemia.45
regions.30 Oxidative stress has a profound effect on
Hyperglycemia-mediated BBB disruption and
stroke pathogenesis due to the high susceptibility of perfusion reduction
the brain to ROS-induced injuries. The brain has high The BBB is formed by endothelial cells, tight junction
concentrations of unsaturated lipids, low levels of proteins, pericytes, and astrocytes. The BBB plays a
antioxidants, high levels of oxygen consumption, and critical role both in maintaining brain homeostasis and
high levels of iron that acts as an oxidizing agent under restricting blood-borne substances from entering the
pathological conditions.31–33 All of these conditions parenchyma. Hyperglycemia is a risk factor for aug-
favor lipid peroxidation. mented BBB injury during ischemia. Hyperglycemia-
There are two known pathways by which ROS may induced cerebral acidosis and superoxide production
be produced following ischemia, one as a consequence are generally considered the mechanisms of enhanced
of mitochondrial membrane hyperpolarization and the brain injury, particularly as pertains to neurons.46,47
other through the use of the nicotinamide adenine However, recent reports suggest that damage to brain
dinucleotide phosphate oxidase (NADPH oxidase) endothelial cells and the BBB has a profound impact
pathway.34 Both are upregulated by hyperglycemia. on overall functional outcome and may even exceed
The accumulation of intracellular calcium and protons the impact on neuronal damage.
during ischemia onset causes hyperpolarization of the First, hyperglycemia has been shown to disrupt the
mitochondrial membrane.35,36 The rate of superoxide stability of the BBB by indirectly upregulating the
production increases exponentially as the mitochon- phosphorylation of occludin fragments, leading to
drial membrane potential increases above 140 mV.37,38 their ubiquitination and subsequent BBB permeabil-
Using an oxygen and glucose deprivation model, ity. Short-term hyperglycemia enhances de novo
Abramov et al. showed ROS production to be elevated synthesis of diacylglycerol (DAG) in capillary en-
1.6-fold during the initial onset of hypoxia and 2.6- dothelial cells from glycolytic intermediates.48 DAG
fold during reoxygenation. is then responsible for activating protein kinase C-
The NADPH oxidase pathway is an alternative beta II, one of nine PKC family proteins altered by
mechanism for ROS production. NADPH oxidase is a DAG during hyperglycemia.49 Activated PKC-beta
multi-subunit enzyme that catalyzes oxygen reduction II then phosphorylates occludin fragments at the
by using NADPH as the electron donor. The five Ser490 position, which leads to the ubiquitination
subunits of NADPH oxidase consist of p47phox, and redistribution of tight junction proteins in
p67phox, p40phox, p22phox, and the catalytic subunit endothelial cells.50 The end result of these molecular
gp91phox.39 In normal brains, p47phox, p67phox, and changes is increased BBB permeability. This mechan-
p40phox exist in the cytosol, while p22phox and ism occurs within minutes of reperfusion, which
gp91phox are attached to the cell membrane. concurs with the time course observed in in vivo
However, upon stimulation, such as during ischemia, models.51 Together, the evidence from human and
p47phox is phosphorylated and the cytosolic subunits animal studies supports the notion that hyperglyce-
form a complex that translocates to the cell membrane, mia worsens cerebrovascular reperfusion injuries
where it associates with p22phox and gp91phox to through the PKC-beta II pathway.
assemble the active oxidase.40 Second, hyperglycemia has been shown to signifi-
High intracellular glucose levels could exacerbate cantly reduce blood flow in the penumbral region. In
reperfusion-induced injuries by upregulating super- animal studies, induction of hyperglycemia by acute
oxide production. Thus, Suh et al.41 demonstrated glucose injection caused changes in cerebral blood
that glucose is central to ROS generation after OGD flow after either 2 or 4 hours of occlusion and
and reoxygenation, and that it acts independent of 2 hours of reperfusion. Cerebral blood flow in the
lactic acidosis in neurons. Glucose sustains the hexose ischemic hemisphere of hyperglycemic animals was
monophosphate shunt, allowing it to regenerate approximately 40% of that in the contralateral non-
NADPH, which serves as a substrate for ROS ischemic hemisphere, compared to 100% in the
production (Fig. 3).42 Superoxide production is normoglycemic controls.52 Additionally, induction
significantly downregulated in the absence of glucose of acute hyperglycemia by glucose infusion decreased
as well as when the hexose monophosphate shunt is penumbral blood flow to 60% of pre-ischemic values
inhibited after OGD.43 Furthermore, the increase in in the hyperglycemic group, compared to 89% in the
superoxide production during hyperglycemia is abol- normoglycemic group.53 Another mechanism for the
ished in p47 knockout mice and after NADPH reduced rate of perfusion in hyperglycemia has been
oxidase inhibition with apocynin.42,44 Additionally, attributed to decreased bioavailability of nitric oxide
2-deoxyglucose, which slows glucose transport and (NO), which is a potent vasodilator and inhibitor of

Neurological Research 2013 VOL . 35 NO . 5 483


Li et al. Hyperglycemia in stroke and possible treatments

platelet aggregation.54,55 Although this mechanism is components of the immune system are not only
not well delineated, it has been suggested that intimately involved in the ischemic cascade, but that
hyperglycemia could upregulate the dephosphoryla- they interact with tight junction proteins and the basal
tion of endothelial nitric oxide synthase (eNOS) at lamina to disrupt the BBB and degrade white matter to
the Ser1177 position and its phosphorylation at the cause parenchymal injuries.69–72 Hyperglycemia has
Thr495 position, both of which decrease eNOS been shown to have a tremendous effect on the
activity.55,56 Indeed, hyperglycemia has been shown inflammatory pathway post ischemia, elevating several
to downregulate eNOS activity by upregulating the transcription pathways to increase leukocyte migra-
hexosamine and PKC pathways, thereby downregu- tion to the brain and their ability to cross into the
lating the phosphoinositide 3-kinase/protein kinase B injured tissue.
(PI3K/AKT) pathway.57,58 This process inhibits Elevated intracellular glucose results in an increase
vasodilation, thus decreasing blood flow to the in nuclear factor-kappaB (NF-kappaB) and a de-
ischemic regions of the brain (Fig. 4). crease in IkappaB expressions in endothelial cells.73,74
Finally, ROS are known to interact with NO to Under quiescent conditions, NF-kappaB is bound to
produce the highly reactive peroxynitrite radical IkappaB in the cytosol. However during IkappaB
which decreases the availability of NO and further degradation, NF-kappaB translocates into the nucleus
impairs the blood vessels’ ability to dilate.56 Even and stimulates the expression of pro-inflammatory
mild acute hyperglycemia (y150 mg/dl) significantly cytokines such as TNF-alpha, interleukin (IL)-1 and
reduces blood flow to the penumbral region and IL-6 and chemokines, such as monocyte chemoattrac-
causes red blood cells to be trapped within areas of tant protein.74–77 These cytokines and chemokines
the ischemic vasculature. The end result of these attract mononuclear cells to the site of injury and
hyperglycemic sequelae is the diminishing or absence upregulate endothelial cell surface receptors to facil-
of oxygen delivery to a portion of the penumbra.59 itate mononuclear cell attachment and their transmi-
These oxygen deprived areas exhibited marked gration into the parenchyma.78,79 Indeed, Morigi
increases in BBB permeability after reperfusion.59 et al.80 found that maintaining a blood glucose level
In summary, vascular injury appears to be an of 174 mg/dl for 24 hours significantly increased levels
important factor associated with hyperglycemia- of NF-kappaB in endothelial cells and adhesion
induced derangements and the consequences of vascu- molecules including intercellular cell adhesion mole-
lar damage may significantly limit the benefit derived cule-1 (ICAM-1) and vascular cell adhesion molecule-
from available protective neuronal treatments. As later 1. The inhibition of NF-kappaB with pyrrolidine
mentioned in this Review, more effective treatments dithiocarbamate and N-tosyl-L-phenylalanine chloro-
may emerge from a better understanding of hypergly- methyl ketone reversed hyperglycemia-induced
cemia’s effects on the cerebrovasculature after ischemia. increases in NF-kappaB expression and subsequent
mononuclear cell adhesion to endothelial cells.80
Hyperglycemia-mediated inflammatory response The second major pathway in hyperglycemia media-
The brain’s inflammatory responses to ischemia are tion of the immune response was elucidated by
characterized by the rapid activation of resident cells. Smolock et al.,81 who demonstrated that elevated
Beginning two hours after stroke onset, microglia are glucose levels activated the PKC pathway. In turn,
activated via CD14 receptors and subsequently by PKC upregulated its downstream molecule calpain in a
toll-like receptor 4.60–62 Activated microglia undergo calcium-dependent manner.81 This upregulation of
morphological transformation into phagocytes and calpain ultimately resulted in the overexpression of
act as cytotoxin-producing factories releasing proin- ICAM-1 on the surface of endothelial cells.81 However,
flammatory cytokines such as tumor-necrosis factor- there is controversy over whether elevated glucose
alpha (TNF-alpha), IL-1, and IL-6 in addition to NO concentrations alone cause the increase in ICAM-1
and ROS.63,64 Similar to microglia, astrocytes are expression. Supporting the direct role of hyperglyce-
also activated during the acute phase to produce pro- mia, Kado et al.82 demonstrated that incubating
inflammatory cytokines, chemokines, and inducible endothelial cells with 30 mM of glucose for 24 hours
nitric oxide synthase.65 Elevated levels of the afore- significantly increased ICAM-1 levels (Fig. 5). Several
mentioned molecules initiate the second step of the other studies have confirmed the above results.80,83
post-ischemic inflammatory response, which includes In contrast, studies conducted by Rasmussen
circulating inflammatory cell chemotaxis and trans- et al.84 suggested that the induction of cellular
migration into the injured brain.66–68 Lymphocytes, adhesion molecules in human endothelial cells could
normally excluded from the neuronal compartment be associated with the upregulation of pro-inflamma-
under quiescent conditions, can be visualized by tory cytokines. Esposito et al.85 further demonstrated
immunohistochemistry in the same compartment in that acute hyperglycemia could induce an increase in
the post-ischemic brain. Recent work suggests that the concentrations of plasma IL-6 and TNF-alpha.

484 Neurological Research 2013 VOL . 35 NO . 5


Li et al. Hyperglycemia in stroke and possible treatments

This last finding has been substantiated by in vitro ischemic injuries, the translation of knowledge gath-
studies.86–89 Taken together, the above observations ered from experimental studies into effective treat-
suggest multiple mechanisms by which hyperglycemia ments has been uniformly disappointing. While tissue
orchestrates pro-inflammatory phenotypes. plasminogen activator (tPA) administration after
Inflammatory mechanisms intrinsic to the brain stroke is considered the gold standard in re-establish-
and its vasculature are crucial mediators of increased ing blood flow to the brain, co-administration of
BBB permeability during ischemia. Hyperglycemia neuroprotective agents that interrupt pathways of
may upregulate activator protein-1, which then ischemic cell death could improve stroke outcomes. A
stimulates the transcription of matrix metalloprotei- therapy that would both salvage ischemic tissue and
nases -2 (MMP-2) and -9 (MMP-9) in leukocytes.90 protect the brain from hyperglycemia-induced injuries
Furthermore, hyperglycemia promotes the recruit- post-ischemia could benefit millions, yet this medical
ment of blood-borne leukocytes, mainly neutrophils, need remains unmet.
which further increase the levels of MMP-9.91
Expression of MMP-2 and MMP-9 has been inver- Insulin therapy
sely correlated with BBB tight junction protein There is an emerging body of evidence supporting the
(zonula occludens-1) expression and basal laminin beneficial effects of normalizing serum glucose levels
levels, both of which are integral to the integrity of within 48 hours of ischemic stroke onset.100 In animal
the BBB.92 Decreases in the latter two protein models, insulin therapy has been shown to reduce
concentrations are thought to affect the integrity of infarct volume and enhance neuroprotection during
the BBB and increase its permeability, resulting in acute focal ischemia.101 However, the translation of
edema and hemorrhagic transformation. these findings from the laboratory to the bedside has
The source of MMP-9 is still a matter of debate. yielded mixed results.102,103
Gasche et al.93 found active MMP-9 to be colocalized Evidence supports the notion that hyperglyce-
with endothelial cells and astrocytic processes. In mia can have a significantly deleterious effect on the
contrast, our initial results show OGD significantly neurovasculature. In rodent MCAO models, insulin
increased the expression of MMP-9 by resident micro- exerts a neuroprotective effect on the brain vasculature
glial cells in the brain. Recent studies using chimeric mice through the upregulation of NO via the PI3K/AKT
with GFP bone marrow cells provided the opportunity pathway.104 The activation of the PI3K/AKT path-
to distinguish brain versus blood macrophages and their way negatively modulates genes that promote vascular
contribution to vascular injury after ischemic stroke. permeability and inflammation, thereby protecting
Although it is generally thought that both resident and vascular integrity.105 eNOS, a molecule downstream
blood-derived macrophages play significant roles in of AKT, is a known catalyst of NO. As previously
the inflammatory pathway, resident microglial cells discussed, NO has a crucial role in regulating vascular
predominate.94 This conclusion is supported by our tone and integrity. Generation of NO could potentially
studies using an in vitro OGD-reoxygenation brain increase vasodilation and improve blood flow to the
slice culture method in which blood-borne macro- penumbra, thus diminishing platelet aggregation.106 In
phages are eliminated from the preparation. addition to increasing NO synthesis, insulin therapy
Compelling evidence suggests that hyperglycemia- also decreases ROS accumulation by downregulating
induced MMP-9 overexpression may also enhance NADPH oxidase activity. This change in NADPH
white matter degeneration following ischemic stroke, oxidase increases the bioavailability of NO.73
leading to poorer outcomes.95 Patients with inade- In addition to lowering glucose levels, insulin may
quately regulated serum glucose levels are more also have a direct neuroprotective effect on the CNS
susceptible to white matter lesions.96 Similarly, dis- parenchyma by reducing inflammation.107 In patients
ruption of the BBB as a result of ischemic onset who received intensive insulin therapy (target glucose
exposes the brain parenchyma to blood-borne macro- level between 80 and 110 mg/dl), the activity of the
phages and has been associated with white matter transcription regulator NF-kappaB and its down-
lesions.97,98 Asahi et al.99 showed that MMP-9 knock- stream targets, ICAM-1 and E-selectin, were signifi-
out mice have reduced myelin degradation after cantly reduced.107 As ICAM-1 and E-selectin are
ischemic stroke. Given the critical role of white matter cardinal mediators of blood macrophage transmigra-
cells in ischemic outcomes, further investigation is tion across the endothelium after stroke onset,
needed to better understand the effects of the decreased expression of both receptors produces an
inflammatory response on white matter degeneration. oppositional effect to that of hyperglycemia on the
vascular endothelium.
Treatments However, a broad-spectrum reduction in the
Despite the ongoing effort to better understand inflammatory response may paradoxically limit recov-
the mechanisms underlying hyperglycemia-enhanced ery since immune cells and inflammation also play a

Neurological Research 2013 VOL . 35 NO . 5 485


Li et al. Hyperglycemia in stroke and possible treatments

crucial role in tissue repair. Although there is evidence increase neuronal survival by interacting with the
to support the beneficial effects of the anti-inflamma- IGF-1 receptor, IGF-1 binds to the receptor with
tory response during the acute phase in ameliorating much higher affinity.118 Thus there is reason to
tissue damage, counteracting this response may believe that IGF-1 treatment may be effective in
actually worsen long-term outcomes by reducing ameliorating the negative effects of hyperglycemia
overall tissue repair and reorganization.108–113 More during stroke onset.
work in the area of the brain’s immune response after
Glucagon-like peptide-1 therapy
injury would likely lead to effective novel therapies.
Glucagon-like peptide-1 (GLP-1) and artificial GLP-
Early insulin based clinical trails have failed to
1 receptor agonists such as exendin-4 (Ex-4) are well-
show significant results. The Glucose Insulin in
established diabetes medications.119 In recent years,
Stroke Trial (GIST) conducted in the UK reported
several studies have shown GLP-1 receptor (GLP-
no evidence of benefit from insulin therapy.102 There
1R) stimulation to ameliorate the adverse effects of
are several possible explanations for this result. First,
ischemic stroke.119,120 GLP-1 and Ex-4 have a wide
the difference in mean glucose levels between the
range of physiological actions in glucose metabolism,
control group and the experimental group was only
including the stimulation of insulin secretion, inhibi-
10 mg/dl after insulin treatment.114 As a result, there
tion of glucagon release, and reduction of appetite
were no significant differences in clinical outcomes
and food intake.120,121 GLP-1R is expressed through-
as measured by mortality rate and neurological
out the brain, and GLP-1 and Ex-4 are able to cross
deficits.102 Second, the optimal range of serum
the BBB due to high lipophilicity.122 GLP-1 admin-
glucose was not well defined and a heterogeneous
istration elicits the combined outcomes of decreased
group of patients was accepted into the study. intracerebral glucose content and increased cerebral
Nonetheless, the GIST study helped establish a 24- metabolic rate.123 Models of transient MCAO stroke
hour glucose-insulin infusion safety parameter com- in rodent have shown that treatment with Ex-4
prised of maintaining glucose levels between 72 and provides marked benefits on locomotor activity and
126 mg/dl in order to avoid the significant risk of infarct volume that are absent in GLP-1R knock out
hypoglycemia or mortality in excess of 40 weeks. mice.124
Currently, the Stroke Hyperglycemia Insulin Net- GLP-1R stimulation has also been shown to reduce
work Effort study, a phase III single-blinded ran- the synthesis of pro-inflammatory cytokine IL-1beta
domized control trial, is looking for a correlation in activated astrocytes via stimulation of adenylate
between normalized serum glucose levels and a cyclase and the accumulation of cAMP.125 The
reduction in thrombolysis-induced intracranial hemor- mechanism by which GLP-1R stimulation drives
rhage associated with hyperglycemia.115 A stratified neuroprotection is unclear, and future research is
result from this trial has the potential to alter future necessary to investigate the potential of GLP-1 as a
treatment decisions for hyperglycemic stroke patients. treatment.
Furthermore, the Continuous Glucose Monitoring
and Insulin Pump Therapy in Diabetic Gastroparesis Hyperbaric oxygen preconditioning
clinical trial, currently conducted at the University of Accumulating evidence indicates that hyperbaric
Michigan, uses continuous glucose monitoring and an oxygen therapy (HBO) may have beneficial effects in
insulin pump to precisely maintain the patient’s stroke. Qin et al.126 demonstrated that HBO precon-
glucose levels. The validated end point has the ditioning decreased hyperglycemia-induced hemorrha-
potential to reduce the resulting complications of gic transformation. While the exact mechanism by
hypoglycemia and would allow physicians to maintain which HBO limits hemorrhagic transformation is not
patients’ glucose at a more consistent level. well-defined, recent reports suggest that HBO pre-
conditioning may ameliorate hyperglycemia-induced
Insulin-like growth factor 1 injuries by reducing BBB disruption. Veltkamp
Insulin-like growth factor I (IGF-1) is structurally et al.127 showed that HBO therapy reduces basal
similar to insulin and thus has many analogous lamina degradation and MMP-9 levels, which are
functions. In animal models, it was found that IGF-1, associated with hemorrhagic transformation.
when injected into the ventricles, reduced ischemic HBO preconditioning also protects against brain
damage.116 IGF-1 displays strong anti-apoptotic cell ischemia–reperfusion injury by upregulating antiox-
survival properties and is a potent neuroprotective idant species.128,129 Evidence suggests that several
agent when administered after stroke induction.117 antioxidant genes may act synergistically to remove
Cheng et al.117 showed neuroprotective properties of ROS through sequential enzymatic reactions. It is
IGF-1 are mediated by the activation of PI3K/AKT believed that nuclear factor erythroid 2 mediates the
as well as FKHRL, both of which improve cell expression of these genes. The relevant enzymes
survival. While both insulin and IGF-1 can directly include heme oxygenase-1, glutathione S-transferase,

486 Neurological Research 2013 VOL . 35 NO . 5


Li et al. Hyperglycemia in stroke and possible treatments

and NADPH quinone oxidoreductase.130 Soejima interferes with pathways of ischemic hemorrhagic
et al.128 found that HBO preconditioning significantly transformation, neuronal cell death, and increased
attenuated transient MCAO-induced neurological blood vessel permeability shows promise. However,
deficits, infarct volume, and BBB permeability in further studies are needed to determine whether
hyperglycemic rodents. ethanol’s effect on these post-ischemic injuries could
be clinically significant.
Hypothermia
Hypothermia has long been suspected to have Conclusion
neuroprotective effects when administered immedi- Acutely elevated glucose levels are associated with
ately after stroke and has been a topic of intense poorer outcomes in ischemic stroke patients.141
research. In temporary ischemic stroke models, Substantial evidence from experimental work and
hypothermia has been shown to reduce infarct size clinical studies indicate that hyperglycemia further
and improve functional outcome.131 The physiological complicates the metabolic state and mitochondrial
effects of hypothermia are multifaceted. Hypothermia function in the ischemic penumbra, increases both
decreases inflammation,132–134 neuronal apoptosis,135 infarct volume and hemorrhagic transformation, and
mitochondrial dysfunction,136 and excitotoxicity.137 worsens outcomes even after thrombolytic therapy.
Mild hypothermia (33C) significantly reduces cyto- Using in vivo (e.g. MCAO models) and in vitro (e.g.
chrome c translocation into the cytoplasm and Bax OGD) models, investigators have shown hyperglyce-
and Bcl-2 levels.135 Furthermore, hypothermia re- mia to exacerbate intracellular calcium imbalance
stores elevated calcium levels after stroke outcome and
and the accumulation of ROS in endothelial cells.
reduces calcium dependent degradative pathways
These intracellular changes are thought to cause
leading to cell death.138
detrimental effects in the penumbral region and the
Ethanol neurovascular system. In addition, the activation of
The efficacy of ethanol therapy as a treatment for both microglia and macrophages in the neuronal
hyperglycemia-induced brain injury post-ischemic compartment after ischemic stroke aggravate the
stroke has yet to be tested. However, there is evidence extent of injury by disrupting the BBB and causing
to suggest that a moderate concentration of etha- degeneration of the white matter.
nol treatment may be neuroprotective and can re- Although experimental studies have elucidated
duce hyperglycemia-associated injuries. Previously several mechanisms by which hyperglycemia influ-
we showed that acute ethanol administration re- ences the survival of ischemic penumbral tissue,
duces both infarct lesion volume and hemorrhagic studies bridging the gap between clinical stroke and
transformation.139 One of the pathways by which experimental models have been limited. Recently,
ethanol exerts its neuroprotective effects may be MRI data have shown a strong correlation between
through a reduction of ROS. Thus, in a MCAO loss of penumbral tissue and elevated blood glucose.3
model, we found that a moderate ethanol treatment Although several treatments have been proposed
(1.5 g/kg) administered immediately upon reperfu- to ameliorate hyperglycemia-induced injury after
sion significantly reduced NADPH oxidase and ischemic stroke, their translation from the laboratory
gp91(phox) in the brain.140 to the bedside has been disappointing thus far. While
Furthermore, acute ethanol treatment reduces insulin therapy to normalize glucose levels has shown
disruption of the BBB caused by reperfusion. In the promise in animal models, clinical application of this
same model, we showed that ischemic rodents gained strategy in the GIST trial failed to show significant
neuroprotection as exhibited by decreased expression improvement. Furthermore, the GIST trial revealed
of aquaporins 4 and 9, two important mediators of the difficulty with managing patient’s glucose levels in
cerebral edema post-ischemic stroke, when adminis- a clinical setting. Lowering the patient’s glucose levels
tered moderate concentrations of ethanol immedi- using insulin has the potential to induce hypoglyce-
ately following reperfusion.92 In addition, using an in mia, causing more harm than good. Anti-inflamma-
vitro oxygen and glucose deprivation and reoxygena- tory therapies have shown paradoxical outcomes; as a
tion model, we found that ethanol administration result, non-specific anti-inflammatory therapy has yet
at a moderate concentration of 30 mM significantly to fulfill its promise. Research is needed to delineate
reduced the levels of MMP-2 and MMP-9 and specific inflammatory pathways and their effects on
increased basal lamina and tight-junction zonula ischemia-induced injuries. A better understanding of
occludins-1 levels.92 These effects likely helped to the inflammatory pathways involved in ischemic
stabilize the BBB after stroke onset. stroke will allow physicians to inhibit destructive
Ethanol-derived neuroprotection may constitute one pathways while promoting those that are neuro-
method of combating hyperglycemiainduced post- restorative. Finally, though in its infancy, ethanol ther-
ischemic injuries. Evidence that ethanol treatment apy is promising, as it exerts significant depressive

Neurological Research 2013 VOL . 35 NO . 5 487


Li et al. Hyperglycemia in stroke and possible treatments

effect on glucose metabolism and influence on the cell death after intracerebral hemorrhage. Stroke.
2003;34:2215–20.
neurovascular system. Ethanol therapy is one of the 10 Silver IA, Deas J, Erecinska M. Ion homeostasis in brain cells:
few treatments that impedes both the metabolic differences in intracellular ion responses to energy limitation
between cultured neurons and glial cells. Neuroscience.
pathway and the inflammatory pathway to protect 1997;78:589–601.
the penumbral tissue. However, more research is 11 Yuan Y, Peng C, Li K, Hussain M, Sikharam C, Guthikonda
M, et al. Ethanol reduces expression of apoptotic proteins
necessary to establish ethanol-derived neuroprotection after hypoxia/reoxygenation in a brain slice model. Neurol
in hyperglycemic conditions. Res. 2012;34:373–8.
The effect of hyperglycemia on ischemic stroke 12 Rosenblum WI. ATP-sensitive potassium channels in the
cerebral circulation. Stroke. 2003;34:1547–52.
outcome is profound and may significantly affect 13 Klaidman L, Morales M, Kem S, Yang J, Chang ML, Adams
treatment outcome. Accumulating data suggests JD, Jr. Nicotinamide offers multiple protective mechanisms in
stroke as a precursor for NADz, as a PARP inhibitor and by
diabetic rodents, when treated with tPA, exhibit partial restoration of mitochondrial function. Pharmacology.
negative side effects including increased BBB perme- 2003;69:150–7.
14 Yang GY, Chen SF, Kinouchi H, Chan PH, Weinstein PR.
ability and an orders of magnitude increase in Edema, cation content, and ATPase activity after middle
intracerebral hemorrhage volumes compared to cerebral artery occlusion in rats. Stroke. 1992;23:1331–6.
non-hyperglycemic rodents treated with tPA.142,143 15 Hansen AJ, Nedergaard M. Brain ion homeostasis in cerebral
ischemia. Neurochem Pathol. 1988;9:195–209.
A recent study published by Chen et al. showed that 16 Hossmann KA. Glutamate-mediated injury in focal cerebral
bone marrow stromal cell treatment of stroke in ischemia: the excitotoxin hypothesis revised. Brain Pathol.
1994;4:23–36.
diabetic rats precipitated adverse effects, including 17 Lipton P. Ischemic cell death in brain neurons. Physiol Rev.
increased BBB leakage, cerebral artery neointimal 1999;79:1431–568.
formation, arteriosclerosis, and worse functional 18 Luetjens CM, Bui NT, Sengpiel B, Munstermann G, Poppe
M, Krohn AJ, et al. Delayed mitochondrial dysfunction in
outcome.144 Traditional methods known to improve excitotoxic neuron death: cytochrome c release and a
functional outcome after ischemic stroke have shown secondary increase in superoxide production. J Neurosci.
2000;20:5715–23.
adverse effects when used in hyperglycemic animal 19 Jiang X, Wang X. Cytochrome c promotes caspase-9
models. activation by inducing nucleotide binding to APAF-1. J Biol
Chem. 2000;275:31199–203.
As the diabetic population continues to expand in 20 Krantic S, Mechawar N, Reix S, Quirion R. Apoptosis-
both developed and developing countries, the effect inducing factor: a matter of neuron life and death. Prog
of hyperglycemia post-ischemic stroke should be Neurobiol. 2007;81:179–96.
21 Li PA, Shuaib A, Miyashita H, He QP, Siesjo BK, Warner
investigated. Successful therapeutic intervention for DS. Hyperglycemia enhances extracellular glutamate accumu-
the large population facing the social, financial, and lation in rats subjected to forebrain ischemia. Stroke.
2000;31:183–92.
health consequences of diabetes and ischemic stroke 22 Araki N, Greenberg JH, Sladky JT, Uematsu D, Karp A,
will depend on understanding the interplay between Reivich M. The effect of hyperglycemia on intracellular
calcium in stroke. J Cereb Blood Flow Metab. 1992;12:469–76.
glucose and different systems of the brain, including
23 Li PA, Rasquinha I, He QP, Siesjo BK, Csiszar K, Boyd CD,
the cerebrovascular system, the inflammatory system, et al. Hyperglycemia enhances DNA fragmentation after
and the metabolic system. 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
References release of cytochrome c and activation of caspase-3 in
1 Rosamond W, Flegal K, Furie K, Go A, Greenlund K, Haase hyperglycemic rats subjected to transient forebrain ischemia.
N, et al. Heart disease and stroke statistics – 2008 update: a Brain Res. 2001;896:69–76.
report from the American Heart Association Statistics 25 Belayev L, Zhao W, Busto R, Ginsberg MD. Transient middle
Committee and Stroke Statistics Subcommittee. Circulation. cerebral artery occlusion by intraluminal suture: I. Three-
2008;117:e25–46. dimensional autoradiographic imageanalysis of local cerebral
2 Capes SE, Hunt D, Malmberg K, Pathak P, Gerstein HC. glucose metabolism–blood flow interrelationships during
Stress hyperglycemia and prognosis of stroke in nondiabetic ischemia and early recirculation. J Cereb Blood Flow
and diabetic patients: a systematic overview. Stroke. Metabolism. 1997;17:1266–80.
2001;32:2426–32. 26 Folbergrova J, Zhao Q, Katsura K, Siesjo BK. N-tert-butyl-
3 Parsons MW, Barber PA, Desmond PM, Baird TA, Darby alpha-phenylnitrone improves recovery of brain energy state
DG, Byrnes G, et al. Acute hyperglycemia adversely affects in rats following transient focal ischemia. Proc Natl Acad Sci
stroke outcome: a magnetic resonance imaging and spectro- USA. 1995;92:5057–61.
scopy study. Ann Neurol. 2002;52:20–8. 27 Goldberg MP, Choi DW. Combined oxygen and glucose
4 Martini SR, Kent TA. Hyperglycemia in acute ischemic deprivation in cortical cell culture: calcium-dependent and
stroke: a vascular perspective. J Cereb Blood Flow Metab. calcium-independent mechanisms of neuronal injury. J
2007;27:435–51. Neurosci. 1993;13:3510–24.
5 Baird TA, Parsons MW, Phanh T, Butcher KS, Desmond PM, 28 Pringle AK, Iannotti F, Wilde GJ, Chad JE, Seeley PJ,
Tress BM, et al. Persistent poststroke hyperglycemia is Sundstrom LE. Neuroprotection by both NMDA and non-
independently associated with infarct expansion and worse nmda receptor antagonists in in vitro ischemia. Brain Res.
clinical outcome. Stroke. 2003;34:2208–14. 1997;755:36–46.
6 Ginsberg MD. Hyperglycemia and stroke outcome: vindica- 29 Turrens JF, Boveris A. Generation of superoxide anion by the
tion of the ischemic penumbra. Ann Neurol. 2002;52:5–6. NADH dehydrogenase of bovine heart mitochondria.
7 Kent TA, Soukup VM, Fabian RH. Heterogeneity affecting Biochem J. 1980;191:421–7.
outcome from acute stroke therapy: making reperfusion 30 Turrens JF, Alexandre A, Lehninger AL. Ubisemiquinone is
worse. Stroke. 2001;32:2318–27. the electron donor for superoxide formation by complex III of
8 Suarez JI. Does hyperglycemia contribute to secondary injury heart mitochondria. Arch Biochem Biophys. 1985;237:408–14.
in subarachnoid hemorrhage? Stroke. 2006;37:8–9. 31 Saeed SA, Shad KF, Saleem T, Javed F, Khan MU. Some new
9 Song EC, Chu K, Jeong SW, Jung KH, Kim SH, Kim M, et al. prospects in the understanding of the molecular basis of the
Hyperglycemia exacerbates brain edema and perihematomal pathogenesis of stroke. Exp Brain Res 2007;182:1–10.

488 Neurological Research 2013 VOL . 35 NO . 5


Li et al. Hyperglycemia in stroke and possible treatments

32 Hewett SJ, Uliasz TF, Vidwans AS, Hewett JA. 56 Forstermann U, Munzel T. Endothelial nitric oxide synthase
Cyclooxygenase-2 contributes to N-methyl-D-aspartate- in vascular disease: from marvel to menace. Circulation.
mediated neuronal cell death in primary cortical cell culture. 2006;113:1708–14.
J Pharmacol Exp Therap. 2000;293:417–25. 57 Federici M, Menghini R, Mauriello A, Hribal ML, Ferrelli F,
33 Sugawara T, Fujimura M, Noshita N, Kim GW, Saito A, Lauro D, et al. Insulindependent activation of endothelial
Hayashi T, et al. Neuronal death/survival signaling pathways nitric oxide synthase is impaired by O-linked glycosylation
in cerebral ischemia. NeuroRx. 2004;1:17–25. modification of signaling proteins in human coronary
34 Miller AA, Dusting GJ, Roulston CL, Sobey CG. NADPH- endothelial cells. Circulation. 2002;106:466–72.
oxidase activity is elevated in penumbral and non-ischemic 58 Fleming I, Busse R. Molecular mechanisms involved in the
cerebral arteries following stroke. Brain Res. 2006;1111:111–6. regulation of the endothelial nitric oxide synthase. Am J
35 Piantadosi CA, Zhang J. Mitochondrial generation of reactive Physiol. 2003;284:R1–12.
oxygen species after brain ischemia in the rat. Stroke. 59 Ennis SR, Keep RF. Effect of sustained-mild and transient-
1996;27:327–31; discussion 332. severe hyperglycemia on ischemia-induced blood–brain barrier
36 Choi K, Kim J, Kim GW, Choi C. Oxidative stress-induced opening. J Cerebral Blood Flow Metab. 2007;27:1573–82.
necrotic cell death via mitochondira-dependent burst of 60 Wood PL. Microglia as a unique cellular target in the
reactive oxygen species. Curr Neurovasc Res. 2009;6:213–22. treatment of stroke: potential neurotoxic mediators produced
37 Starkov AA, Fiskum G. Regulation of brain mitochondrial by activated microglia. Neurol Res. 1995;17:242–8.
H2O2 production by membrane potential and NAD(P)H 61 Saito S, Matsuura M, Tominaga K, Kirikae T, Nakano M.
redox state. J Neurochem. 2003;86:1101–7. Important role of membrane-associated CD14 in the induction
38 Liu SS. Mitochondrial q cycle-derived superoxide and of IFN-beta and subsequent nitric oxide production by murine
chemiosmotic bioenergetics. Ann NY Acad Sci. macrophages in response to bacterial lipopolysaccharide. Eur
2010;1201:84–95. J Biochem. 2000;267:37–45.
39 Chabrashvili T, Tojo A, Onozato ML, Kitiyakara C, Quinn 62 Beschorner R, Schluesener HJ, Gozalan F, Meyermann R,
MT, Fujita T, et al. Expression and cellular localization of Schwab JM. Infiltrating CD14z monocytes and expression of
classic NADPH oxidase subunits in the spontaneously CD14 by activated parenchymal microglia/macrophages con-
hypertensive rat kidney. Hypertension. 2002;39:269–74. tribute to the pool of CD14z cells in ischemic brain lesions. J
40 Cifuentes ME, Pagano PJ. C-Src and smooth muscle Nad(P)H Neuroimmunol. 2002;126:107–15.
oxidase: assembling a path to hypertrophy. Arterioscler 63 Ekdahl CT, Kokaia Z, Lindvall O. Brain inflammation and
Thromb Vasc Biol. 2003;23:919–21. adult neurogenesis: the dual role of microglia. Neuroscience.
41 Suh SW, Shin BS, Ma H, van Hoecke M, Brennan AM, 2009;158:1021–9.
Yenari MA, et al. Glucose and NADPH oxidase drive 64 Lucas SM, Rothwell NJ, Gibson RM. The role of inflamma-
neuronal superoxide formation in stroke. Ann Neurol. tion in CNS injury and disease. Br J Pharmacol.
2008;64:654–63. 2006;147(Suppl 1):S232–40.
42 Tang XN, Cairns B, Kim JY, Yenari MA. Nadph oxidase in 65 Endoh M, Maiese K, Wagner J. Expression of the inducible
stroke and cerebrovascular disease. Neurol Res. 2012;34:338– form of nitric oxide synthase by reactive astrocytes after
45. transient global ischemia. Brain Res. 1994;651:92–100.
43 Suh SW, Gum ET, Hamby AM, Chan PH, Swanson RA. 66 Huang J, Upadhyay UM, Tamargo RJ. Inflammation in
Hypoglycemic neuronal death is triggered by glucose reperfu- stroke and focal cerebral ischemia. Surg Neurol. 2006;66:232–
sion and activation of neuronal NADPH oxidase. J Clin 45.
Invest. 2007;117:910–8. 67 Springer TA. Traffic signals for lymphocyte recirculation and
44 Brennan AM, Suh SW, Won SJ, Narasimhan P, Kauppinen leukocyte emigration: the multistep paradigm. Cell.
TM, Lee H, et al. NADPH oxidase is the primary source of 1994;76:301–14.
superoxide induced by NMDA receptor activation. Nat 68 Colgan SP, Dzus AL, Parkos CA. Epithelial exposure to
Neurosci. 2009;12:857–63. hypoxia modulates neutrophil transepithelial migration. J Exp
45 Bertoni JM. Competitive inhibition of rat brain hexokinase by Med. 1996;184:1003–15.
2-deoxyglucose, glucosamine, and metrizamide. J Neurochem. 69 Emsley HC, Smith CJ, Tyrrell PJ, Hopkins SJ. Inflammation
1981;37:1523–8. in acute ischemic stroke and its relevance to stroke critical
46 Dietrich WD, Alonso O, Busto R. Moderate hyperglycemia care. Neurocrit Care. 2008;9:125–38.
worsens acute blood–brain barrier injury after forebrain 70 Jin R, Yang G, Li G. Inflammatory mechanisms in ischemic
ischemia in rats. Stroke. 1993;24:111–6. stroke: role of inflammatory cells. J Leuk Biol. 2010;87:779–
47 Siemkowicz E. Brain uptake of mannitol and sucrose after 89.
cerebral ischemia: effect of hyperglycemia. Acta Physiol 71 Fernandez-Lopez D, Faustino J, Daneman R, Zhou L, Lee
Scand. 1981;112:359–63. SY, Derugin N, et al. Blood–brain barrier permeability is
48 Lee TS, Saltsman KA, Ohashi H, King GL. Activation of increased after acute adult stroke but not neonatal stroke in
protein kinase C by elevation of glucose concentration: the rat. J Neurosci. 2012;32:9588–600.
proposal for a mechanism in the development of diabetic 72 Belayev L, Busto R, Zhao W, Ginsberg MD. Quantitative
vascular complications. Proc Natl Acad Sci USA. evaluation of blood brain barrier permeability following
1989;86:5141–5. middle cerebral artery occlusion in rats. Brain Res.
49 Clarke H, Marano CW, Peralta Soler A, Mullin JM. 1996;739:88–96.
Modification of tight junction function by protein kinase C 73 Dhindsa S, Tripathy D, Mohanty P, Ghanim H, Syed T,
isoforms. Adv Drug Delivery Rev. 2000;41:283–301. Aljada A, et al. Differential effects of glucose and alcohol on
50 Murakami T, Frey T, Lin C, Antonetti DA. Protein kinase reactive oxygen species generation and intranuclear nuclear
Cbeta phosphorylates occludin regulating tight junction factor-kappaB in mononuclear cells. Metabolism. 2004;53:
trafficking in vascular endothelial growth factorinduced 330–4.
permeability in vivo. Diabetes. 2012;61:1573–83. 74 Barnes PJ, Karin M. Nuclear factor-kappaB: a pivotal
51 Geraldes P, King GL. Activation of protein kinase C isoforms transcription factor in chronic inflammatory diseases. N
and its impact on diabetic complications. Circ Res. Engl J Med. 1997;336:1066–71.
2010;106:1319–31. 75 Stein B, Baldwin AS, Jr. Distinct mechanisms for regulation of
52 Kawai N, Keep RF, Betz AL. Hyperglycemia and the vascular the interleukin-8 gene involve synergism and cooperativity
effects of cerebral ischemia. Stroke. 1997;28:149–54. between C/EBP and NF-kappa B. Mol Cell Biol.
53 Venables GS, Miller SA, Gibson G, Hardy JA, Strong AJ. The 1993;13:7191–8.
effects of hyperglycaemia on changes during reperfusion 76 Schreck R, Rieber P, Baeuerle PA. Reactive oxygen inter-
following focal cerebral ischaemia in the cat. J Neurol mediates as apparently widely used messengers in the
Neurosurg Psychiatry. 1985;48:663–9. activation of the NF-kappa B transcription factor and HIV-
54 Yilmaz G, Granger DN. Leukocyte recruitment and ischemic 1. EMBO J. 1991;10:2247–58.
brain injury. Neuromol Med. 2010;12:193–204. 77 Xie QW, Kashiwabara Y, Nathan C. Role of transcription
55 Du XL, Edelstein D, Dimmeler S, Ju Q, Sui C, Brownlee M. factor NF-kappa B/REL in induction of nitric oxide synthase.
Hyperglycemia inhibits endothelial nitric oxide synthase J Biol Chem. 1994;269:4705–8.
activity by posttranslational modification at the AKT site. J 78 Okada Y, Copeland BR, Mori E, Tung MM, Thomas WS, del
Clin Invest. 2001;108:1341–8. Zoppo GJ. Pselectin and intercellular adhesion molecule-1

Neurological Research 2013 VOL . 35 NO . 5 489


Li et al. Hyperglycemia in stroke and possible treatments

expression after focal brain ischemia and reperfusion. Stroke. 96 Roriz-Filho SJ, Sa-Roriz TM, Rosset I, Camozzato AL,
1994;25:202–11. Santos AC, Chaves ML, et al. (Pre)diabetes, brain aging, and
79 Clark WM, Lauten JD, Lessov N, Woodward W, Coull BM. cognition. Biochim Biophys Acta. 2009;1792:432–43.
Time course of ICAM-1 expression and leukocyte subset 97 Nakaji K, Ihara M, Takahashi C, Itohara S, Noda M,
infiltration in rat forebrain ischemia. Mol Chem Neuropathol. Takahashi R, et al. Matrix metalloproteinase-2 plays a critical
1995;26:213–30. role in the pathogenesis of white matter lesions after chronic
80 Morigi M, Angioletti S, Imberti B, Donadelli R, Micheletti G, cerebral hypoperfusion in rodents. Stroke. 2006;37:2816–23.
Figliuzzi M, et al. Leukocyte–endothelial interaction is 98 Wallin A, Sjogren M, Edman A, Blennow K, Regland B.
augmented by high glucose concentrations and hyperglycemia Symptoms, vascular risk factors and blood–brain barrier
in a NF-kB-dependent fashion. J Clin Invest. 1998;101:1905– function in relation to CT white-matter changes in dementia.
15. Eur Neurol. 2000;44:229–35.
81 Smolock AR, Mishra G, Eguchi K, Eguchi S, Scalia R. 99 Asahi M, Wang X, Mori T, Sumii T, Jung JC, Moskowitz
Protein kinase C upregulates intercellular adhesion molecule-1 MA, et al. Effects of matrix metalloproteinase-9 gene knock-
and leukocyte–endothelium interactions in hyperglycemia via out on the proteolysis of blood–brain barrier and white matter
activation of endothelial expressed calpain. Arterioscler components after cerebral ischemia. J Neurosci. 2001;21:7724–
Thromb Vasc Biol. 2011;31:289–96. 32.
82 Kado S, Wakatsuki T, Yamamoto M, Nagata N. Expression 100 Gentile NT, Seftchick MW, Huynh T, Kruus LK, Gaughan J.
of intercellular adhesion molecule-1 induced by high glucose Decreased mortality by normalizing blood glucose after acute
ischemic stroke. Acad Emerg Med. 2006;13:174–80.
concentrations in human aortic endothelial cells. Life Sci.
101 Hamilton MG, Tranmer BI, Auer RN. Insulin reduction of
2001;68:727–37.
cerebral infarction due to transient focal ischemia. J
83 Piconi L, Quagliaro L, Da Ros R, Assaloni R, Giugliano D,
Neurosurg. 1995;82:262–8.
Esposito K, et al. Intermittent high glucose enhances ICAM-1,
102 Gray CS, Hildreth AJ, Sandercock PA, O’Connell JE,
VCAM-1, E-selectin and interleukin-6 expression in human
Johnston DE, Cartlidge NE, et al. Glucose-potassium-insulin
umbilical endothelial cells in culture: the role of poly(adpri- infusions in the management of post-stroke hyperglycaemia:
bose) polymerase. J Thromb Haemost. 2004;2:1453–9. the UK Glucose Insulin in Stroke Trial (GIST-UK). Lancet
84 Rasmussen LM, Schmitz O, Ledet T. Increased expression of Neurol. 2007;6:397–406.
vascular cell adhesion molecule-1 (VCAM-1) in cultured 103 Bruno A, Kent TA, Coull BM, Shankar RR, Saha C, Becker
endothelial cells exposed to serum from type 1 diabetic KJ, et al. Treatment of hyperglycemia in ischemic stroke
patients: no effects of high glucose concentrations. Scand J (THIS): a randomized pilot trial. Stroke. 2008;39:384–9.
Clin Lab Invest. 2002;62:485–93. 104 Hartell NA, Archer HE, Bailey CJ. Insulin-stimulated
85 Esposito K, Nappo F, Marfella R, Giugliano G, Giugliano F, endothelial nitric oxide release is calcium independent and
Ciotola M, et al. Inflammatory cytokine concentrations are mediated via protein kinase B. Biochem Pharmacol.
acutely increased by hyperglycemia in humans: role of 2005;69:781–90.
oxidative stress. Circulation. 2002;106:2067–72. 105 Takahashi S, Mendelsohn ME. Synergistic activation of
86 Morohoshi M, Fujisawa K, Uchimura I, Numano F. Glucose- endothelial nitric-oxide synthase (eNOS) by HSP90 and
dependent interleukin 6 and tumor necrosis factor production AKT: calcium-independent eNOS activation involves forma-
by human peripheral blood monocytes in vitro. Diabetes. tion of an HSP90–AKT–CAM-bound eNOS complex. J Biol
1996;45:954–9. Chem. 2003;278:30821–7.
87 Morohoshi M, Fujisawa K, Uchimura I, Numano F. The 106 Butterworth RJ, Cluckie A, Jackson SH, Buxton-Thomas M,
effect of glucose and advanced glycosylation end products on Bath PM. Pathophysiological assessment of nitric oxide (given
IL-6 production by human monocytes. Ann NY Acad Sci. as sodium nitroprusside) in acute ischaemic stroke.
1995;748:562–70. Cerebrovasc Dis. 1998;8:158–65.
88 Pampfer S, Vanderheyden I, de Hertogh R. Increased 107 Dandona P, Aljada A, Mohanty P, Ghanim H, Hamouda W,
synthesis of tumor necrosis factor-alpha in uterine explants Assian E, et al. Insulin inhibits intranuclear nuclear factor
from pregnant diabetic rats and in primary cultures of uterine kappaB and stimulates IkappaB in mononuclear cells in obese
cells in high glucose. Diabetes. 1997;46:1214–24. subjects: evidence for an anti-inflammatory effect? J Clin
89 Coughlan MT, Oliva K, Georgiou HM, Permezel JM, Rice Endocrinol Metab. 2001;86:3257–65.
GE. Glucose-induced release of tumour necrosis factor-alpha 108 Genovese T, Mazzon E, Paterniti I, Esposito E, Bramanti P,
from human placental and adipose tissues in gestational Cuzzocrea S. Modulation of NADPH oxidase activation in
diabetes mellitus. Diabet Med. 2001;18:921–7. cerebral ischemia/reperfusion injury in rats. Brain Res.
90 Kamada H, Yu F, Nito C, Chan PH. Influence of 2011;1372:92–102.
hyperglycemia on oxidative stress and matrix metalloprotei- 109 Chamorro A, Meisel A, Planas AM, Urra X, van de Beek D,
nase-9 activation after focal cerebral ischemia/reperfusion in Veltkamp R. The immunology of acute stroke. Nat Rev.
rats: relation to blood–brain barrier dysfunction. Stroke. 2012;8:401–10.
2007;38:1044–9. 110 Moalem G, Leibowitz-Amit R, Yoles E, Mor F, Cohen IR,
91 Gidday JM, Gasche YG, Copin JC, Shah AR, Perez RS, Schwartz M. Autoimmune T cells protect neurons from
Shapiro SD, et al. Leukocyte-derived matrix metalloprotei- secondary degeneration after central nervous system axotomy.
nase-9 mediates blood–brain barrier breakdown and is Nat Med. 1999;5:49–55.
proinflammatory after transient focal cerebral ischemia. Am 111 Lewitus GM, Kipnis J, Avidan H, Ben-Nun A, Schwartz M.
J Phys. 2005;289:H558–68. Neuroprotection induced by mucosal tolerance is epitope-
dependent: conflicting effects in different strains. J
92 Zeng X, Asmaro K, Ren C, Gao M, Peng C, Ding JY, et al.
Neuroimmunol. 2006;175:31–8.
Acute ethanol treatment reduces blood–brain barrier dysfunc-
112 Takeda H, Spatz M, Ruetzler C, McCarron R, Becker K,
tion following ischemia/reperfusion injury. Brain Res.
Hallenbeck J. Induction of mucosal tolerance to E-selectin
2012;1437:127–33.
prevents ischemic and hemorrhagic stroke in spontaneously
93 Gasche Y, Copin JC, Sugawara T, Fujimura M, Chan PH. hypertensive genetically stroke-prone rats. Stroke.
Matrix metalloproteinase inhibition prevents oxidative stress- 2002;33:2156–63.
associated blood–brain barrier disruption after transient focal 113 Chen Y, Ruetzler C, Pandipati S, Spatz M, McCarron RM,
cerebral ischemia. J Cereb Blood Flow Metab. 2001;21:1393– Becker K, et al. Mucosal tolerance to E-selectin provides cell-
400. mediated protection against ischemic brain injury. Proc Natl
94 Schilling M, Besselmann M, Leonhard C, Mueller M, Acad Sci USA. 2003;100:15107–12.
Ringelstein EB, Kiefer R. Microglial activation precedes and 114 Bruno A, Liebeskind D, Hao Q, Raychev R. Diabetes
predominates over macrophage infiltration in transient focal mellitus, acute hyperglycemia, and ischemic stroke. Curr
cerebral ischemia: a study in green fluorescent protein Treat Options Neurol. 2010;12:492–503.
transgenic bone marrow chimeric mice. Exp Neurol. 115 Southerland AM, Johnston KC. Considering hyperglycemia
2003;183:25–33. and thrombolysis in the stroke hyperglycemia insulin network
95 Chen J, Cui X, Zacharek A, Cui Y, Roberts C, Chopp M. effort (SHINE) trial. Ann NY Acad Sci. 2012;1268:72–8.
White matter damage and the effect of matrix metalloprotei- 116 Zhu CZ, Auer RN. Intraventricular administration of insulin
nases in type 2 diabetic mice after stroke. Stroke. 2011;42:445– and IGF-1 in transient forebrain ischemia. J Cereb Blood
52. Flow Metab. 1994;14:237–42.

490 Neurological Research 2013 VOL . 35 NO . 5


Li et al. Hyperglycemia in stroke and possible treatments

117 Cheng HL, Steinway M, Delaney CL, Franke TF, Feldman transient middle cerebral artery occlusion: a comparative
EL. IGF-I promotes Schwann cell motility and survival via study under various treatment paradigms. Brain Res.
activation of AKT. Mol Cell Endocrinol. 2000;170:211–5. 2000;863:94–105.
118 Guan J, Bennet L, Gluckman PD, Gunn AJ. Insulin-like 132 Dietrich WD, Chatzipanteli K, Vitarbo E, Wada K, Kinoshita
growth factor-1 and post-ischemic brain injury. Prog K. The role of inflammatory processes in the pathophysiology
Neurobiol. 2003;70:443–62. and treatment of brain and spinal cord trauma. Acta
119 Darsalia V, Mansouri S, Ortsater H, Olverling A, Nozadze N, Neurochirurg. 2004;89:69–74.
Kappe C, et al. Glucagon-like peptide-1 receptor activation 133 Suehiro E, Fujisawa H, Akimura T, Ishihara H, Kajiwara K,
reduces ischaemic brain damage following stroke in type 2 Kato S, et al. Increased matrix metalloproteinase-9 in blood in
diabetic rats. Clin Sci (Lond). 2012;122:473–83. association with activation of interleukin-6 after traumatic
120 Drucker DJ, Nauck MA. The incretin system: glucagon-like brain injury: influence of hypothermic therapy. J
peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibi- Neurotrauma. 2004;21:1706–11.
tors in type 2 diabetes. Lancet. 2006;368:1696–705. 134 Deng H, Han HS, Cheng D, Sun GH, Yenari MA. Mild
121 Holst JJ. On the physiology of gip and GLP-1. Horm Metab hypothermia inhibits inflammation after experimental stroke
Res. 2004;36:747–54. and brain inflammation. Stroke. 2003;34:2495–501.
122 Kastin AJ, Akerstrom V. Entry of exendin-4 into brain is 135 Xu L, Yenari MA, Steinberg GK, Giffard RG. Mild
rapid but may be limited at high doses. Int J Obesity Relat hypothermia reduces apoptosis of mouse neurons in vitro
Metab Disord. 2003;27:313–8. early in the cascade. J Cereb Blood Flow Metab. 2002;22:21–8.
123 Gejl M, Egefjord L, Lerche S, Vang K, Bibby BM, Holst JJ, 136 Gong P, Li CS, Hua R, Zhao H, Tang ZR, Mei X, et al. Mild
et al. Glucagon-like peptide-1 decreases intracerebral glucose hypothermia attenuates mitochondrial oxidative stress by
content by activating hexokinase and changing glucose protecting respiratory enzymes and upregulating MNSOD in
clearance during hyperglycemia. J Cereb Blood Flow Metab. a pig model of cardiac arrest. PloS ONE. 2012;7:e35313.
2012;32:2146–52. 137 Tseng EE, Brock MV, Lange MS, Troncoso JC, Blue ME,
124 Teramoto S, Miyamoto N, Yatomi K, Tanaka Y, Oishi H, Lowenstein CJ, et al. Glutamate excitotoxicity mediates
Arai H, et al. Exendin-4, a glucagon-like peptide-1 receptor neuronal apoptosis after hypothermic circulatory arrest. Ann
agonist, provides neuroprotection in mice transient focal Thorac Surg. 2010;89:440–5.
cerebral ischemia. J Cereb Blood Flow Metab. 2011; 138 Phillips KF, Deshpande LS, Delorenzo RJ. Hypothermia
31:1696–705. reduces calcium entry via the N-methyl-D-aspartate and
125 Iwai T, Ito S, Tanimitsu K, Udagawa S, Oka J. Glucagon-like ryanodine receptors in cultured hippocampal neurons. Eur J
peptide-1 inhibits LPS-induced IL-1beta production in cul- Pharmacol. 2012;698:186–92.
tured rat astrocytes. Neurosci Res. 2006;55:352–60. 139 Wang F, Wang Y, Geng X, Asmaro K, Peng C, Sullivan JM,
126 Qin Z, Karabiyikoglu M, Hua Y, Silbergleit R, He Y, Keep et al. Neuroprotective effect of acute ethanol administration in
RF, et al. Hyperbaric oxygen-induced attenuation of hemor- a rat with transient cerebral ischemia. Stroke. 2012;43:205–
rhagic transformation after experimental focal transient 10.
cerebral ischemia. Stroke. 2007;38:1362–7. 140 Kochanski R, Peng C, Higashida T, Geng X, Hüttemann M,
127 Veltkamp R, Siebing DA, Sun L, Heiland S, Bieber K, Marti Guthikonda M, Ding Y. Neuroprotection conferred by post-
HH, et al. Hyperbaric oxygen reduces blood–brain barrier ischemia ethanol therapy in experimental stroke: an inhibitory
damage and edema after transient focal cerebral ischemia. effect on hyperglycolysis and NADPH oxidase activation. J
Stroke. 2005;36:1679–83. Neurochem. 2013; 125(2), doi: 10.1111/jnc.12169. Epub 2013
128 Soejima Y, Ostrowski RP, Manaenko A, Fujii M, Tang J, Jan 27.
Zhang JH. Hyperbaric oxygen preconditioning attenuates 141 Poppe AY, Majumdar SR, Jeerakathil T, Ghali W, Buchan
hyperglycemia enhanced hemorrhagic transformation after AM, Hill MD, et al. Admission hyperglycemia predicts a
transient MCAO in rats. Med Gas Res. 2012;2:9. worse outcome in stroke patients treated with intravenous
129 Wada K, Miyazawa T, Nomura N, Tsuzuki N, Nawashiro H, thrombolysis. Diabetes Care. 2009;32:617–22.
Shima K. Preferential conditions for and possible mechanisms 142 Won SJ, Tang XN, Suh SW, Yenari MA, Swanson RA.
of induction of ischemic tolerance by repeated hyperbaric Hyperglycemia promotes tissue plasminogen activator-
oxygenation in gerbil hippocampus. Neurosurgery. induced hemorrhage by increasing superoxide production.
2001;49:160–6; discussion 166–7. Ann Neurol. 2011;70:583–90.
130 Godman CA, Chheda KP, Hightower LE, Perdrizet G, Shin 143 Ning R, Chopp M, Yan T, Zacharek A, Zhang C, Roberts C,
DG, Giardina C. Hyperbaric oxygen induces a cytoprotective et al. Tissue plasminogen activator treatment of stroke in type-
and angiogenic response in human microvascular endothelial 1 diabetes rats. Neuroscience. 2012;222:326–32.
cells. Cell Stress Chaper. 2010;15:431–42. 144 Chen J, Ye X, Yan T, Zhang C, Yang XP, Cui X, et al.
131 Zausinger S, Hungerhuber E, Baethmann A, Reulen H, Adverse effects of bone marrow stromal cell treatment of
Schmid-Elsaesser R. Neurological impairment in rats after stroke in diabetic rats. Stroke. 2011;42:3551–8.

Neurological Research 2013 VOL . 35 NO . 5 491

Potrebbero piacerti anche