Diabetes, Stroke, and Neuroresilience Looking Beyond

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 21

Ann. N.Y. Acad. Sci.

ISSN 0077-8923

A N N A L S O F T H E N E W Y O R K A C A D E M Y O F SC I E N C E S
Special Issue: The Year in Diabetes and Obesity

Review

Diabetes, stroke, and neuroresilience: looking beyond


hyperglycemia
Matthew J. Krinock and Neel S. Singhal
Department of Neurology, University of California - San Francisco, San Francisco, California

Address for correspondence: Neel S. Singhal, MD, PhD, Department of Neurology, University of California – San Francisco,
555 Mission Bay Blvd S., MS3118, San Francisco, CA 94158. neel.singhal@ucsf.edu

Ischemic stroke is a leading cause of morbidity and mortality among type 2 diabetic patients. Preclinical and trans-
lational studies have identified critical pathophysiological mediators of stroke risk, recurrence, and poor outcome
in diabetic patients, including endothelial dysfunction and inflammation. Most clinical trials of diabetes and stroke
have focused on treating hyperglycemia alone. Pioglitazone has shown promise in secondary stroke prevention for
insulin-resistant patients; however, its use is not yet widespread. Additional research into clinical therapies directed
at diabetic pathophysiological processes to prevent stroke and improve outcome for diabetic stroke survivors is
necessary. Resilience is the process of active adaptation to a stressor. In patients with diabetes, stroke recovery is
impaired by insulin resistance, endothelial dysfunction, and inflammation, which impair key neuroresilience path-
ways maintaining cerebrovascular integrity, resolving poststroke inflammation, stimulating neural plasticity, and
preventing neurodegeneration. Our review summarizes the underpinnings of stroke risk in diabetes, the clinical
consequences of stroke in diabetic patients, and proposes hypotheses and new avenues of research for therapeutics
to stimulate neuroresilience pathways and improve stroke outcome in diabetic patients.

Keywords: diabetes; stroke; neuroprotection; endothelial; neuroinflammation; neuroplasticity

Introduction reducing patients’ risk and severity of the first-time


stroke, as diabetic patients with adequate glycemic
Stroke is one of the most predominant causes of
control have lower rates of recurrent stroke.7,8 Of
morbidity and mortality in the world. In the United
even greater importance for individual diabetic
States, approximately 800,000 Americans per year
stroke patients is that pathophysiological features
suffer a stroke, leading to over $30 billion in hospital
associated with diabetes, such as hyperglycemia,
and lost productivity costs per year.1 Strokes cause
endothelial dysfunction, poor collateral circula-
sudden paralysis or motor impairments, speech dis-
tion, and immune dysregulation, interfere with pro-
turbances, and/or loss of vision, among other neu-
cesses promoting brain cell survival and neuro-
rological symptoms. This is most often due to an
logical recovery during and following a stroke. In
interruption of blood flow caused by an embolism
this review, we outline the clinical implications
to arteries supplying the brain. Aging, hyperten-
of T2D in stroke. We focus on ischemic stroke,
sion, diabetes, smoking, and hypercholesterolemia
the most common cerebrovascular manifestation of
are the most important risk factors for stroke. Dia-
diabetes, which is associated with even higher mor-
betes increases an individual’s risk of stroke by
bidity among patients with diabetes. We also explore
1.5–3-fold.2–4 Importantly, the increasing preva-
the mechanisms linking the two diseases and sug-
lence of type 2 diabetes (T2D) is a key factor under-
gest research avenues to improve stroke preven-
lying increasing rates of stroke in patients under
tion, recovery, and neuroresilience in patients with
65 years old.2,5,6 Controlling diabetes is crucial to
diabetes.
doi: 10.1111/nyas.14583
Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences. 1
Diabetes and stroke Krinock & Singhal

Figure 1. Ischemic stroke subtypes. (A) Large-vessel occlusion (LVO) stroke. The left panel shows a brain diffusion–weighted
(DW) MRI image from a patient with newly diagnosed atrial fibrillation suffering from a large left hemisphere infarction due
to middle cerebral artery (MCA) occlusion. The right panel schematic illustrates that LVO can quickly lead to an area of core
infarction as well as a larger area of ischemic but viable brain tissue termed the penumbra. Patients with a large penumbra benefit
from endovascular thrombectomy (EVT) as an acute stroke treatment. Patients with poor collaterals, a complication of diabetes,
may suffer larger core infarct growth over time and benefit less from EVT. (B) Lacunar stroke. The left panel demonstrates a DW-
MRI image from a patient with untreated T2D presenting with acute weakness due a lacunar stroke in the internal capsule. The
right panel schematic illustrates that lacunar infarctions result from small-vessel atherosclerotic disease in end arterioles without
collateral flow causing a smaller “lacunar” infarction, often in brain regions necessary for motor control.

Diabetic patients are predisposed to stroke prising the remaining balance.1 Ischemic strokes
and suffer worse outcomes can be further classified into large- or small-vessel
strokes (Fig. 1). Large-vessel strokes can lead to
In the United States, the majority of strokes
profound neurological deficits and even death due
are ischemic in nature (87%), with hemorrhagic
to large areas of brain infarction from embolic
strokes, often associated with hypertension, com-
occlusion of the carotid or proximal cerebral

2 Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences.
Krinock & Singhal Diabetes and stroke

arteries. This is often the result of cardioembolism stroke treatment since 1995 has been recanaliza-
from atrial fibrillation, structural heart disease, tion of blocked arteries with tissue plasminogen
atherosclerotic disease, or a hypercoagulable state. activator.22 Over the last 6 years, there has been
Strokes resulting from vascular disease of end arte- a paradigm shift with the advent of mechani-
rioles result in “lacunar” infarctions of deep brain cal endovascular thrombectomy (EVT) for patients
structures. This includes brain regions supplied by with occlusion of large vessels presenting with
end arterioles, such as the lenticular nucleus, inter- favorable imaging characteristics indicative of brain
nal capsule, thalamus, and pons.9,10 These “small- tissue that is likely to be salvaged by reperfusion.23
vessel strokes” are typically associated with small Although this has revolutionized acute stroke treat-
areas of brain infarction (often <1.5 cm) but result ment, diabetic patients with LVO treated with
in highly morbid deficits due to their localization EVT achieve favorable neurological outcomes at
near prominent motor tracts. Lacunar strokes are 3 months only 35% of the time, compared with
usually associated with chronic hypertension or dia- 50–55% for nondiabetic patients.24,25 This does not
betes and are characterized by pathological thick- appear to be related to differences in EVT reper-
ening of the arterial media by fibrinoid deposition fusion success. The reasons for this marked dis-
(lipohyalinosis) or obstruction of penetrating end crepancy are not fully understood but may include
arteries by intimal plaques.10,11 poor collateral circulation, comorbid intracranial
There are clear differences in stroke patterns in atherosclerotic disease, increased susceptibility to
patients with diabetes. Patients with diabetes have cell death during ischemia and reperfusion, and
a higher proportion of ischemic stroke compared impaired recovery mechanisms. Intriguingly, the
with hemorrhagic strokes. Epidemiological studies efficacy of EVT may unmask further disparity in the
are mixed as to whether the risk of hemorrhagic outcomes of nondiabetic compared with diabetic
stroke is elevated in diabetic patients compared patients as nearly two-thirds of diabetic stroke sur-
with nondiabetic patients.3,4 Intracranial stenoses vivors will still suffer permanent and severe func-
are more common in patients with diabetes and tional disabilities. Thus, a more detailed under-
are proportional to its severity.12,13 Interestingly, standing of the mechanisms underlying poor stroke
among diabetic patients suffering ischemic stroke, outcomes in diabetic patients is essential. We pro-
lacunar stroke may be more prevalent than large pose that further understanding how the systemic
vessel stroke.14 This may be particularly impact- consequences of diabetes, including endothelial
ful for diabetic patients suffering from stroke, as dysfunction and inflammation, affect stroke risk
the risk of lacunar stroke recurrence is higher than and neurorecovery pathophysiology will lead to
with other stroke types, as is the risk of devel- important new insights to promote resilience and
oping cognitive impairment or dementia.15,16 As improve stroke outcomes in diabetic patients.
with numerous other health outcomes, diabetes
negatively impacts the care of hospitalized stroke
Physiological mechanisms mediating
patients. Diabetic patients suffering a stroke experi-
stroke risk and poor outcomes in diabetes
ence increased length of hospital stay, worse func-
tional outcomes, and have a higher risk of stroke Diabetes increases cardiovascular disease risk,
recurrence.17–19 Diabetics also have two-fold higher including stroke, through systemic metabolic and
1-year mortality compared with nondiabetic stroke inflammatory effects that alter the structure and
patients.7,20 Glycemic control is extremely critical to function of blood vessels and modulate immune
secondary stroke prevention, as each 1% increase in function. Over time, arteries, arterioles, and cap-
hemoglobin A1c increases the odds of death follow- illaries become increasingly stiff, tortuous, and
ing a stroke by approximately 33%; however, tight narrowed as a result of diabetes.26 Atheroscle-
glycemic control does not completely abate the ele- rosis and thrombogenesis are accelerated, and
vated risk of stroke recurrence in diabetic patients.21 physiological processes critical to endogenous
Diabetes may also be an important determi- neuroresilience and recovery, such as cerebral
nant of the efficacy of new and highly efficacious vasoreactivity, blood–brain barrier (BBB) perme-
acute ischemic stroke treatments for large-vessel ability, neuroplasticity, and neuroinflammation, are
occlusion (LVO). The mainstay of acute ischemic compromised (Fig. 2).

Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences. 3
Diabetes and stroke Krinock & Singhal

Figure 2. Diabetes exacerbates stroke injury and impairs neuroresilience. The temporal cellular and physiological events fol-
lowing stroke are altered by diabetic pathophysiology. (A) In particular, diabetic patients suffer increased neuronal injury
and degeneration due to increased susceptibility to bioenergetic failure, blood–brain barrier (BBB) dysfunction, and increased
M1 proinflammatory microglial/macrophage polarization. (B) Diabetes also reduces endogenous compensatory and neurore-
silience mechanisms, including increased cerebral blood flow from collateral circulation compensation, anti-inflammatory M2
microglial/macrophage polarization, and neural plasticity.

Although many clinical studies and trials sur- infarct growth, poor outcome, and hemorrhagic
rounding stroke and diabetes focus on hyper- conversion.32–35 Animal models of stroke and tis-
glycemia, endothelial dysfunction coupled with sue culture models of ischemia similarly demon-
inflammation has emerged as central to the cascade strate links between hyperglycemia at the time of
of pathophysiological events predisposing diabetic infarct and increased cell death, BBB dysfunction,
patients to stroke and poor neurological recovery.27 and impaired fibrinolysis. The presence of hyper-
Endothelial dysfunction and increased inflamma- glycemia during brain ischemia promotes lactic
tion occur with aging, but their consequences are acid production and worsens tissue acidosis. The
accelerated and exacerbated by diabetes. For exam- production of advanced glycosylation end-products
ple, functional MRI studies in diabetic patients (AGE) and reactive oxygen species (ROS) also con-
have found reductions in cerebral blood flow, par- tributes to BBB breakdown.36,37 Increased infarct
ticularly in subcortical regions perfused by small size and levels of inflammatory markers are also
vessels, associated with diabetes.28,29 Endothelial observed in hyperglycemic Zucker obese rats sub-
dysfunction and inflammation also increase the sus- jected to experimental stroke.38 Importantly, acute
ceptibility of the BBB to damage, which contributes hyperglycemia also impairs cerebral autoregulation,
to an increased risk of hemorrhagic transformation, which can be a critical compensatory mechanism
reperfusion injury, and cerebral edema following preventing the progression of ischemic penum-
stroke.30 Diabetic patients exhibit altered cerebral bral tissues into infarcted tissue. Studies in stroke
vasoreactivity, limiting autoregulatory control of patients have demonstrated greater infarct growth
cerebral blood flow and stunting the development in patients with hyperglycemia or diabetes or both
of crucial collateral blood supply, which exacerbates using serial MRI.39–41 Taken together, the clinical
stroke injury in diabetics.31 Finally, inflammatory and mechanistic studies provide a strong scien-
processes underlying diabetes counter neural tific rationale for the American Heart Association/
plasticity necessary for optimal stroke recovery American Stroke Association recommendations of
(Fig. 3). treating hyperglycemia to achieve blood glucose
levels in the range of 140–180 mg/dL. To understand
Acute hyperglycemia if further patient benefit could be derived from
Hyperglycemia is present in approximately 40% more aggressive blood glucose targets, the Stroke
of patients with acute ischemic stroke. Many ret- Hyperglycemia Insulin Network Effect (SHINE)
rospective studies have found compelling asso- study was carried out from 2012 to 2018.42 SHINE
ciations between admission blood glucose and was a multicenter randomized clinical trial of 1151

4 Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences.
Krinock & Singhal Diabetes and stroke

Figure 3. Mechanisms of stroke risk and poor stroke outcome in diabetes. Diabetes increases stroke risk, and its underlying
pathophysiological mechanisms exacerbate stroke injury and limit neuroplasticity, leading to poor stroke outcomes. Targeting the
mechanistic underpinnings of endothelial dysfunction and inflammation in diabetic stroke patients may restore neuroresilience
mechanisms and improve outcomes following stroke for diabetic patients.

patients conducted to assess the efficacy of intensive more likely to yield clinical benefit than addressing
versus standard blood glucose control in ischemic acute hyperglycemia alone.
stroke patients with diabetes or admission hyper-
glycemia (>150 mg/dL). Patients were randomized Endothelial and microvascular dysfunction
to either intensive (80–130 mg/dL) or standard glu- The microvasculature is a dynamic and highly
cose targets (80–170 mg/dL). The study did not regulated component of the cardio- and cere-
show clinical benefit to intensive insulin treatment, brovascular systems. Endothelial cells (ECs) and
despite the intensive group achieving significantly the extracellular matrix (ECM) comprising the
lower blood glucose values. This indicates that the microvasculature are integral to cerebral function
current clinical standard of avoiding extreme hyper- and nutrient homeostasis. In the brain, ECs play an
glycemia and hypoglycemia may be adequate. It important role in the maintenance and regulation of
also suggests that the underlying pathophysiolog- the cerebral vasculature. T2D mellitus and IR alter
ical mechanisms of diabetes may be the underly- endothelial signaling pathways, resulting in dys-
ing poor outcome rather than hyperglycemia itself. functional regulation of protective vascular signals,
Of note, recent studies investigating outcomes of such as nitric oxide (NO) and prostacyclins.27,45
diabetic patients undergoing embolectomy have ECs metabolize l-arginine via endothelial nitric
demonstrated markedly worse outcomes related to oxide synthase (eNOS) to form NO. Once NO is
elevated admission blood glucose.43 However, more produced by the cell, it rapidly exerts its biolog-
definitive studies are still required as other analy- ical actions by diffusing across cell membranes.
ses suggest that outcome is independent of admis- NO induces vasodilation by regulating vascu-
sion blood glucose.25,44 On the basis of these results lar smooth muscle through activation of soluble
as well as the SHINE trial, we propose that tar- guanylyl cyclase, which produces cyclic guano-
geting underlying diabetic pathophysiological pro- sine monophosphate. NO prevents thrombosis
cesses, such as insulin resistance (IR), endothelial through inhibition of platelet surface glycoproteins,
dysfunction, and aberrant immune function, will be notably the glycoprotein IIb/IIIa complex. Thus, the

Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences. 5
Diabetes and stroke Krinock & Singhal

production of NO by ECs regulates vasomotor tone Diabetes-related immune dysfunction and


and acts on adjacent smooth muscle cells (SMCs) inflammation
and circulating platelets and leukocytes to discour-
Systemic and local release of proinflammatory
age adhesion and thrombus formation.
cytokines exacerbates endothelial dysfunction and
Oxidative stress (OS) is a key mediator of
also independently contributes to the pathogene-
endothelial dysfunction in diabetes and metabolic
sis of diabetes and its cardiovascular complications.
syndrome (MetS) by antagonizing NO signaling.
Hyperglycemia and excess nutrient flux, resulting in
Exposure of vascular tissues to increased glucose
OS, activate the innate immune system, including
or free fatty acid concentrations induces the pro-
Toll-like receptors (TLRs).66 TLR stimulation pro-
duction of superoxide, an ROS, from NADPH
motes the production of proinflammatory cytokines
oxidase, mitochondrial sources, and uncoupling
in tissues throughout the body. The proinflamma-
of eNOS.46,47 NADPH oxidase is a major source
tory milieu coupled with endothelial dysfunction in
of superoxide in animal models of diabetes and
diabetes exacerbates stroke risk by leading to the
human vascular grafts.48 Superoxide reacts with
formation of thrombogenic plaques. Interestingly,
NO to form peroxynitrite, which uncouples the
emerging evidence links poor cognitive outcomes
biological activity of eNOS by oxidizing tetrahy-
after stroke to immune dysregulation, suggesting
drobiopterin, a critical cofactor, leading to further
that diabetic inflammation may impair mechanisms
reductions in NO synthesis.49 The loss of the bene-
of neural repair and plasticity, and also contribute to
ficial NO signaling in ECs leads to reduced vasoreg-
neurodegeneration (discussed further below).
ulatory responses, and together with the chron-
Diabetes is associated with increased activ-
ically increased inflammatory milieu in diabetic
ity of the proinflammatory transcription factor
patients, leads to impaired vascular collateraliza-
NF-κB. Hyperglycemia leads to the formation
tion, atherosclerosis, and platelet aggregation.
of AGE and activation of the mitogen-activated
Increased mitochondrial production of ROS
protein kinase pathway, both of which exacerbate
as a result of chronic hyperglycemia is another
inflammatory signaling. The IKK/NF-κB signaling
contributor to OS and inflammation in the diabetic
pathway is an important mediator connecting
vasculature.50,51 At the cellular level, increased
inflammatory and metabolic pathways in IR.67,68
ROS is driven by hyperglycemia-induced increased
The family of NF-κB transcription factors includes
diacylglycerol, which stimulates protein kinase
a collection of dimeric proteins formed from the
C (PKC) and NADPH oxidase. Overproduction
subunits p50, p52, RalA/p65, RelB, and c-Rel.
of ROS has pathological effects in diabetes by
NF-κB is located in the cytoplasm and is associated
disrupting the normal cellular regulation of adeno-
with its inhibitor IκB, but in response to cellular
sine monophosphate kinase (AMPK), leading to
stressors or inflammatory effectors (such as inter-
reduced insulin sensitivity and decreased Akt acti-
leukin (IL)-1β, tumor necrosis factor (TNF)-α,
vation in addition to the decreasing NO signaling as
or lipopolysaccharide), IκB is phosphorylated by
discussed above.52–56 Furthermore, increased ROS
IKK kinase. After ubiquitination, IκB becomes
also leads to a decline in mitochondrial biogenesis
a substrate for the proteasome, which releases
and impaired fission/fusion, a process regulated
an NF-κB dimer that can then enter the nucleus
by peroxisomal proliferator activator receptor
and regulate its target genes. In particular, in
(PPAR)-γ coactivator-1α and nuclear respiratory
vulnerable ECs, NF-κB regulates numerous inflam-
factor-1, which are dependent on eNOS and NO
matory effector molecules to express chemokines,
bioavailability.57–60 Interestingly, the mitochondrial
cytokines, and metalloproteases. This inflamma-
shift toward fission further exacerbates mitochon-
tory microenvironment stimulates the binding of
drial dysfunction and OS leading to a positive
adhesion molecules and recruits T lymphocytes
feedback cycle on further ROS generation.61–64
and monocytes/macrophages, which promote ECM
PKC activation by ROS is also a key regulator of the
remodeling, intimal hyperplasia, foam cell trans-
proinflammatory transcription factor nuclear factor
formation, and atherosclerotic lesion formation.69
kappa B (NF-κB), which acts at many target tissues
Proliferating vascular SMCs further exacerbates
to contribute to diabetes-related complications
plaque formation and contributes to a fibrous cap
discussed further below.65

6 Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences.
Krinock & Singhal Diabetes and stroke

covering the plaque. The release of macrophage reducing infections and mortality in stroke patients.
proteases in advanced plaques breaks down the However, it is unclear if this protective effect holds
fibrous cap and leads to plaque rupture, which can for diabetic stroke patients.89,90 Preclinical studies
precipitate thromboses and stroke. suggest that TLR4 antagonism may reduce “hyper-
In addition to mediating atherogenesis, inflam- inflammatory” responses; however, whether this
matory signals, such as NF-κB, may also be involved can also reduce poststroke immunodepression is
in mediating poststroke immune dysfunction and unknown.86
neuronal degeneration.70–72 In the brain, activation
How does the diabetic milieu impair
of NF-κB can promote survival by stimulating anti-
neuroresilience mechanisms?
apoptotic proteins in the context of conditioning
stimuli.73,74 In diabetes, however, excess TLR4 acti- Poor collateral circulation leads to larger core
vation and NF-κB signaling lead to exaggerated cen- infarcts and reduces reperfusion efficacy
tral and peripheral inflammatory responses, which Neurons rely almost entirely on oxidative
exacerbates neuronal injury and poor recovery in metabolism to sustain their homeostatic func-
animal studies. TLR4 is upregulated after stroke, tions. Owing to this energetic requirement, even
and TLR4-deficient mice have smaller areas of brain just a few minutes after blood vessel occlusion
infarction and lower inflammatory response after from a stroke, millions of neuronal cells within the
middle cerebral artery occlusion (MCAO).75,76 Sim- ischemic core rapidly die. The tissue surround-
ilarly, inhibition of TLR4 signaling in mouse models ing the infarcted core, known as the ischemic
of stroke reduces neuronal cell apoptosis, decreases penumbra (Fig. 1), can remain viable with the
hemorrhagic transformation, and improves neuro- restoration of normal blood flow, as happens with
logical outcomes in diabetic animals.77,78 Although EVT or thrombolytic reperfusion therapies. The
a definitive link between TLR4 or NF-κB and post- presence of prominent leptomeningeal collateral
stroke neuronal degeneration remains to be shown, vessels plays a key role in maintaining enough
sustained glial and microglial activation resulting blood flow to the ischemic penumbra following
from CNS inflammation is strongly associated with stroke and is strongly associated with functional
neuronal degeneration in numerous neurodegen- outcome.91–93 Leptomeningeal collateral vessels are
erative and brain injury models.79–82 Prior stud- anastomotic connections between the distal-most
ies have also linked NF-κB expression to neuronal cerebral arterioles providing redundant blood
degeneration in patients with Alzheimer’s disease supply to branches of the middle, anterior, and
(AD). Thus, additional research focused on uncov- posterior cerebral arteries. Age, hypertension,
ering the mechanistic links between poststroke neu- and MetS or diabetes all impact the robustness
ral degeneration and inflammation may yield widely of cerebral collateral circulation.93 In particular,
applicable insights.83,84 chronic endothelial dysfunction and inflamma-
Altered immune responses in diabetic stroke tion accompanying diabetes negatively influence
patients are also an important contributor to the cerebrovascular reactivity, and preexisting diabetic
poor outcome following stroke. Even in nondi- microvascular disease impedes vascular remod-
abetic patients, poststroke immunodepression is eling, both of which are required to compensate
observed to underly the high rates of pneumo- for acute ischemic stroke.94 For example, diabetic
nia and other infections following stroke.85 Sup- mouse models demonstrate impaired recruitment
pression of neutrophil phagocytic actions has been of collaterals following MCAO compared with
posited to underly susceptibility to infections in dia- nondiabetic animals, which influences the extent
betic patients; however, more recent investigations of ischemic injury and behavioral outcomes.95 In
point to markedly exaggerated cytokine responses patients, poor collateral circulation reduces eligi-
and “hyperinflammation” in diabetes.86 Diabetic bility for EVT, as poor collaterals may reduce the
stroke patients suffer infectious consequences fol- salvageable penumbra at the time of presentation.96
lowing stroke more often than nondiabetic stroke Although it is clear that collateral status impacts
patients,87,88 reflecting a potentiation of the diabetic stroke infarct size and outcome, treatments
immunocompromised state following stroke. Ret- to augment blood volume and flow through
rospective studies point to a role for β-blockers in collateral circulation, such as albumin, have not

Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences. 7
Diabetes and stroke Krinock & Singhal

proven to be effective for stroke in clinical trials.97 burden of preexisting cerebral microvascular
This failure may be related to poor patient selection lesions, which alters imaging features of regional
criteria in clinical trials, as many patients with connectivity.107–109 This may significantly reduce
poor collaterals or nonsalvageable penumbras are the recovery potential of diabetic stroke patients
unlikely to benefit from further volume expansion relative to control subjects. Supporting this hypoth-
by albumin. Further studies are needed to define esis, Huynh and colleagues,110 using transcranial
specific patients subgroups that may benefit from magnetic stimulation, found that patients with
collateral flow augmentation treatments.96 diabetes have impaired capacity for cortical plas-
Reperfusion hemorrhage and cellular injury fol- ticity after an acute stroke. Similarly, diffusion
lowing the restoration of blood flow to infarcted tracer tractography has found that lacunar strokes
and ischemic tissues is a significant clinical con- involving the corona radiata lead to more perma-
cern associated with high morbidity and mor- nent disruptions in motor tracts in patients with
tality. Patients with the poor collateral flow and diabetes.111
diabetes suffer much higher rates of reperfusion The underlying mechanisms by which diabetes
hemorrhage.34,98 Poorer outcomes after EVT in dia- modulates brain regional connectivity and influ-
betic patients despite adequate large vessel reperfu- ences recovery from injury are the areas of active
sion may reflect increased microvascular dysfunc- investigation, but several candidate processes have
tion and susceptibility to “no-reflow.” No-reflow emerged. Studies in diverse animal models of
refers to the absence of brain tissue perfusion diabetes have shown suppressed brain plasticity
despite adequate recanalization of the large vessel processes, including neurogenesis, oligodendroge-
and is thought to be mediated by constriction of nesis, synaptogenesis, and axonal sprouting.112–113
pericytes. Pericytes are smooth muscle–containing Exacerbated white matter injury, suppressed neu-
cells located at the periphery of the microvessel rogenesis and oligodendrogenesis, and impaired
wall that perform critical functions, including the dendritic/spine plasticity have been observed in
maintenance of ECs, the BBB, and microvascu- middle-aged nicotinamide-streptozotocin dia-
lar blood flow.99,100 The ischemic death of peri- betic rats following stroke.114 Neurogenesis and
cytes during stroke leads to smooth muscle con- synaptic reorganization are important for func-
traction of microvessels and impairment of tissue tional improvement after stroke.115 Neurogen-
perfusion.101 Hyperglycemia-induced microvascu- esis in animals occurs in dense clusters around
lar dysfunction leads to a loss of pericytes, which actively dividing ECs, where vascular endothelial
has been described in diabetic retinopathy and growth factor (VEGF) expression is high.116 Fol-
AD.102,103 Interestingly, in addition to contributing lowing stroke, the newly activated vascular ECs
to neuronal ischemia and vascular dysfunction, per- increase the release of brain-derived neurotrophic
icyte dysfunction in diabetes may also have implica- factor (BDNF), whose induction in rodents is
tions for poststroke neural degeneration, as pericyte both spatially and temporally associated with
loss exacerbates BBB dysfunction, neuroinflamma- recruitment of new neurons, axonal growth, and
tion, and tau pathology in mouse models of aging synaptogenesis.117–120 Thus, impaired neural plas-
and AD.99,105 ticity in diabetes may stem from dysfunctional
EC function. Interestingly, HMG-CoA reductase
Diabetic pathophysiology impedes inhibitors (statins), which are indicated and widely
neuroplasticity and stroke recovery prescribed after stroke, are associated with sig-
Cerebral endothelial dysfunction. Diabetes nificant improvement in functional neurologic
slows the patient’s stroke recovery trajectory, recovery in mice.121 The authors find that this may
exacerbates poststroke cognitive decline, and not just be due to vascular benefits of statins, but
increases the risk of poststroke dementia.104,105 also through the promotion of neurogenesis and
Even when adjusting for stroke severity and age, neuronal plasticity by increasing the expression of
diabetes is associated with a slower and poorer VEGF, VEGFR2, and BDNF in the ischemic border
recovery of function.8,106 Interestingly, in diabetic after stroke in mice, providing further rationale
patients without stroke, structural and func- for the benefits of statins in diabetic patients with
tional imaging studies demonstrate an increased stroke.

8 Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences.
Krinock & Singhal Diabetes and stroke

Insulin resistance. The brain is not only glucose- and stimulating trophic factors, but in pathological
sensitive but also insulin-sensitive. Beyond the states, these cells can also lead to neural injury and
effects of insulin in cellular metabolism and the impede repair.139 The opposing proinflammatory
regulation of nutrient intake, insulin also affects (M1) and inflammation-resolving (M2) functions
cognition and neural plasticity.122 Activation of are reflected in the spectrum of immune cellular
insulin receptor and insulin-like growth factor phenotypes acquired by microglia/macrophages
(IGF) receptor signaling pathways improves recov- based on the time course of injury/repair and
ery from brain injury by activating neuronal antiox- microenvironmental milieu. Animal studies link
idant defense and engaging synaptic plasticity diabetes to exacerbated M1 macrophage polariza-
mechanisms.123,124 Brain insulin resistance (BIR) in tion and microglial responses after stroke.113,140
diabetes is a critical modulator of neural metabolic Exacerbated release of proinflammatory cytokines
functions, restorative processes, and susceptibility and M1 polarization of microglia limit benefi-
to neurodegeneration in human patients and ani- cial inflammatory-resolving responses typically
mal models of disease.125,126 A rat model of diabetes observed following injury.141–144 Treatments that
was found to have decreased long-term potentia- promote M2 polarization after stroke result in
tion (LTP) in the hippocampus, a critical process less neuroinflammation and improve stroke
for the formation of memories.127 Rodent models of outcome.113,140 Studies have also linked M1
diabetes have decreased synaptogenesis and axonal microglial and macrophage polarization with
sprouting, which are two key mechanisms under- reduced neurogenesis, axonal regeneration, and
lying diabetes-induced cognitive deficits.128 Ani- synaptic density.145 However, simply ablating M1
mal models of disease also indicate that BIR affects or enhancing M2 responses does not improve out-
stroke outcome. IGF-1 given to mice following come after stroke in animal models,146 and further
MCAO results in improved recovery with improved research is required to refine the time course and
angiogenic and neuroplasticity markers.129 Interest- extent of M1/M2 activation required to optimize
ingly, while systemic administration of insulin to patient recovery. Additional studies with longer-
target lower blood glucose is not beneficial to stroke term follow-up are also required to directly establish
patients, intranasal insulin to overcome BIR has a link between specific inflammatory pathways in
been posited as a potential treatment for demen- diabetes and poststroke dementia. Mechanistically,
tia and stroke. Although no results are yet available animal studies have indicated that stroke in diabetic
on human trials of intranasal insulin in poststroke models leads to Aβ deposition and tau hyperphos-
recovery, it has been shown to be safe and effective phorylation; however, the relationship of this with
at improving cognition in small human studies of macrophage/microglial polarization and specific
patients with AD.130 inflammatory pathways is unknown.147,148 In an
intriguing artificial intelligence–aided review of
Inflammation. Diabetes and MetS are closely medical records of 56 million patients with autoim-
linked with inflammation, cognitive decline, and mune diseases, patients prescribed TNF blockers
AD in human epidemiological studies.131–134 Inter- had a markedly lower risk of developing AD.149
estingly, markers of inflammation associated with Further clinical trials of numerous immunomodu-
AD activity are also found in blood and post- lators for stroke treatment are ongoing (see below),
mortem brains of patients months and even over 1 and specific attention should be placed on dia-
year after stroke.135–137 AD-like pathology, includ- betic populations given their known underlying
ing amyloid (A) β deposition, has also been found pathological inflammation.
in postmortem tissues of stroke survivors with cog-
nitive impairment, suggesting a mechanistic overlap
Beyond glycemic control: treatments with
in diabetic inflammation, poststroke dementia, and
multifaceted actions are needed to harness
AD.138
neuroresilience mechanisms and reverse
Inflammatory cells and cytokines stimulated by
diabetic pathophysiology
stroke or neurodegeneration have both beneficial
and detrimental effects. Activated macrophages or Neuroresilience following brain injury has been
microglia help brain recovery by clearing debris described in numerous laboratory paradigms, such

Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences. 9
Diabetes and stroke Krinock & Singhal

Figure 4. Therapeutics to reverse diabetic pathophysiology and enhance neuroresilience are needed to improve stroke outcome
for diabetic patients. Diabetes worsens stroke injury and antagonizes neuroresilience pathways. In particular, by increasing proin-
flammatory immune function, cellular OS, and even contributing to amyloid (A)-β deposition, diabetes leads to poor stroke
outcomes. Targeting endothelial dysfunction and inflammation with pleotropic therapies like immunomodulators, thiazolidine-
diones (TZDs), adiponectin mimetics, or exercise can stimulate neuroresilience via increased nitric oxide (NO), vascular endothe-
lial growth factor (VEGF), and brain-derived neurotrophic factor (BDNF) signaling.

as following brain injury in animals, evolved to Targeting PPARγ to enhance insulin


withstand harsh environmental conditions, or those sensitivity reduces stroke risk
exposed to prior “conditioning” stimuli.150–152 Clin- Glycemic control is critical to reducing the risk of
icians also frequently observe marked variability in recurrent stroke. However, glycemic control alone
the improvement of patients with similar strokes may not optimize recovery from stroke in diabetic
or brain injuries. Young patients, in particular, pos- patients.156 Pioglitazone is an antidiabetic thiazo-
sess a remarkable degree of capacity for recovery lidinedione (TZD) that promotes normoglycemia
following brain injuries, which has been attributed by activating PPARγ signaling in adipocytes,
to fewer medical comorbidities facilitating less immune and endothelial cells, and other tissues.
detrimental immune responses and vigorous Owing to its direct beneficial effects on metabolism
mechanisms of neural plasticity.153 Specific single and anti-inflammatory properties, pioglitazone has
nucleotide polymorphisms have also been impli- also been investigated as a potential therapeutic
cated in improved outcomes following stroke or agent in patients with IR. The Insulin Resistance
traumatic brain injury, suggesting a genetic basis for Intervention after Stroke (IRIS) trial was designed
neuroresilience mechanisms.154,155 Despite inten- to test the hypothesis that pioglitazone would
sive research efforts, these studies have not trans- reduce the rates of stroke or myocardial infarction
lated into a specific therapeutic to reduce the size in a high-risk group of insulin-resistant patients
of the stroke acutely or to enhance stroke recovery with a history of stroke or transient ischemic attack
and outcome. This failure reflects the complexity of in the last 6 months.157–159 Pioglitazone was found
the multifaceted neural recovery response and its to reduce rates of stroke or myocardial infarction by
heterogeneity among individual patients. Given the approximately one-quarter. Importantly, post-hoc
clear pathophysiological consequences of diabetes analyses revealed that in patients with prediabetes
to stroke risk and recovery, targeting the pleiotropic (HbA1c 5.7–6.4%), good adherence to pioglitazone
pathways discussed above with treatments, such was achieved in 44.2% and resulted in significant
as antidiabetic agents, immunomodulators, and risk reductions in stroke and myocardial infarction
even exercise, to improve outcomes in diabetic (hazard ratio [HR] 0.57, 95% confidence interval
stroke patients presents a significant opportunity in (CI): 0.39–0.84).160 Even more encouraging, in this
precision medicine treatment (Fig. 4). subset of prediabetic patients with good treatment

10 Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences.
Krinock & Singhal Diabetes and stroke

adherence, there was a marked reduction in the published rodent studies have examined the effects
5-year risk of progression to diabetes (HR 0.18, 95% of pioglitazone as a neuroprotective agent in
CI: 0.10–0.33). Real-world retrospective data seem various models of MCAO or focal ischemia. A
to corroborate the beneficial effects of pioglitazone meta-analysis (of studies before 2010) determined
on stroke severity or recovery as well, finding that a cumulative beneficial effect of pioglitazone as
patients admitted for stroke experienced shorter well as rosiglitazone, another TZD.168 However,
median hospitalizations (3 versus 7 days). In spite of despite this relatively rich clinical and preclinical
these positive results, stroke neurologists have been literature, a human neuroprotection trial of acutely
reluctant to adopt TZDs for stroke prevention given administered pioglitazone has not been completed.
safety concerns observed in the trial, including Mechanistically, further research is required to
weight gain and bone fractures requiring hospital- understand the mediators of TZD actions. A variety
ization observed in the treatment group. However, it of effects have been postulated, including broad
is important to note that historical concerns regard- anti-inflammatory and immunomodulatory prop-
ing side effects of weight gain and bladder cancer erties, such as the suppression of proinflammatory
have been overstated in light of post-hoc and real- cytokines and macrophage infiltration. TZDs also
world analyses, and clinicians’ neglect of pioglita- lead to enhanced NOS expression and reduced ROS
zone should be reconsidered.161,162 In an important in target tissues, promote neovascularization, and
post-hoc analysis of the IRIS trial, Spence and increase adiponectin secretion by adipocytes.168
colleagues found only a slight increase in patients
suffering from fractures in the subgroup of predia-
betic patients with good adherence to treatment. In Adiponectin as a candidate for engaging
this subgroup, the number needed to treat (NNT) neuroresilience pathways
to prevent one stroke or myocardial infarction is 24, Another important effect of PPARγ stimula-
and the NNT to prevent diabetes is 12. Conversely, tion and adipogenesis is increasing circulating
the number needed to harm to cause a fracture is adiponectin.169 Adiponectin is a hormone derived
125.160 Interestingly, smaller clinical studies have from adipose tissue and one of the most abundant
also found that low-dose pioglitazone (7.5 mg, one- proteins found in plasma. Its levels are usually
sixth of the dose used in the IRIS trial) is effective higher in women than in men and are down-
in improving metabolic parameters.163,164 Thus, regulated with increasing IR. Adiponectin exerts
additional research is warranted to understand anti-inflammatory, antiatherogenic, and insulin-
whether lower dose pioglitazone regimens also sensitizing effects as well as promotes healthier
reduce long-term cardiovascular risk and whether lipid profiles.170 Total adiponectin circulates in the
specific patient populations with reduced risk of bloodstream in globular, low- (LMW), medium-
TZD-induced weight gain and fracture risk could (MMW), and high-molecular-weight (HMW)
significantly increase the utilization of pioglitazone forms. The biological activity of HMW adiponectin
and improve the clinical outcomes of diabetic stroke is higher for its insulin-sensitizing effects, but
patients.165 other fractions have also been associated with anti-
Interestingly, animal studies have also impli- inflammatory effects. Meta-analyses of prospective
cated TZDs as having a neuroprotective effect cohort studies have not conclusively determined
in acute stroke. In the two preclinical trials of that plasma adiponectin levels are a risk factor
pioglitazone’s acute neuroprotective effects fol- for stroke.171 Low plasma adiponectin levels,
lowing transient MCAO adhering to consensus however, predict an increased risk of 5-year mor-
guidelines for experimental rigor, animals given tality after a first-ever ischemic stroke, although
pioglitazone in the perireperfusion period demon- further studies are required to further understand
strated a marked reduction in histological infarct whether a particular form of adiponectin could
volume and improvement in short-term motor serve as an important therapeutic biomarker for
function.166,167 Gamboa and colleagues found TZD treatment in stroke patients.172 Thus, given
that even with a longer ischemic time, if piogli- the human correlational data and strong mechanis-
tazone is given before reperfusion, infarct size is tic links to vasculo- and neuroprotective actions,
limited.162 In addition to these studies, 13 other adiponectin is a promising target to effect multiple

Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences. 11
Diabetes and stroke Krinock & Singhal

neuroresilience-promoting benefits in diabetic Modulating aberrant immune responses


stroke patients. Inflammation after stroke can persist in the brain
Protective actions of adiponectin in stroke may for months or even years. Chronic inflammatory
also be due to pleiotropic actions, including pro- responses following stroke may represent a link
tection against neurotoxic insults and enhancement between impaired immunity, poor outcomes, and
of EC function. Adiponectin improves endothe- increased rates of poststroke dementia after stroke
lial function in pathological inflammatory states in diabetic patients.187–190 Although our knowledge
by suppressing ROS, activating eNOS, and increas- about poststroke neuroinflammation has improved,
ing the bioavailability of NO at the vascular further clinical research is necessary to establish
endothelium.173–175 Adiponectin also reduces the a clear link between diabetes, the time course
NF-κB activation induced by hyperglycemia and of poststroke inflammatory processes, and neuro-
TNF-α in ECs, which prevents atherogenesis.176 logical and neuropsychiatric outcomes. Expand-
In addition, adiponectin is protective in numer- ing our understanding of the impact of diabetes
ous models of neurotoxicity, including stroke,177,178 on poststroke inflammation will open the door
kainate-induced excitotoxicity,179 and Aβ toxicity for immunomodulation tailored to patient-specific
during OS in vitro and in vivo.180,181 Interest- variables. Numerous clinical trials of immunomod-
ingly, studies in mice have shown that the loss ulators have been performed in stroke patients;191
of adiponectin–adiponectin receptor-1 signaling in unfortunately, definitive phase 3 trials testing rele-
the brain results in poor performance on cog- vant neuropsychiatric outcome measures are lack-
nitive tests and expression of proinflammatory ing. However, if targeted specifically toward patients
cytokines and activated microglia.182 This high- with aberrant immune responses, such as patients
lights an important role for diabetes and MetS in with diabetes, and outcome measures expanded to
modulating the development of neurodegenerative include depression and cognition, previously trialed
conditions. A further delineation of the role of therapies may yield more promising results.
adiponectin signaling in the brain, particularly fol- Dampening diabetes-induced activation of
lowing stroke and brain injury, will yield important innate and adaptive immunity may be an impor-
insights into new potential treatments. tant target to encourage improved neurorecovery.
Adiponectin peptide mimetics and receptor ago- Animal studies have noted brain atrophy and
nists are being developed, but none has under- necrosis following stroke is dependent on proin-
gone extensive study, and further research will be flammatory cytokines leading to excessive protease
required to understand their potential uses for treat- activity.192,193 Interestingly, high levels of IL-1β and
ing diabetes and its complications. Drugs target- glucose have been linked to poststroke fatigue,194
ing AMPK, a major effector of adiponectin action, while the elevation of IL-6 and TNF-α, which are
maybe another avenue to target numerous antidia- downstream of NF-κB, has been linked to stroke
betic pathways.183 AMPK stimulation by metformin severity and poststroke depression.195,196 Two
or other drugs has been associated with improved FDA-approved immunomodulating medications
outcomes in animal models of stroke by improv- that reduce leukocyte infiltration into the brain,
ing mitochondrial function, promoting beneficial fingolimod and anakinra, have demonstrated some
vascular effects, and decreasing inflammation.184 encouraging biological activity in clinical trials,
However, other drugs that stimulate AMPK more though additional trials are needed to firmly estab-
broadly, such as the adenosine analog AICAR, lish clinical efficacy.197–199 Additionally, fasudil,
have been associated with poorer outcomes after an inhibitor of neutrophil and monocyte infiltra-
stroke, which may reflect cell- and time-specific tion into the brain that improves M2 polarization,
windows of therapeutic effect.185,186 Thus, further has also shown minor functional improvement
filling in knowledge gaps in our understanding of in small clinical trials.200 Tocilizumab, a mono-
AMPK in the regulation of energy homeostasis, clonal antibody directed against the IL-6 receptor
immunity, and neural plasticity is necessary to sup- FDA-approved for the treatment of cytokine
port the use of activators of AMPK in ischemic release syndrome and other immune disorders,
stroke. has demonstrated efficacy in a small number of

12 Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences.
Krinock & Singhal Diabetes and stroke

animal models of stroke201 Of note, fingolimod, increased serum levels of BDNF are most consis-
fasudil, and tocilizumab are all currently being tently observed following aerobic exercise sessions
tested in a multicenter preclinical trial sponsored that last more than 30 min and at a frequency of
by the NINDS. This information will be partic- 4 days per week,215 which is very similar to 150
ularly valuable when considering human clinical min weekly recommended by clinical practice
trials, as IL-6 and subacute inflammatory responses guidelines.216 In animal models of stroke, exercise
mediated by leukocytes, in particular, have been promotes angiogenesis, synaptic plasticity, and
implicated in early detrimental but subacute ben- even neurogenesis via VEGF and BDNF signal-
eficial neurotrophic effects in stroke and brain ing pathways that are highly dependent on NO
injury.191,202 signaling.217,218 Physical activity is a potent inducer
of eNOS expression in the vasculature and protects
Exercise enhances vascular function and against ischemic stroke in part by helping establish
neuroplasticity poststroke robust cerebral collateral blood supplies.219–221
Exercise and physical activity are some of the most Mice deficient in eNOS also demonstrate reduced
powerful predictors of reduced cardiovascular risk BDNF expression and poorer functional recovery
in diabetic patients. Long-term longitudinal data after stroke.118 BDNF improves synaptic matu-
from the Nurses’ Health Study suggest that in dia- ration and density via regulation of synapsin I
betics, >4 h of exercise per week reduces the and postsynaptic density protein-95.222,223 These
risk of stroke by almost 50%.203 Prospective and interactions also facilitate excitatory glutamate
randomized clinical studies of exercise interven- neurotransmission which supports activity depen-
tions have found that various regimens of exer- dent plasticity.224–226 eNOS also plays a prominent
cise improve glycemic control204 as well as restore role in poststroke VEGF-induced proliferation of
healthier endothelial and metabolic phenotypes in ECs and angiogenesis.227,228 Thus, exercise acti-
diabetic patients.205,206 Importantly, emerging data vates eNOS and BDNF pathways to counter the
also suggest that exercise and physical activity aid endothelial dysfunction of diabetes229 and activate
in the poststroke recovery process by promoting neuroresilience pathways to improve stroke recov-
both angiogenesis and neuroplasticity. In fact, there ery. Interestingly, the use of electrical or magnetic
is increasing evidence that exercise-induced neu- brain stimulation protocols to improve plasticity
roplasticity is highly dependent on angiogenesis following brain injury is an area of active investi-
in experimental models of recovery after ischemic gation that may also take advantage of pathways
brain injury.207 similar to exercise.230–232 Plasticity induced by
Neurogenesis is extremely limited in adult transcranial magnetic or electrical stimulation is
humans if present at all.208 Thus, the ability of the thought to involve mechanisms of LTP, including
brain to adapt to injuries like stroke is through activation of eNOS.233 Thus, of great interest is
the regeneration or growth of neuronal axons, ongoing research aimed at optimizing exercise reg-
synapses, and dendrites.209 The poststroke func- imens based on individual patient characteristics as
tional recovery phase is characterized by the well as enhancing its effects with adjunctive brain
resolution of inflammation coupled with coordi- stimulation.
nated neurotrophic and angiogenic processes.210
Conclusions
As outlined above, BDNF plays a central role in
increased poststroke neuroplasticity along with Diabetes is one of the most important risk factors
its receptor tyrosine kinase B. BDNF coordinates for stroke. Diabetic patients suffer worse stroke
structural remodeling at synaptic, axonal, and den- outcomes, and have a poorer prognosis for recov-
dritic levels with the participation of downstream ery compared with patients without diabetes. The
synaptic and endothelial targets.211 Remarkably, highest profile and largest clinical trials of diabetes
human and animal studies strongly link aerobic and stroke have focused on treating hyperglycemia;
exercise training with amplified BDNF signaling as however, treating hyperglycemia with insulin alone
well as synaptic proteins, such as growth-associated is simply not enough to improve outcomes in dia-
protein-43, and angiogenic factors, such as IGF-1 betic or insulin-resistant patients. The pathophysio-
and VEGF.210,212–214 Evidence suggests that logical processes resulting from and accompanying

Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences. 13
Diabetes and stroke Krinock & Singhal

diabetes, including IR, endothelial dysfunction, the American Heart Association. Circulation 137: e67–
and inflammation, directly counter endogenous e492.
2. Kissela, B.M., J. Khoury, D. Kleindorfer, et al. 2005. Epi-
resilience and recovery mechanisms activated fol-
demiology of ischemic stroke in patients with diabetes: the
lowing stroke. A pressing need exists to intensify Greater Cincinnati/Northern Kentucky Stroke Study. Dia-
translational research efforts to take advantage of betes Care 28: 355–359.
the plethora of this basic science knowledge and 3. Janghorbani, M., F.B. Hu, W.C. Willett, et al. 2007. Prospec-
understand which treatments to prioritize to limit tive study of type 1 and type 2 diabetes and risk of stroke
subtypes: the Nurses’ Health Study. Diabetes Care 30:
stroke injury and optimize recovery in diabetic
1730–1735.
stroke patients. These efforts will require both 4. The Emerging Risk Factors Collaboration. 2010. Diabetes
improving animal models of stroke and performing mellitus, fasting blood glucose concentration, and risk
rigorous multicenter preclinical trials (as is being of vascular disease: a collaborative meta-analysis of 102
pursued through the NINDS Stroke Preclinical prospective studies. Lancet 375: 2215–2222.
5. Hall, M.J., S. Levant & C.J. DeFrances. 2012. NCHS Data
Assessment Network initiative). Well-designed
Brief No. 95.
clinical trials with relevant outcomes focusing on 6. Centers for Disease Control and Prevention. 2020. National
patients with diabetes and IR are also needed. In Diabetes Statistics Report: Estimates of Diabetes and Its
addition, for treatments that have demonstrated Burden in the United States.
promise in reducing stroke and cardiovascular risk 7. Olsson, T., M. Viitanen, K. Asplund, et al. 1990. Prognosis
after stroke in diabetic patients. A controlled prospective
in diabetic patients, such as pioglitazone, efforts to
study. Diabetologia 33: 244–249.
increase physician adoption and further optimize 8. Jørgensen, H., H. Nakayama, H.O. Raaschou, et al. 1994.
patient response are critical. Recent advances in Stroke in patients with diabetes. The Copenhagen Stroke
the primary and secondary prevention of stroke, as Study. Stroke 25: 1977–1984.
well as the acute treatment of highly morbid strokes 9. Hachinski, V.C. & J.W. Norris. 1985. The vascular infras-
tructure. In The Acute Stroke. F.A. Davis, Ed.: 27–40.
resulting from LVO, have improved the treatment
Philadelphia, PA: F.A. Davis.
landscape for stroke patients. However, these 10. Caplan, L.R. 2015. Lacunar infarction and small vessel dis-
treatments have not resulted in similar gains for ease: pathology and pathophysiology. J. Stroke 17: 2–6.
diabetic patients. Thus, many important unresolved 11. Campbell, B.C.V. & P. Khatri. 2020. Stroke. Lancet North
questions remain, and we are optimistic that the Am. Ed. 396: 129–142.
12. Elmore, E.M., A. Mosquera & J. Weinberger. 2003. The
combined insights from animal models and clinical
prevalence of asymptomatic intracranial large-vessel occlu-
trials will ultimately provide treatments to encour- sive disease: the role of diabetes. J. Neuroimaging 13: 224–
age neuroresilience and minimize the burden of 227.
disability from stroke in the diabetic population. 13. Fujiyoshi, A., M.F.K. Suri, A. Alonso, et al. 2020. Hyper-
glycemia, duration of diabetes, and intracranial atheroscle-
Acknowledgments rotic stenosis by magnetic resonance angiography: the
ARIC-NCS study. J. Diabetes Complications 34: 107605.
We thank the American Heart Association 14. Tuttolomondo, A., A. Pinto, G. Salemi, et al. 2008. Dia-
(18CDA3403044) and the Hellman Family Fund betic and nondiabetic subjects with ischemic stroke: dif-
for financial support (N.S.S.). ferences, subtype distribution and outcome. Nutr. Metab.
Cardiovasc. Dis. 18: 152–157.
Author contributions 15. Arboix, A., A. Font, C. Garro, et al. 2007. Recurrent lacu-
nar infarction following a previous lacunar stroke: a clin-
Both authors participated in conceptualizing the ical study of 122 patients. J. Neurol. Neurosurg. Psychiatry
review, drafting the manuscript, revising its intellec- 78: 1392–1394.
tual content, and approved the final version of the 16. Hart, R.G., L.A. Pearce, M.F. Bakheet, et al. 2014. Pre-
dictors of stroke recurrence in patients with recent lacu-
submitted manuscript.
nar stroke and response to interventions according to
Competing interests risk status: Secondary Prevention of Small Subcortical
Strokes (SPS3) trial. J. Stroke Cerebrovasc. Dis. 23: 618–
The authors declare no competing interests. 624.
17. Hankey Graeme, J., K. Jamrozik, J. Broadhurst Robyn, et al.
1998. Long-term risk of first recurrent stroke in the Perth
Community Stroke Study. Stroke 29: 2491–2500.
References 18. Tuomilehto, J., D. Rastenytė, P. Jousilahti, et al. 1996. Dia-
1. Benjamin, E.J., S.S. Virani, C.W. Callaway, et al. 2018. Heart betes mellitus as a risk factor for death from stroke. Stroke
disease and stroke statistics—2018 update: a report from 27: 210–215.

14 Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences.
Krinock & Singhal Diabetes and stroke

19. Megherbi, S.-E., C. Milan, D. Minier, et al. 2003. Associa- 37. Martini, S.R. & T.A. Kent. 2007. Hyperglycemia in acute
tion between diabetes and stroke subtype on survival and ischemic stroke: a vascular perspective. J. Cereb. Blood Flow
functional outcome 3 months after stroke. Stroke 34: 688– Metab. 27: 435–451.
694. 38. Rehni, A.K., A. Liu, M.A. Perez-Pinzon, et al. 2017. Dia-
20. Sprafka, J.M., B.A. Virnig, E. Shahar, et al. 1994. Trends in betic aggravation of stroke and animal models. Exp. Neurol.
diabetes prevalence among stroke patients and the effect of 292: 63–79.
diabetes on stroke survival: the Minnesota Heart Survey. 39. Parsons, M.W., P.A. Barber, P.M. Desmond, et al. 2002.
Diabet. Med. 11: 678–684. Acute hyperglycemia adversely affects stroke outcome: a
21. Stevens, R.J., R.L. Coleman, A.I. Adler, et al. 2004. Risk fac- magnetic resonance imaging and spectroscopy study. Ann.
tors for myocardial infarction case fatality and stroke case Neurol. 52: 20–28.
fatality in type 2 diabetes. Diabetes Care 27: 201–207. 40. Baird Tracey, A., W. Parsons Mark, T. Phan, et al. 2003.
22. 1995. Tissue plasminogen activator for acute ischemic Persistent poststroke hyperglycemia is independently asso-
stroke. N. Engl. J. Med. 333: 1581–1588. ciated with infarct expansion and worse clinical outcome.
23. Nogueira, R.G. & M. Ribó. 2019. Endovascular treatment Stroke 34: 2208–2214.
of acute stroke. Stroke 50: 2612–2618. 41. Shimoyama, T., K. Kimura, J. Uemura, et al. 2014. Elevated
24. Arnold, M., S. Mattle, A. Galimanis, et al. 2014. Impact of glucose level adversely affects infarct volume growth and
admission glucose and diabetes on recanalization and out- neurological deterioration in nondiabetic stroke patients,
come after intra-arterial thrombolysis for ischaemic stroke. but not diabetic stroke patients. Eur. J. Neurol. 21: 402–410.
Int. J. Stroke 9: 985–991. 42. Johnston, K.C., A. Bruno, Q. Pauls, et al. 2019. Intensive vs
25. Lu, G.-D., Z.-Q. Ren, J.-X. Zhang, et al. 2018. Effects of dia- standard treatment of hyperglycemia and functional out-
betes mellitus and admission glucose in patients receiving come in patients with acute ischemic stroke. JAMA 322:
mechanical thrombectomy: a systematic review and meta- 326–335.
analysis. Neurocrit. Care 29: 426–434. 43. Chamorro, Á., S. Brown, S. Amaro, et al. 2019. Glu-
26. Yahagi, K., D. Kolodgie Frank, C. Lutter, et al. 2017. Pathol- cose modifies the effect of endovascular thrombectomy in
ogy of human coronary and carotid artery atherosclerosis patients with acute stroke. Stroke 50: 690–696.
and vascular calcification in diabetes mellitus. Arterioscler. 44. Osei, E., M. den Hertog Heleen, A. Berkhemer Olvert, et al.
Thromb. Vasc. Biol. 37: 191–204. 2017. Admission glucose and effect of intra-arterial treat-
27. Tabit, C.E., W.B. Chung, N.M. Hamburg, et al. 2010. ment in patients with acute ischemic stroke. Stroke 48:
Endothelial dysfunction in diabetes mellitus: molecular 1299–1305.
mechanisms and clinical implications. Rev. Endocr. Metab. 45. Creager, M.A. & T.F. Lüscher. 2003. Diabetes and vascular
Disord. 11: 61–74. disease: pathophysiology, clinical consequences, and med-
28. Last, D., D.C. Alsop, A.M. Abduljalil, et al. 2007. Global and ical therapy: part I. Circulation 108: 1527–1532.
regional effects of type 2 diabetes on brain tissue volumes 46. Tesfamariam, B. & R.A. Cohen. 1992. Free radicals medi-
and cerebral vasoreactivity. Diabetes Care 30: 1193. ate endothelial cell dysfunction caused by elevated glucose.
29. Jansen, J.F.A., F.C.G. van Bussel, H.J. van de Haar, et al. Am. J. Physiol. 263: H321–H326.
2016. Cerebral blood flow, blood supply, and cognition in 47. Gao, L. & G.E. Mann. 2009. Vascular NAD(P)H oxidase
type 2 diabetes mellitus. Sci. Rep. 6: 10. activation in diabetes: a double-edged sword in redox sig-
30. Venkat, P., M. Chopp & J. Chen. Blood–brain barrier dis- nalling. Cardiovasc. Res. 82: 9–20.
ruption, vascular impairment, and ischemia/reperfusion 48. Guzik Tomasz, J., M. Shafi, G. Daniela, et al. 2002. Mech-
damage in diabetic stroke. J. Am. Heart Assoc. 6: e005819. anisms of increased vascular superoxide production in
31. Iadecola, C. & R.L. Davisson. 2008. Hypertension and cere- human diabetes mellitus. Circulation 105: 1656–1662.
brovascular dysfunction. Cell Metab. 7: 476–484. 49. Chen, C.-A., T.-Y. Wang, S. Varadharaj, et al. 2010. S-
32. Bruno, A., S.R. Levine, M.R. Frankel, et al. 2002. Admis- glutathionylation uncouples eNOS and regulates its cellular
sion glucose level and clinical outcomes in the NINDS rt- and vascular function. Nature 468: 1115–1118.
PA Stroke Trial. Neurology 59: 669–674. 50. Balaban, R.S., S. Nemoto & T. Finkel. 2005. Mitochondria,
33. Sacco, R.L., T. Shi, M.C. Zamanillo, et al. 1994. Predictors of oxidants, and aging. Cell 120: 483–495.
mortality and recurrence after hospitalized cerebral infarc- 51. Volpe, C.M.O., P.H. Villar-Delfino, P.M.F. dos Anjos, et al.
tion in an urban community. Neurology 44: 626–634. 2018. Cellular death, reactive oxygen species (ROS) and
34. Demchuk Andrew, M., B. Morgenstern Lewis, W. Krieger diabetic complications. Cell Death Dis. 9: 119.
Derk, et al. 1999. Serum glucose level and diabetes predict 52. Brownlee, M. 2001. Biochemistry and molecular cell biol-
tissue plasminogen activator–related intracerebral hemor- ogy of diabetic complications. Nature 414: 813–820.
rhage in acute ischemic stroke. Stroke 30: 34–39. 53. Midaoui Adil, E.L. & J. de Champlain. 2002. Prevention of
35. Capes Sarah, E., D. Hunt, K. Malmberg, et al. 2001. Stress hypertension, insulin resistance, and oxidative stress by α-
hyperglycemia and prognosis of stroke in nondiabetic and lipoic acid. Hypertension 39: 303–307.
diabetic patients. Stroke 32: 2426–2432. 54. Hagen, T.M., R.T. Ingersoll, J. Lykkesfeldt, et al. 1999. (R)-
36. Suh, S.W., B.S. Shin, H. Ma, et al. 2008. Glucose and alpha-lipoic acid-supplemented old rats have improved
NADPH oxidase drive neuronal superoxide formation in mitochondrial function, decreased oxidative damage, and
stroke. Ann. Neurol. 64: 654–663. increased metabolic rate. FASEB J. 13: 411–418.

Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences. 15
Diabetes and stroke Krinock & Singhal

55. Smith, A.R. & T.M. Hagen. 2003. Vascular endothelial dys- 73. Chu, Z.-L., T.A. McKinsey, L. Liu, et al. 1997. Suppression
function in aging: loss of Akt-dependent endothelial nitric of tumor necrosis factor-induced cell death by inhibitor of
oxide synthase phosphorylation and partial restoration apoptosis c-IAP2 is under NF-κB control. Proc. Natl. Acad.
by (R)-alpha-lipoic acid. Biochem. Soc. Trans. 31: 1447– Sci. USA 94: 10057–10062.
1449. 74. Tamatani, M., Y.H. Che, H. Matsuzaki, et al. 1999. Tumor
56. Heitzer, T., B. Finckh, S. Albers, et al. 2001. Benefi- necrosis factor induces Bcl-2 and Bcl-x expression through
cial effects of alpha-lipoic acid and ascorbic acid on NFkappaB activation in primary hippocampal neurons. J.
endothelium-dependent, nitric oxide-mediated vasodila- Biol. Chem. 274: 8531–8538.
tion in diabetic patients: relation to parameters of oxidative 75. Caso, J.R., J.M. Pradillo, O. Hurtado, et al. 2007. Toll-like
stress. Free Radic. Biol. Med. 31: 53–61. receptor 4 is involved in brain damage and inflammation
57. Nisoli, E., E. Clementi, C. Paolucci, et al. 2003. Mitochon- after experimental stroke. Circulation 115: 1599–1608.
drial biogenesis in mammals: the role of endogenous nitric 76. Hyakkoku, K., J. Hamanaka, K. Tsuruma, et al. 2010. Toll-
oxide. Science 299: 896–899. like receptor 4 (TLR4), but not TLR3 or TLR9, knock-
58. Nisoli, E., S. Falcone, C. Tonello, et al. 2004. Mitochondrial out mice have neuroprotective effects against focal cerebral
biogenesis by NO yields functionally active mitochondria ischemia. Neuroscience 171: 258–267.
in mammals. Proc. Natl. Acad. Sci. USA 101: 16507–16512. 77. Li, C., L.-H. Che, T.-F. Ji, et al. 2017. Effects of the TLR4
59. Nisoli, E., E. Clementi, M.O. Carruba, et al. 2007. Defec- signaling pathway on apoptosis of neuronal cells in dia-
tive mitochondrial biogenesis: a hallmark of the high car- betes mellitus complicated with cerebral infarction in a rat
diovascular risk in the metabolic syndrome? Circ. Res. 100: model. Sci. Rep. 7: 43834.
795–806. 78. Abdul, Y., M. Abdelsaid, W. Li, et al. 2019. Inhibition
60. Nisoli, E., C. Tonello, A. Cardile, et al. 2005. Calorie restric- of toll-like receptor-4 (TLR-4) improves neurobehavioral
tion promotes mitochondrial biogenesis by inducing the outcomes after acute ischemic stroke in diabetic rats: pos-
expression of eNOS. Science 310: 314–317. sible role of vascular endothelial TLR-4. Mol. Neurobiol. 56:
61. Twig, G., B. Hyde & O.S. Shirihai. 2008. Mitochondrial 1607–1617.
fusion, fission and autophagy as a quality control axis: 79. Nguyen, M.D., J.-P. Julien & S. Rivest. 2002. Innate immu-
the bioenergetic view. Biochim. Biophys. Acta 1777: 1092– nity: the missing link in neuroprotection and neurodegen-
1097. eration? Nat. Rev. Neurosci. 3: 216–227.
62. Twig, G., A. Elorza, A.J.A. Molina, et al. 2008. Fission 80. Smith, D.H., V.E. Johnson & W. Stewart. 2013. Chronic
and selective fusion govern mitochondrial segregation and neuropathologies of single and repetitive TBI: substrates of
elimination by autophagy. EMBO J. 27: 433–446. dementia? Nat. Rev. Neurol. 9: 211–221.
63. Yu, T., S.-S. Sheu, J.L. Robotham, et al. 2008. Mitochondrial 81. Ransohoff, R.M., D. Schafer, A. Vincent, et al. 2015. Neu-
fission mediates high glucose-induced cell death through roinflammation: ways in which the immune system affects
elevated production of reactive oxygen species. Cardiovasc. the brain. Neurotherapeutics 12: 896–909.
Res. 79: 341–351. 82. Shi, K., D.-C. Tian, Z.-G. Li, et al. 2019. Global brain
64. Yu, T., J.L. Robotham & Y. Yoon. 2006. Increased produc- inflammation in stroke. Lancet Neurol. 18: 1058–1066.
tion of reactive oxygen species in hyperglycemic condi- 83. Terai, K., A. Matsuo & P.L. McGeer. 1996. Enhancement
tions requires dynamic change of mitochondrial morphol- of immunoreactivity for NF-kappa B in the hippocampal
ogy. Proc. Natl. Acad. Sci. USA 103: 2653–2658. formation and cerebral cortex of Alzheimer’s disease. Brain
65. Lowell, B.B. & G.I. Shulman. 2005. Mitochondrial dysfunc- Res. 735: 159–168.
tion and type 2 diabetes. Science 307: 384–387. 84. Boissière, F., S. Hunot, B. Faucheux, et al. 1997. Nuclear
66. Nogueira-Machado, J.A., C.M. de O. Volpe, C.A. Veloso, translocation of NF-kappaB in cholinergic neurons of
et al. 2011. HMGB1, TLR and RAGE: a functional tripod patients with Alzheimer’s disease. Neuroreport 8: 2849–
that leads to diabetic inflammation. Expert Opin. Ther. Tar- 2852.
gets 15: 1023–1035. 85. Katzan, I.L., R.D. Cebul, S.H. Husak, et al. 2003. The effect
67. Hotamisligil, G.S. & E. Erbay. 2008. Nutrient sensing and of pneumonia on mortality among patients hospitalized for
inflammation in metabolic diseases. Nat. Rev. Immunol. 8: acute stroke. Neurology 60: 620–625.
923–934. 86. Nielsen, T.B., P. Pantapalangkoor, J. Yan, et al. 2017. Dia-
68. Medzhitov, R. 2008. Origin and physiological roles of betes exacerbates infection via hyperinflammation by sig-
inflammation. Nature 454: 428–435. naling through TLR4 and RAGE. mBio 8: e00818–17.
69. Hansson Göran, K. 2001. Immune mechanisms in 87. Liao, C.-C., C.-C. Shih, C.-C. Yeh, et al. 2015. Impact of
atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 21: diabetes on stroke risk and outcomes. Medicine (Baltimore)
1876–1890. 94: e2282.
70. Hayden, M.S., A.P. West & S. Ghosh. 2006. NF-κB and the 88. Liu, D.-D., S.-F. Chu, C. Chen, et al. 2018. Research
immune response. Oncogene 25: 6758–6780. progress in stroke-induced immunodepression syndrome
71. Harari, O.A. & J.K. Liao. 2010. NF-κB and innate immunity (SIDS) and stroke-associated pneumonia (SAP). Neu-
in ischemic stroke. Ann. N.Y. Acad. Sci. 1207: 32–40. rochem. Int. 114: 42–54.
72. Downes, C.E. & P.J. Crack. 2010. Neural injury following 89. Dziedzic, T., A. Slowik, J. Pera, et al. 2007. Beta-blockers
stroke: are Toll-like receptors the link between the immune reduce the risk of early death in ischemic stroke. J. Neurol.
system and the CNS? Br. J. Pharmacol. 160: 1872–1888. Sci. 252: 53–56.

16 Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences.
Krinock & Singhal Diabetes and stroke

90. Vogelgesang, A. & A. Dressel. 2011. Immunological con- 106. Sweetnam, D., A. Holmes, K.A. Tennant, et al. 2012. Dia-
sequences of ischemic stroke: immunosuppression and betes impairs cortical plasticity and functional recovery fol-
autoimmunity. J. Neuroimmunol. 231: 105–110. lowing ischemic stroke. J. Neurosci. 32: 5132–5143.
91. Miteff, F., C.R. Levi, G.A. Bateman, et al. 2009. The inde- 107. Brundel, M., L.J. Kappelle & G.J. Biessels. 2014. Brain imag-
pendent predictive utility of computed tomography angio- ing in type 2 diabetes. Eur. Neuropsychopharmacol. 24:
graphic collateral status in acute ischaemic stroke. Brain J. 1967–1981.
Neurol. 132: 2231–2238. 108. Chen, Y.-C., Y. Jiao, Y. Cui, et al. 2014. Aberrant brain
92. Maas, M.B., M.H. Lev, H. Ay, et al. 2009. Collateral vessels functional connectivity related to insulin resistance in type
on CTA predict outcome in acute ischemic stroke. Stroke J. 2 diabetes: a resting-state fMRI study. Diabetes Care 37:
Cereb. Circ. 40: 3001–3005. 1689–1696.
93. Menon, B.K., E.E. Smith, J. Modi, et al. 2011. Regional 109. Zhang, D., J. Gao, X. Yan, et al. 2020. Altered functional
leptomeningeal score on CT angiography predicts clinical connectivity of brain regions based on a meta-analysis
and imaging outcomes in patients with acute anterior cir- in patients with T2DM: a resting-state fMRI study. Brain
culation occlusions. AJNR Am. J. Neuroradiol. 32: 1640– Behav. 10: e01725.
1645. 110. Huynh, W., N. Kwai, R. Arnold, et al. 2017. The effect of dia-
94. Poittevin, M., P. Bonnin, C. Pimpie, et al. 2015. Diabetic betes on cortical function in stroke: implications for post-
microangiopathy: impact of impaired cerebral vasoreactiv- stroke plasticity. Diabetes 66: 1661.
ity and delayed angiogenesis after permanent middle cere- 111. Moon, J.S., S.M. Chung, S.H. Jang, et al. 2019. Effects of
bral artery occlusion on stroke damage and cerebral repair diabetes on motor recovery after cerebral infarct: a diffu-
in mice. Diabetes 64: 999–1010. sion tensor imaging study. J. Clin. Endocrinol. Metab. 104:
95. Akamatsu, Y., Y. Nishijima, C.C. Lee, et al. 2015. Impaired 3851–3858.
leptomeningeal collateral flow contributes to the poor out- 112. Stranahan, A.M., T.V. Arumugam, R.G. Cutler, et al.
come following experimental stroke in the type 2 diabetic 2008. Diabetes impairs hippocampal function through
mice. J. Neurosci. 35: 3851. glucocorticoid-mediated effects on new and mature neu-
96. Bang Oh, Y., M. Goyal & S. Liebeskind David. 2015. Collat- rons. Nat. Neurosci. 11: 309–317.
eral circulation in ischemic stroke. Stroke 46: 3302–3309. 113. Ma, S., J. Wang, Y. Wang, et al. 2018. Diabetes melli-
97. Martin Renee’, H., D. Yeatts Sharon, D. Hill Michael, et al. tus impairs white matter repair and long-term functional
2016. ALIAS (Albumin in Acute Ischemic Stroke) Trials. deficits after cerebral ischemia. Stroke 49: 2453–2463.
Stroke 47: 2355–2359. 114. Zhang, L., M. Chopp, Y. Zhang, et al. 2016. Diabetes melli-
98. Bang Oh, Y., L. Saver Jeffrey, J. Kim Suk, et al. 2011. Collat- tus impairs cognitive function in middle-aged rats and neu-
eral flow averts hemorrhagic transformation after endovas- rological recovery in middle-aged rats after stroke. Stroke
cular therapy for acute ischemic stroke. Stroke 42: 2235– 47: 2112–2118.
2239. 115. Hallett, M. 2001. Plasticity of the human motor cortex and
99. Bell, R.D., E.A. Winkler, A.P. Sagare, et al. 2010. Pericytes recovery from stroke. Brain Res. Rev. 36: 169–174.
control key neurovascular functions and neuronal pheno- 116. Palmer, T.D., A.R. Willhoite & F.H. Gage. 2000. Vascular
type in the adult brain and during brain aging. Neuron 68: niche for adult hippocampal neurogenesis. J. Comp. Neurol.
409–427. 425: 479–494.
100. Hall, C.N., C. Reynell, B. Gesslein, et al. 2014. Capillary per- 117. Nakahashi, T., H. Fujimura, C.A. Altar, et al. 2000. Vascular
icytes regulate cerebral blood flow in health and disease. endothelial cells synthesize and secrete brain-derived neu-
Nature 508: 55–60. rotrophic factor. FEBS Lett. 470: 113–117.
101. Kloner, R.A., K.S. King & M.G. Harrington. 2018. No- 118. Chen, J., A. Zacharek, C. Zhang, et al. 2005. Endothelial
reflow phenomenon in the heart and brain. Am. J. Physiol. nitric oxide synthase regulates brain-derived neurotrophic
Heart Circ. Physiol. 315: H550–H562. factor expression and neurogenesis after stroke in mice. J.
102. Romeo, G., W.-H. Liu, V. Asnaghi, et al. 2002. Activation Neurosci. 25: 2366–2375.
of nuclear factor-kappaB induced by diabetes and high glu- 119. Cui, X., M. Chopp, A. Zacharek, et al. 2013. Endothelial
cose regulates a proapoptotic program in retinal pericytes. nitric oxide synthase regulates white matter changes via
Diabetes 51: 2241–2248. the BDNF/TrkB pathway after stroke in mice. PLoS One 8:
103. Sagare, A.P., R.D. Bell, Z. Zhao, et al. 2013. Pericyte loss e80358.
influences Alzheimer-like neurodegeneration in mice. Nat. 120. Madinier, A., N. Bertrand, M. Rodier, et al. 2013. Ipsilat-
Commun. 4: 2932. eral versus contralateral spontaneous post-stroke neuro-
104. Leibson, C.L., W.A. Rocca, V.A. Hanson, et al. 1997. plastic changes: involvement of BDNF? Neuroscience 231:
Risk of dementia among persons with diabetes mellitus: a 169–181.
population-based cohort study. Am. J. Epidemiol. 145: 301– 121. Chen, J., C. Zhang, H. Jiang, et al. 2005. Atorvastatin induc-
308. tion of VEGF and BDNF promotes brain plasticity after
105. Cukierman, T., H.C. Gerstein & J.D. Williamson. 2005. stroke in mice. J. Cereb. Blood Flow Metab. 25: 281–290.
Cognitive decline and dementia in diabetes—systematic 122. Fernandez, A.M. & I. Torres-Alemán. 2012. The many faces
overview of prospective observational studies. Diabetolo- of insulin-like peptide signalling in the brain. Nat. Rev.
gia 48: 2460–2469. Neurosci. 13: 225–239.

Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences. 17
Diabetes and stroke Krinock & Singhal

123. Martin-Montañez, E., J. Pavia, L.J. Santin, et al. 2014. 140. Yan, T., P. Venkat, M. Chopp, et al. 2015. Neurorestora-
Involvement of IGF-II receptors in the antioxidant and tive therapy of stroke in type two diabetes rats treated with
neuroprotective effects of IGF-II on adult cortical neuronal human umbilical cord blood cells. Stroke J. Cereb. Circ. 46:
cultures. Biochim. Biophys. Acta 1842: 1041–1051. 2599–2606.
124. Martín-Montañez, E., C. Millon, F. Boraldi, et al. 2017. 141. Kigerl, K.A., J.C. Gensel, D.P. Ankeny, et al. 2009. Identi-
IGF-II promotes neuroprotection and neuroplasticity fication of two distinct macrophage subsets with divergent
recovery in a long-lasting model of oxidative damage effects causing either neurotoxicity or regeneration in the
induced by glucocorticoids. Redox. Biol. 13: 69–81. injured mouse spinal cord. J. Neurosci. 29: 13435–13444.
125. Anthony, K., L.J. Reed, J.T. Dunn, et al. 2006. Attenuation 142. Li, D., C. Wang, Y. Yao, et al. 2016. mTORC1 pathway dis-
of insulin-evoked responses in brain networks controlling ruption ameliorates brain inflammation following stroke
appetite and reward in insulin resistance: the cerebral basis via a shift in microglia phenotype from M1 type to M2 type.
for impaired control of food intake in metabolic syndrome? FASEB J. 30: 3388–3399.
Diabetes 55: 2986–2992. 143. Sapkota, A., B.P. Gaire, M.-G. Kang, et al. 2019. S1P2 con-
126. Arnold, S.E., Z. Arvanitakis, S.L. Macauley-Rambach, et al. tributes to microglial activation and M1 polarization fol-
2018. Brain IR in type 2 diabetes and Alzheimer disease: lowing cerebral ischemia through ERK1/2 and JNK. Sci.
concepts and conundrums. Nat. Rev. Neurol. 14: 168–181. Rep. 9: 12106.
127. Kamal, A., G.-J. Biessels, I.J.A. Urban, et al. 1999. Hip- 144. Yang, H.-C., M. Zhang, R. Wu, et al. 2020. C-C chemokine
pocampal synaptic plasticity in streptozotocin-diabetic receptor type 2-overexpressing exosomes alleviated exper-
rats: impairment of long-term potentiation and facilitation imental post-stroke cognitive impairment by enhancing
of long-term depression. Neuroscience 90: 737–745. microglia/macrophage M2 polarization. World J. Stem Cells
128. McNay, E.C. & A.K. Recknagel. 2011. Brain insulin signal- 12: 152–167.
ing: a key component of cognitive processes and a potential 145. Hu, X., R.K. Leak, Y. Shi, et al. 2015. Microglial and
basis for cognitive impairment in type 2 diabetes. Neuro- macrophage polarization—new prospects for brain repair.
biol. Learn. Mem. 96: 432–442. Nat. Rev. Neurol. 11: 56–64.
129. Zhu, W., Y. Fan, T. Frenzel, et al. 2008. Insulin growth 146. Desestret, V., A. Riou, F. Chauveau, et al. 2013. In vitro
factor-1 gene transfer enhances neurovascular remodeling and in vivo models of cerebral ischemia show discrepancy
and improves long-term stroke outcome in mice. Stroke J. in therapeutic effects of M2 macrophages. PLoS One 8:
Cereb. Circ. 39: 1254–1261. e67063.
130. Reger, M.A., G.S. Watson, P.S. Green, et al. 2008. Intranasal 147. Zhang, T., B.-S. Pan, G.-C. Sun, et al. 2010. Diabetes syner-
insulin improves cognition and modulates beta-amyloid in gistically exacerbates poststroke dementia and tau abnor-
early AD. Neurology 70: 440–448. mality in brain. Neurochem. Int. 56: 955–961.
131. Desmond, D., J. Moroney, M. Sano, et al. 2002. Incidence 148. Zhang, T., X. Liu, Q. Li, et al. 2010. Exacerbation of
of dementia after ischemic stroke: results of a longitudinal ischemia-induced amyloid-β generation by diabetes is
study. Stroke J. Am. Heart Assoc. 33: 2254–2262. associated with autophagy activation in mice brain. Neu-
132. Yaffe, K., A. Kanaya, K. Lindquist, et al. 2004. The metabolic rosci. Lett. 479: 215–220.
syndrome, inflammation, and risk of cognitive decline. 149. Zhou, M., R. Xu, D.C. Kaelber, et al. 2020. Tumor necrosis
JAMA 292: 2237–2242. factor (TNF) blocking agents are associated with lower risk
133. Bettcher, B.M., J. Neuhaus, M.J. Wynn, et al. 2019. Increases for Alzheimer’s disease in patients with rheumatoid arthri-
in a pro-inflammatory chemokine, MCP-1, are related to tis and psoriasis. PLoS One 15: e0229819.
decreases in memory over time. Front. Aging Neurosci. 150. Gidday, J.M. 2006. Cerebral preconditioning and ischaemic
11: 25. tolerance. Nat. Rev. Neurosci. 7: 437–448.
134. Pillai, J.A., J. Bena, G. Bebek, et al. 2020. Inflammatory 151. Anrather, J. & J.M. Hallenbeck. 2013. Biological networks
pathway analytes predicting rapid cognitive decline in MCI in ischemic tolerance — rethinking the approach to clinical
stage of Alzheimer’s disease. Ann. Clin. Transl. Neurol. 7: conditioning. Transl. Stroke Res. 4: 114–129.
1225–1239. 152. Bhowmick, S. & K.L. Drew. 2019. Mechanisms of innate
135. Akiyama, H., S. Barger, S. Barnum, et al. 2000. Inflamma- preconditioning towards ischemia/anoxia tolerance:
tion and Alzheimer’s disease. Neurobiol. Aging 21: 383–421. lessons from mammalian hibernators. Cond. Med. 2:
136. Dziewulska, D. & M. Mossakowski. 2003. Cellular expres- 134–141.
sion of tumor necrosis factor a and its receptors in human 153. Ballantyne, A.O., A.M. Spilkin, J. Hesselink, et al. 2008.
ischemic stroke. Clin. Neuropathol. 22: 35–40. Plasticity in the developing brain: intellectual, language and
137. Schulze, J., D. Zierath, P. Tanzi, et al. 2013. Severe stroke academic functions in children with ischaemic perinatal
induces long-lasting alterations of high-mobility group box stroke. Brain 131: 2975–2985.
1. Stroke 44: 246–248. 154. Söderholm, M., A. Pedersen, E. Lorentzen, et al. 2019.
138. Bennett, D., J. Schneider, J. Bienias, et al. 2005. Mild cogni- Genome-wide association meta-analysis of functional out-
tive impairment is related to Alzheimer disease pathology come after ischemic stroke. Neurology 92: e1271.
and cerebral infarctions. Neurology 64: 834–841. 155. Zeiler, F.A., C. McFadyen, V.F.J. Newcombe, et al. 2019.
139. Anrather, J. & C. Iadecola. 2016. Inflammation and stroke: Genetic influences on patient-oriented outcomes in
an overview. Neurotherapeutics 13: 661–670. traumatic brain injury: a living systematic review of

18 Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences.
Krinock & Singhal Diabetes and stroke

non-apolipoprotein e single-nucleotide polymorphisms. J. 173. Motoshima, H., X. Wu, K. Mahadev, et al. 2004.
Neurotrauma https://doi.org/10.1089/neu.2017.5583. Adiponectin suppresses proliferation and superoxide
156. Ray, K.K., S.R.K. Seshasai, S. Wijesuriya, et al. 2009. Effect generation and enhances eNOS activity in endothelial
of intensive control of glucose on cardiovascular out- cells treated with oxidized LDL. Biochem. Biophys. Res.
comes and death in patients with diabetes mellitus: a meta- Commun. 315: 264–271.
analysis of randomised controlled trials. Lancet North Am. 174. Ouedraogo, R., X. Wu, S.-Q. Xu, et al. 2006. Adiponectin
Ed. 373: 1765–1772. suppression of high-glucose–induced reactive oxygen
157. Kernan, W.N., C.M. Viscoli, K.L. Furie, et al. 2016. Pioglita- species in vascular endothelial cells. Diabetes 55: 1840.
zone after ischemic stroke or transient ischemic attack. N. 175. Ouedraogo, R., Y. Gong, B. Berzins, et al. 2007.
Engl. J. Med. 374: 1321–1331. Adiponectin deficiency increases leukocyte–endothelium
158. Lee, M., L. Saver Jeffrey, H.-W. Liao, et al. 2017. Pioglita- interactions via upregulation of endothelial cell adhesion
zone for secondary stroke prevention. Stroke 48: 388–393. molecules in vivo. J. Clin. Invest. 117: 1718–1726.
159. Yaghi, S., L. Furie Karen, M. Viscoli Catherine, et al. 2018. 176. Wu, X., K. Mahadev, L. Fuchsel, et al. 2007. Adiponectin
Pioglitazone prevents stroke in patients with a recent tran- suppresses IκB kinase activation induced by tumor necrosis
sient ischemic attack or ischemic stroke. Circulation 137: factor-α or high glucose in endothelial cells: role of cAMP
455–463. and AMP kinase signaling. Am. J. Physiol. Endocrinol.
160. Spence, J.D., C.M. Viscoli, S.E. Inzucchi, et al. 2019. Piogli- Metab. 293: E1836–E1844.
tazone therapy in patients with stroke and prediabetes: a 177. Li, X., H. Guo, L. Zhao, et al. 2017. Adiponectin attenuates
post hoc analysis of the IRIS randomized clinical trial. NADPH oxidase-mediated oxidative stress and neuronal
JAMA Neurol. 76: 526–535. damage induced by cerebral ischemia-reperfusion injury.
161. Pantoni, L. 2019. Potential new horizons for the prevention Biochim. Biophys. Acta 1863: 3265–3276.
of cerebrovascular diseases and dementia. JAMA Neurol. 178. Song, W., T. Huo, F. Guo, et al. 2013. Globular adiponectin
76: 521–522. elicits neuroprotection by inhibiting NADPH oxidase-
162. Jesse, D. 2018. Pioglitazone use after stroke. Circulation mediated oxidative damage in ischemic stroke. Neuro-
138: 1221–1223. science 248: 136–144.
163. Rajagopalan, S., P. Dutta, D. Hota, et al. 2015. Effect of low 179. Lee, E.B., G. Warmann, R. Dhir, et al. 2011. Metabolic dys-
dose pioglitazone on glycemic control and insulin resis- function associated with adiponectin deficiency enhances
tance in Type 2 diabetes: a randomized, double blind, clin- kainic acid-induced seizure severity. J. Neurosci. 31: 14361–
ical trial. Diabetes Res. Clin. Pract. 109: e32–e35. 14366.
164. Adachi, H., H. Katsuyama & H. Yanai. 2017. The low 180. Chan, K.-H., K.S.-L. Lam, O.-Y. Cheng, et al. 2012.
dose (7.5 mg/day) pioglitazone is beneficial to the improve- Adiponectin is protective against oxidative stress induced
ment in metabolic parameters without weight gain and an cytotoxicity in amyloid-beta neurotoxicity. PLoS One 7:
increase of risk for heart failure. Int. J. Cardiol. 227: 247– e52354.
248. 181. Ng, R.C.-L., O.-Y. Cheng, M. Jian, et al. 2016. Chronic
165. Viscoli Catherine, M., M. Kent David, R. Conwit, et al. adiponectin deficiency leads to Alzheimer’s disease-like
2019. Scoring system to optimize pioglitazone therapy after cognitive impairments and pathologies through AMPK
stroke based on fracture risk. Stroke 50: 95–100. inactivation and cerebral insulin resistance in aged mice.
166. Gamboa, J., D.A. Blankenship, J.P. Niemi, et al. 2010. Exten- Mol. Neurodegener. 11: 71.
sion of the neuroprotective time window for thiazolidine- 182. Kim, M.W., N. bin Abid, M.H. Jo, et al. 2017. Suppression of
diones in ischemic stroke is dependent on time of reperfu- adiponectin receptor 1 promotes memory dysfunction and
sion. Neuroscience 170: 846–857. Alzheimer’s disease-like pathologies. Sci. Rep. 7: 12435.
167. Culman, J., M. Nguyen-Ngoc, T. Glatz, et al. 2012. Treat- 183. Shetty, S., C.M. Kusminski & P.E. Scherer. 2009.
ment of rats with pioglitazone in the reperfusion phase of Adiponectin in health and disease: evaluation of
focal cerebral ischemia: a preclinical stroke trial. Exp. Neu- adiponectin-targeted drug development strategies.
rol. 238: 243–253. Trends Pharmacol. Sci. 30: 234–239.
168. White, A.T. & A.N. Murphy. 2010. Administration of thi- 184. Jiang, S., T. Li, T. Ji, et al. 2018. AMPK: potential therapeutic
azolidinediones for neuroprotection in ischemic stroke; a target for ischemic stroke. Theranostics 8: 4535–4551.
preclinical systematic review. J. Neurochem. 115: 845–853. 185. Manwani, B. & L.D. McCullough. 2013. Function of
169. Yu, J.G., S. Javorschi, A.L. Hevener, et al. 2002. The effect of the master energy regulator adenosine monophosphate-
thiazolidinediones on plasma adiponectin levels in normal, activated protein kinase in stroke. J. Neurosci. Res. 91:
obese, and type 2 diabetic subjects. Diabetes 51: 2968–2974. 1018–1029.
170. Turer, A.T. & P.E. Scherer. 2012. Adiponectin: mechanistic 186. McCullough, L.D., Z. Zeng, H. Li, et al. 2005. Pharma-
insights and clinical implications. Diabetologia 55: 2319– cological inhibition of AMP-activated protein kinase pro-
2326. vides neuroprotection in stroke. J. Biol. Chem. 280: 20493–
171. Arregui, M., B. Buijsse, A. Fritsche, et al. 2014. Adiponectin 20502.
and risk of stroke. Stroke 45: 10–17. 187. Pendlebury, S.T. & P.M. Rothwell. 2009. Prevalence, inci-
172. Efstathiou Stamatis, P., D.I. Tsioulos, G. Tsiakou Aphrodite, dence, and factors associated with pre-stroke and post-
et al. 2005. Plasma adiponectin levels and five-year survival stroke dementia: a systematic review and meta-analysis.
after first-ever ischemic stroke. Stroke 36: 1915–1919. Lancet Neurol. 8: 1006–1018.

Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences. 19
Diabetes and stroke Krinock & Singhal

188. Doyle, K.P., L.N. Quach, M. Solé, et al. 2015. B- 205. Maiorana, A., G. O’Driscoll, C. Cheetham, et al. 2001. The
lymphocyte-mediated delayed cognitive impairment fol- effect of combined aerobic and resistance exercise training
lowing stroke. J. Neurosci. 35: 2133. on vascular function in type 2 diabetes. J. Am. Coll. Cardiol.
189. Surawan, J., S. Areemit, S. Tiamkao, et al. 2017. Risk factors 38: 860–866.
associated with post-stroke dementia: a systematic review 206. Mayhan, W.G., D.M. Arrick, K.P. Patel, et al. 2010. Exercise
and meta-analysis. Neurol. Int. 9: 7216. training normalizes impaired NOS-dependent responses
190. Tsai, A.S., K. Berry, M.M. Beneyto, et al. 2019. A year-long of cerebral arterioles in type 1 diabetic rats. Am. J. Physiol.
immune profile of the systemic response in acute stroke Heart Circ. Physiol. 300: H1013–H1020.
survivors. Brain 142: 978–991. 207. Ergul, A., M. Abdelsaid, A.Y. Fouda, et al. 2014. Cere-
191. Iadecola, C., M.S. Buckwalter & J. Anrather. 2020. Immune bral neovascularization in diabetes: implications for stroke
responses to stroke: mechanisms, modulation, and thera- recovery and beyond. J. Cereb. Blood Flow Metab. 34: 553–
peutic potential. J. Clin. Invest. 130: 2777–2788. 563.
192. Yang, Y. & G.A. Rosenberg. 2015. Matrix metallopro- 208. Sorrells, S.F., M.F. Paredes, A. Cebrian-Silla, et al. 2018.
teinases as therapeutic targets for stroke. Brain Res. 1623: Human hippocampal neurogenesis drops sharply in chil-
30–38. dren to undetectable levels in adults. Nature 555: 377–381.
193. Chung, A.G., J.B. Frye, J.C. Zbesko, et al. 2018. Liquefac- 209. Fuchs, E. & G. Flügge. 2014. Adult neuroplasticity: more
tion of the brain following stroke shares a similar molec- than 40 years of research. Neural Plast. 2014: 1–10.
ular and morphological profile with atherosclerosis and 210. Xing, Y. & Y. Bai. 2020. A review of exercise-induced neu-
mediates secondary neurodegeneration in an osteopontin- roplasticity in ischemic stroke: pathology and mechanisms.
dependent mechanism. eNeuro 5. https://doi.org/10.1523/ Mol. Neurobiol. 57: 4218–4231.
ENEURO.0076-18.2018. 211. Brown, C.E., P. Li, J.D. Boyd, et al. 2007. Extensive turnover
194. Ormstad, H., H.C.D. Aass, K.-F. Amthor, et al. 2011. Serum of dendritic spines and vascular remodeling in cortical tis-
cytokine and glucose levels as predictors of poststroke sues recovering from stroke. J. Neurosci. 27: 4101–4109.
fatigue in acute ischemic stroke patients. J. Neurol. 258: 212. Voss, M.W., K.I. Erickson, R.S. Prakash, et al. 2013. Neu-
670–676. robiological markers of exercise-related brain plasticity in
195. Su, J.-A., S.-Y. Chou, C.-S. Tsai, et al. 2012. Cytokine older adults. Brain. Behav. Immun. 28: 90–99.
changes in the pathophysiology of poststroke depression. 213. El-Tamawy, M.S., F. Abd-Allah, S.M. Ahmed, et al. 2014.
Gen. Hosp. Psychiatry 34: 35–39. Aerobic exercises enhance cognitive functions and brain
196. Basic Kes, V., A.-M. Simundic, N. Nikolac, et al. 2008. Pro- derived neurotrophic factor in ischemic stroke patients.
inflammatory and anti-inflammatory cytokines in acute NeuroRehabilitation 34: 209–213.
ischemic stroke and their relation to early neurological 214. Alcantara, C.C., L.F. García-Salazar, M.A. Silva-Couto,
deficit and stroke outcome. Clin. Biochem. 41: 1330–1334. et al. 2018. Post-stroke BDNF concentration changes fol-
197. Emsley, H.C.A., C.J. Smith, R.F. Georgiou, et al. 2005. A lowing physical exercise: a systematic review. Front. Neurol.
randomised phase II study of interleukin-1 receptor antag- 9: 637.
onist in acute stroke patients. J. Neurol. Neurosurg. Psychi- 215. Berretta, A., Y.-C. Tzeng & A.N. Clarkson. 2014. Post-
atry 76: 1366–1372. stroke recovery: the role of activity-dependent release of
198. Fu, Y., N. Zhang, L. Ren, et al. 2014. Impact of an immune brain-derived neurotrophic factor. Expert Rev. Neurother.
modulator fingolimod on acute ischemic stroke. Proc. Natl. 14: 1335–1344.
Acad. Sci. USA 111: 18315–18320. 216. Colberg, S.R., R.J. Sigal, J.E. Yardley, et al. 2016. Phys-
199. Smith Craig, J., S. Hulme, A. Vail, et al. 2018. SCIL- ical activity/exercise and diabetes: a position statement
STROKE (Subcutaneous Interleukin-1 Receptor Antago- of the American Diabetes Association. Diabetes Care 39:
nist in Ischemic Stroke). Stroke 49: 1210–1216. 2065.
200. Shibuya, M., S. Hirai, M. Seto, et al. 2005. Effects of fasudil 217. Gao, Y., Y. Zhao, J. Pan, et al. 2014. Treadmill exercise pro-
in acute ischemic stroke: results of a prospective placebo- motes angiogenesis in the ischemic penumbra of rat brains
controlled double-blind trial. J. Neurol. Sci. 238: 31–39. through caveolin-1/VEGF signaling pathways. Brain Res.
201. Wang, S., J. Zhou, W. Kang, et al. 2016. Tocilizumab inhibits 1585: 83–90.
neuronal cell apoptosis and activates STAT3 in cerebral 218. Li, F., X. Geng, C. Huber, et al. 2020. In search of a dose: the
infarction rat model. Bosn. J. Basic Med. Sci. 16: 145–150. functional and molecular effects of exercise on post-stroke
202. Suzuki, S., K. Tanaka & N. Suzuki. 2009. Ambivalent rehabilitation in rats. Front. Cell. Neurosci. 14: 186.
aspects of interleukin-6 in cerebral ischemia: inflammatory 219. Lo, E.H., H. Hara, J. Rogowska, et al. 1996. Temporal corre-
versus neurotrophic aspects. J. Cereb. Blood Flow Metab. 29: lation mapping analysis of the hemodynamic penumbra in
464–479. mutant mice deficient in endothelial nitric oxide synthase
203. Hu, F.B., M.J. Stampfer, C. Solomon, et al. 2001. Physi- gene expression. Stroke 27: 1381–1385.
cal activity and risk for cardiovascular events in diabetic 220. Endres, M., K. Gertz, U. Lindauer, et al. 2003. Mechanisms
women. Ann. Intern. Med. 134: 96–105. of stroke protection by physical activity. Ann. Neurol. 54:
204. Umpierre, D. 2011. Physical activity advice only or struc- 582–590.
tured exercise training and association with HbA 1c levels 221. Gertz, K., J. Priller, G. Kronenberg, et al. 2006. Physical
in type 2 diabetes: a systematic review and meta-analysis. activity improves long-term stroke outcome via endothe-
JAMA 305: 1790. lial nitric oxide synthase–dependent augmentation of

20 Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences.
Krinock & Singhal Diabetes and stroke

neovascularization and cerebral blood flow. Circ. Res. 99: 228. Ziche, M., L. Morbidelli, R. Choudhuri, et al. 1997. Nitric
1132–1140. oxide synthase lies downstream from vascular endothe-
222. Gómez-Pinilla, F., Z. Ying, R.R. Roy, et al. 2002. Voluntary lial growth factor-induced but not basic fibroblast growth
exercise induces a BDNF-mediated mechanism that pro- factor-induced angiogenesis. J. Clin. Invest. 99: 2625–2634.
motes neuroplasticity. J. Neurophysiol. 88: 2187–2195. 229. Spartano, N.L., K.L. Davis-Plourde, J.J. Himali, et al. 2019.
223. Qiao, H., S.-C. An, C. Xu, et al. 2017. Role of proBDNF Self-reported physical activity and relations to growth and
and BDNF in dendritic spine plasticity and depressive-like neurotrophic factors in diabetes mellitus: the Framingham
behaviors induced by an animal model of depression. Brain Offspring Study. J. Diabetes Res. 2019: 2718465.
Res. 1663: 29–37. 230. Kim, Y.J., J. Ku, S. Cho, et al. 2014. Facilitation of cor-
224. Tang, Q., Q. Yang, Z. Hu, et al. 2007. The effects of willed ticospinal excitability by virtual reality exercise following
movement therapy on AMPA receptor properties for adult anodal transcranial direct current stimulation in healthy
rat following focal cerebral ischemia. Behav. Brain Res. 181: volunteers and subacute stroke subjects. J. Neuroeng. Reha-
254–261. bil. 11: 124.
225. Gao, B.-Y., D.-S. Xu, P.-L. Liu, et al. 2020. Modified 231. Fried, P.J., A. Jannati, P. Davila-Pérez, et al. 2017. Repro-
constraint-induced movement therapy alters synaptic plas- ducibility of single-pulse, paired-pulse, and intermittent
ticity of rat contralateral hippocampus following middle theta-burst TMS measures in healthy aging, type-2 dia-
cerebral artery occlusion. Neural Regen. Res. 15: 1045. betes, and Alzheimer’s disease. Front. Aging Neurosci. 9:
226. Schwenk, J., S. Boudkkazi, M.K. Kocylowski, et al. 2019. 263.
An ER assembly line of AMPA-receptors controls excita- 232. VanDerwerker, C.J., R.E. Ross, K.H. Stimpson, et al. 2018.
tory neurotransmission and its plasticity. Neuron 104: 680– Combining therapeutic approaches: rTMS and aerobic
692.e9. exercise in post-stroke depression: a case series. Top. Stroke
227. Fukumura, D., T. Gohongi, A. Kadambi, et al. 2001. Pre- Rehabil. 25: 61–67.
dominant role of endothelial nitric oxide synthase in vas- 233. Klomjai, W., R. Katz & A. Lackmy-Vallée. 2015. Basic
cular endothelial growth factor-induced angiogenesis and principles of transcranial magnetic stimulation (TMS) and
vascular permeability. Proc. Natl. Acad. Sci. USA 98: 2604– repetitive TMS (rTMS). Ann. Phys. Rehabil. Med. 58: 208–
2609. 213.

Ann. N.Y. Acad. Sci. xxxx (2021) 1–21 © 2021 New York Academy of Sciences. 21

You might also like