You are on page 1of 9

Neurochemistry International 146 (2021) 105015

Contents lists available at ScienceDirect

Neurochemistry International
journal homepage: www.elsevier.com/locate/neuint

Diagnostic and prognostic blood biomarkers in vascular dementia: From the


viewpoint of ischemic stroke
Satoshi Hosoki, Tomotaka Tanaka, Masafumi Ihara *
Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan

A R T I C L E I N F O A B S T R A C T

Keywords: Reliable quantitative blood biomarkers are important in vascular dementia (VaD) because early diagnosis and
Vascular dementia therapeutic intervention are effective in preventing progression of dementia. Although many blood biomarkers
Acute ischemic stroke for acute ischemic stroke (AIS) or VaD have been reported, there are few reliable blood biomarkers. VaD and AIS
Biomarker
have similar pathological conditions that are associated with small vessel disease (SVD) such as oxidative stress,
Noncoding RNA
Multimarker
inflammation, endothelial dysfunction, and neuronal injury. Therefore, it may be possible to find superior blood
biomarkers of VaD among AIS blood biomarkers. Owing to recent developments, noncoding RNAs such as
microRNA and long noncoding RNA, which can be analyzed using a single drop of blood, are also particularly
reliable VaD markers because they stably reflect brain tissue damage. A multimarker combining several blood
biomarkers or artificial intelligence technology may also be beneficial to compensate for insufficiencies of a
single blood biomarker. This review describes the blood biomarkers of VaD and how they are related to blood
biomarkers of AIS.

1. Introduction population (Pendlebury and Rothwell, 2019). According to a


meta-analysis (Pendlebury and Rothwell, 2009), approximately 10% of
Vascular dementia (VaD) is the second most common cognitive patients develop dementia within a few months to a year after stroke,
impairment following Alzheimer’s disease (AD) (Gorelick et al., 2011) and >30% develop dementia after stroke recurrence. However, the
and often complicated by co-existent AD (Kalaria and Ballard, 1999). remaining stroke cases also have SVDs such as white matter hyper­
Management of vascular risk factors is directly associated with a intensities (WMH), cerebral microbleeds (CMBs), and lacunar infarction.
decrease in incidence of VaD (Fotuhi et al., 2009). Therefore, an early These SVDs are expected to cause dementia (Béjot et al., 2011; Pend­
diagnosis of VaD by blood biomarkers can contribute to early inter­ lebury and Rothwell, 2009; Yatawara et al., 2020). Therefore, it is
vention and prevention of dementia. Blood biomarkers may be optimal possible to find new blood biomarkers of VaD among candidate blood
candidates for identifying VaD in terms of the accessibility, minimal biomarkers of AIS. In addition, owing to the recent developments,
invasiveness, and cost-effectiveness. However, there are few reliable noncoding RNAs such as microRNA and long noncoding RNA, which can
blood biomarkers of VaD. Meanwhile, many blood biomarkers for acute be analyzed using a single drop of blood, are also particularly reliable
ischemic stroke (AIS) have been explored for a long time (Whiteley et al., VaD markers. They are caused by transcriptional changes induced in the
2008). VaD and AIS have similar pathological conditions that are asso­ pathological changes in VaD and stably reflect brain tissue damage. This
ciated with small vessel disease (SVD), such as oxidative stress, review describes the blood biomarkers of VaD and how they are related
inflammation, endothelial dysfunction, and neuronal injury. Approxi­ to AIS blood biomarkers.
mately one-fourth of the patients with AIS have SVD, mainly in the form
of lacunar infarction (Pasi and Cordonnier, 2020). In addition, SVD is 2. Pathomechanism and diagnosis of VaD
the major pathological alteration of VaD (Iadecola, 2013). Thus, SVD is a
shared pathological mechanism between VaD and AIS. Moreover, AIS VaD accounts for at least 20% of dementia cases (Gorelick et al.,
itself is a risk factor of VaD. The incidence of dementia is nearly 50 times 2011), and vascular factors are known to accelerate AD pathology
higher in patients in the year after a major stroke than in the general (Kalaria et al., 2012; Snowdon et al., 1997). Inducing cerebral

* Corresponding author. Department of Neurology, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan.
E-mail address: ihara@ncvc.go.jp (M. Ihara).

https://doi.org/10.1016/j.neuint.2021.105015
Received 31 December 2020; Received in revised form 25 February 2021; Accepted 2 March 2021
Available online 26 March 2021
0197-0186/© 2021 The Author(s). Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
S. Hosoki et al. Neurochemistry International 146 (2021) 105015

circulatory failure in AD model mice that overexpress human mutant resulting in neuronal injury (Brouns and De Deyn, 2009). Although VaD
amyloid precursor protein increases the polymerized amyloid β protein is generally caused by slower response to more chronic cerebral hypo­
in the soluble fraction, causing synergistic cognitive deterioration perfusion, oxidative stress, inflammation, endothelial dysfunction, and
function (Viswanathan et al., 2009). Moreover, an 8-month brain neuronal injury are shared pathological mechanisms between VaD and
hypoperfusion of wild-type mice caused hippocampal atrophy and AIS. The next section will outline the blood biomarkers of VaD and how
decreased glucose metabolism, resulting in impairment of episodic they are related to the blood biomarkers of AIS (Fig. 1, Table 1).
memory (Nishio et al., 2010). Inducing cerebral circulatory insufficiency
in a mouse model of cerebral amyloid angiopathy significantly increases 4. Oxidative stress
the amount of amyloid β protein deposited on the blood vessel wall and
exacerbates cognitive dysfunction (Okamoto et al., 2012). These explain Brain ischemia generates reactive oxygen species (ROS), such as
the significant effects of vascular factors on neurodegeneration (Ihara superoxide anion, hydroxyl radical, hydroperoxyl radical, hydrogen
and Tomimoto, 2011). In patients with AD, cerebral amyloid angiopathy peroxide, and singlet oxygen, and reactive nitrogen species (RNS), such
and cortical vascular abnormalities are frequently observed (Kitaguchi as nitric oxide and peroxinitrite anion. Oxidative stress refers to a situ­
et al., 2007), and peripheral vascular resistance correlates with amyloid ation in which cells’ antioxidant capacity is exceeded in the state where
accumulation in the brain observed by amyloid imaging (Hughes et al., ROS and RNS are excessively generated (Brouns and De Deyn, 2009).
2013). Therefore, VaD and AD may be linked bidirectionally and syn­ Excessive ROS and RNS production causes DNA damage, protein
ergistically to exacerbation of dementia (Kalaria and Ihara, 2013). In destruction, lipid peroxidation, and tissue damage, resulting in the death
fact, the pathological changes with VaD and AD frequently occur of the nerve cell (Chan, 2001). ROS and RNS also trigger necrosis and
together, which is termed as mixed dementia (Saito and Ihara, 2016; apoptosis of nerve cells (Rana and Singh, 2018) (Fig. 1A).
Zekry et al., 2002). In retrospective and prospective autopsy studies, the A product of membrane phospholipid peroxidation is oxidized low-
prevalence of mixed dementia ranges from 2% to 58%, with reasonable density lipoprotein (LDL). The pathological roles of oxidized LDL
means ranging from 6% to 12% (Jellinger and Attems, 2007). Clinically, (oxLDL) are endothelial cell injury, promotion of foam cell formation,
mixed dementia also includes AD with cerebrovascular disease, vascular proliferation of vascular smooth muscle cells, and platelet adhesion
lesions, and vascular risk factors (Kalaria, 2000). In the Rush Memory aggregation (Wang et al., 2017). Elevated oxLDL levels are associated
and Aging Project, the frequency of mixed dementia was 40% among the with lower MMSE scores in patients with AIS in a large cohort (VaD
total dementia cases (Schneider et al., 2007). Understanding the pa­ group, 2711; control group, 1015) (Wang et al., 2018a). OxLDL is
thology of vascular dementia can also aid in better management of AD. reportedly related to AIS (Yokota et al., 2016) and the prognosis of AIS
The Framingham study found a 44% reduction in dementia in 30 years (Uno et al., 2003).
(Satizabal et al., 2016). The most significant decrease was in VaD, which Malondialdehyde (MDA) is produced as a reactive oxygen peroxide
suggests that control of vascular risk factors is associated with the and degrades membrane phospholipids in cell membranes (Cano et al.,
reduction of VaD. In addition, the incidence of AD also declined, 2003). MDA is a blood biomarker of AIS (Cano et al., 2003) and has been
although not statistically significant, suggesting that control of vascular associated with early AIS outcome (Polidori et al., 2002) and related to
risk factors is related to the decrease in incidence of AD (Satizabal et al., VaD.
2016). Thus, in the treatment or prevention of dementia, even AD, DJ-1, a translation product of DJ-1, which is the causative gene of
physicians should determine vascular contribution to dementia and familial Parkinson’s disease PARK7, is implicated in oxidative stress
consider early management of vascular risk factors, if present, such as after ischemia and has an antioxidant effect by promoting self-oxidation
hypertension, diabetes, hypertension, and obesity (Fotuhi et al., 2009). and transcription of antioxidant stress factors (Ariga et al., 2013).
The current diagnosis of VaD is mainly based on cognitive and neuro­ Elevated blood DJ-1 level can distinguish patients with acute stroke
imaging evaluation, and the sensitivity and specificity of diagnosis are from controls (Allard et al., 2005).
inadequate. Diagnosis by neuroimaging is difficult because similar Nicotinamide adenine dinucleotide phosphate oxidase (NOXs) is a
neuroimaging findings are associated with dementia in some patients molecular source of ROS during aging, hypoperfusion, stroke, and hy­
but with normal cognition in others (van der Flier et al., 2018). There is pertension (Drummond et al., 2011). Several in vivo experiments
no consensus on dementia screening tools (Ghafar et al., 2019). There­ demonstrate that NOXs are involved in cognitive dysfunction (Choi and
fore, the development of objective blood biomarkers for VaD has great Lee, 2017). Thus, NOXs can be a promising blood biomarker for VaD.
clinical significance. Several microRNAs associated with apoptosis are blood biomarkers
for VaD and AIS. miR-502-3P regulates SET, which is a potent and
3. Shared pathology of VaD and AIS specific inhibitor of protein phosphatase 2A, and is associated with
many cellular processes, such as cell cycle control, migration, and
The vascular alterations that cause VaD are diverse and include apoptosis (Madeira et al., 2005), while miR-486-5p is associated with
systemic conditions that influence global cerebral perfusion, or alter­ proliferation, survival, apoptosis, and differentiation of endothelial
ations involving cerebral blood vessels, most commonly SVD (Iadecola, progenitor cells (Jiang et al., 2019). In a cohort of 204 patients with VaD
2013). SVD includes various pathological changes that affect arterioles, and 200 healthy and age- and sex-matched controls, elevated blood
venules, and capillaries (Iadecola, 2013), which are associated with miR-502-3p levels can distinguish patients with VaD with 75% sensi­
imaging findings of SVD, including lacunar infarction, WMH, CMBs, and tivity and 89% specificity, while elevated blood miR-486-5p levels can
enlarged perivascular spaces (Staals et al., 2014). Despite the diversity distinguish patients with VaD from 200 control patients with 75%
of the underlying brain pathology, VaD and AIS have similar pathogenic sensitivity and 83% specificity (Prabhakar et al., 2017). As for other
bases associated with SVD, resulting from hypoperfusion, oxidative apoptosis-related microRNAs, miR-16 promotes ischemic brain damage
stress, and inflammation, which in turn lead to endothelial damage, by negatively regulating anti-apoptotic Bcl-2 and Bcl-w (Yang et al.,
blood-brain barrier (BBB) breakdown, activation of innate immunity, 2017b), while miR-335 promotes stress granule formation and inhibits
disruption of trophic coupling between vascular and brain cells, and apoptosis by reducing Rho-associated protein kinase 2 expression in AIS
neuronal injury (Iadecola, 2013). Such markers in each pathological (Si et al., 2019). Moreover, miR-30a-5p regulates the expression of RhoB
mechanism may be candidates for blood biomarkers in VaD. AIS begins and beclin-1 genes and promotes apoptosis (Wang et al., 2018b). With
with sudden brain hypoperfusion, resulting in cell energy deficiency, regard to the association between apoptosis-related microRNA and AIS,
neurotoxicity, oxidative stress within minutes, microangiopathy, acti­ blood levels of miR-16, miR-335, and miR-30a-5p, which are
vation of hemostatic mechanisms, inflammatory response, endothelial apoptosis-related microRNAs, are diagnostic blood biomarkers of AIS
damage, and dysfunction of the blood–brain barrier within hours, (Leung et al., 2014; Wang et al., 2018b; Zhao et al., 2016). High blood

2
S. Hosoki et al. Neurochemistry International 146 (2021) 105015

Fig. 1. The association between the pathology of VaD and the biomarkers. Many biomarkers are associated with the pathology of VaD, oxidative stress (A),
inflammation (B), endothelial dysfunction (C), and neuronal injury (D). ADMA: asymmetric dimethylarginine, AM: adrenomedullin, BBB: blood–brain barrier, DAMP:
damage-associated molecular patterns, GFAP: Glial fibrillary acidic protein, H-FABP: heart-type fatty acid-binding protein, ICAM-1: intercellular adhesion molecule-
1, IL-6: interleukin-6, IL-10: interleukin-10, MDA: malondialdehyde, MIAT: myocardial infarction-associated transcript, MMP-9: matrix metalloproteinase-9, NO:
nitric oxide, NOS: nitric oxide synthase, NOX: nicotinamide adenine dinucleotide phosphate oxidase, NSE: neuron-specific enolase, ox LDL: oxidized low-density
lipoprotein, TM: thrombomodulin, TNF-α: tumor necrosis factor-α, ROS: reactive oxygen species, RNS: reactive nitrogen species, UCH-L1: ubiquitin C-terminal
hydrolase L1, VCAM-1: vascular cell adhesion protein, vWF: von Willebrand factor.

miR-16 level is also associated with poor outcomes (Tian et al., 2016). Akt-eNOS signaling pathway (Yang et al., 2017a). miR-126 can be a
These microRNAs are promising blood biomarkers of VaD. diagnostic marker of AIS (Long et al., 2013) and is also associated with
the National Institutes of Health Stroke Scale (NIHSS) score on admis­
5. Inflammation sion and modified Rankin Scale (mRS) score at 3 months (Qi et al.,
2020). Myocardial infarction-associated transcript (MIAT), a long non­
Damage-associated molecular patterns, released during cell death, coding RNA, is related to vascular endothelial cell inflammation. The
activate macrophages and neutrophils and produce inflammatory cy­ function of MIAT is to prolong inflammation by suppressing effer­
tokines. T cells are further activated to prolong inflammation (Naka­ ocytosis and inhibiting clearance of apoptotic cells (Ye et al., 2019).
mura and Shichita, 2019). Inflammation exacerbates leukoaraiosis MIAT is a diagnostic marker for AIS (Zhu et al., 2018) and also associ­
through demyelination, loss of axons, and oligodendrocyte degenera­ ated with NIHSS at admission and mRS at 3 months (Zhu et al., 2018).
tion, causing neurodegeneration and cell death, and promoting the miR-126 and MIAT are candidate blood biomarkers of VaD.
inflammation of glial cells (Venkat et al., 2015) (Fig. 1B).
Interleukin-6 (IL-6) is a potent pro-inflammatory factor secreted by T 6. Endothelial dysfunction
cells, macrophages, and neutrophils. A positive correlation has been
detected between IL-6 and WMH. IL-6 is significantly related to WMH In the inflammatory response, leukocytes are recruited from the
severity independent of age and systolic blood pressure (Nagai et al., circulation into the extravascular space. Activation of endothelial cells
2011). allows them to express adhesion molecules on their surface and bind to
Matrix metalloproteinase-9 (MMP-9) activated by ROS attacks the interacting molecules on the surface of circulating white blood cells
basal lamina and tight junctions of the cerebral endothelial cells, causing (Fig. 1C).
BBB dysfunction. MMP-9 levels are significantly increased in the cere­ The cell adhesion molecules include intercellular adhesion molecule-
brospinal fluid of patients with VaD compared to those with AD (Adair 1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and selectins
et al., 2004). With regard to the blood biomarker of AIS, high plasma (P- and E-selectin) (Cipollini et al., 2019). Upregulated expression of
MMP-9 level in AIS is associated with hemorrhagic transformation ICAM-1 is associated with progression of WMH. After adjusting for age,
(Castellanos et al., 2003). sex, cardiovascular risk factors, and C-reactive protein, ICAM-1 is
Several microRNAs and long noncoding RNA associated with associated with WMH lesion progression at both 3 and 6 years (Markus
inflammation are blood biomarkers for VaD and AIS. miR-126 inhibits et al., 2005). As for the blood biomarker of AIS, increased ICAM-1 levels
the secretion of tumor necrosis factor-α (TNF-α) and IL-6 in human are associated with an increased risk of AIS in patients with coronary
gingival fibroblasts under high-glucose conditions by targeting TNF heart disease (Tanne et al., 2002) and with early death in middle-aged
receptor-associated factor 6; and protects human cardiac microvascular patients with AIS (Rallidis et al., 2009).
endothelial cells from hypoxia-reoxygenation-induced injury and in­ VCAM-1 is a ligand expressed on the vascular endothelium that binds
flammatory response (IL-6, IL-10, and TNF-α) by activating the PI3K- to α4 integrin, which plays an essential role in the infiltration of T cells

3
S. Hosoki et al. Neurochemistry International 146 (2021) 105015

Table 1
Association between VaD and AIS biomarkers.
VaD AIS

Assoc VaD Ctrl Sn Sp ref Assoc AIS Ctrl OTMT Sn Sp ref


(n) (n) (%) (%) (n) (n) (%) (%)

Oxidative oxLDL Higher 2711 1015 – – Wang et al. AIS 250 250 <72 h N/ N/ Yokota et al. (2016)
Stress prevalence of (2018a) Dx A A
lower MMSE Associated with the prognosis of AIS Uno et al. (2003)
score
MDA not reported AIS 15 15 <24 h N/ N/ Cano et al. (2003)
Dx A A
Associated with the early outcomes of AIS Polidori et al. (2002)
DJ-1 AIS 622 31 <3 h N/ N/ Allard et al. (2005)
Dx A A
miR-502- VaD Dx 204 200 75 89 Prabhakar not reported
3p et al. (2017)
miR-486- 75 83
5p
miR-16 not reported AIS 74 19 <24 h 95 35 Leung et al. (2014)
Dx
Associated with the early prediction of mortality Tian et al. (2016)
and mRS
miR-335 AIS 168 104 <24 h 98 69 Zhao et al. (2016)
miR-30a- Dx 15 24 <6 h N/ N/ Wang et al. (2018b)
5p A A
Inflammation IL-6 WMH severity 137 – – – Nagai et al. not reported
(2011)
MMP-9 not reported Associated with the hemorrhagic transformation Castellanos et al.
in acute ischemic stroke (2003)
miR-126 AIS 197 50 <24 h 92 84 Long et al. (2013)
Dx
Associated with mRS at 3 months Qi et al. (2020)
MIAT AIS 189 189 <24 h 74 80 Zhu et al. (2018)
Dx
Associated with mRS at 3 months
Endothelial ICAM-1 Progression of 296 – – – Markus AIS 134 134 – – – Tanne et al. (2002)
Dysfunction WMH et al. (2005) Dx
Associated with early death in middle-aged Rallidis et al. (2009)
patients with AIS
VCAM-1 Severe WMH 11 18 – – de Leeuw Associated with outcome at 3 months in patients Richard et al. (2015)
et al. (2002) with CVD
P-selectin – – Associated with the atherothrombotic properties Pawelczyk et al.
of hyperglycemia and hyperlipidemia in stroke (2018)
patients
vWF WMH severity 198 – – – Nagai et al. Associated with the severity, outcome, and infarct Sato et al. (2006)
(2012) size in ischemic stroke
TM Severe WMH 110 50 – – Hassan Associated with fatal outcomes in patients with Olivot et al. (2004)
and the number et al. (2003) AIS
of lacunar
infarction on
MRI
ADMA Prevalence of 2013 – – – Pikula et al. AIS 52 48 <24 h – – Ercan et al. (2019)
silent brain (2009) Dx
infarcts
Fibrinogen Increased risk of 185 – – – Xu et al. Associated with the prognosis of AIS Kernagis and
both AD and (2008) Laskowitz (2012)
VaD
mid- WMH severity 288 – – – Kuriyama not reported
regional and cognitive et al. (2017)
proAM function test
scores
Neuronal NSE VaD Dx 42 38 – – Shen and AIS 66 N/A <3 h N/ N/ Wunderlich et al.
injury Gao (2015) Dx A A (2006)
Associated with the prognosis of LAA and IS after Wang et al. (2018c)
IVT
S100β Cognitive 210 207 – – Gao et al. Distinguishing between AIS and ICH (Wunderlich et al.,
impairment in (2015) Associated with the prognosis of AIS 2004; Zhou et al.,
patients with 2016)
WMH
GFAP not reported Distinguishing between AIS and ICH in 4.5 h Ren et al. (2016)
H-FABP AIS 22 22 <24 h 68 100 Zimmermann-Ivol
Dx et al. (2004)
Associated with poor outcomes Lyang et al. (2019)
miR-134 AIS 50 50 <24 h 75 73 Zhou et al. (2018)
Dx
Associated with the poor outcomes of AIS
miR-451a VaD Dx 204 200 70 75 not reported
(continued on next page)

4
S. Hosoki et al. Neurochemistry International 146 (2021) 105015

Table 1 (continued )
VaD AIS

Assoc VaD Ctrl Sn Sp ref Assoc AIS Ctrl OTMT Sn Sp ref


(n) (n) (%) (%) (n) (n) (%) (%)

Miscellaneous miR-409- 76 89 Prabhakar


markers 3p et al. (2017)
Multimarker 4-protein not reported AIS 65 157 <6 h 90 90 Lynch et al. (2004)
panel Dx

ADMA: asymmetric dimethylarginine, AIS: acute ischemic stroke, Assoc: association, AM: adrenomedullin, Ctrl: control, Dx: diagnosis, GFAP: Glial fibrillary acidic
protein, H-FABP: heart-type fatty acid-binding protein, ICAM-1: intercellular adhesion molecule-1, ICH: intracerebral hemorrhage, IL-6: interleukin-6, IVT: intrave­
nous thrombolysis, LAA: large-artery atherosclerosis, MDA: malondialdehyde, mid-regional proAM: mid-regional pro-adrenomedullin, MIAT: myocardial infarction-
associated transcript, MMP-9: matrix metalloproteinase-9, MMSE: Mini Mental State Examination, MRI: magnetic resonance imaging, mRS: modified Rankin Scale,
NO: nitric oxide, NOS: nitric oxide synthase, NOX: nicotinamide adenine dinucleotide phosphate oxidase, NSE: neuron-specific enolase, OTMT: onset to the mea­
surement time, ox LDL: oxidized low-density lipoprotein, Sn: Sensitivity, Sp: Specificity, ref: references, TM: thrombomodulin, VaD: vascular cognitive impairment,
VCAM-1: vascular cell adhesion protein, vWF: von Willebrand factor, WMH: white matter hyperintensity.

to the brain. The plasma P-selectin and VCAM-1 levels were associated prognosis of AIS (Tombul et al., 2005).
with severe WMH. Those with severe periventricular WMH and symp­ Adrenomedullin (AM) is a potent vasodilator and bioactive natri­
tomatic carotid artery stenosis had higher VCAM-1 and P-selectin levels uretic peptide that is secreted by endothelial cells (Shindo et al., 2000)
than those without (de Leeuw et al., 2002). With regard to the blood in response to hypoxic invasion (Nakayama et al., 1998), shear stress
biomarker of AIS, the blood VCAM-1 level is associated with 3-month (Chun et al., 1997), and inflammation (Ueda et al., 1999). Cerebral
outcomes in patients with cerebrovascular disease (Richard et al., endothelial cells are a significant source of AM (Kis et al., 2002). AM
2015), and the blood P-selectin level can be useful in assessment of suppresses leukocyte infiltration, oxidative stress, and apoptosis, and
atherothrombotic properties of hyperglycemia and hyperlipidemia in simultaneously mobilizes CD34+ mononuclear cells to promote vascular
AIS (Pawelczyk et al., 2018). regeneration and compensate for brain protection in AIS (Koyama et al.,
When subcutaneous matrix proteins are exposed to the bloodstream, 2015; Miyashita et al., 2006). The levels of mid-regional proAM, which
platelets attach to the vessel wall by binding platelet surface receptors to is a stable fragment of the AM precursor, are significantly increased with
the endothelial von Willebrand factor (vWF). High plasma vWF levels the severity of deep white matter lesions and inversely correlated with
expressed by endothelial cells after tissue damage are associated with cognitive function test scores (Kuriyama et al., 2017), which suggests
WMH severity (Nagai et al., 2012). Likewise, the vWF level correlated that AM can be an effective marker of AIS and cognitive decline.
with severity, outcome, and infarct size of AIS and is significantly higher
in patients with stroke than in those without atrial fibrillation (Sato 7. Neuronal injury
et al., 2006).
Thrombomodulin (TM) is present on the surface of vascular endo­ Endothelial dysfunction leads to increased BBB permeability, sub­
thelial cells and forms a complex with thrombin that appears in the sequent exposure of neurons to inflammatory factors, white matter
blood to activate protein C, a coagulation inhibitor. TM is induced by damage (demyelination, axonal loss, and oligodendrocyte degenera­
endothelial damage and degraded by intracellular proteases and tion), neurodegeneration and cell death, increased glial inflammation,
released into the blood, mainly due to damage or destruction of glial cell activation, and ultimately neuronal injury (Cipollini et al.,
microvessel walls. Therefore, blood TM level measurement estimates 2019) (Fig. 1D).
endothelial cell production function and degree of damage, so it is useful Neuron-specific enolase (NSE) is an intracellular dimer enzyme of
as a marker for damage to vascular endothelial cells. Blood TM level is glucose metabolism localized predominantly in neuronal cytoplasm
associated with severe WMH and the number of lacunar infarctions on (Påhlman et al., 1984). Serum NSE levels in patients with VaD are
magnetic resonance imaging (Hassan et al., 2003). Increased TM level significantly higher than those in patients without VaD (Shen and Gao,
also predicts a fatal outcome in patients with AIS (Olivot et al., 2004). 2015). Moreover, serum NSE levels are associated with the prognosis of
Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of large-artery atherosclerotic strokes (Wang et al., 2018c) and AIS after
nitric oxide synthase (NOS). L-arginine is the substrate of NOS to pro­ intravenous thrombolysis (Lu et al., 2015). With regard to the blood
duce NO (nitric oxide). As the structure of ADMA is similar to that of L- biomarker of AIS, serum NSE levels initially increase in 2–3 h after AIS
arginine, ADMA competes with L-arginine for NOS binding, thereby (Wunderlich et al., 2006).
inhibiting NOS function and impairing NO formation, which impairs Ubiquitin C-terminal hydrolase L1 (UCH-L1) is a cytoplasmic deu­
normal vascular endothelial cell function, and is associated with endo­ biquitinating enzyme of neurons that have been associated with synaptic
thelial dysfunction (Vallance et al., 1992). High plasma ADMA levels are plasticity and homeostasis and the brain’s self-repair mechanisms after
associated with silent brain infarcts (SBIs). The presence of SBIs is a risk injury (Papa et al., 2010) (Rojo et al., 2011). The blood UCH-L1 level
factor for developing symptomatic stroke (Vermeer et al., 2003a) and cannot distinguish AIS from intracerebral hemorrhage (Ren et al., 2016).
cognitive dysfunction (Vermeer et al., 2003b, 2007). In multivariable Protein S100 beta (S100β) is a calcium-binding protein expressed
analyses adjusting for age, sex, and traditional stroke risk factors, higher mainly in human astroglial cells. S100β levels are elevated in patients
ADMA levels are associated with an increased risk of prevalent SBIs with SVD and associated with cognitive impairment in patients with
(Pikula et al., 2009). As for the blood biomarker of AIS, ADMA levels in WMH (Gao et al., 2015). Moreover, S100β discriminates AIS from
patients with AIS are significantly higher than those in the control group intracranial hemorrhage with high sensitivity (Zhou et al., 2016) and is
(Ercan et al., 2019). related to prognosis of AIS (Wunderlich et al., 2004).
Fibrinogen converts to fibrin at the site of vascular endothelial cell Glial fibrillary acidic protein (GFAP) is a brain-specific astrocytic
deficiency in the process of coagulation. An abnormal fibrinogen level is intermediate filament protein found almost exclusively in the central
a parameter of an abnormality in this hemostatic mechanism. It is also nervous system (Eng et al., 2000). Higher GFAP levels distinguish be­
involved in wound healing as a substrate for factor XIII and increases tween AIS and intracerebral hemorrhage (Ren et al., 2016).
during inflammation as an acute phase reactant. High fibrinogen levels Heart-type fatty acid-binding protein (H-FABP), which is a fatty acid-
are correlated with an increased risk of AD and VaD (Xu et al., 2008). As binding protein in the heart, is located in the brain. Serum H-FABP levels
for the blood biomarker of AIS, fibrinogen level is associated with the could help distinguish AIS from the control group with 68% sensitivity

5
S. Hosoki et al. Neurochemistry International 146 (2021) 105015

and 100% specificity in a study that examined 22 cases within 3 h after single markers.
stroke onset (Zimmermann-Ivol et al., 2004) and are also associated The clinical application of artificial intelligence (AI) is also expected
with poor outcomes in patients with AIS (Lyang et al., 2019). to be appropriate. In 2017, a diagnostic accuracy comparison was made
Several microRNAs associated with neuronal injury are blood bio­ between pathologists and machines in detecting lymph node metastases
markers for VaD and AIS. A brain-specific microRNA, miR-134, is in breast cancer. It was reported that machines performed better than
associated with dendrite spines. In a rodent model with VaD, histo­ pathologists under certain conditions (Ehteshami Bejnordi et al., 2017).
pathological analysis reveals that miR-134-5p aggravates cognitive In addition, the clinical application of deep learning is also expected, in
impairment in rats with VaD through damage to cortical neurons and which AI independently extracts features from learning data and ac­
loss of synaptic proteins (Liu et al., 2019). As for the blood biomarker of quires rules and knowledge. The expected areas where deep learning
AIS, a study that assessed stroke within 24 h of onset showed that may be imminently applied to stroke management are image segmen­
increased expression levels of miR-134 differentiated stroke from the tation, automated featurization, and multimodal prognostication (Feng
control group, with 75% sensitivity and 73% specificity (Zhou et al., et al., 2018). One of the largest barriers is said to be the complexity of
2018). Serum miR-134 levels are also associated with NIHSS, cerebral the underlying deep learning frameworks, which are largely inaccessible
infarction volume, and poor prognosis (Zhou et al., 2018). to individuals without advanced training (Feng et al., 2018). AI will also
be applied to evaluations of biomarkers by combining such technology
8. Miscellaneous markers with biomarkers and background factors, which will make VaD diag­
nosis more precise.
miR-451-a is associated with reduced natural killer cells in AIS (Kong VaD is potentially preventable and treatable by managing the
et al., 2020), while miR-409-3p is associated with a tumor suppressor vascular risk factors in the preclinical or early stages. Hypertension is a
(Song et al., 2018). In a cohort of 204 patients with VaD and 200 healthy strong risk factor of VaD (Novak and Hajjar, 2010). Antihypertensive
and age- and sex-matched controls, elevated blood miR-451-a levels can therapy for middle-aged individuals with hypertension is likely to sub­
distinguish patients with VaD with 70% sensitivity and 75% specificity sequently suppress the onset of dementia (Livingston et al., 2017).
with VaD, while decreased blood miR-409-3p levels can distinguish However, its effectiveness has not been proven in intervention studies
patients with VaD with 76% sensitivity and 89% specificity (Prabhakar owing to the difficulty of conducting long-term trials. Furthermore,
et al., 2017). microRNAs have the potential to be considered as therapeutic targets
and may be important in VaD treatment (Yuan and Bi, 2020). In a rodent
9. Multimarkers study, post-ischemic treatment with intravenous miR-7 mimic admin­
istration decreased the ischemic volume and improved motor and
The use of multiple markers has been investigated since there is a cognitive impairments with minimal peripheral toxicity, and minimized
limit to a single marker’s diagnostic power. As a diagnostic blood the apoptosis and autophagic death of neurons (Kim et al., 2018). Thus,
biomarker of AIS, four-protein panel (combination of S100β, vWF, miR-7 could be used as a potential therapeutic target for VaD. Diagnostic
MMP-9, and VCAM) (Lynch et al., 2004) has been reported. This mul­ biomarkers of VaD that are secreted as a defense mechanism can also be
timarker is useful in the diagnosis of acute stroke with high sensitivity considered as a therapeutic approach. The administration of AM as a
and specificity of >90%. The multimarker of VaD is expected to achieve possible therapeutic agent for VaD and AIS can be evaluated owing to
a high diagnostic accuracy. VaD is attributed to various causes, and the ability of AM to promote arteriogenesis and angiogenesis, inhibit
different pathological pathways underlie the development of VaD oxidative stress, preserve white matter integrity, and prevent cognitive
(Iadecola, 2013). Thus, a single marker is unlikely useful for the diag­ decline after chronic cerebral hypoperfusion in a rodent model (Maki
nosis or prediction of a disease with such heterogeneity; instead, mul­ et al., 2011). Indeed, AM is being tested for its potential application in
tiple markers that encompass the different aspects of the disease patients with AIS in a Phase II clinical trial (jRCT ID: jRCT2051190092)
physiopathology may be more useful. (Yoshimoto et al., 2021).

10. Perspective 11. Conclusion

Many blood biomarkers were found in both VaD and AIS. However, This review has outlined the increasing lines of evidence and possi­
some blood biomarkers have been reported in only AIS, and these are bilities of blood biomarkers in VaD from the viewpoint of AIS pathol­
candidates for new blood biomarkers for VaD. Among the VaD blood ogies and noncoding RNA multimarkers combined with multiple
biomarkers, oxLDL and ADMA are promising biomarkers because they biomarkers. Future studies are warranted to validate these promising
were reported in a prospective study with a large sample size. Some biomarkers.
noncoding RNAs were able to diagnose VaD with relatively high sensi­
tivity and specificity. Noncoding RNAs are present in human plasma in a Funding
stable form, can be analyzed using a single drop of blood, regulate gene
expression at the post-translational level, and play an important role in This study is funded by the Japan Agency for Medical Research and
the pathogenesis of VaD and thus have the potential to be diagnostic Development (19lk0201096h0001 and 20lk0201096h0002).
blood biomarkers for VaD (Mitchell et al., 2008; Yuan and Bi, 2020).
Currently, no blood biomarkers have yet to be proven useful in Authors’ Contributions
clinical practice (Mijajlović et al., 2017). Factors that make clinical
application of blood biomarkers difficult are the BBB, limiting the SH, TT, and MI were involved in writing of the manuscript, and made
leakage of proteins from the brain parenchyma into the blood and critical revisions of the manuscript for important intellectual content.
presence of these markers in other pathological conditions, such as ep­ All authors approved the final version of the manuscript.
ilepsy, encephalitis, and myocardial infarction (Whiteley et al., 2008).
By contrast, microRNAs have potential limitations related to reproduc­ Declaration of competing interest
ibility. One such limitation is that various methods can be used for
measuring microRNAs, and no gold standard has been established The authors declare that they have no conflict of interest.
(Witwer and Halushka, 2016).
Multimarkers of VaD are expected to encompass the different aspects
of the disease physiopathology and to compensate for the demerits of

6
S. Hosoki et al. Neurochemistry International 146 (2021) 105015

Acknowledgments Hughes, T.M., Kuller, L.H., Barinas-Mitchell, E.J., Mackey, R.H., McDade, E.M.,
Klunk, W.E., Aizenstein, H.J., Cohen, A.D., Snitz, B.E., Mathis, C.A., Dekosky, S.T.,
Lopez, O.L., 2013. Pulse wave velocity is associated with β-amyloid deposition in the
We would like to thank Dr. Ahmad Khundakar for critical editing of brains of very elderly adults. Neurology 81, 1711–1718.
this manuscript. Iadecola, C., 2013. The pathobiology of vascular dementia. Neuron 80, 844–866.
Ihara, M., Tomimoto, H., 2011. Lessons from a mouse model characterizing features of
vascular cognitive impairment with white matter changes. J Aging Res 2011,
References 978761.
Jellinger, K.A., Attems, J., 2007. Neuropathological evaluation of mixed dementia.
Adair, J.C., Charlie, J., Dencoff, J.E., Kaye, J.A., Quinn, J.F., Camicioli, R.M., Stetler- J. Neurol. Sci. 257, 80–87.
Stevenson, W.G., Rosenberg, G.A., 2004. Measurement of gelatinase B (MMP-9) in Jiang, H., Toscano, J.F., Song, S.S., Schlick, K.H., Dumitrascu, O.M., Pan, J., Lyden, P.D.,
the cerebrospinal fluid of patients with vascular dementia and Alzheimer disease. Saver, J.L., Gonzalez, N.R., 2019. Differential expression of circulating exosomal
Stroke 35, e159–162. microRNAs in refractory intracranial atherosclerosis associated with
Allard, L., Burkhard, P.R., Lescuyer, P., Burgess, J.A., Walter, N., Hochstrasser, D.F., antiangiogenesis. Sci. Rep. 9, 19429.
Sanchez, J.C., 2005. PARK7 and nucleoside diphosphate kinase A as plasma markers Kalaria, R.N., 2000. The role of cerebral ischemia in Alzheimer’s disease. Neurobiol.
for the early diagnosis of stroke. Clin. Chem. 51, 2043–2051. Aging 21, 321–330.
Ariga, H., Takahashi-Niki, K., Kato, I., Maita, H., Niki, T., Iguchi-Ariga, S.M., 2013. Kalaria, R.N., Akinyemi, R., Ihara, M., 2012. Does vascular pathology contribute to
Neuroprotective function of DJ-1 in Parkinson’s disease. Oxid Med Cell Longev Alzheimer changes? J. Neurol. Sci. 322, 141–147.
2013, 683920. Kalaria, R.N., Ballard, C., 1999. Overlap between pathology of Alzheimer disease and
Béjot, Y., Aboa-Eboulé, C., Durier, J., Rouaud, O., Jacquin, A., Ponavoy, E., Richard, D., vascular dementia. Alzheimer Dis. Assoc. Disord. 13 (Suppl. 3), S115–S123.
Moreau, T., Giroud, M., 2011. Prevalence of early dementia after first-ever stroke: a Kalaria, R.N., Ihara, M., 2013. Dementia: vascular and neurodegenerative pathways-will
24-year population-based study. Stroke 42, 607–612. they meet? Nat. Rev. Neurol. 9, 487–488.
Brouns, R., De Deyn, P.P., 2009. The complexity of neurobiological processes in acute Kernagis, D.N., Laskowitz, D.T., 2012. Evolving role of biomarkers in acute
ischemic stroke. Clin. Neurol. Neurosurg. 111, 483–495. cerebrovascular disease. Ann. Neurol. 71, 289–303.
Cano, C.P., Bermúdez, V.P., Atencio, H.E., Medina, M.T., Anilsa, A., Souki, A., Molina, O. Kim, T., Mehta, S.L., Morris-Blanco, K.C., Chokkalla, A.K., Chelluboina, B., Lopez, M.,
M., Restrepo, H., Vargas, M.E., Núñez, M., Ambard, M., Toledo, A., Contreras, F., Sullivan, R., Kim, H.T., Cook, T.D., Kim, J.Y., Kim, H., Kim, C., Vemuganti, R., 2018.
Velasco, M., 2003. Increased serum malondialdehyde and decreased nitric oxide The microRNA miR-7a-5p ameliorates ischemic brain damage by repressing
within 24 hours of thrombotic stroke onset. Am. J. Therapeut. 10, 473–476. α-synuclein. Sci. Signal. 11.
Castellanos, M., Leira, R., Serena, J., Pumar, J.M., Lizasoain, I., Castillo, J., Dávalos, A., Kis, B., Kaiya, H., Nishi, R., Deli, M.A., Abraham, C.S., Yanagita, T., Isse, T., Gotoh, S.,
2003. Plasma metalloproteinase-9 concentration predicts hemorrhagic Kobayashi, H., Wada, A., Niwa, M., Kangawa, K., Greenwood, J., Yamashita, H.,
transformation in acute ischemic stroke. Stroke 34, 40–46. Ueta, Y., 2002. Cerebral endothelial cells are a major source of adrenomedullin.
Chan, P.H., 2001. Reactive oxygen radicals in signaling and damage in the ischemic J. Neuroendocrinol. 14, 283–293.
brain. J. Cerebr. Blood Flow Metabol. 21, 2–14. Kitaguchi, H., Ihara, M., Saiki, H., Takahashi, R., Tomimoto, H., 2007. Capillary beds are
Choi, D.H., Lee, J., 2017. A mini-review of the NADPH oxidases in vascular dementia: decreased in Alzheimer’s disease, but not in Binswanger’s disease. Neurosci. Lett.
correlation with NOXs and risk factors for VaD. Int. J. Mol. Sci. 18. 417, 128–131.
Chun, T.H., Itoh, H., Ogawa, Y., Tamura, N., Takaya, K., Igaki, T., Yamashita, J., Doi, K., Kong, Y., Li, S., Cheng, X., Ren, H., Zhang, B., Ma, H., Li, M., Zhang, X.A., 2020. Brain
Inoue, M., Masatsugu, K., Korenaga, R., Ando, J., Nakao, K., 1997. Shear stress ischemia significantly alters microRNA expression in human peripheral blood
augments expression of C-type natriuretic peptide and adrenomedullin. natural killer cells. Front. Immunol. 11, 759.
Hypertension 29, 1296–1302. Koyama, T., Sakurai, T., Kamiyoshi, A., Ichikawa-Shindo, Y., Kawate, H., Shindo, T.,
Cipollini, V., Troili, F., Giubilei, F., 2019. Emerging biomarkers in vascular cognitive 2015. Adrenomedullin-RAMP2 system in vascular endothelial cells.
impairment and dementia: from pathophysiological pathways to clinical application. J. Atherosclerosis Thromb. 22, 647–653.
Int. J. Mol. Sci. 20. Kuriyama, N., Ihara, M., Mizuno, T., Ozaki, E., Matsui, D., Watanabe, I., Koyama, T.,
de Leeuw, F.E., de Kleine, M., Frijns, C.J., Fijnheer, R., van Gijn, J., Kappelle, L.J., 2002. Kondo, M., Tokuda, T., Tamura, A., Yamada, K., Akazawa, K., Takeda, K.,
Endothelial cell activation is associated with cerebral white matter lesions in patients Takada, A., Mizuno, S., Nakagawa, M., Watanabe, Y., 2017. Association between
with cerebrovascular disease. Ann. N. Y. Acad. Sci. 977, 306–314. mid-regional proadrenomedullin levels and progression of deep white matter lesions
Drummond, G.R., Selemidis, S., Griendling, K.K., Sobey, C.G., 2011. Combating oxidative in the brain accompanying cognitive decline. J Alzheimers Dis 56, 1253–1262.
stress in vascular disease: NADPH oxidases as therapeutic targets. Nat. Rev. Drug Leung, L.Y., Chan, C.P., Leung, Y.K., Jiang, H.L., Abrigo, J.M., Wang de, F., Chung, J.S.,
Discov. 10, 453–471. Rainer, T.H., Graham, C.A., 2014. Comparison of miR-124-3p and miR-16 for early
Ehteshami Bejnordi, B., Veta, M., Johannes van Diest, P., van Ginneken, B., diagnosis of hemorrhagic and ischemic stroke. Clin. Chim. Acta 433, 139–144.
Karssemeijer, N., Litjens, G., van der Laak, J., the, C.C., Hermsen, M., Manson, Q.F., Liu, X., Zhang, R., Wu, Z., Si, W., Ren, Z., Zhang, S., Zhou, J., Chen, D., 2019. miR-134-
Balkenhol, M., Geessink, O., Stathonikos, N., van Dijk, M.C., Bult, P., Beca, F., 5p/Foxp2/Syn1 is involved in cognitive impairment in an early vascular dementia
Beck, A.H., Wang, D., Khosla, A., Gargeya, R., Irshad, H., Zhong, A., Dou, Q., Li, Q., rat model. Int. J. Mol. Med. 44, 1729–1740.
Chen, H., Lin, H.J., Heng, P.A., Hass, C., Bruni, E., Wong, Q., Halici, U., Oner, M.U., Livingston, G., Sommerlad, A., Orgeta, V., Costafreda, S.G., Huntley, J., Ames, D.,
Cetin-Atalay, R., Berseth, M., Khvatkov, V., Vylegzhanin, A., Kraus, O., Shaban, M., Ballard, C., Banerjee, S., Burns, A., Cohen-Mansfield, J., Cooper, C., Fox, N., Gitlin, L.
Rajpoot, N., Awan, R., Sirinukunwattana, K., Qaiser, T., Tsang, Y.W., Tellez, D., N., Howard, R., Kales, H.C., Larson, E.B., Ritchie, K., Rockwood, K., Sampson, E.L.,
Annuscheit, J., Hufnagl, P., Valkonen, M., Kartasalo, K., Latonen, L., Ruusuvuori, P., Samus, Q., Schneider, L.S., Selbæk, G., Teri, L., Mukadam, N., 2017. Dementia
Liimatainen, K., Albarqouni, S., Mungal, B., George, A., Demirci, S., Navab, N., prevention, intervention, and care. Lancet 390, 2673–2734.
Watanabe, S., Seno, S., Takenaka, Y., Matsuda, H., Ahmady Phoulady, H., Long, G., Wang, F., Li, H., Yin, Z., Sandip, C., Lou, Y., Wang, Y., Chen, C., Wang, D.W.,
Kovalev, V., Kalinovsky, A., Liauchuk, V., Bueno, G., Fernandez-Carrobles, M.M., 2013. Circulating miR-30a, miR-126 and let-7b as biomarker for ischemic stroke in
Serrano, I., Deniz, O., Racoceanu, D., Venancio, R., 2017. Diagnostic assessment of humans. BMC Neurol. 13, 178.
deep learning algorithms for detection of lymph node metastases in women with Lu, K., Xu, X., Cui, S., Wang, F., Zhang, B., Zhao, Y., 2015. Serum neuron specific enolase
breast cancer. J. Am. Med. Assoc. 318, 2199–2210. level as a predictor of prognosis in acute ischemic stroke patients after intravenous
Eng, L.F., Ghirnikar, R.S., Lee, Y.L., 2000. Glial fibrillary acidic protein: GFAP-thirty-one thrombolysis. J. Neurol. Sci. 359, 202–206.
years (1969-2000). Neurochem. Res. 25, 1439–1451. Lyang, O.V., Kochetov, A.G., Zhirova, I.A., Novozhenova, Y.V., Ivoylov, O.O.,
Ercan, M., Mungan, S., Güzel, I., Celik, H.T., Bal, C., Abusoglu, S., Akbulut, D., Oguz, E. Stakhovskaya, L.V., 2019. Heart - type fatty acid binding protein in prognosis of
F., Yilmaz, F.M., 2019. Serum asymmetric dimethylarginine and nitric oxide levels in ischemic stroke at the hospital stage. Ter. Arkh. 91, 35–40.
Turkish patients with acute ischemic stroke. Adv. Clin. Exp. Med. 28, 693–698. Lynch, J.R., Blessing, R., White, W.D., Grocott, H.P., Newman, M.F., Laskowitz, D.T.,
Feng, R., Badgeley, M., Mocco, J., Oermann, E.K., 2018. Deep learning guided stroke 2004. Novel diagnostic test for acute stroke. Stroke 35, 57–63.
management: a review of clinical applications. J. Neurointerventional Surg. 10, Madeira, A., Pommet, J.M., Prochiantz, A., Allinquant, B., 2005. SET protein (TAF1beta,
358–362. I2PP2A) is involved in neuronal apoptosis induced by an amyloid precursor protein
Fotuhi, M., Hachinski, V., Whitehouse, P.J., 2009. Changing perspectives regarding late- cytoplasmic subdomain. Faseb. J. 19, 1905–1907.
life dementia. Nat. Rev. Neurol. 5, 649–658. Maki, T., Ihara, M., Fujita, Y., Nambu, T., Miyashita, K., Yamada, M., Washida, K.,
Gao, Q., Fan, Y., Mu, L.Y., Ma, L., Song, Z.Q., Zhang, Y.N., 2015. S100B and ADMA in Nishio, K., Ito, H., Harada, H., Yokoi, H., Arai, H., Itoh, H., Nakao, K., Takahashi, R.,
cerebral small vessel disease and cognitive dysfunction. J. Neurol. Sci. 354, 27–32. Tomimoto, H., 2011. Angiogenic and vasoprotective effects of adrenomedullin on
Ghafar, M., Miptah, H.N., O’Caoimh, R., 2019. Cognitive screening instruments to prevention of cognitive decline after chronic cerebral hypoperfusion in mice. Stroke
identify vascular cognitive impairment: a systematic review. Int. J. Geriatr. 42, 1122–1128.
Psychiatr. 34, 1114–1127. Markus, H.S., Hunt, B., Palmer, K., Enzinger, C., Schmidt, H., Schmidt, R., 2005. Markers
Gorelick, P.B., Scuteri, A., Black, S.E., Decarli, C., Greenberg, S.M., Iadecola, C., of endothelial and hemostatic activation and progression of cerebral white matter
Launer, L.J., Laurent, S., Lopez, O.L., Nyenhuis, D., Petersen, R.C., Schneider, J.A., hyperintensities: longitudinal results of the Austrian Stroke Prevention Study. Stroke
Tzourio, C., Arnett, D.K., Bennett, D.A., Chui, H.C., Higashida, R.T., Lindquist, R., 36, 1410–1414.
Nilsson, P.M., Roman, G.C., Sellke, F.W., Seshadri, S., 2011. Vascular contributions Mijajlović, M.D., Pavlović, A., Brainin, M., Heiss, W.D., Quinn, T.J., Ihle-Hansen, H.B.,
to cognitive impairment and dementia: a statement for healthcare professionals from Hermann, D.M., Assayag, E.B., Richard, E., Thiel, A., Kliper, E., Shin, Y.I., Kim, Y.H.,
the american heart association/american stroke association. Stroke 42, 2672–2713. Choi, S., Jung, S., Lee, Y.B., Sinanović, O., Levine, D.A., Schlesinger, I., Mead, G.,
Hassan, A., Hunt, B.J., O’Sullivan, M., Parmar, K., Bamford, J.M., Briley, D., Brown, M. Milošević, V., Leys, D., Hagberg, G., Ursin, M.H., Teuschl, Y., Prokopenko, S.,
M., Thomas, D.J., Markus, H.S., 2003. Markers of endothelial dysfunction in lacunar Mozheyko, E., Bezdenezhnykh, A., Matz, K., Aleksić, V., Muresanu, D., Korczyn, A.
infarction and ischaemic leukoaraiosis. Brain 126, 424–432.

7
S. Hosoki et al. Neurochemistry International 146 (2021) 105015

D., Bornstein, N.M., 2017. Post-stroke dementia - a comprehensive review. BMC Satizabal, C.L., Beiser, A.S., Chouraki, V., Chêne, G., Dufouil, C., Seshadri, S., 2016.
Med. 15, 11. Incidence of dementia over three decades in the framingham heart study. N. Engl. J.
Mitchell, P.S., Parkin, R.K., Kroh, E.M., Fritz, B.R., Wyman, S.K., Pogosova- Med. 374, 523–532.
Agadjanyan, E.L., Peterson, A., Noteboom, J., O’Briant, K.C., Allen, A., Lin, D.W., Sato, M., Suzuki, A., Nagata, K., Uchiyama, S., 2006. Increased von Willebrand factor in
Urban, N., Drescher, C.W., Knudsen, B.S., Stirewalt, D.L., Gentleman, R., Vessella, R. acute stroke patients with atrial fibrillation. J. Stroke Cerebrovasc. Dis. 15, 1–7.
L., Nelson, P.S., Martin, D.B., Tewari, M., 2008. Circulating microRNAs as stable Schneider, J.A., Arvanitakis, Z., Bang, W., Bennett, D.A., 2007. Mixed brain pathologies
blood-based markers for cancer detection. Proc. Natl. Acad. Sci. U. S. A. 105, account for most dementia cases in community-dwelling older persons. Neurology
10513–10518. 69, 2197–2204.
Miyashita, K., Itoh, H., Arai, H., Suganami, T., Sawada, N., Fukunaga, Y., Sone, M., Shen, Y., Gao, H.M., 2015. Serum somatostatin and neuron-specific enolase might be
Yamahara, K., Yurugi-Kobayashi, T., Park, K., Oyamada, N., Sawada, N., Taura, D., biochemical markers of vascular dementia in the early stage. Int. J. Clin. Exp. Med.
Tsujimoto, H., Chao, T.H., Tamura, N., Mukoyama, M., Nakao, K., 2006. The 8, 19471–19475.
neuroprotective and vasculo-neuro-regenerative roles of adrenomedullin in ischemic Shindo, T., Kurihara, H., Maemura, K., Kurihara, Y., Kuwaki, T., Izumida, T.,
brain and its therapeutic potential. Endocrinology 147, 1642–1653. Minamino, N., Ju, K.H., Morita, H., Oh-hashi, Y., Kumada, M., Kangawa, K.,
Nagai, K., Kozaki, K., Sonohara, K., Akishita, M., Toba, K., 2011. Relationship between Nagai, R., Yazaki, Y., 2000. Hypotension and resistance to lipopolysaccharide-
interleukin-6 and cerebral deep white matter and periventricular hyperintensity in induced shock in transgenic mice overexpressing adrenomedullin in their
elderly women. Geriatr. Gerontol. Int. 11, 328–332. vasculature. Circulation 101, 2309–2316.
Nagai, M., Hoshide, S., Kario, K., 2012. Association of prothrombotic status with markers Si, W., Ye, S., Ren, Z., Liu, X., Wu, Z., Li, Y., Zhou, J., Zhang, S., Li, Y., Deng, R., Chen, D.,
of cerebral small vessel disease in elderly hypertensive patients. Am. J. Hypertens. 2019. miR-335 promotes stress granule formation to inhibit apoptosis by targeting
25, 1088–1094. ROCK2 in acute ischemic stroke. Int. J. Mol. Med. 43, 1452–1466.
Nakamura, K., Shichita, T., 2019. Cellular and molecular mechanisms of sterile Snowdon, D.A., Greiner, L.H., Mortimer, J.A., Riley, K.P., Greiner, P.A., Markesbery, W.
inflammation in ischaemic stroke. J. Biochem. 165, 459–464. R., 1997. Brain infarction and the clinical expression of Alzheimer disease. The Nun
Nakayama, M., Takahashi, K., Murakami, O., Shirato, K., Shibahara, S., 1998. Induction Study. J. Am. Med. Assoc. 277, 813–817.
of adrenomedullin by hypoxia and cobalt chloride in human colorectal carcinoma Song, Q., Ji, Q., Xiao, J., Li, F., Wang, L., Chen, Y., Xu, Y., Jiao, S., 2018. miR-409 inhibits
cells. Biochem. Biophys. Res. Commun. 243, 514–517. human non-small-cell lung cancer progression by directly targeting SPIN1. Mol.
Nishio, K., Ihara, M., Yamasaki, N., Kalaria, R.N., Maki, T., Fujita, Y., Ito, H., Oishi, N., Ther. Nucleic Acids 13, 154–163.
Fukuyama, H., Miyakawa, T., Takahashi, R., Tomimoto, H., 2010. A mouse model Staals, J., Makin, S.D., Doubal, F.N., Dennis, M.S., Wardlaw, J.M., 2014. Stroke subtype,
characterizing features of vascular dementia with hippocampal atrophy. Stroke 41, vascular risk factors, and total MRI brain small-vessel disease burden. Neurology 83,
1278–1284. 1228–1234.
Novak, V., Hajjar, I., 2010. The relationship between blood pressure and cognitive Tanne, D., Haim, M., Boyko, V., Goldbourt, U., Reshef, T., Matetzky, S., Adler, Y.,
function. Nat. Rev. Cardiol. 7, 686–698. Mekori, Y.A., Behar, S., 2002. Soluble intercellular adhesion molecule-1 and risk of
Okamoto, Y., Yamamoto, T., Kalaria, R.N., Senzaki, H., Maki, T., Hase, Y., Kitamura, A., future ischemic stroke: a nested case-control study from the Bezafibrate Infarction
Washida, K., Yamada, M., Ito, H., Tomimoto, H., Takahashi, R., Ihara, M., 2012. Prevention (BIP) study cohort. Stroke 33, 2182–2186.
Cerebral hypoperfusion accelerates cerebral amyloid angiopathy and promotes Tian, C., Li, Z., Yang, Z., Huang, Q., Liu, J., Hong, B., 2016. Plasma MicroRNA-16 is a
cortical microinfarcts. Acta Neuropathol. 123, 381–394. biomarker for diagnosis, stratification, and prognosis of hyperacute cerebral
Olivot, J.M., Labreuche, J., Aiach, M., Amarenco, P., 2004. Soluble thrombomodulin and infarction. PLoS One 11, e0166688.
brain infarction: case-control and prospective study. Stroke 35, 1946–1951. Tombul, T., Atbas, C., Anlar, O., 2005. Hemostatic markers and platelet aggregation
Påhlman, S., Esscher, T., Bergvall, P., Odelstad, L., 1984. Purification and factors as predictive markers for type of stroke and neurological disability following
characterization of human neuron-specific enolase: radioimmunoassay development. cerebral infarction. J. Clin. Neurosci. 12, 429–434.
Tumour Biol 5, 127–139. Ueda, S., Nishio, K., Minamino, N., Kubo, A., Akai, Y., Kangawa, K., Matsuo, H.,
Papa, L., Akinyi, L., Liu, M.C., Pineda, J.A., Tepas 3rd, J.J., Oli, M.W., Zheng, W., Fujimura, Y., Yoshioka, A., Masui, K., Doi, N., Murao, Y., Miyamoto, S., 1999.
Robinson, G., Robicsek, S.A., Gabrielli, A., Heaton, S.C., Hannay, H.J., Demery, J.A., Increased plasma levels of adrenomedullin in patients with systemic inflammatory
Brophy, G.M., Layon, J., Robertson, C.S., Hayes, R.L., Wang, K.K., 2010. Ubiquitin C- response syndrome. Am. J. Respir. Crit. Care Med. 160, 132–136.
terminal hydrolase is a novel biomarker in humans for severe traumatic brain injury. Uno, M., Kitazato, K.T., Nishi, K., Itabe, H., Nagahiro, S., 2003. Raised plasma oxidised
Crit. Care Med. 38, 138–144. LDL in acute cerebral infarction. J. Neurol. Neurosurg. Psychiatry 74, 312–316.
Pasi, M., Cordonnier, C., 2020. Clinical relevance of cerebral small vessel diseases. Stroke Vallance, P., Leone, A., Calver, A., Collier, J., Moncada, S., 1992. Endogenous
51, 47–53. dimethylarginine as an inhibitor of nitric oxide synthesis. J. Cardiovasc. Pharmacol.
Pawelczyk, M., Glabiński, A., Kaczorowska, B., Baj, Z., 2018. sP- and sE-selectin in stroke 20 (Suppl. 12), S60–S62.
patients with metabolic disorders. Neurol. Neurochir. Pol. 52, 599–605. van der Flier, W.M., Skoog, I., Schneider, J.A., Pantoni, L., Mok, V., Chen, C.L.H.,
Pendlebury, S.T., Rothwell, P.M., 2009. Prevalence, incidence, and factors associated Scheltens, P., 2018. Vascular cognitive impairment. Nat Rev Dis Primers 4, 18003.
with pre-stroke and post-stroke dementia: a systematic review and meta-analysis. Venkat, P., Chopp, M., Chen, J., 2015. Models and mechanisms of vascular dementia.
Lancet Neurol. 8, 1006–1018. Exp. Neurol. 272, 97–108.
Pendlebury, S.T., Rothwell, P.M., 2019. Incidence and prevalence of dementia associated Vermeer, S.E., Hollander, M., van Dijk, E.J., Hofman, A., Koudstaal, P.J., Breteler, M.M.,
with transient ischaemic attack and stroke: analysis of the population-based Oxford 2003a. Silent brain infarcts and white matter lesions increase stroke risk in the
Vascular Study. Lancet Neurol. 18, 248–258. general population: the Rotterdam Scan Study. Stroke 34, 1126–1129.
Pikula, A., Böger, R.H., Beiser, A.S., Maas, R., DeCarli, C., Schwedhelm, E., Himali, J.J., Vermeer, S.E., Longstreth Jr., W.T., Koudstaal, P.J., 2007. Silent brain infarcts: a
Schulze, F., Au, R., Kelly-Hayes, M., Kase, C.S., Vasan, R.S., Wolf, P.A., Seshadri, S., systematic review. Lancet Neurol. 6, 611–619.
2009. Association of plasma ADMA levels with MRI markers of vascular brain injury: Vermeer, S.E., Prins, N.D., den Heijer, T., Hofman, A., Koudstaal, P.J., Breteler, M.M.,
framingham offspring study. Stroke 40, 2959–2964. 2003b. Silent brain infarcts and the risk of dementia and cognitive decline. N. Engl.
Polidori, M.C., Cherubini, A., Stahl, W., Senin, U., Sies, H., Mecocci, P., 2002. Plasma J. Med. 348, 1215–1222.
carotenoid and malondialdehyde levels in ischemic stroke patients: relationship to Viswanathan, A., Rocca, W.A., Tzourio, C., 2009. Vascular risk factors and dementia:
early outcome. Free Radic. Res. 36, 265–268. how to move forward? Neurology 72, 368–374.
Prabhakar, P., Chandra, S.R., Christopher, R., 2017. Circulating microRNAs as potential Wang, A., Liu, J., Meng, X., Li, J., Wang, H., Wang, Y., Su, Z., Zhang, N., Dai, L.,
biomarkers for the identification of vascular dementia due to cerebral small vessel Wang, Y., Wang, Y., 2018a. Association between oxidized low-density lipoprotein
disease. Age Ageing 46, 861–864. and cognitive impairment in patients with ischemic stroke. Eur. J. Neurol. 25,
Qi, R., Liu, H., Liu, C., Xu, Y., Liu, C., 2020. Expression and short-term prognostic value 185–191.
of miR-126 and miR-182 in patients with acute stroke. Exp Ther Med 19, 527–534. Wang, A., Yang, Y., Su, Z., Yue, W., Hao, H., Ren, L., Wang, Y., Cao, Y., Wang, Y., 2017.
Rallidis, L.S., Zolindaki, M.G., Vikelis, M., Kaliva, K., Papadopoulos, C., Kremastinos, D. Association of oxidized low-density lipoprotein with prognosis of stroke and stroke
T., 2009. Elevated soluble intercellular adhesion molecule-1 levels are associated subtypes. Stroke 48, 91–97.
with poor short-term prognosis in middle-aged patients with acute ischaemic stroke. Wang, W., Li, D.B., Li, R.Y., Zhou, X., Yu, D.J., Lan, X.Y., Li, J.P., Liu, J.L., 2018b.
Int. J. Cardiol. 132, 216–220. Diagnosis of hyperacute and acute ischaemic stroke: the potential utility of exosomal
Rana, A.K., Singh, D., 2018. Targeting glycogen synthase kinase-3 for oxidative stress MicroRNA-21-5p and MicroRNA-30a-5p. Cerebrovasc. Dis. 45, 204–212.
and neuroinflammation: opportunities, challenges and future directions for cerebral Wang, Y., Xu, S., Pan, S., Ouyang, H., Zang, Z., Tan, J., 2018c. Association of serum
stroke management. Neuropharmacology 139, 124–136. neuron-specific enolase and bilirubin levels with cerebral dysfunction and prognosis
Ren, C., Kobeissy, F., Alawieh, A., Li, N., Li, N., Zibara, K., Zoltewicz, S., Guingab- in large-artery atherosclerotic strokes. J. Cell. Biochem. 119, 9685–9693.
Cagmat, J., Larner, S.F., Ding, Y., Hayes, R.L., Ji, X., Mondello, S., 2016. Assessment Whiteley, W., Tseng, M.C., Sandercock, P., 2008. Blood biomarkers in the diagnosis of
of serum UCH-L1 and GFAP in acute stroke patients. Sci. Rep. 6, 24588. ischemic stroke: a systematic review. Stroke 39, 2902–2909.
Richard, S., Lagerstedt, L., Burkhard, P.R., Debouverie, M., Turck, N., Sanchez, J.C., Witwer, K.W., Halushka, M.K., 2016. Toward the promise of microRNAs - enhancing
2015. E-selectin and vascular cell adhesion molecule-1 as biomarkers of 3-month reproducibility and rigor in microRNA research. RNA Biol. 13, 1103–1116.
outcome in cerebrovascular diseases. J. Inflamm. 12, 61. Wunderlich, M.T., Lins, H., Skalej, M., Wallesch, C.W., Goertler, M., 2006. Neuron-
Rojo, D.R., Prough, D.S., Falduto, M.T., Boone, D.R., Micci, M.A., Kahrig, K.M., specific enolase and tau protein as neurobiochemical markers of neuronal damage
Crookshanks, J.M., Jimenez, A., Uchida, T., Cowart, J.C., Hawkins, B.E., Avila, M., are related to early clinical course and long-term outcome in acute ischemic stroke.
DeWitt, D.S., Hellmich, H.L., 2011. Influence of stochastic gene expression on the Clin. Neurol. Neurosurg. 108, 558–563.
cell survival rheostat after traumatic brain injury. PLoS One 6, e23111. Wunderlich, M.T., Wallesch, C.W., Goertler, M., 2004. Release of neurobiochemical
Saito, S., Ihara, M., 2016. Interaction between cerebrovascular disease and Alzheimer markers of brain damage is related to the neurovascular status on admission and the
pathology. Curr. Opin. Psychiatr. 29, 168–173. site of arterial occlusion in acute ischemic stroke. J. Neurol. Sci. 227, 49–53.

8
S. Hosoki et al. Neurochemistry International 146 (2021) 105015

Xu, G., Zhang, H., Zhang, S., Fan, X., Liu, X., 2008. Plasma fibrinogen is associated with Double-Blind, Placebo-Controlled, Phase-II Trial: AdrenoMedullin for Ischemic
cognitive decline and risk for dementia in patients with mild cognitive impairment. Stroke Study. J Stroke Cerebrovasc Dis 105761. doi:10.1016/j.
Int. J. Clin. Pract. 62, 1070–1075. jstrokecerebrovasdis.2021.105761. In press.
Yang, H.H., Chen, Y., Gao, C.Y., Cui, Z.T., Yao, J.M., 2017a. Protective effects of Yuan, M., Bi, X., 2020. Therapeutic and diagnostic potential of microRNAs in vascular
MicroRNA-126 on human cardiac microvascular endothelial cells against hypoxia/ cognitive impairment. J. Mol. Neurosci. 70, 1619–1628.
reoxygenation-induced injury and inflammatory response by activating PI3K/Akt/ Zekry, D., Hauw, J.J., Gold, G., 2002. Mixed dementia: epidemiology, diagnosis, and
eNOS signaling pathway. Cell. Physiol. Biochem. 42, 506–518. treatment. J. Am. Geriatr. Soc. 50, 1431–1438.
Yang, X., Tang, X., Sun, P., Shi, Y., Liu, K., Hassan, S.H., Stetler, R.A., Chen, J., Yin, K.J., Zhao, B., Zhu, Z., Hao, J., Wan, Z., Guo, X., 2016. Decreased plasma miR-335 expression
2017b. MicroRNA-15a/16-1 antagomir ameliorates ischemic brain injury in in patients with acute ischemic stroke and its association with calmodulin
experimental stroke. Stroke 48, 1941–1947. expression. J. Int. Med. Res. 44, 1331–1338.
Yatawara, C., Guevarra, A.C., Ng, K.P., Chander, R., Lam, B.Y.K., Wong, A., Mok, V., Zhou, J., Chen, L., Chen, B., Huang, S., Zeng, C., Wu, H., Chen, C., Long, F., 2018.
Kandiah, N., 2020. The role of cerebral microbleeds in the incidence of post-stroke Increased serum exosomal miR-134 expression in the acute ischemic stroke patients.
dementia. J. Neurol. Sci. 412, 116736. BMC Neurol. 18, 198.
Ye, Z.M., Yang, S., Xia, Y.P., Hu, R.T., Chen, S., Li, B.W., Chen, S.L., Luo, X.Y., Mao, L., Zhou, S., Bao, J., Wang, Y., Pan, S., 2016. S100β as a biomarker for differential diagnosis
Li, Y., Jin, H., Qin, C., Hu, B., 2019. LncRNA MIAT sponges miR-149-5p to inhibit of intracerebral hemorrhage and ischemic stroke. Neurol. Res. 38, 327–332.
efferocytosis in advanced atherosclerosis through CD47 upregulation. Cell Death Dis. Zhu, M., Li, N., Luo, P., Jing, W., Wen, X., Liang, C., Tu, J., 2018. Peripheral blood
10, 138. leukocyte expression of lncRNA MIAT and its diagnostic and prognostic value in
Yokota, C., Sawamura, T., Watanabe, M., Kokubo, Y., Fujita, Y., Kakino, A., Nakai, M., ischemic stroke. J. Stroke Cerebrovasc. Dis. 27, 326–337.
Toyoda, K., Miyamoto, Y., Minematsu, K., 2016. High levels of soluble lectin-like Zimmermann-Ivol, C.G., Burkhard, P.R., Le Floch-Rohr, J., Allard, L., Hochstrasser, D.F.,
oxidized low-density lipoprotein receptor-1 in acute stroke: an age- and sex-matched Sanchez, J.C., 2004. Fatty acid binding protein as a serum marker for the early
cross-sectional study. J. Atherosclerosis Thromb. 23, 1222–1226. diagnosis of stroke: a pilot study. Mol. Cell. Proteomics 3, 66–72.
Yoshimoto, T., Saito, S., Omae, K., Hattori, Y., Fukuma, K., Kitamura, K., Kakuta, R., Kita,
T., Maruyama, H., Yamamoto, H., Ihara, M., n.d.. Study Protocol for a Randomized,

You might also like