Abstract
Ischemic stroke (IS) is a leading disease with high mortality and disability, as well as with limited therapeutic window. Biomarkers for earlier diagnosis of IS have long been pursued. Family and twin studies confirm that genetic variations play an important role in IS pathogenesis. Besides DNA mutations found previously by genetic linkage analysis for monogenic IS (Mendelian inheritance), recent studies using genome-wide associated study (GWAS) and microRNA expression profiling have resulted in a large number of DNA and microRNA biomarkers in polygenic IS (sporadic IS), especially in different IS subtypes and imaging phenotypes. The present review summarizes genetic markers discovered by clinical studies and discusses their pathogenic molecular mechanisms involved in developmental or regenerative anomalies of blood vessel walls, neuronal apoptosis, excitotoxic death, inflammation, neurogenesis and angiogenesis. The possible impact of environment on genetics is addressed as well. We also include a perspective on further studies and clinical application of these IS biomarkers.
Keywords: Biomarkers, Genetics, Ischemic stroke, MicroRNA, Monogenic, Polygenic
Introduction
Stroke is one of the leading causes of mortality and disability, annually affecting 16.9 million people worldwide [1]. Approximately 80-90% of cases are ischemic stroke (IS) [2, 3]. Cerebral lesions due to ischemia and hypoxia can cause acute, irreversible neuronal death. The short therapeutic window and poor prognosis bring a sense of urgency to developing earlier diagnosis and preventive treatment [4-8]. Recent revolutionary advances in microarray and computational methodologies have facilitated high-throughput screening of biomarkers in complex diseases, as evidenced by recent successes in gene discovery for coronary artery disease [9]. Here we summarize the potential genetic markers including DNA mutations, single nucleotide polymorphisms (SNP) and microRNAs that have been discovered so far in both single-gene and sporadic IS patients.
IS Subtypes, Heredity and Modes of Inheritance
According to Trial of Org 10172 in Acute Stroke Treatment (TOAST) criteria, IS is classified into five subtypes: large artery atherosclerosis (LAA), small-vessel disease (SVD), cardioembolic stroke (CES), other determined etiology (ODE), and cryptogenic ischemia stroke (CIS) [10]. It is reported that the distribution of these 5 subtypes among UK residents of European background is 16%, 14%, 38%, 5% and 27%, respectively [11] (Fig. 1a).
Fig. 1.

Etiology, subtype and heredity of ischemic stroke (IS). a, Incidence and genetics of different IS subtypes. The percentages in the boxes indicate incidences and those outside the boxes denote genetics; OD: other determined etiology. b, Etiological factors of IS. The percentages in the boxes indicate incidences and the content in the text boxes indicates risk factors. These investigative data are extracted from references studying on European residents [11, 13, 14, 16].
The heredity of IS has been confirmed by twin and family-history studies [12] and extended by genome-wide association studies (GWAS) [13]. In general, hereditary factors cause IS by three pathways, i.e. monogenic, polygenetic and epigenetic modes. Although monogenic IS has high penetrance, it only occurs in 7% of IS patients [14]. In contrast, about 38% of IS incidences result from genetic polymorphisms in multiple genes with each risk allele making minor effects to the pathogenesis. The various combinations of these risk alleles lead to different heredity of stroke subtypes, which is reported to be 40% for LAA, 33% for CES and 16% for SVD (Fig. 1a) [13]. These results are consistent with the current common consensus that stroke is not a disorder but rather a syndrome. The involvement of epigenetic alterations in environmentally triggered phenotypes and diseases has generated a lot of interest [15] and this is also true for IS where environmental risk factors such as aging, smoking, alcohol and other such factors account for non-genetic etiologies of IS incidence [16] (Fig. 1b).
Genetic Markers for Monogenic IS
There are dozens of established monogenic disorders to date that can cause IS as the predominant clinical phenotype or as part of a systemic disease. The majority of monogenic forms of stroke are associated with single IS subtypes, although a few can present with more than one subtype [17]. Genetic linkage analysis has identified most of the pathogenic genes [18] as presented in Table 1 [19-49], which can act as robust diagnostic bases for clinical suspicion in young adults with stroke due to unknown causes [50]. According to targeted components of these genetic defects, most monogenic IS is caused by structural deficiencies of blood vessel walls due to developmental or regenerative anomalies (Fig. 2). Of note, the severity and onset age of IS phenotype are affected by genetic diversity and life styles [19].
Table 1.
Genetic markers for monogenic cause of ischemic stroke
| Genes | Gene products | Diseases | Mode of inheritance |
Mutaition Loci |
IS Subtype | No. of Mutations |
Mechanisms | IS onset Rates* |
Age of IS Presentation |
Refs |
|---|---|---|---|---|---|---|---|---|---|---|
| NOTCH3 | Notch3 | CADASIL | AD | 19p13.2-p13.1 | SVD | 230 | The Notch3 ectodomain misfolding and accumulation lead to intimal hyperplasia. | 51-74% | 46±9.7 years | [15-23] |
| HTRA1 | HtrA serine peptidase/proteas e 1 | CARASIL | AR | 10q25 | SVD | 11 | De-repression of TGF-β1 and fibrous intimal proliferation. | 50% | 20-30 years | [20, 24, 25] |
| TREX1 | TREXl 3’-5’ exonuclease | RVCL | AD | 3p21.31 | SVD-like | 5 frame-shift mutations | Lose of perinuclear localization and apoptosis induction leading to intimal hyperplasia | 97% | 40-50 years | [26-28] |
| COL4A1/COL4A2 | Procollagen Type VI αl | HANAC , ARALES | AD | 13q24 | SVD | 14 | Interruption and thickening of capillary basement membrane. | 6% | 35-50 years | [29-31] |
| α-GAL A | α-galactosidase A | Fabry disease | X-linked | Xq22.1 | LAA, SVD | 245 | Accumulation of globotriaosylceramide in lysosomes of vascular endothelial cells and smooth muscle cells. | Male: 6.0%; Female: 3.7% | 40-55 years | [32-34] |
| HBB | β-hemoglobin | Sickle-cell anemia | AR | 11p15.4 | LAA, SVD | Defective Beta-chain hemoglobin molecule. | 3.2% | < 45 years | [35, 36] | |
| FBN1 | Fibrillin-1 | Marfan syndrome | AD | 15q21.1 | CES | 637 | Deficiency of fibrillin-1 disrupts the elastic lamina of aorta. | 2.5% | < 40 years | [37-39] |
| CBS | Cystathionev β-synthase | Homocystinuria | AR | 21q22.3 | LAA, SVD | 48 | High homocysteine leading to vascular endothelial injuries. | 13% | childhood | [40-42] |
| NF1 | Neurofibromin | Neurofibromat osis type 1 | AD | 17q11.2 | LAA, SVD | 553 | Increasing mitogenic signaling and leading to cellular proliferation or differentiation | 0.49% | < 30 years | [43-46] |
| MTLL1 | MELAS | tRNALeu(UUR) | Maternally inherited | mtDNA | IS-like | 5 | Protein synthese obstacle affect energy metabolism, reducing oxidation stress tolerance | 84-99% | 2-40 years | [47-49] |
IS onset rates in patients carrying monogenic mutations.
Fig. 2.

Targeted components of blood vessels by genetic defects in monogenic ischemic stroke. The genetic mutations affecting integrity of basement membrane in capillaries and intima elastica in arterioles can lead to cerebral small vessel diseases (SVD); activation of endothelial cells and smooth muscle cells in arterioles and large arteries by dysfunctional expression of growth factors or metabolic enzymes can cause cerebral SVD and large artery atherosclerotic (LAA) stroke; mutations in FBN1 leading to deficient tunica elastica are the main causes of cardioembolic stroke (CES) in Marfan syndrome. The italic scripts in the figure denote genetic markers for monogenic ischemic stroke, the lines indicate the targeted components of blood vessel walls by genetic markers.
Genetic Markers for Polygenic IS
Studies to identify genetic markers for common sporadic IS have used linkage-association analysis, genome-wide association (GWS) analysis and rare variation association analysis (RVA) based on next-generation genetic sequencing (NGGS) [51]. The use of these innovative approaches has already identified a number of novel variants associated with risk of IS (Table 3) and has provided important insights into the genetic basis of this disease.
Table 3.
MicroRNA markers in patients with ischemic stroke
| Category | Sourcea | Timee | Markersf | Refs | Category | Markers | Changesg | AUC | Target genes | Refs |
|---|---|---|---|---|---|---|---|---|---|---|
| Profiling | Whole blood | 21-72h | Up: miRNA-125b-2, 27a, 422a, 488, and 627. | [100] | Apoptosis | miRNA-15a | + 8.3 | 0.70 | Bcl-2, Akt-3, NT-κB | [115] |
| Whole blood (Youngb) | 6-18 m 1290, let-7e; | Up: miRNA-25, 181a, 513a-5p, 550, 602, 665, 891a, 923, 933, 939, 1184, 1246, 1261, 1275, 1285; Down: 15b, 126, 142-3p, 186, 519e, 768-5p, 1259, let-7f. | [101] | Inflammation | −16 −21 −24 −29b miRNA-let-7b | + 42.0 + 3.3 + 4.9 - + 7.5 | 0.82 NA NA NA 0.93 | Bcl-2, VEGF-A Bcl-2 XIAP AQP-4 LOX-1 | [114-116] [118] [118] [120] [125] | |
| Plasma | 24h | Up: miRNA-106b-5P, 4306; Down: 320e, 320d. | [102] | −let-7e | + 1.5 | 0.86 | CRP | [100,126] | ||
| Plasma (Low riskc) | 2-24 m | Up: miRNA-1258, 125a-5p, 1260, 1273, 149,220b,
23a, 26b, 29b-1, 302e, 488, 490-3p, 506, 659, 890, 920, 934; Down: 25, 34b, 483-5p, 498. |
[103] | −let-7c-5p −let-7f −let-7i −124 | −1.9 - − 2.1 + 1.3 | NA NA NA 0.76 | Caspase 3 IL-6 CD86, CXCL8, HMGB1 hs-CRP, MMP-9 | [127] [128] [105,129] [131] | ||
| Plasma (Depressiond) | 2w | Up: miRNA-22-3p, 4476, 486-5p, 92a-3p; Down:
1185- 1-3p, 1234-5p, 1247-3p, 133a, 183-3p, 187-5p, 3184-3p, 3202, 3615,
3667-5p, 4310, 4714-5p, 4716-3p, 4738-3p, 4769-5p,548a, 5571-5p, 629-3p, 636, 665, 887. |
[104] | Excitotoxicity | −145 −221 miRNA-223 −107 −128b | + 2.1 + 10.4 + 3.5 + 2.8 + 2.1 | NA NA NA 0.96 0.90 | KLF4/5 PTEN, AKT GluR2, NR2B GLT-1 SP1, GLT-1 | [134-136] [138] [143] [146] [146] | |
| Serum | 24h | Up: miRNA-32-3p, 106-5p, 1246; Down: 532-5p. | [105] | −125b | + 2.5 | 0.95 | NR2A | [148,149] | ||
| PBC | 72h | Up: miRNA-363, 487b; Down: miRNA-122, 148a, 19a, 320d, 4429, let-7i. | [106] | Neurogenesis and −210 angiogenesis | miRNA-9 | - − 1.4 | NA 0.80 | hs-CRP, MMP-9 EFNA3 | [131,152] [157-159] | |
| CSF | 72h | Up: miRNA-let-7c, 221-3p. | [107] −126 | −126 | − 8.3 | 0.92 | VCAM-1, DLK1 | [125,160] |
sample source;
young IS patients;
low IS risk people;
IS patients with depression;
deteting time from IS onset;
WRISTA markers( Up:up-regulated expression; Down: down-regulated expression);
References refer to supplemental materials;
changing magnifications ( “+”up-regulated expression, “−”down-regulated expression); NA:not available.
AKT:protein kinase B; Akt-3:AKT serine/threonine kinase 3; AQP-4:aquaporin-4; Bcl-2:B-cell lymphoma-2; CSF:erebral spinal fluid; CXCL8:C-X-C motif chemokine ligand 8; DLK-1:delta like 1 homolog; EFNA3:Eph family receptor interacting proteins-A3; GLT-1:glutamate transporter-1; GluR2: glutamate receptor 2; HMGB1:high mobility group box-1; hs-CRP:high sensitive C-reactive protein; IGF-1:insulin-like growth factor-1; KLF4/5:kruppel-like factor 4/5; IL-6:interleukin 6; LOX-1:lectin-like oxidized low density lipoprotein receptor-1; MMP-9:matrix metalloprotein-9; NF-KB:nuclear factor kappa-B; NR2A:NMDA receptor 2A; NR2B:NMDA receptor 2B; PBC:peripheral blood cell; PT:prothrombin; PTEN:phosphatase and tensin homolog deleted on chromosome ten; SP1:transcription factor spl; TLR: toll-like receptor; VCAM-1: vascular cell adhesion molecule 1; VEGF-A: vascular endothelial growth factor A; XIAP:x-linked inhibitor of apoptosis protein.
Genetic Markers Discovered by Linkage-Association Studies
Linkage-association studies are based on the initial linkage analysis in stroke families to scan for susceptibility genes and subsequent association analysis in unrelated stroke individuals using microsatellite markers. Recent Linkage-association studies have found two predisposed genes: arachidonate 5-lipoxygenase activating protein (ALOX5AP) and phosphodiesterase 4D (PDE4D), for common stroke and IS respectively [52, 53].
The ALOX5AP gene encodes a major regulator for 5-lipoxygenase that catalyzes the oxidation of arachidonic acid to leukotrienes A4 (LTA4), which is released by inflammatory cells at injured sites and thus plays an important role in atherosclerosis and other vascular damage [54]. Three SNPs (SG13S114A/T, SG13S89A/G and SG13S32A/C) of ALOX5AP with a relationship to IS incidence have been extensively studied in Asians and Caucasians; however, conflicting results have been reported across different ethnic backgrounds [55]. Recent meta-analysis of the previous studies indicated SG13S114T as a risk factor of IS in the Caucasian population but a protective factor of LAA stroke in the Chinese population [55, 56],
The PDE4D gene encodes a cAMP-specific 3’, 5’-cyclic phosphodiesterase 4D that can degrade the signal transduction molecule cAMP in multiple cell types, including vascular cells [57]. Linkage-association studies identified an association of SNP 83 and AC008818-1 polymorphism in the PDE4D gene with carotid stroke in an Icelandic population [47]. But these results have not been completely replicated in other ethnicities. Recent meta-analysis concluded that the allele 0 of AC008818-1 was associated with only 1.12-fold risk increase of IS in Caucasians [58] and SNP83 polymorphism was associated with 1.69-fold risk increase of CE subtype of IS under the dominant model [59].
Genetic Markers Discovered by GWAS
Earlier studies utilizing GWAS focused on genetic variants identified in other cardiovascular diseases. Variants in two genes associated with atrial fibrillation (AF), PITX2 and ZFHX3, were found to be independent risk factors for IS. Further analysis determined that these associations were specific to CE stroke, which was confirmed by several subsequent studies [60-65]. Furthermore, a genetic variant in CDKN2A/CDKN2B located at chromosome 9p21 that has a widely reported association with myocardial infarction and coronary artery disease [66] was associated with LAA stroke across multiple cohorts [62, 67, 68].
The first widely replicated novel genetic association with IS subtype was the 7q21 locus near histone deacetylase 9 (HDAC9), which was found to be associated especially with the LAA stroke (OR 1.42) [60]. This association was replicated in the same study in additional patients from Europe, America and Australia and in subsequent studies [62, 65]. Further GWAS analyses have reported that the genetic variants located at chromosome 6p21.1 and MMP12 and TSPAN2 were associated with LAA stroke, but not other subtypes of stroke among white populations [62, 65, 69, 70]. A genetic variant near ABCC1 was associated with CG-type IS in European and African populations [62].
Several GWAS studies performed in Asian populations identified an association of CELSR1 with IS in Japanese and the LAA subtype of IS in the Chinese Han population [71, 72]. A GWAS conducted in a group of Japanese IS patients identified a genetic variant in PRKCH especially associated with SVD stroke [73]. This result was replicated in both an independent Japanese cohort and a Chinese population [73, 74]. Additionally, a genetic variant at chromosome 14q13.3 nearing PTCSC3 was also reported to be associated with LAA stroke in the Chinese Han population [75]. A SNP locus (rs505922) in ABO gene increases the risk of LAA, CE and overall IS in Europeans while another locus in the same gene seems to be a protective factor for LAA stroke in the Chinese Han population [76, 77].
The first genetic variants to be associated with all types of IS were reported to lie in the chromosome 12p 13 region near NINJ2 [78]. However, this association has not been replicated in large case–control meta-analyses [61, 62 65, 79]. The first association of a genetic variant with all types of IS to be replicated convincingly was reported with a locus at 12q24 near ALDH2 [62, 80]. Recently, a novel variant near FOXF2 was also found to be associated with overall stroke [81]
Genetic Markers Based on Imaging Phenotypes
White matter hyperintensities (WMH)
WMH on T2-weighted MRI are reported to be a risk factor and predictor of lacunar stroke in prospective community populations [82, 83]. Severe confluent WMH are often found in patients with the SVD stroke subtype [84]. Twin and family history studies suggest a heritability of 55% to 80% to WMH [85] and common SNPs contribute between 13% and 45% to the heritability [86]. Previous candidate gene studies investigated variants in 19 genes and found associations between WMH extent and polymorphisms in APOE, MTHFR, ACE and ATG genes [87]. In 2011, the CHARGE Consortium performed the first GWAS on WMH in the general population and identified 6 SNPs mapping to a locus on chromosome 17q25 [88]. A recent meta-analysis of community populations and of stroke patients indicated 6 common genetic polymorphism loci that were associated with WMH at the genome-wide level: rs72934505 (NBEAL1); rs941898 (EVL); rs962888 (C1QL1); rs9515201 (COL4A2); rs7214628 (TRIM65); rs78857879 (EFEMP1) [89].
Carotid Intima-Media Thickness (CIMT)
The CIMT based on high-resolution carotid duplex ultrasound is a surrogate marker of subclinical atherosclerosis and a strong predictor of stroke [90]. Family studies have demonstrated IMT heritability ranging from 30% to 65% [91]. A larger meta-analysis conducted by the CHARGE collaborators in over 31,000 European subjects has identified three novel carotid IMT associations that reach genome-wide significance (p < 1×10−8): rs11781551 (ZHX2); rs445925 (APOC1, APOE, APOC2, APOC4); rs6601530 (PINX1) [92]. The first large-scale GWAS of carotid IMT in a non-European population found one suggestive association (p = 2.3×10−7): rs17356664 (19q13, EXOC3L2, MARK4) [93]. While the first GWAS in an Asian population identified two novel loci that were significantly associated with CIMT at the genomic level (p < 1×10−7): rs36071027 (EBF1) and rs975809 ( PCDH15) [94].
MicroRNA Markers for IS
miRNAs are small endogenously expressed noncoding RNAs approximately 22 nucleotides in length that regulate gene expression, mainly at the post-transcriptional level. The human genome encodes over 1000 miRNAs, each of which may regulate the level of hundreds of mRNAs [95]. Therefore, miRNAs are likely to be involved in many biologic and pathogenic processes [96-98]. The discovery of circulating miRNAs in peripheral blood, which are unexpectedly stable [99], has allowed the recent completion of several studies in human stroke patients that have confirmed the differential expression of specific miRNAs following stroke and have addressed their potential use as diagnostic and prognostic markers.
MiRNA Profiling
The miRNA profiling studies have been conducted in succession in whole blood [100, 101], plasma [102-104], serum [105], peripheral blood cell [106] and cerebral spinal fluid [107] samples. There are substantial differences in miRNA expressions in young IS patients [101], IS with lower stroke risk [103], IS with post-stroke depression [104] or patients with other neurological diseases [107] compared with normal healthy people or control IS patients. Differences also exist between pre-stroke samples and post-stroke samples within various times after onset [100] (Table 3). Considering the different time periods examined in reference to injury and recovery and different sample types, these studies have led to little overlap in terms of consistency of finding.
Candidate MiRNA Markers
Besides miRNA profiling, several miRNAs documented to involve in apoptosis, excitotoxic neuronal death, inflammation, neurogenesis and angiogenesis have showed significant dysregulation in IS patients. (Table 3).
Apoptosis
Apoptosis is the main cause of neuronal death in the penumbra region following acute brain infarction [2, 108-110]. In addition, the apoptosis of endothelial cells caused by hyperlipoidemia, hyperhomo-cystinemia and hypertension could increase risk of stroke especially for LAA stroke. Increasing evidences support the involvement of miRNAs in the regulation of these apoptotic processes before and after IS [111].
MiR-16 belongs to cluster miR-15/miR-16 that has been well documented as an apoptosis-related miRNA [112]. It has been identified as tumor-suppressor gene and downregulation of miR-16 contributes to cancer development [113]. In contrast, miR-16 is increased in the serum of patients with critical limb ischemia and stroke [114]. The serum expression levels of miR-15a and miR-16 is found to be elevated 8.3- and 42-fold respectively in IS patients compared to controls. Diagnostic efficiency analysis by Receiver operating characteristic (ROC) curves revealed areas under the curves (AUCs) of 0.698, and 0.82 for these two miRNAs respectively [115]. Moreover, plasma miR-16 concentration in IS patients is higher than that in hemorrhagic stroke (HS) patients. The odds ratio (OR) for discriminating HS and IS with miR-16 is 9.75, suggesting miR-16 be a good discrimination markers between HS and IS [116].
MiR-21 is initially reported to have a protective role in ischemia reperfusion-induced cardiocyte apoptosis via inhibiting the phosphatase and tensin homolog (PTEN)/Akt-dependent pathway [117]. Plasma miR-21 and miR-24 are significantly lower in acute cerebral infarction (ACI) patients within 24 hours onset than in the controls, and a negative correlation is revealed between miR-21, miR-24 and the National Institutes of Health Scales Score (NIHSS). Therefore plasma miR-21 and miR-24 are suggested as potential early stage markers of acute cerebral infarction [118]. Moreover, the data of N2A neuroblastoma cells following oxygen glucose deprivation (OGD) and reoxygenation indicates that miR-21 may have an anti-apoptotic effect, while miR-24 may have a pro-apoptotic effect [118].
MiR-29b belongs to the miR-29 family that is reported to target both pro- and anti-apoptotic BCL-2 family members [111]. It is markedly induced during neuronal maturation and functioned as a strong inhibitor of neuronal apoptosis through targeting BH3-only genes [119]. The levels of miR-29b from the infarct site and blood are both decreased in middle cerebral artery occlusion (MCAO) mice. Overexpression of miR-29b by gene transfer to mice brains reduces infarct volume, edema, and blood–brain barrier (BBB) disruption via downregulating aquaporin-4 (AQP-4) [120]. Clinical studies show that the level of miR-29b in white blood cells in stroke patients within 72 hours after stroke onset is significantly lower compared with the controls, and is negatively associated with NIHSS scores and brain infarct volume, suggesting miR-29b as a circulating biomarker to predict stroke outcomes [120].
Inflammation
The inflammatory response is not only an important etiological factor in IS, but also plays a pivotal role in early ischemia-hypoxia injuries and long-term neuronal recovery. It has been reported that inhibition of inflammatory cells reduces ischemic brain injury [121], but also increases the risk of diminished neurological outcome and death [122]. Studies on IS patients have disclosed significant changes of several miRNAs targeting inflammation factors: Let-7, miRNA-124, −145, −181.
Let-7 is a miRNA family containing 12 members in humans, which are highly abundant regulators of gene expression in the CNS [123]. Extracellular let-7 activates the RNA-sensing Toll-like receptor (TLR) and induces neurodegeneration [124]. Clinical studies show that the circulating let-7b is at a higher level in IS patients than healthy volunteers up through 24 weeks and the AUC of let-7b at 24 h, 1, 4 and 24w is 0.93, 0.92, 0.92 and 0.91 respectively [125]. Serum let-7e is also significantly increased in IS patients within 24 h onset and is positively associated with serum C reactive protein (CRP) levels. It has been demonstrated that let-7e had a specificity up to 73.4 % and a sensitivity of 82.8 % for IS diagnosis at the acute stage [126]. In contrast, the content of let-7c-5p is significantly reduced in the plasma of IS patients and the ipsilateral cortex and striatum of MCAO mice at 24 hours reperfusion, which concurs with increased activation of microglia [127]. While another member of let-7 family, let-7f, is reported to be significantly down-regulated in sera of massive cerebral infarction without hemorrhagic transformation (HT) patients within 48 hours of a stroke compared with controls, leading to an increased serum IL-6 level [128]. Additionally, let-7i is reported to be decreased in circulating leukocytes of patients with acute IS within 72 hours and to be associated with increased expression of its mRNA targets including CD86, CXCL8, HMGB1, indicating its involvement in leukocyte activation, recruitment and proliferation [129].
MiR-124 is the most abundant miRNA of the CNS and is commonly recognized as a modulator on polarization of activated microglia and infiltrating macrophages towards the anti-inflammatory M2 phenotype during focal cerebral ischemia [130]. The serum level of miR-124 in IS patients is significantly decreased within 24 h after IS onset, which is negatively correlated with infarct volume, plasma high-sensitivity C-reactive protein (hs-CRP) and MMP-9 levels [131]. The miR-124 expression is also found in atheromatous plaque of acute IS patients compared with intact tissue [132]. It seems that miR-124 is suppressed in acute IS thus facilitating inflammation and brain injury.
MiR-145 is recognized as a marker and modulator of vascular smooth muscle cell phenotype [133]. Serum miR-145 is significantly upregulated within 24h after stroke onset, which is strongly and positively correlated with hs-CRP [134, 135]. It is also found to be significantly overexpressed in symptomatic versus asymptomatic human carotid plaques [136]. The antagomir to miR-145 has been found to be neuroprotective in vivo [137], indicating the potential use of miR-145 as a candidate biomarker or therapeutic target for stroke. In contrast, serum miR-221 is significant decreased in IS patients at 1 day and 7 days after stroke onset and is negatively associated with hs-CRP. However, it is found to be significantly overexpressed in symptomatic versus asymptomatic human carotid plaques [135,136,138].
Excitotoxicity
Excitotoxicity is an important trigger of neuronal damage in early-stage of cerebral ischemia. It stems from excessive accumulation of excitatory amino acids such as glutamate, which leads to toxic increases in intracellular calcium and zinc [139, 140]. Recent data indicated that specific miRNAs such as miRNA-223, −107, −125b regulate glutamate neurotransmission and excitotoxicity during stroke [141].
miR-223 expression is increased in circulating blood samples of patients with acute IS, and the severity and volume of infarct is lesser in patients who had higher expression of miR-223 [142]. Overexpression of miR-223 is found to attenuate neuronal loss after excitotoxic insult by lowering the level of glutamate receptor subunits GluR2 and NMDA receptor 2B (NR2B) in brain [143]. However, several studies have shown that AMPAR lacking GluR2 subunit increases neuronal vulnerability to excitotoxicity [144, 145]. In addition, the circulating levels of miR-107 and miR-128b is increased 2.78-fold and 2.13-fold respectively in IS patients in comparison to the healthy volunteers and positively correlated with the severity of stroke as defined by NIHSS classes. The AUC for circulating miR-107 and miR-128b is 0.97 and 0.903 respectively [146]. At the same time, the elevation of both miR-107 and glutamate in IS patients are accounted at least partially by suppression of GLT-1 expression [147].
On the other hand, miR-125b exhibits maximum expression levels within the acute phase of stroke in humans [104, 148]. Together with evidence that the NR2A expression in hippocampal neurons is negatively regulated through its 3’UTR by FMRP, miR-125b and Argonaute 1 [149], it can be inferred that overexpression of miR-125b after IS can downregulate the level of NR2ARs, reduce post-stroke excitotoxicity and protect cell from death.
Neurogenesis and angiogenesis
Angiogenesis and neurogenesis are crucial processes for brain tissue repair and remodeling after brain injury. MiR-9 has a key effect on differentiation of oligodendrocyte progenitor cells and myelinogenesis [150]. There is apparently disordered expression of miR-9 in patients with Alzheimer’s disease or Huntington’s disease [151]. miRNA-9 is down-regulated in OGD neurons and MCAO mice brain and application of miR-9 agomir can restore the neurological scores and reduce infarct volume, brain water content, and behavioral impairments by promoting the repair of myelin sheath and suppressing neuronal apoptosis [152]. Serum level of miR-9 is reported to be negatively correlated with blood hsCRP, MMP-9 levels, infarct volume and NIHSS scores in acute IS patients with lesion volume > 4 cm3 indicating that the protective role of miR-9 is also associated with inhibition of neuroinflammation [131].
MiR-210 is a pleiotropic hypoxiamir activated by hypoxia inducible factor-alpha (HIF-α) for hypoxic induction [153]. It is the only miRNA that is up-regulated under hypoxia in several cell types in vitro [152]. Intracerebral injection of miR-210 could substantively promote endothelial cell proliferation and new microvessel formation, and increase the number of neural progenitor cells in the subventricular zone of normal adult mouse brain via the vascular endothelial growth factor (VEGF) pathway [155]. Furthermore, miR-210 is significantly up-regulated in the adult rat ischemic cerebral cortex, leading to enhanced Notchl signaling [156]. miR-210 in human atherosclerotic plaques is four-fold higher than that in control arteries [157]. However, acute IS patients show significantly decreased blood miR-210 level at 4 days and 7 days of stroke onset compared to healthy controls and patients with higher circulating blood miR-210 display better clinical outcomes, indicating blood miR-210 is a sensitive biomarker for clinical prognosis in acute cerebral ischemia [158, 159].
MiR-126 has been found to be specifically and highly expressed in human endothelial cells and enhances the proangiogenic actions of VEGF and FGF and promotes blood vessel formation by repressing the expression of Spred-1, an intracellular inhibitor of angiogenic signaling [158]. Targeted deletion of miR-126 in mice displays leaky vessels, hemorrhage and defective cardiac neovascularization following myocardial infarction [160]. A miRNA profiling study discloses that miR-126 is down-regulated in young stroke patients. The AUC of plasma miR-126 at 24h, 1, 4 and 24w is 0.92, 0.94, 93 and 0.92 respectively after symptoms onset [125].
Conclusion
Overall, the current evidences support that genetic factor is an important etiology of IS. Two genes, TREX1 and MTLL1, can serves as markers for monogenic IS due to high IS incidence in mutation carriers and have potential usefulness for IS prediction in people under 45 year old. For polygenic IS, the use of GWAS techniques has identified a large number of markers. Four loci—PITX2, ZFHX3, HDAC9, and 12q24.12—have been repeatedly identified to exceed genome-scale significance (p < 5.0x10−8) in residents with European background. However, findings of several prospective studies indicate that improvements in prediction ability by genetic risk score (GRS) are limited [160, 161]. It seems that polygenic IS is associated with excessive susceptible genes, each only exerting a minor effect, leading to its vulnerability to environmental, nutritional, and other non-genetic factors.
Considering the high heredity of polygenic IS and the present low predicting ability of GRS, novel genetic markers still need to be explored by using improved method. One possible improvement is to focus on rare genetic variations by using next generation gene sequencing technique. Based on the present finding that indicates “different subtypes, different variants”, refined clinical subtyping is also necessary. A recent study [162] showed significant enrichment in low-frequency variants (allele frequency < 5%) for both LAA and SVD, and an enrichment of higher frequency variants (allele frequency 10% and 30%) for CE (all p < 10−5). Larger IS samples and more detailed stratification sampling are also needed to exclude the interference of non-inherited elements and to allow for well-powered studies that link genotype to phenotype. Prediction models may have to be changed to personalized medicine due to the complex nature of polygenic IS. The possible impact of environment on epigenetic regulation needs to be validated in large scale human studies to help understand the effects of extrinsic factors on IS over time and across generations.
Table 2.
Genetic markers for polygenic cause of ischemic stroke by Genome-Wide Associated Studies and meta-analysis
| Genes | Chr | Marker Loci |
Minor Alleles |
Subtypes | Ethnic | Case (n) |
OR | P value | Gene Product | Gene Functions | Shared diseases |
Refs |
|---|---|---|---|---|---|---|---|---|---|---|---|---|
| PITX2 | 4q25 | rs2200733 | T | CE | Europe | 1498 | 1.52 | 5.8×10−12 | Paired like | Involving in left-right | AF | [60-63] |
| T | EuropeR | 2322 | 1.32 | 5.1×10−8 | homeodomain 2 | asymmetry during development. | ||||||
| T | SiGNa | 7418 | 1.36 | 8.1×10−30 | ||||||||
| rs1906591 | A | Europe | 1376 | 1.48 | 1.2×10−7 | |||||||
| HDAC9 | 7p21.1 | rs11984041 | A | LAA | Europe | 1780 | 1.42 | 1.9×10−11 | Histone | Involving in heart and muscle | CAD, | [61, 62, 65] |
| A | SiGNa | 4595 | 1.23 | 4.5×10−8 | deacetylase 9 | tissue development. | ICA | |||||
| rs2107595 | A | Metastroke | 2167 | 1.39 | 2.0×10−16 | |||||||
| TSPAN2 | 1p13.2 | rs12122341 | G | LAA | SiGNa | 4964 | 1.19 | 1.3×10−9 | Tetraspanin 2 | Regulating cell development, activation, growth and motility. | Migraine , lung cancer | [62] |
| ABCC1 | 16p13.1 | rs74475935 | G | CIS | SiGNa | 5861 | 4.63 | 4.7×10−11 | ATP-binding cassette C1 | Functioning as an organic anion transporter | Cancer | [62] |
| ZFHX3 | 16q22.3 | rs7193343 | T | CE | Europe | 1454 | 1.22 | 2.1×10−4 | Zinc finger | Regulating myogenic and | AF | [62, 64, 65] |
| T | SiGNa | 7418 | 1.16 | 8.9×10−9 | homeobox protein | neuronal differentiation. | ||||||
| rs879324 | A | Metastroke | 2365 | 1.25 | 1.5×10−7 | |||||||
| CDKN2 | 9p21.3 | rs23832073 | G | LAA | Europe | 961 | 1.16 | 8.3×10−3 | Cyclin-dependent | Regulating CDK4 and p53 in | CAD, | [62, 67, 68] |
| A/B | G | SiGNa | 2346 | 1.09 | 8.1×10−3 | kinase inhibitor | cell cycle G1 progression | MI, ICA | ||||
| rs2383206 | G | Chineseb | 122 | 2.09 | 2.0×10−3 | |||||||
| 6p21 | 6p21.1 | rs556621 | T | LAA | Europe | 2136 | 1.21 | 4.7×10−8 | ||||
| T | Metastroke | 4601 | 1.03 | 5.3×10−5 | Cancer | [62, 65, 69] | ||||||
| T | SiGNa | 2346 | 1.11 | 2.6×10−3 | ||||||||
| MMP12 | 11q22.2 | rs660599 | T | LAA | Europe | 6778 | 1.18 | 2.6×10−8 | Matrix metallo-proteinase 12 | Involving in the breakdown of extracellular matrix | CAD, cancer | [70] |
| CELSR1 | 22ql3.3 | rs6007897 | G | IS | Japanese | 750 | 1.85 | 4.7×l0−4 | Cadherin EGF | Involving in contact- | Spina | [71, 72] |
| 1 | rs4044210 | G G | LAA IS | Chinese Japanese | 422 748 | 2.84 1.78 | 2.0×10−3 1.0×10−3 | LAG sevenpass G-type receptor 1 | mediated communication in early embryo genesis | bifida | ||
| LAA | Chinese | 422 | 2.93 | 1.0×10−3 | ||||||||
| PRKCH | 14q23.1 | rs2230500 | A | SVD | Japanese | 1628 | 1.40 | 5.1×l0−7 | protein kinase Cη | Involving in vascular | HS | [73, 74] |
| A | IS | Chinese | 873 | 1.31 | 5.8×l0−3 | endothelium injury | ||||||
| PTCSC3 | 14q13.3 | rs934075 | G | LAA | Chinese- | 444 | 1.41 | 4.0×l0−9 | Papillary thyroid | Noncoding RNA involving in | Thyroid | [75] |
| rs2415317 | C | Han | 1.39 | 3.1×l0−8 | carcinoma susceptibility | carcinoma development through modifying relative | tumors | |||||
| rs1952706 | T | 1.37 | 2.9×l0−8 | candidate 3 | gene expression. | |||||||
| ABO | 9q34.2 | rs505922 | c | LAA | Europe | 2113 | 1.23 | 1.0×10−3 | Glycosyltransferas | Converting H antigen into A | CAD, | [76, 77] |
| C | CES | Europe | 2326 | 1.13 | 2.0×l0−4 | es | or B antigen of blood group. | MI, VTE | ||||
| rs532436 | A | IS | Europe | 16820 | 1.09 | 4.3×l0−8 | ||||||
| rs2073824 | A | LAA | Chinese | 644 | 0.61 | 1.7×l0−4 | ||||||
| NINJ2 | 12p13.3 | rs12425791 | A | IS | Europe | 1161 | 1.29 | 1.1×10−9 | Nerve injury | Promoting neurite outgrowth | Prostate | [61, 62, 70, |
| 3 | A | Asianb | 8626 | 1.08 | 2.5×10−2 | induced protein 2 | and nerve regeneration after nerve injury. | cancer | 78, 79] | |||
| rs11833579 | A | EuropeR | 9407 | 1.00 | 0.98 | |||||||
| A | Metastroke | 12389 | 1.00 | 0.81 | ||||||||
| A | SiGNa | 16851 | 1.02 | 0.22 | ||||||||
| ALDH2 | 12q24.1 | rs10744777 | T | IS | Europe | 15448 | 1.10 | 1.2×10−9 | Aldehyde | Involving in alcohol | CAD, | [62, 80] |
| 2 | SiGNa | 16851 | 1.07 | 4.2×10−9 | dehydrogenase 2 | metabolism | MI, HT | |||||
| FOXF2 | 6p25 | rs12204590 | A | Stroke | SiGNa | 19816 | 1.08 | 1.48×10−8 | Forkhead box F2 | ARS | [81] |
cases including European, African and Americas using GWAS;
meta-analysis only;
replication study in population of Europen ancestry.
SiGN:Stroke Genetics Network, stroke cases were from American, European and African; CAD:coronary artery disease; MI: myocardial infarction; VTE:venous thromboembolism; CSA: coronary spastic angina; HT:hypertension; LAA:large artery atherosclerosis; SVD:cerebral small vessel disease; CIS:cryptogenic ischemia stroke; ICA:intracranial aneurysm; CES:cardioembolic stroke.
Acknowledgments
This work was supported by grants from the Bill and Melinda Gates Foundation (to X. W.), Grant/Award Number: 01075000191 and OPP1099070; the Brigham and Women’s Hospital BRI Fund to Sustain Research Excellence; the National Institutes of Health/National Institute of Neurological Disorders and Stroke (to X.W.), Grant/Award Number: NS055072 and NS051756; National Natural Science Foundation of China (to Z. Z.), Grant Number:81344438.
Footnotes
Conflicts of interest
The authors declare that this work contains no potential conflicts in terms of commercial interests.
References
- 1.Feigin VL, Norrving B, Mensah GA (2017) Global burden of stroke. Circ Res 120:439–448 [DOI] [PubMed] [Google Scholar]
- 2.Pandya RS, Mao L, Zhou H et al. (2011) Central nervous system agents for ischemic stroke: neuroprotection mechanisms. Cent Nerv Syst Agents Med Chem 11:81–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Go AS, Mozaffarian D, Roger VL et al. (2014) Heart disease and stroke statistics-2014 update a report from the American Heart Association. Circulation 131:29–322. [DOI] [PubMed] [Google Scholar]
- 4.Lorenz MW, Lauer A, Foerch C (2015) Quantifying the benefit of prehospital rapid treatment in acute stroke: benchmark for future innovative Clinical Trials. Stroke 46:3168–3176. [DOI] [PubMed] [Google Scholar]
- 5.Chen P, Goldberg DE, Kolb B et al. (2002) Inosine induces axonal rewiring and improves behavioral outcome after stroke. Proc Natl Acad Sci U S A 99:9031–9036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Zai L et al. (2011) Inosine augments the effects of a Nogo receptor blocker and of environmental enrichment to restore skilled forelimb use after stroke. J Neurosci 31:5977–5988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Zhou H, Wang J, Jiang J et al. (2014) N-acetyl-serotonin offers neuroprotection through inhibiting mitochondrial death pathways and autophagic activation in experimental models of ischemic injury. J Neurosci 34:2967–2978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Wang X, Figueroa BE, Stavrovskaya IG et al. (2009) Methazolamide and melatonin inhibit mitochondrial cytochrome C release and are neuroprotective in experimental models of ischemic injury. Stroke 40:1877–1885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Müller B, Wilcke A, Boulesteix AL et al. (2016) Improved prediction of complex diseases by common genetic markers: state of the art and further perspectives. Hum Genet 135: 259–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Adams HP, Bendixen BH, Kappelle LJ et al. (1993) Classification of subtype of acute ischemic stroke. Definitions for use in a multicenter clinical trial. TOAST. Trial of Org 10172 in Acute Stroke Treatment. Stroke 24:35–41. [DOI] [PubMed] [Google Scholar]
- 11.Gulli G, Rutten-Jacobs LC, Kalra L et al. (2016) Differences in the distribution of stroke subtypes in a UK black stroke population-final results from the South London Ethnicity and Stroke Study. BMC Med 14:77. doi: 10.1186/s12916-016-0618-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Flossmann E, Schulz UG, Rothwell PM (2004) Systematic review of methods and results of studies of the genetic epidemiology of ischemic stroke. Stroke 35:212–227. [DOI] [PubMed] [Google Scholar]
- 13.Bevan S, Traylor M, Adibsamii P et al. (2012) Genetic heritability of ischemic stroke and the contribution of previously reported candidate gene and genomewide associations. Stroke 43:3161–3167. [DOI] [PubMed] [Google Scholar]
- 14.Bersano A, Markus HS, Quaglini S et al. (2016) Clinical pregenetic screening for stroke monogenic diseases: results from Lombardia GENS Registry. Stroke 47:1702–1709. [DOI] [PubMed] [Google Scholar]
- 15.Feil R, Fraga MF (2012) Epigenetics and the environment: emerging patterns and implications. Nat Rev Genet 13:97–109. [DOI] [PubMed] [Google Scholar]
- 16.Fox CS, Polak JF, Chazaro I et al. (2003) Genetic and environmental contributions to atherosclerosis phenotypes in men and women: heritability of carotid intima-media thickness in the Framingham Heart Study. Stroke 34:397–401. [DOI] [PubMed] [Google Scholar]
- 17.Tan RYY, Markus HS (2015) Monogenic causes of stroke: now and the future. J Neurol 262: 2601–2616. [DOI] [PubMed] [Google Scholar]
- 18.Lindgren A (2014) Stroke genetics: a review and update. J Stroke 16:114–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Opherk C, Peters N, Holtmannspötter M et al. (2006) Heritability of MRI lesion volume in CADASIL: evidence for genetic modifiers. Stroke 37:2684–2689. [DOI] [PubMed] [Google Scholar]
- 20.Tikka S, Baumann M, Siitonen M et al. (2014) CADASIL and CARASIL. Brain Pathol 24: 525–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Kilarski LL, Ruttenjacobs LC, Bevan S et al. (2015) Prevalence of CADASIL and Fabry disease in a cohort of MRI defined younger onset lacunar stroke. PLoS One 10:21–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ince B, Benbir G, Siva A et al. (2014) Clinical and radiological features in CADASIL and NOTCH3-negative patients: a multicenter study from Turkey. Eur Neurol 72:125–131. [DOI] [PubMed] [Google Scholar]
- 23.Liu X, Zuo Y, Sun W et al. (2015) The genetic spectrum and the evaluation of CADASIL screening scale in Chinese patients with NOTCH3 mutations. J Neurol Sci 354:63–69. [DOI] [PubMed] [Google Scholar]
- 24.Hara K, Shiga A, Fukutake T et al. (2009) Association of HTRA1 mutations and familial ischemic cerebral small-vessel disease. New Engl J Med 360:1729–1739. [DOI] [PubMed] [Google Scholar]
- 25.Nozaki H, Nishizawa M, Onodera O (2014) Features of cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy. Stroke 45:3447–3453. [DOI] [PubMed] [Google Scholar]
- 26.Richards A, Am VDM, Jen JC et al. (2007) C-terminal truncations in human 3'-5' DNA exonuclease TREX1 cause autosomal dominant retinal vasculopathy with cerebral leukodystrophy. Nat Genet 39:1068–1070. [DOI] [PubMed] [Google Scholar]
- 27.Kolar GR, Kothari PH, Khanlou N et al. (2014) Neuropathology and genetics of cerebroretinal vasculopathies. Brain Pathol 24:510–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Stam AH, Kothari PH, Shaikh A et al. (2016) Retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations. Brain 139:2909–2922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Alamowitch S, Plaisier E, Favrole P et al. (2009) Cerebrovascular disease related to COL4A1 mutations in HANAC syndrome. Neurology 73:1873–1882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Sibon I, Coupry I, Menegon P et al. (2007) COL4A1, mutation in Axenfeld–Rieger anomaly with leukoencephalopathy and stroke. Ann Neurol 62:177–184. [DOI] [PubMed] [Google Scholar]
- 31.Lanfranconi S, Markus HS (2010) COL4A1 mutations as a monogenic cause of cerebral small vessel disease: a systematic review. Stroke 41:513–518. [DOI] [PubMed] [Google Scholar]
- 32.Peters FP, Vermeulen A, Kho TL (2001) Anderson-fabry's disease: α galactosidase deficiency. Lancet 357:138–140. [DOI] [PubMed] [Google Scholar]
- 33.Moore DF, Kaneski CR, Askari H et al. (2007) The cerebral vasculopathy of Fabry disease. J Neurol Sci 257:258–263. [DOI] [PubMed] [Google Scholar]
- 34.Sims K, Politei J, Banikazemi M et al. (2009) Stroke in Fabry disease frequently occurs before diagnosis and in the absence of other clinical events: natural history data from the Fabry Registry. Stroke 40:788–794. [DOI] [PubMed] [Google Scholar]
- 35.Ashley-Koch A, Yang Q, Olney RS (2000) Sickle hemoglobin (HbS) allele and sickle cell disease: A HuGE Review. Am J Epidemiol 151:839–845. [DOI] [PubMed] [Google Scholar]
- 36.Ohenefrempong K, Weiner SJ, Sleeper LA et al. (1998) Cerebrovascular accidents in sickle cell disease: rates and risk factors. Blood 91:288–294. [PubMed] [Google Scholar]
- 37.Handford PA (2000) Fibrillin-1, a calcium binding protein of extracellular matrix. Biochim Biophys Acta 1498:84–90. [DOI] [PubMed] [Google Scholar]
- 38.Dietz HC (1991) Marfan syndrome caused by a recurrent de novo missense mutation, in the fibrillin gene. Nature 352:337–339. [DOI] [PubMed] [Google Scholar]
- 39.Wityk RJ, Zanferrari C, Oppenheimer S (2002) Neurovascular complications of marfan syndrome: a retrospective, hospital-based study. Stroke 33:680–684. [DOI] [PubMed] [Google Scholar]
- 40.Testai FD, Gorelick PB (2010) Inherited metabolic disorders and stroke part 2: homocystinuria, organic acidurias, and urea cycle disorders. Arch Neurol 67:148–153. [DOI] [PubMed] [Google Scholar]
- 41.Kraus J P, Janosík M, Kozich V et al. (1999) Cystathionine beta-synthase mutations in homocystinuria. Hum Mutat 13:362–375. [DOI] [PubMed] [Google Scholar]
- 42.Alehan F, Saygi S, Gedik S et al. (2010) Stroke in early childhood due to homocystinuria. Pediatr Neurol 43:294–296. [DOI] [PubMed] [Google Scholar]
- 43.Shen MH, Harper PS, Upadhyaya M (1996) Molecular genetics of neurofibromatosis type 1 (NF1). J Med Genet 33:2–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ballabio E, Bersano A, Bresolin N et al. (2007) Monogenic vessel diseases related to ischemic stroke: a clinical approach. J Cereb Blood Flow Metab 27:1649–1662. [DOI] [PubMed] [Google Scholar]
- 45.Créange A, Zeller J, Rostaing-Rigattieri S et al. (1999) Neurological complications of neurofibromatosis type 1 in adulthood. Brain 122 :473–481. [DOI] [PubMed] [Google Scholar]
- 46.Terry AR, Jordan JT, Schwamm L et al. (2016) Increased risk of cerebrovascular disease among patients with neurofibromatosis type 1: population-based approach. Stroke 47:60–65. [DOI] [PubMed] [Google Scholar]
- 47.Sakuta R, Goto Y, Horai S et al. (1993) Mitochondrial DNA mutations at nucleotide positions 3243 and 3271 in mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes: a comparative study. J Neurol Sci 115:158–160. [DOI] [PubMed] [Google Scholar]
- 48.El-Hattab AW, Adesina AM, Jones J et al. (2015) MELAS syndrome: Clinical manifestations, pathogenesis, and treatment options. Mol Genet Metab 116:4–12. [DOI] [PubMed] [Google Scholar]
- 49.Li R, Xiao HF, Lyu JH et al. (2016) Differential diagnosis of mitochondrial encephalopathy with lactic acidosis and stroke-like episodes (MELAS) and ischemic stroke using 3D pseudocontinuous arterial spin labeling. J Magn Resone Imaging 45:199–2064. [DOI] [PubMed] [Google Scholar]
- 50.Falcone GJ, Malik R, Dichgans M et al. (2014) Current concepts and clinical applications of stroke genetics. Lancet Neurol 13:405–418. [DOI] [PubMed] [Google Scholar]
- 51.Maasz A, Melegh B (2010) Three periods of one and a half decade of ischemic stroke susceptibility gene research: lessons we have learned. Genome Med. 10.1186/gm185. [DOI] [PMC free article] [PubMed]
- 52.Helgadottir A, Manolescu A, Thorleifsson G et al. (2004) The gene encoding 5-lipoxygenase activating protein confers risk of myocardial infarction and stroke. Nat Genet 36:233–239. [DOI] [PubMed] [Google Scholar]
- 53.Gretarsdottir S, Thorleifsson G, Reynisdottir ST et al. (2003) The gene encoding phosphodiesterase 4D confers risk of ischemic stroke. Nat Genet 35:131–138. [DOI] [PubMed] [Google Scholar]
- 54.Dixon RA, Diehl RE, Opas E et al. (1990) Requirement of a 5-lipoxygenase-activating protein for leukotriene synthesis. Nature 343:282–284. [DOI] [PubMed] [Google Scholar]
- 55.Zintzaras E, Rodopoulou P, Sakellaridis N (2009) Variants of the arachidonate 5-lipoxygenase-activating protein (ALOX5AP) gene and risk of stroke: a HuGE gene-disease association review and meta-analysis. Am J Epidemiol 169:523–532. [DOI] [PubMed] [Google Scholar]
- 56.Zhang R, Guo X, Li X et al. (2014) Arachidonate 5-lipoxygenase-activating protein (ALOX5AP) gene rs4073259 polymorphism not associated with ischemic stroke in the northeastern Chinese Han population. Clin Neurol Neurosur 119:64–69. [DOI] [PubMed] [Google Scholar]
- 57.Rampersad SN, Ovens JD, Huston E et al. (2010) Cyclic AMP phosphodiesterase 4D (PDE4D) tethers EPAC1 in a vascular endothelial cadherin (VE-Cad)-based signaling complex and controls cAMP-mediated vascular permeability. J Biol Chem 285:33614–33622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Bevan S, Dichgans M, Gschwendtner A et al. (2008) Variation in the PDE4D gene and ischemic stroke risk: a systematic review and meta-analysis on 5200 cases and 6600 controls. Stroke 39:1966–1971. [DOI] [PubMed] [Google Scholar]
- 59.Liu X, Zhu R, Li L et al. (2013) Genetic polymorphism in PDE4D gene and risk of ischemic stroke in Chinese population: a meta-analysis. PLoS One 8:2344–2348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Gretarsdottir S, Thorleifsson G, Manolescu A et al. (2008) Risk variants for atrial fibrillation on chromosome 4q25 associate with ischemic stroke. Ann Neurol 64:402–409. [DOI] [PubMed] [Google Scholar]
- 61.Bellenguez C, Bevan S, Gschwendtner A et al. (2012) Genome-wide association study identifies a variant in HDAC9 associated with large vessel ischemic stroke. Nat Genet 44:328–333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Neurol L (2016) Loci associated with ischaemic stroke and its subtypes (SiGN): a genome-wide association study. Lancet Neurol 15:174–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Lemmens R, Buysschaert I, Geelen V et al. (2010) The association of the 4q25 susceptibility variant for atrial fibrillation with stroke is limited to stroke of cardioembolic etiology. Stroke 41:54–61. [DOI] [PubMed] [Google Scholar]
- 64.Gudbjartsson DF, Holm H, Gretarsdottir S et al. (2009) A sequence variant in ZFHX3 on 16q22 associates with atrial fibrillation and ischemic stroke. Nat Genet 41:876–878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Traylor M, Farrall M, Holliday EG et al. (2012) Genetic risk factors for ischaemic stroke and its subtypes (the METASTROKE collaboration): a meta-analysis of genome-wide association studies. Lancet Neurol 11:951–962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Helgadottir A, Thorleifsson G, Manolescu A et al. (2007) A common variant on chromosome 9p21 affects the risk of myocardial infarction. Science 316:1491–1493. [DOI] [PubMed] [Google Scholar]
- 67.Gschwendtner A, Bevan S, Cole JW et al. (2009) Sequence variants on chromosome 9p21.3 confer Risk of atherosclerotic stroke. Arm Neurol 65:531–539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Hu W L, Li SJ, Liu DT et al. (2009) Genetic variants on chromosome 9p21 and ischemic stroke in Chinese. Brain Res Bull 79:431–435. [DOI] [PubMed] [Google Scholar]
- 69.Holliday EG, Maguire JM, Evans TJ et al. (2012) Common variants at 6p21.1 are associated with large artery atherosclerotic stroke. Nat Genet 44:1147–1151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Traylor M, Mäkelä KM, Kilarski LL et al. (2014) A novel MMP12 locus is associated with large Artery atherosclerotic stroke using a genome-wide age-at-onset informed approach. PLoS Genet. 10.1371/journal.pgen.1004469. [DOI] [PMC free article] [PubMed]
- 71.Yamada Y, Fuku N, Tanaka M et al. (2009) Identification of CELSR1 as a susceptibility gene for ischemic stroke in Japanese individuals by a genome-wide association study. Atherosclerosis 207:144–149. [DOI] [PubMed] [Google Scholar]
- 72.Zhan YH, Lin Y, Tong SJ et al. (2015) The CELSR1 polymorphisms rs6007897 and rs4044210 are associated with ischaemic stroke in Chinese Han population. Ann Hum Biol 42:26–30. [DOI] [PubMed] [Google Scholar]
- 73.Kubo M, Hata J, Ninomiya T et al. (2007) A nonsynonymous SNP in PRKCH (protein kinase Cη) increases the risk of cerebral infarction. Nat Genet 39:212–217. [DOI] [PubMed] [Google Scholar]
- 74.Wu L, Shen Y, Liu X et al. (2009) The 1425G/A SNP in PRKCH is associated with ischemic stroke and cerebral hemorrhage in a Chinese population. Stroke 40:2973–2976. [DOI] [PubMed] [Google Scholar]
- 75.Lee TH, Ko TM, Chen CH et al. (2016) Identification of PTCSC3 as a novel locus for large - vessel ischemic stroke: a genome-wide association study. J Am Heart Assoc, 10.1161/JAHA.115.003003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Williams FM, Carter AM, Hysi P G et al. (2013) Ischemic stroke is associated with the ABO locus: the EuroCLOT study. Ann Neurol 73:16–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Zhang H, Zhang Z, Zhang J et al. (2017) Fine-mapping of ABO gene identifies two novel SNPs associated with large artery atherosclerotic stroke in a Chinese Han population. Mol Neurobiol 54:2107–2113. [DOI] [PubMed] [Google Scholar]
- 78.Ikram MA, Seshadri S, Bis JC et al. (2009) Genomewide association studies of stroke. J Vase Surg 50:1718–1728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Zhang Z, Xu G, Wei Y et al. (2015) Impact of chromosome 12p13 variants on ischemic stroke risk. Int J Neurosci 126:856–862. [DOI] [PubMed] [Google Scholar]
- 80.Kilarski LL, Achterberg S, Devan WJ et al. (2014) Meta-analysis in more than 17,900 cases of ischemic stroke reveals anovel association at 12q24.12. Neurology 83:678–685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Neurology Working Group of the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium (2016) Identification of additional risk loci for stroke and small vessel disease: a meta-analysis of genome-wide association studies. Lancet Neurol 15:695–707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Traylor M, Ruttenjacobs LC, Thijs V et al. (2016) Genetic associations with white matter hyperintensities confer risk of lacunar stroke. Stroke 47:1174–1179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Debette S, Markus HS (2010) The clinical importance of white matter hyperintensities on brain magnetic resonance imaging: systematic review and meta-analysis. BMJ. 10.1136/bmj.c3666. [DOI] [PMC free article] [PubMed]
- 84.Rost NS, Rahman RM, Biffi A et al. (2010) White matter hyperintensity volume is increased in small vessel stroke subtypes. Neurology 75:1670–1677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Atwood LD, Wolf PA, Heardcosta NL et al. (2004) Genetic variation in white matter hyperintensity volume in the Framingham Study. Stroke 35:1609–1613. [DOI] [PubMed] [Google Scholar]
- 86.Adibsamii P, Devan W, Traylor M et al. (2015) Genetic architecture of white matter hyperintensities differs in hypertensive and nonhypertensive ischemic stroke. Stroke 46:348–353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Paternoster L, Chen W, Sudlow CLM et al. (2009) Genetic determinants of white matter hyperintensities on brain scans: a systematic assessment of 19 candidate gene polymorphisms in 46 studies in 19 000 subjects. Stroke 40:2020–2026. [DOI] [PubMed] [Google Scholar]
- 88.Myriam Fornage, Stephanie Debette, Bis JC et al. (2011) Genome-wide association studies of cerebral white matter lesion burden. Ann Neurol 69:928–939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Traylor M, Zhang CR, Adibsamii P et al. (2015) Genome-wide meta-analysis of cerebral white matter hyperintensities in patients with stroke. Neurology 86:146–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.O'Leary DH, Polak JF, Kronmal RA et al. (1999) Carotid-artery intima and media thickness as a risk factor for myocardial infarction and stroke in older adults. Cardiovascular Health Study Collaborative Research Group. N Engl J Med 340:14–22. [DOI] [PubMed] [Google Scholar]
- 91.Moskau S, Golla A, Grothe C et al. (2005) Heritability of carotid artery atherosclerotic lesions: an ultrasound study in 154 families. Stroke 36:5–8. [DOI] [PubMed] [Google Scholar]
- 92.Bis JC, Kavousi M, Franceschini N et al. (2011) Meta-analysis of genome-wide association studies from the CHARGE consortium identifies common variants associated with carotid intima media thickness and plaque. Nat Genet 43:940–947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Melton PE, Carless MA, Curran JE et al. 2013() Genetic architecture of carotid artery intima-media thickness in Mexican Americans. Circ Cardiovasc Genet 6:211–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Xie G, Myint PK, Voora D et al. (2015) Genome-wide association study on progression of carotid artery intima media thickness over 10 years in a Chinese cohort. Atherosclerosis 243:30–37. [DOI] [PubMed] [Google Scholar]
- 95.Friedman RC, Farh KH, Burge CB et al. (2009) Most mammalian mRNAs are conserved targets of microRNAs. Genome Res 19:92–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Liu N, Zhang L, Wang Z et al. (2017) MicroRNA-101 inhibits proliferation, migration and invasion of human glioblastoma by targeting SOX9. Oncotarget 8:19244–19254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Kefas B, Godlewski J, Comeau L et al. (2008) microRNA-7 Inhibits the Epidermal Growth Factor Receptor and the Akt Pathway and Is Down-regulated in Glioblastoma. Cancer Res 68:3566–3572. [DOI] [PubMed] [Google Scholar]
- 98.Zhou F, Guan Y, Chen Y et al. (2013) miRNA-9 expression is upregulated in the spinal cord of G93A-SOD1 transgenic mice. Int J Clin Exp Pathol 6:1826–1838. [PMC free article] [PubMed] [Google Scholar]
- 99.Hunter MP, Ismail N, Zhang X et al. (2008) Detection of microRNA Expression in Human Peripheral Blood Microvesicles. PLoS One. 10.1371/annotation/b15ca816-7b62-4474-a568-6b60b8959742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Sepramaniam S, Tan JR, Tan KS et al. (2014) Circulating microRNAs as biomarkers of acute stroke. IntJMol Sci 15:1418–1432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Tan KS, Armugam A, Sepramaniam S et al. (2009) Expression profile of microRNAs in young stroke patients. PLoS One 4:1067–1078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Wang W, Sun G, Zhang L et al. (2014) Circulating microRNAs as novel potential biomarkers for early diagnosis of acute stroke in humans. J Stroke Cerebrovasc Dis 23:2607–2613. [DOI] [PubMed] [Google Scholar]
- 103.Tan JR, Tan KS, Koo YX et al. (2013) Blood microRNAs in low or no risk ischemic stroke patients. Int J Mol Sci 14:2072–2084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Zhang Y, Cheng L, Chen Y et al. (2016) Clinical predictor and circulating microRNA profile expression in patients with early onset post-stroke depression. J Affect Disord 193:51–58. [DOI] [PubMed] [Google Scholar]
- 105.Li P, Teng F, Gao F et al. (2015) Identification of circulating microRNAs as potential biomarkers for detecting acute ischemic stroke. Cell Mol Neurobiol 35:433–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Glen C. Jickling, Bradley P et al. (2014) MicroRNA expression in peripheral blood cells following acute ischemic stroke and their predicted gene targets. PLoS One. 10.1371/journal.pone.0099283. [DOI] [PMC free article] [PubMed]
- 107.Sorensen SS, Nygaard AB, Nielsen MY et al. (2014) MiRNA expression profiles in cerebrospinal fluid and blood of patients with acute ischemic stroke. Transl Stroke Res 5:711–718. [DOI] [PubMed] [Google Scholar]
- 108.Broughton BR, Reutens DC, Sobey CG (2009) Apoptotic mechanisms after cerebral ischemia. Stroke 40:331–339. [DOI] [PubMed] [Google Scholar]
- 109.Wang X (2009) The antiapoptotic activity of melatonin in neurodegenerative diseases. CNS Neurosci Ther 15:345–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Wang X (2014) The antiapoptotic effects of melatonin in neonatal hypoxic-ischemic brain injury and adult ischemic stroke. JSM Neurosurgery and Spine 2, 1033. [Google Scholar]
- 111.Ouyang YB, Giffard RG (2014) MicroRNAs affect BCL-2 family proteins in the setting of cerebral ischemia. Neurochem Int 77:2–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Cimmino A, Calin GA, Fabbri M et al. (2005) miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci 102:13944–13949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Musumeci M, Coppola V, Addario A et al. (2011) Control of tumor and microenvironment cross-talk by miR-15a and miR-16 in prostate cancer. Oncogene 30:4231–4242. [DOI] [PubMed] [Google Scholar]
- 114.Spinetti G, Fortunato O, Caporali A et al. (2013) MicroRNA-15a and microRNA-16 impair human circulating proangiogenic cell functions and are increased in the proangiogenic cells and serum of patients with critical limb ischemia. Circ Res 112:707–715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Wu J, Du K, Lu X (2015) Elevated expressions of serum miR-15a, miR-16, and miR-17-5p are associated with acute ischemic stroke. Int J Clin Exp Med 8:21071–21079. [PMC free article] [PubMed] [Google Scholar]
- 116.Leung LY, Chan CP, Leung YK et al. (2014) Comparison of miR-124-3p and miR-16 for early diagnosis of hemorrhagic and ischemic stroke. Clin Chim Acta 433:139–144. [DOI] [PubMed] [Google Scholar]
- 117.Yang Q, Yang K, Li A (2014) MicroRNA-21 protects against ischemia-reperfusion and hypoxia-reperfusion-induced cardiocyte apoptosis via the phosphatase and tensin homolog/ Akt-dependent mechanism. Mol Med Rep 9:2213–2220. [DOI] [PubMed] [Google Scholar]
- 118.Zhou J, Zhang J (2014) Identification of miRNA-21 and miRNA-24 in plasma as potential early stage markers of acute cerebral infarction. Mol Med Rep 10:971–976. [DOI] [PubMed] [Google Scholar]
- 119.Kole AJ, Swahari V, Hammond SM et al. (2011) MiR29b is activated during neuronal maturation and targets BH3-only genes to restrict apoptosis. Genes Dev 25:125–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Wang Y, Huang J, Ma Y et al. (2015) MicroRNA-29b is a therapeutic target in cerebral ischemia associated with aquaporin 4. J Cereb Blood Flow Metab 35:1977–1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Zhang Y, Wang X, Baranov SV et al. (2011) Dipyrone inhibits neuronal cell death and diminishes hypoxic/ischemic brain injury. Neurosurgery 69:942–956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Picascia A, Grimaldi V, Iannone C et al. (2015) Innate and adaptive immune response in stroke: focus on epigenetic regulation. J Neuroimmunol 289:111–120. [DOI] [PubMed] [Google Scholar]
- 123.Lee H, Han S, Chang SK et al. (2016) Biogenesis and regulation of the let-7, miRNAs and their functional implications. Protein Cell 7:100–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Shamsuzzama, Kumar L, Haque R et al. (2016) Role of microRNA Let-7 in modulating multifactorial aspect of neurodegenerative diseases: an overview. Mol Neurobiol 53:2787–2793. [DOI] [PubMed] [Google Scholar]
- 125.Long G, Wang F, Li H et al. (2013) Circulating miR-30a, miR-126 and let-7b as biomarker for ischemic stroke in humans. BMC Neurol 13:178–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Peng G, Yuan Y, Wu S et al. (2015) MicroRNA let-7e is a potential circulating biomarker of acute stage ischemic stroke. Transl Stroke Res 6:437–445. [DOI] [PubMed] [Google Scholar]
- 127.Ni J, Wang X, Chen S et al. (2015) MicroRNA let-7c-5p protects against cerebral ischemia injury via mechanisms involving the inhibition of microglia activation. Brain Behav Immun 49:75–85. [DOI] [PubMed] [Google Scholar]
- 128.Gong Z, Zhao S, Zhang J et al. (2016) Initial research on the relationship between let-7 family members in the serum and massive cerebral infarction. J Neurol Sci 361:150–157. [DOI] [PubMed] [Google Scholar]
- 129.Jickling GC, Ander BP, Shroff N et al. (2016) Leukocyte response is regulated by microRNA let7i in patients with acute ischemic stroke. Neurology 87:2198–2205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Hamzei Taj S, Kho W, Aswendt M et al. (2016) Dynamic modulation of microglia/macrophage polarization by miR-124 after focal cerebral ischemia. J Neuroimmune Pharmacol 11:733–748 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Liu Y, Zhang J, Han R et al. (2015) Downregulation of serum brain specific microRNA is associated with inflammation and infarct volume in acute ischemic stroke. J Clin Neurosci 22:291–295. [DOI] [PubMed] [Google Scholar]
- 132.Zeng Y, Liu JX, Yan ZP et al. (2015) Potential microRNA biomarkers for acute ischemic stroke. Int J Mol Med 36:1639–1647. [DOI] [PubMed] [Google Scholar]
- 133.Cordes KR, Sheehy NT, White MP et al. (2009) MiR-145 and miR-143 regulate smooth muscle cell fate decisions. Nature 460:705–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Gan CS, Wang CW, Tan KS (2012) Circulatory microRNA-145 expression is increased in cerebral ischemia. Genet Mol Res 11:147–152. [DOI] [PubMed] [Google Scholar]
- 135.Jia L, Fang H, Wang W et al. (2015) Circulating miR-145 is associated with plasma high-sensitivity c-reactive protein in acute ischemic stroke patients. Cell Biochem Funct 33:314–319. [DOI] [PubMed] [Google Scholar]
- 136.Maitrias P, Meuth ML, Massy ZA et al. (2015) MicroRNA deregulation in symptomatic carotid plaque. J Vase Surg 62:144–145. [DOI] [PubMed] [Google Scholar]
- 137.Dharap A, Bowen K, Place R et al. (2009) Transient focal ischemia induces extensive temporal changes in rat cerebral microRNAome. J Cereb Blood Flow Metab 29:675–687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Tsai PC, Liao YC, Wang YS et al. (2013) Serum microRNA-21 and microRNA-221 as potential biomarkers for cerebrovascular disease. J Vase Res 50:346–354. [DOI] [PubMed] [Google Scholar]
- 139.Lai TW, Shu Z, Yu TW (2014) Excitotoxicity and stroke: identifying novel targets for neuroprotection. Prog in Neurobiol 115:157–188. [DOI] [PubMed] [Google Scholar]
- 140.Koh JY, Sang WS, Gwag BJ et al. (1996) The Role of Zinc in Selective Neuronal Death After Transient Global Cerebral Ischemia. Science 272:1013–1016. [DOI] [PubMed] [Google Scholar]
- 141.Majdi A, Mahmoudi J, Sadigh-Eteghad S et al. (2016) The interplay of microRNAs and post-ischemic glutamate excitotoxicity: an emergent research field in stroke medicine. Neurol Sci 37:1765–1771. [DOI] [PubMed] [Google Scholar]
- 142.Wang Y, Zhang Y, Huang J et al. (2014) Increase of circulating miR-223 and insulin-like growth factor-1 is associated with the pathogenesis of acute ischemic stroke in patients. BMC Neurol. 10.1186/1471-2377-14-77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Harraz MM, Eacker SM, Wang X et al. (2012) MicroRNA-223 is neuroprotective by targeting glutamate receptors. Proc Natl Acad SciU SA 109:18962–18967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Min W, Li S, Zhang H et al. (2012) Direct interaction between GluR2 and GAPDH regulates AMPAR-mediated excitotoxicity. Mol Brain, 10.1186/1756-6606-5-13.. [DOI] [PMC free article] [PubMed]
- 145.Liu Z, Chen X, Gao Y et al. (2014) Involvement of GluR2 up-regulation in neuroprotection by electroacupuncture pretreatment via cannabinoid CB1 receptor in mice. Sci Rep. 10.1038/srep09490. [DOI] [PMC free article] [PubMed]
- 146.Yang ZB, Li TB, Zhang Z et al. (2016) The diagnostic value of circulating brain-specific microRNAs for ischemic stroke. Intern Med 55:1279–1286. [DOI] [PubMed] [Google Scholar]
- 147.Yang ZB, Zhang Z, Li TB et al. (2014) Up-regulation of brain-enriched miR-107 promotes excitatory neurotoxicity through down-regulation of glutamate transporter-1 expression following ischaemic stroke. Clin Sci 127:679–689. [DOI] [PubMed] [Google Scholar]
- 148.Tiedt S, Prestel M, Malik R et al. (2017) RNA-seq identifies circulating miR-125a-5p, miR-125b-5p and miR-143-3p as potential biomarkers for acute Ischemic stroke. Circ Res 121: 970–980. [DOI] [PubMed] [Google Scholar]
- 149.Edbauer D, Neilson JR, Foster KA et al. (2010) Regulation of synaptic structure and function by FMRP-associated microRNAs miR-125b and miR-132. Neuron 65:373–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Buller B, Chopp M, Ueno Y et al. (2012) Regulation of serum response factor by miRNA-200 and miRNA-9 modulates oligodendrocyte progenitor cell differentiation. Glia 60:1906–1914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Packer AN, Xing Y, Harper SQ et al. (2009) The bifunctional microRNA miR-9/miR-9* regulates REST and CoREST and is downregulated in Huntington's disease. J Neurosci 28:14341–14346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Wei N, Xiao L, Xue R et al. (2016) MicroRNA-9 mediates the cell apoptosis by targeting Bcl2l11 in ischemic stroke. Mol Neurobiol 53:6809–6817. [DOI] [PubMed] [Google Scholar]
- 153.Kelly TJ, Souza AL, Clish CB et al. (2011) A hypoxia-induced positive feedback loop promotes hypoxia-inducible factor 1 alpha stability through miR-210 suppression of glycerol-3-phosphate dehydrogenase 1-like. Mol Cell Biol 31:2696–2706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Chan SY, Loscalzo J (2010) MicroRNA-210: a unique and pleiotropic hypoxamir. Cell Cycle 9:1072–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Zeng L, He X, Wang Y et al. (2014) MicroRNA-210 overexpression induces angiogenesis and neurogenesis in the normal adult mouse brain. Gene Ther 21:37–43. [DOI] [PubMed] [Google Scholar]
- 156.Lou YL, Guo F, Liu F et al. (2012) miR-210 activates notch signaling pathway in angiogenesis induced by cerebral ischemia. Mol Cell Biochem 370:45–51. [DOI] [PubMed] [Google Scholar]
- 157.Eken SM, Osterholm C, Chernogubova E et al. (2013) Array-based profiling reveals biomarker and therapeutic potential for different microRNAs in patients with symptomatic carotid stenosis. Eur Heart J 34:394–394. [Google Scholar]
- 158.Zeng L, Liu J, Wang Y et al. (2011) MicroRNA-210 as a novel blood biomarker in acute cerebral ischemia. Front Biosci 3:1265–1272. [DOI] [PubMed] [Google Scholar]
- 159.Zeng L, Liu J, Wang Y et al. (2013) Cocktail blood biomarkers: prediction of clinical outcomes in patients with acute ischemic stroke. Eur Neurol 69:68–75. [DOI] [PubMed] [Google Scholar]
- 160.Ibrahimverbaas CA, Fornage M, Bis JC et al. (2014) Predicting stroke through genetic risk functions: The CHARGE risk score project. Stroke 45:403–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Malik R, Bevan S, Nalls MA et al. (2014) Multilocus genetic risk score associates with ischemic stroke in case-control and prospective cohort studies. Stroke 45:394–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Malik R, Traylor M, Pulit SL et al. (2016) Low-frequency and common genetic variation in ischemic stroke: the METASTROKE collaboration. Neurology 86:1217–1226. [DOI] [PMC free article] [PubMed] [Google Scholar]
