Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Aug 1.
Published in final edited form as: Biomed Pharmacother. 2021 May 25;140:111753. doi: 10.1016/j.biopha.2021.111753

New epigenetic players in stroke pathogenesis: From non-coding RNAs to exosomal non-coding RNAs

Maryam Mahjoubin-Tehran a,b,#, Samaneh Rezaei a,b,#, Amin Jesmani c, Nafise Birang d, Korosh Morshedi e, Hashem Khanbabaei f, Haroon Khan g, Ashkan Piranviseh h,*,#, Majid Nejati i,*,#, Michael Aschner j, Hamed Mirzaei k,*,#
PMCID: PMC8222190  NIHMSID: NIHMS1708146  PMID: 34044272

Abstract

Non-coding RNAs (ncRNAs) have critical role in the pathophysiology as well as recovery after ischemic stroke. ncRNAs, particularly microRNAs, and the long non-coding RNAs (lncRNAs) are critical for angiogenesis and neuroprotection, and they have been suggested to be therapeutic, diagnostic and prognostic tools in cerebrovascular diseases, including stroke. Moreover, exosomes have been considered as nanocarriers capable of transferring various cargos, such as lncRNAs and miRNAs to recipient cells, with prominent inter-cellular roles in the mediation of neuro-restorative events following strokes and neural injuries. In this review, we summarize the pathogenic role of ncRNAs and exosomal ncRNAs in the stroke.

Keywords: Stroke, Long non-coding RNAs, MicroRNAs, Pathogenesis

1. Introduction

Stroke is a debilitating disease, and the 2nd main cause of global mortality. As stated by the WHO, almost 15 million people experience strokes worldwide, of which 5.5 million die each year [1]. Stroke prevalence, particularly in developing countries continues to grow, causing major social and economic burdens [2].

LncRNAs have key functions in various disorders. Numerous abnormally expressed lncRNAs are detected in patients with ischemic stroke or animal models of ischemia [3]. LncRNAs play a role in angiogenesis, inflammation, cell apoptosis and cell death. MicroRNAs (miRNAs) are a group of small non-coding RNAs [4]. Following stroke, miRNAs affect various pathophysiological mechanisms, such as hematopoiesis, proliferation or rapid growth, immune function, depression as well as metabolism [5]. Indeed, many investigations have corroborated the aberrant expression of miRNAs in stroke [5,6]. These studies establish miRNAs as key mediators in pathological and pathogenic features of ischemic stroke.

Exosomes are extra-cellular vesicles formed by fusion of multi-vesicular bodies with plasma membrane with subsequent release from cells. Exosomes are 30–100 nm in size, and express several cell-surface markers such as CD-63 and Alix. They are characterized by 1.13–1.19 g/mL density in sucrose and thus can be sedimented at 100,000 g [7]. Non-coding RNAs are released from cells in exosomes. Exosomal cargos, such as cellular proteins, non-coding RNAs(lncRNAs and mRNAs)and lipids act as mediators of inter-cellular cross-talk between recipient and effector cells [8]. Additionally, cargo preservation from degradation and transport across the blood-brain barrier (BBB) into the systemic circulation renders exosomes as potential disease biomarkers [9]. Exosomes contribute to neuro-inflammatory stress response as well as neurodegenerative diseases such as Parkinson’s disease (PD), stroke, Alzheimer ‘s disease (AD) and Schizophrenia [10]. Herein, we highlight the pathogenic role of non-coding RNAs in the stroke. Moreover, we summarize the role of exosomal non-coding RNAs in the stroke pathogenesis.

2. Biogenesis of lncRNAs

LncRNAs are important mediators of health and disease [1117]. Several classes of lncRNAs exist, which are transcribed from several DNA elements including intergenic regions, promoters, and enhancers in eukaryotes [18] (Fig. 1). A variety of mechanisms are involved in the biogenesis of lncRNAs. including generation of mature ends by ribonuclease P (RNaseP) cleavage, creation of small nucleolar RNA (snoRNA), the formation of protein (snoRNP) complex caps at their ends, as well as creation of circular structures[19,20]. During biogenesis of lncRNAs, sub-nuclear structures, termed “paraspeckles”, are found in close proximity to specific lncRNAs [21]. Four specific proteins (PSPs) are required for paraspeckle formation [21,22]. The exact mechanism of synthesis and regulation of lncRNAs has yet to be detailed. Techniques, such as cross linking immunoprecipitation (CLIP), ChIRP-Seq (Chromatin Isolation via RNA purification), phylogenetic lineage tracing, RNA structure mapping, ribosome profiling, genetic screens, and genome engineering through CRISPR [2325], are likely to help in this task.

Fig. 1.

Fig. 1.

Biogenesis of lncRNAs. LncRNA transcripts are classified based on their genomic location in relation to the closest gene including antisense lncRNAs, sense lncRNAs, intronic lncRNAs, enhancer lncRNAs and intergenic lncRNAs.

3. lncRNAs and stroke

Long non-coding RNAs play a significant role in ischemic stroke. Numerous aberrantly expressed lncRNAs have been identified in ischemic stroke samples of human or animal models [26,27]. Among them, anti-sense non-coding RNA in INK4 locus (ANRIL), MALAT1,H19, N1LR, the maternally expressed gene 3 (MEG3), CaMK2D-associated transcript 1 (C2dat1), taurine up-regulated gene 1 (TUG1),small nucleolar RNA host gene 14 (SNHG14), and Fos downstream transcript (FosDT) were shown to affect angiogenesis, cell death, cell apoptosis, as well as inflammation in the course of ischemic stroke (Table 1, Fig. 2).

Table 1.

Selected lncRNAs involved in stroke pathogenesis.

LncRNA Target (s) Effect (s) Study models Ref
N1LR Maybe inactivates p53 Inhibits neural death and apoptosis probably via reducing p53 phosphorylation In vitro: N2a cells In vivo: SD rats, mice [76]
MALAT1 E-selectin and Bim Inhibits inflammation and endothelial cell death and defends cerebral parenchyma and microvasculature from brain ischemic injury. In vitro: BMECs In vivo: C57BL/6 mice [83]
H19 DUSP5-ERK1/2 Silenced autophagy which activated by OGD/R via DUSP5-ERK1/2 axis. In vitro: SH-SY5Y cells In vivo: SD rats [84]
TUG1 miR-9/Bcl2l11 Inhibits miR-9 and weakened suppression of bcl2l11 expression and so, leads a neurotoxicity after stroke. In vitro: SH-SY5Y cells In vivo: SD rats [85]
C2dat1 CaMKIIδ Increases the CaMKIIδ expression and activates NF-κB signal impacting neuronal survival after ischemic stroke. n vitro: N2a cells In vivo: C57BL/6 mice [86]
ANRIL VEGF/NF-κB Increases VEGF expression and activates angiogenesis via activation of NF-κB pathway In vivo: male Wistar rats [87]

Fig. 2.

Fig. 2.

Different lncRNAs involved in stroke pathogenesis. This figure is adapted from [82].

3.1. Angiogenesis

One of the first identified lncRNAs is MALAT1, which promotes cancer proliferation and metastasis via gene expression and alternative splicing [2831]. MALAT1 is a stable and conserved lncRNA (~7 kb ([32], and is expressed in vascular endothelial cells, skeletal muscle, cardiomyocytes. MALAT1 thought to be involved in angiogenesis and pathological myogenesis [3336]. In addition, high expression of MALAT1 is inherent to cell nucleus speckles, a pre-mRNA processing domain with a role in gene expression regulation or organization[37]. MALAT1 can also affect pre-mRNA splicing by interacting with splicing factors [38], and in cancers, MALAT1 expression is upregulated by HIF-1αor hypoxia [39,40].

In animal stroke models, such as middle cerebral artery occlusion (MCAO) and oxygen-glucose deprivation (OGD), MALAT1 expression is significantly increased (6.05-fold) [26]. MALAT1knockout mouse leads to a larger size brain infarct, reduced sensorimotor function, and decreased neurological scores after MCAO [41]. Knockout of MALAT1is also associated with reduced vascular growth mice retinas [42], consistent with studies on angiogenesis in experimental cancer models [4244]. In the post-ischemic phase, cerebral vasculature has an important role in the recovery outcomes. Thus, increasing expression of MALAT1 in ischemic stroke affords protection. Further, MALAT1 knockout is characterized by increased expression of Bim, apro-apoptotic factor, pro-inflammatory cytokines, inter-leukin 6 (IL-6), E-selectin, as well as monocyte chemotactic protein-1 (MCP-1),both in ischemic murine brain and brain microvascular endothelial cells (BMECs) [41]. These findings suggest that MALAT1 has protective effect on cerebral ischemic injuries by inhibiting inflammation and endothelial cell death.

3.2. Inflammation

ANRIL, an anti-sense RNAs co-clustered with the p15/CDKN2B-p16/CDKN2A-p14/ARF locus in chromosome 9p21, is involved in cancers and cardiovascular diseases [4547]. ANRIL has several splice variants with a length of ~3.9 kb [48]. In cancers, ANRIL activates by factor-1α and c-Myc which are hypoxia-inducible [49,50]. ANRIL can bind to PRC1/2 and mediates silence gene expression of INK4b-ARF-INK4a locus [51]. The enhanced expression of ANRIL is correlated with coronary artery disease, atherosclerosis, as well as stroke [5255]. ANRIL expression, associated to variants of chromosome 9p21.3, has been suggested as a novel genetic marker for stroke [56]. Its expression is increased in rat cerebral infarcted cortex [57], resulting in activation of IκB/NF-κB pathways and vascular-endothelial growth factor (VEGF)/-VEGF receptor 1 (FLT-1) [57], thus promoting inflammation and angiogenesis. Binding the VEGF to FLT-1 acts as a stimulus for fetal angiogenesis, as well as maintaining endothelial function in adult animals [58]. NF-κB dissociates from IκB in stresses e.g. oxidative stress (OS), cytokines, ultra-violet irradiation, and viral or bacterial antigens. NF-κB translocates to the nucleus and modulates adaptive immune response genes [59]. By activating the VEGF/FLT-1 as well as IκB/NF-κB pathways, ANRIL may significantly contribute to pro-inflammation and angiogenesis.

In addition, caspase recruitment domain (CARD) family member 8, referred to as TUCAN/CARDINAL, is another ANRIL target [60]. rs2043211, an SNP in CARD8 decreases CARD8 expression, and is associated with reduced risks of ischemic stroke [61]. Furthermore, increase or decrease in ANRIL stimulates or inhibits CARD8expression in HepG2 cells, respectively [62]. ANRIL activation suppresses NF-κB by activating CARD8; therefore, likely inhibiting inflammation. Overall, the increased ANRIL levels enhance angiogenesis via VEGF/FLT-1 pathway, and regulate inflammation through NF-κB pathway in ischemic stroke.

3.3. Autophagy

LncRNA-H19 is a 2.3 kb conserved RNA coded by the H19 gene and expressed only in the maternal allele [63]. H19 control growth and development in the embryo [64]. At the first stage (6–8 weeks gestation) of embryonal development, both H19 alleles (paternal and maternal) are expressed. After 10-weeks of gestation, only the maternal H19 alleles are expressed [65]. By targeting another imprinted gene, Igf2, H19 can gain function in controlling the embryo’s growth [66]. The hypermethylation of the H19 promoter and methylation of the 3′ side of H19 in a specific allele is related to the change in H19 expression [65]. Indeed, in pathological conditions, for example, oxidative stress and cancer, the expression of H19 is upregulated [6769]. H19 levels increased in stroke subjects compared to healthy subjects [70] affording diagnostic tool for ischemic stroke [70]. Up-regulation of H19 expression has been also been shown in OGD/reoxygenation in SH-SY5Y cells and ischemia/reperfusion rat brain [71]. Variation in H19 alleles, such as rs217727, has been shown to increase ischemic stroke risk [71]. Additional investigations showed that inhibiting H19 protects SH-SY5Y cells from autophagy and cell death induced by OGD/R. The dual specificity phosphatase 5 (DUSP5)-ERK1/2axis has a role in pro-autophagy impact on H19. DUSP5, a protein kinase phosphatase activated by mitogens, inhibits the ERK1/2 pathway and autophagy [7173]. Increased levels of H19 suppress DUSP5, and therefore activate autophagy and ERK1/2.

3.4. Apoptosis

N1LR, is a lncRNA, located on chromosome 9, with 1.8 kb length; its sequence has overlap with 5′-UTR of Nck1 gene sequence. To date, only one study examined N1LR’s function [74]. N1LR is expressed in cerebral ischemia/reperfusion rat model [75]. Decreased N1LR expression affects infarct volume. Reduction of N1LR expression is inherent toN2a cells undergoing OGD/R. Upregulation of N1LR is associated with a reduction in OGD/R-induced apoptosis in N2a cells by inhibiting the p53 activation [75]. Analysis of the genome location as well as RACE assay has shown overlapping between the N1LR sequences with 5′-UTR of Nck1. The Nck1 gene encodes a protein that plays an important role in glucose tolerance, insulin signaling, and cellular remodeling. Nck1 is increases in the brain of ischemic rats. Furthermore, the genetic ablation of lncRNA-N1LR increases Nck1 expression. However, increasing of lncRNA-N1LR expression did not impactNck1expression [74,76].

FosDT, a 604nt lncRNA, also termed MRAK159688, has an overlapping sequence with the down-stream of Fos [77]. The expression of FosDT is up-regulated (about 13 folds compared to the control) in the acute period of MCAO after focal ischemia [78]. Increased FosDT expression is associated with neurological dysfunction and post-stroke brain damage. Suppressing FosDT causes a reduction in infarct volume and improved recovery of post ischemia motor function in comparison with controls [78]. Bio-informatics analyses has shown that FosDT and Fos are congenic on chromosome 6q31 in rats [78]. Fos rapidly increased after brain injury [79]. The correlation between increased Fos with increased FosDT suggests transcriptional and/or regulatory interactions between them. FosDT attaches to the chromatin modifying proteins (CMPs) including Sin3a as well as co-repressors of transcription factor REST (coREST). These proteins are co-repressors for the transcription factor repressor element-1 silencing transcription factor (REST) [80]. In fact, REST represses neuronal function such as synaptic transmission and neural differentiation [81]. Furthermore, REST forms a complex consisting of Sin3a (REST-coREST-Sin3a) as well as coREST in rats with transient focal ischemia. This complex inhibits the expression of downstream genes such as NF-κB2, GluR2 and N-methyl-D-aspartate 1 and increases ischemic brain damages[78].

4. MicroRNA biogenesis

MicroRNAs are short (~22-nucleotide) non-coding RNAs which silence gene expression post-transcriptionally. The first miRNA has been identified in Caenorhabditis elegans in 1993 [88]; but the regulatory function of miRNAs was first recognized in 2001 [8991]. miRNA binds to the target mRNA degrades it or blocks translation [92]. RNA polymerase II (Pol II) the most of miRNA genes in the nucleus to primary miRNAs (pri-miRNAs). Next, capping, splicing and polyadenylation are carried out by pri-miRNAs [93]. Most of the miRNAs are transcribed from dedicated miRNA gene loci and only 30% of miRNAs are expressed from introns. A single pri-miRNA may produce one or several miRNAs.

Long pri-miRNA is cleaved via Micro-processor consisting of DRO-SHA, an RNase III enzyme, as well as the respective co-factor; namely, DiGeorge syndrome critical region 8 (DGCR8) [94,95]. Two RNase III domains have been identified in DROSHA; each domain processes one strand of the dsRNA to cleave almost 60–70-nucleotide hairpin-shaped precursor miRNAs (pre-miRNAs) [9497]. Microprocessor identifies the single-stranded RNA (ssRNA)–stem junction along with distance from the terminal loop. Therefore, it cleaves dsRNA from the junction with the flanking ssRNA and pre-miRNAs in the shape of the hairpin produce with a 1 or 2 nucleotide overhang at the 3′ end (group I and II miRNAs, respectively) [98101]. Though the core components of DROSHA as well as DGCR8 have been regarded as essential for nearly all miRNA biogenesis, and micro-processor activities may be re-constituted with the recombinant DGCR8 and DROSHA proteins in vitro [95,96],

A variety of factors play important roles in processing of pri-miRNA (more details are illustrated later). Pre-miRNAs are exported to the cytoplasm through exportin 5 (XPO5)[102104]. In the cytoplasm, DICER1 (RNase III enzyme) further processes pre-miRNAs [105]. In addition, DICER1 attachment to the end of pre-miRNA and a-symmetrical cleavage of dsRNA stem. DICER1cleavages near the terminal loop and creates a mature miRNA duplex; while, 2-nucleotide are over hanged in the 3′ end [106]. Hence, DICER1 correlates with the transactivation responsive RNA-binding protein (TRBP),TARBP2, for binding to dsRNA [107]. TRBP increases the DICER1 cleavage fidelity of pre-miRNAs in a structure-dependent manner. TRBP also changes the selection of guide-strand of miRNA by promotion the formation of isomiRNAs, which are one nucleotide longer than the usual miRNAs; but TRBP is not essential for processing of pre-miRNA by DICER1 [108,109]. Moreover, TRBP physically links DICER1 and Argonaute proteins, including AGO2, AGO1, AGO4 and AGO3 in order to involve in formation of a miRNA-induced silencing complex (miRISC) [107]. The guide strand of the miRNA through an Argonaute protein retains in the miRISC. Then miRNA guides the complex to target mRNAs and post-transcriptionally silence gene expression. Such a condition is found in cytoplasmic foci, processing bodies (P-bodies) induced by mRNA silencing; however, P-bodies are not essential for gene silencing mediated by miRNA [110112].

5. MicroRNAs and stroke

Expression of miRNAs after stroke has an important role in the disease. For example, three miRNAs including miR-497, miRNA 21 and miR-99a have been shown to attenuate ischemic volumes and save the neuronal cells from apoptosis, preserving neurological functions [113115]. In addition, overexpression of miR-let-7c-5p and miR-424 showed reduced activation of microglia in cerebral ischemia [116]. Up-regulation of miR-103, miR-103, miR-132 as well as miR-126 diminish neurobehavioral as well as neuropathological alterations in hemorrhagic stroke. These effects occur by protection of BBB integrity, attenuating neuroinflammation and decreasing neuronal apoptosis [117119]. Moreover, neurogenesis and angiogenesis in mouse brain is ameliorated by up-regulation of miR-210, promoting repair [120]. However, miRNA can also have a negative role. For instance, Liu et al. reported that miR-155 silencing increases migration, proliferation, and angiogenesis due to reduced cellular apoptosis, and production of reactive oxygen species (ROS) [121]. In agreement, in hemorrhagic stroke, miR-27b silencing has been shown to mitigate neurological loss by reducing cell death and repressing neuro-inflammation. Additionally, miR-124 and miR-155 have been shown to play a role in macrophage polarization [122]. Furthermore, miRNAs have an opposite effect in modulation of the synaptic plasticity. For example, miR-134 has been shown to improve the remodeling of neuronal structures through translational repression of Limk1-mRNA,a protein kinase with a role in dendritic spine development [123].

miR-212-5p is involved in ferroptotic neuronal death in traumatic brain injury (TBI)murine model. Overexpression of miR-212-5p attenuated ferroptosis while downregulation of miR-212-5p promoted ferroptotic cell death partially by targeting prostaglandin-endoperoxide synthase-2 in HT-22 and Neuro-2a cell lines. In addition, administration of miR-212-5p in controlled cortical impact mice significantly improved learning and spatial memory [124].

Compared with non-DM stroke mice, T2DM-stroke mice exhibit significantly decreased serum and brain tissue miR-126 expression. Endothelial cells and EC-Exo contain high levels of miR-126 compared with other cell types or exosomes derived from other types of cells, respectively (smooth muscle cells, astrocytes, and marrow stromal cells). EC-Exo treatment of T2DM-stroke mice significantly improves neurological and cognitive function, increases axon density, myelin density, vascular density, arterial diameter, as well as induces M2 macrophage polarization in the ischemic boundary zone [125].

5.1. Inflammation

Xie et al. reported on the ability of miR-181a to promote the cellular survival in vitro through repressing inflammation processes in macrophages as well as monocytes [126]. However, Moon has shown that miR-181a silencing attenuated neuronal apoptosis induced by forebrain ischemia [127].

Inflammation is an intricate response after injury and promotes repair process [128]. Enhanced immune responses may be detrimental [129], and can be an important determinant of stroke prognosis [117, 130132]. Both hemorrhagic and ischemic stroke trigger microglial activation and inflammatory factors release, including TNF-α [133,134], promoting brain injury [135137]. Moreover, T-lymphocytes, natural killer cells, polymorpho-nuclear leukocytes and mononuclear phagocytes produce and release several peripherally-derived cytokines which participate in neuroinflammation after stroke [138,139].

Numerous genes function to regulate neuroinflammation. These genes are targeted by miRNAs [140142]. Lentiviral over-expression of miR-424 diminishes brain injuries following the ischemic stroke by repressing microglial activity [116]. miR-let-7c-5p has a neuro-protective effect against neuroinflammation after ischemic stroke, preventing the translational repression of caspase-3 and microglia activation [143]. miR-124 is known as ‘brain-specific miRNA’ because it is expressed particularly in the central nervous system (CNS) [144,145]. Laterza et al. demonstrated that miR-124 is overexpressed in plasma following the brain injuries caused by MCAO [144]. Additionally, this microRNA suppressed the CCAAT/enhancer-binding protein alpha (C/EBP-α) and PU.1 directly as its down-stream element, resulting in microglia quiescence. Furthermore, miR-124 inhibited experimental autoimmune encephalomyelitis (EAE) by macrophage deactivation.

Toll-like receptors (TLRs) have been shown to have major functions in neuro-inflammation following stroke [146148]. Zhang et al. reported that miR-181c inhibited expressing TLR4 via attachment to 3′UTR of its gene, thus reducing levels of the nuclear factor kappa light chain enhancer of the activated B cells (NF-κB) as well as generation of the down-stream pro-inflammatory factors [149]. Furthermore, up-regulation of miR132 in mice hemorrhagic stroke leads to a better prognosis in comparison to controls. In addition, over-expression of miR132 represses activated microglia and pro-inflammatory cytokine production [119]. Yuan and colleagues in an experimental model of ICH have shown that miR-367 reduced IRAK4 levels by direct binding to its 3′-UTR. MiR-367 also inhibits NF-κB activation and synthesis of its downstream pro-inflammatory elements. Another study has shown that miR-223 improves the neurological functions by down-regulating NLRP3 as well as inflammation inhibition via caspase-1 and IL-1beta [150].

MiRNAs have key roles in anti-inflammatory impact in brain due to the regulation of microglia and microphage polarization. MiR-155, by targeting M2-associated genes, promotes the M1 phenotype. Indeed, this microRNA targets different genes related to the M2 phenotype that mitigate production of M2-induced proinflammatory factors such as IL13Rα1, CD206 and IL-10 and Arg-1 [151,152].

5.2. Apoptosis

Apoptosis is energy-dependent and also known as programmed cell death [153]. Apoptosis has a key role in physiological metabolism, growth, and development. [154,155]. However, uncontrolled apoptosis may lead to different disorders such as cancers, Alzheimer’s disease as well as stroke [156,157]. Apoptosis is triggered by either internal or external pathways. External pathways occur surface death receptors activation. These receptors include Fas and TNF-related apoptosis like tumor necrosis factor (TNF)-α and ligand receptors, whereas interior pathways are associated with mitochondrial signaling pathways [158, 159]. Following stroke, a mass influx of Ca2+ into the cell is triggered, resulting in apoptosis-inducing factor (AIF) release or mitochondrial cytochrome c (Cytc) [160]. Binding Cytc to the apoptotic protease-activating factor-1 as well as procaspase-9 forms an apoptosome, in turn activating caspase-9. Eventually, caspase-3 leads to damages to nDNA as well as the ensuing cell death. In addition, AIF translocates to the nucleus, causing a large-scale (50 kb) DNA-fragmentation and cell death mediated by caspase activation [161].

miRNAs regulate post-stroke neuronal survival through regulation of the level of target genes [162,163]. miR-298 has been overexpressed in blood and brain specimens in experimental models of cerebral ischemia as well as ICH models [164]. An anti-apoptotic factor is miR-21. Buller et al. studied the levels of miR-21 expression both in vivo and in vitro. Levels of this microRNA increased following ischemic stroke secondary to decreased Fas ligand (FasL) G, a cell death-inducing ligand [115]. MiR-155 is responsible for regulating \cellular apoptosis via modulation of caspase-3 expression. Knocking down miR-155 decreased apoptosis in brain microvessel endothelial cells [121]. Moreover, miR-99a has been shown to prevent pro-caspase-3 activation and caspase-3 expression and decreased neuronal apoptosis following ischemic stroke. In addition, miR-99a mitigated neuronal injury after the cerebral Ischemia/Reperfusion (I/R), via cell cycle as well as the cellular apoptosis regulation. This suggests miR-99a as a novel treatment factor targeting neuronal cell cycle re-entry after ischemic stroke [114]. Additionally, the bcl-2 family contributes to apoptosis modulation. MiR-106b-5p, miR-181a, miR-497, miR-384-5p and miR134 increase apoptosis by reducing the levels of bcl-2 proteins [113,126,127,165,166].

MiR-132 has been shown to reduce neuronal mortality in ICH mice, following hemorrhagic stroke. Ip-regulation of miR-132 caused decreased likelihood of neurological deficits [119]. Lentivirus induced up-regulation of miR-126 was shown to be protective in ICH by triggering anti-apoptotic mediators secondary to reduced levels of caspase-3 [118]. Increased miR-103-3p expression in an experimental model of the sub-arachnoid hemorrhage associated with decreased levels of caveolin-1 has also been shown [167].

5.3. Oxidative stress

OS contributes to the pathogenesis of many disorders. It represents over-balance of pro-oxidants (ROS/RNS) and or deficiencies in the antioxidant systems in the cells [168170]. Hence, free-radicals as well as ROS generation in the course of the stroke cause N-methyl-D-aspartic acid (NMDA) glutamate receptors [171], mitochondrial impairment, excessing Ca2+ [172174], and activation of neuronal nitric oxide synthase (nNOS) [175]. Detoxifying enzymes and antioxidants include superoxide dismutase (SOD), glutathione reductase, glutathione-S-transferase and glutathione peroxidase, which sustain redox homeostasis [176,177]. Erythroid-2-related factor-2 (Nrf2) plays a neuroprotective role against brain damage after stroke, hydrogen peroxide (H2O2) exposure, glutamate excitotoxicity and Ca2+ overload [178].

miRNAs modulate Nrf2 mRNA levels [179]. Following ischemic stroke, miR-93 suppresses Nrf2 and hemeoxygenase-1 (HO-1) [180]. During cerebral ischemia, miR-424 reduces infarct volume by decreasing ROS levels in cortex and elevating manganese SOD (MnSOD) and extra-cellular SOD. miR-424 has also been shown to decrease H2O2 induced injuries in the neuronal cultures, by increasing cell viability as well as MnSOD activities, and decrease levels of lactate dehydrogenase leakage and malondialdehyde [181]. Moreover, miR-23a-3p and miR-106b-5p may have a neuro-protective impact against the post-ischemic oxidative injuries via overexpressing MnSOD [182,183]. Additionally, miR-145 suppresses SOD2 following ischemic stroke [184]. Xu et al. reported suppression of miR-27b reduces brain injury and results in overexpression of Hmox1, Nrf2, Nqo1 as well as SOD1-following ICH through Nrf2/ARE pathway [185]. Nonetheless, few investigations focused on the anti-oxidative activity of miRNAs in the hemorrhagic stroke. More studies are needed to investigate the contribution of the miRNAs in the hemorrhagic stroke.

5.4. Angiogenesis

Zhang et al. explored structural modifications following stroke, reporting increased vascular volumes from 3% to 6% at 90th days after stroke [186]. Angiogenesis regulator miRNAs is a possible therapeutic target in ischemic stroke [187]. MiR-210 has a significant function in angiogenesis following cerebral ischemia, partially by increasing vascular endothelial growth factor (VEGF) expression. In vitro, under hypoxic conditions, miR-210 mediates tube formation and migration of vascular endothelial cells [120]. Ma and colleagues reported that in a rat model of ICH, miR-129-5p represses the HMGB1-RAGE signaling pathway and consequently re-vascularization [188]. Up-regulation of miR126 is protective against ICH, increasing angiogenesis by elevating VEGF-A protein levels [160]. Hence, improved angiogenesis may represent a therapeutic modality for stroke through the pharmacological regulation of miRNAs.

Neurotrophic agents, representing small polypeptide molecules, have a key part in cell differentiation, proliferation, development and migration of the nervous system. Earlier investigations indicated that neuro-trophic agents such as brain derived neuro-trophic factor (BDNF), nerve growth factor, ciliary neurotrophic factor, insulin-like growth factor-1 (IGF-1) and glial-derived neurotrophic factor mitigate neuronal death as well as the brain lesions [189,190].

miR-Let7f has been shown in an experimental model of cerebral ischemia to support IGF-1-like neuroprotection [190]. Additionally, suppression of miR-134 diminishes ischemic damages by promoting Bcl-2as well as BDNF expression. Moreover, miR-30-5p and miR-107 modulates BDNF expression [191]. In addition, neural precursor cells (NPCs) and endogenous neural stem cells (NSCs) can be switched on and migrate toward the injured location[192]. In this regard, miR-21 can modulate NPC’s activity via Wnt and transforming growth factor (TGF)-β signaling pathways. Furthermore, miR-34a modulates NPC proliferation negatively following cerebral ischemia [193]. Based on studies in type-2 diabetic mice following stroke, miR-126 may also promote the neuro-restorative function induced with umbilical cord blood cells [194]. Table 1 and Fig. 3 listed different miRNAs involved in stroke pathogenesis (Table 2).

Fig. 3.

Fig. 3.

Various microRNAs involved in stroke pathogenesis.

Table 2.

Different microRNAs involving in stroke.

MicroRNA (s) Expression status Target (s)/Mechanism (s) Study model Ref
miR-296-5p Down CD73, PKN2 Mouse/MCAO [195]
miR-686, miR-1224 Up CD73, PKN2 Mouse/MCAO [195]
miR-498, miR-25, miR-483-5p miR-34b Down BCL-2 modifying factor (BMF) and p53 Human/Ischemic stroke [196]
miR-21 Up SOD3, TNF-α Rat/MCAO [197]
mir-30a Down RhoB, beclin-1 Human [198]
mir-126, mir-146 Up VCAM-1, TLR4 Human [199]
MiR-1259, miR-142-3p, miR-15b, miR-186, miR-519e, miR-768-5p Down Arl2 Human [199]
miR-1184, Let-7e, miR-1246, miR-1261, miR-1275, miR-1285, miR-1290, miR-181a, miR-25, miR-513a-5p, miR-550, miR-602, miR-665, miR-891, miR-933, miR-923 Up GPx1, NOX4 Human [199]
miR-16, miR-23a, miR-150, miR-107, miR-185, miR-191, miR-292-5p, Let-7, miR-451, miR-327 and miR-494 Up XIAP, APAF-1/caspase-9 Rat/MCAO [200]
miR-26a, miR-26b, miR-29b, miR-140, miR-214, miR-328, miR-352, miR-320, miR-137 Down NCX1 Rat/MCAO [200]
miR-19b, miR-136, miR- 199a-3p, miR-32 Up PI3K/Akt/mTOR signaling Rat/MCAO [201]
miR-290, miR-218, miR-133, miR-145 Up caspase-9, -3 superoxide dismutase-2 Rat/MCAO [202]
miR-27a, miR-204, miR-301, miR-338, miR-7, miR-137, miR-335, miR-148b, miR-98, miR-30e Down COXIV, SOD2, Txnrd2 Rat/MCAO [202]
miR-155 Down SHIP1, p53 Rat/MCAO [203]
miR- 424 Up Cullin 2, PU.1, RUNX-1 and C/EBPα Mice/MCAO [204]
miR-124 Up VILIP-1 Rat/MCAO [205, 206]

6. Exosome biogenesis

Exosomes are largely conserved amongst eukaryotic organisms [207]. Exosomes originate via an endocytic route, and they are generated by the inward budding of plasma membranes [208].

Exosomes can be derived from stem cells isolated from different cell sources such as mesenchymal stem cells, endothelial progenitor cells, and fibroblasts.The mesenchymal stem cells (MSCs) are the most commonly used on research which have usually been isolated from synovium, bone marrow, and adipose tissue. The three major categories of extracellular vesicles are (a) apoptotic bodies, (b) microparticles or microvesicles, and (c) exosomes or nanovesicles [209].

Exosomes consist of the conserved proteins such as CD63 (membrane-related proteins like LAMP-3), CD9, Alix, CD81, as well as tumor susceptibility gene 101 protein [210], and the tissue- or cell-type-specific proteins, which indicate their cellular sources[211]. Sphingomyelin, ceramide and cholesterol are used to enrich exosome membranes [212]. On the other hand, exosomes consist of several biologically active molecules like proteins, deoxyribonucleic acids, ribonucleic acids (RNAs), microRNAs (miRs) and lipids [213]. Exosomal miRs or exosomal engineered miRs function in regulating the progression of various diseases, such as cancers, cardiovascular disease, and stroke [214216].

Exosomal intercellular communications are mediated by bio-active molecules and are capable of targeting certain types of cells and modifying their target cell functions by delivering lipids, nucleic acids and proteins [217]. The majority of proteins in exosomes derive from the parent cell membranes, Golgi and the cytosol but seldom from endoplasmic reticulum or mitochondria [207]. Hence, cytosolic proteins will remain in exosomes and the ones derived from the plasma membranes will be kept in the vesicle membrane and maintain a similar topology of the cells with the potent contribution to sequestering the soluble ligands [218].

Exosomal proteins have a role in the antigen presentation, cell structure and motility as well as cell adhesion. Moreover, they modulate stress and contribute to transcription, protein synthesis, membrane fusion and trafficking [219]. Several functional impacts of exosomes could be ascribed to RNA and miR content transfer [217]. miRs and RNAs have been proposed as the most related cargo in the exosomes with regard to their abilities of a little number of molecules for influencing diverse proteins or enzymes in 1 or more cellular pathways in the targeted cells [220].

7. Exosomal microRNAs and stroke

The ever-increasing information suggested the involvement of the exosome-mediated inter-cellular communication in the brain re-modeling via transfer of the cargo from the source cells to the targeted cells (Fig. 4) [221]. It is possible to isolate the exosomes from bio-fluids such as CSF and from the supernatant of the cells that have been cultured in the exosome-free medium via centrifugation and the other procedures [222]. Exosomes are commonly enriched with tetra-spanin proteins (CD81& CD63), the regulator of the endosomal trafficking Alix, as well as chaperone protein HSP70, though exosomes’ volume is variable according to the cell origin and pathological and physiological conditions [222,223]. Analyses of the proteomic and RNA analyses also showed that exosomes are the carriers of cargoes of proteins, RNAs and lipids such as miRNAs and mRNAs [222,223]. Nonetheless, there is insufficient information on the loading of biological substances into the individual exosomes. In fact, each brain cell releases exosomes [222,224, 225]. In comparison to exosomes separated from the wild type mice brains, exosomes from brains of transgenic mice overexpress human amyloid-β (Aβ) precursor protein (APP) [224]. In addition, the full-length APP cleavage via β-secretase takes place into the endosomes that is one of the fractions of Aβ peptides sorted for multi-vesicular bodies, and release of such Aβ peptides is seen in conjunction with exosomes [226]. Furthermore, exosomes isolated from the pre-frontal cortices of the cases suffering from schizophrenia and bi-polar disorder indicated diverse profiles of the exosomal miRNAs in comparison to the exosomes from control brains [225]. Collectively, these studies suggest that exosomes released by both human and mouse brains under disease conditions change profiles of exosomal cargo proteins and miRNAs and that exosomes enriched with neurotoxic C-terminal fragments of APP may contribute to the spread of Aβ peptides to the brain. Even though in vivo investigations could not specify the cellular source of the exosomes, information obtained from cultured cells suggested that exosomes released by neurons as well as the astrocytes consist of Aβ peptides [226,227].

Fig. 4.

Fig. 4.

Potent exosome mediated inter-cellular communications in the brain re-modeling following the strokes. As seen, the exosomes transfer RNAs as well as proteins for influencing angiogenesis, oligo-dendrogenesis, EVs, extra-cellular vesicles and neurogenesis.

The ever-increasing information also demonstrated miRNAs are the essential modulators in ICH [228,229]. Exosomal miRNAs expression has a wide variance in diverse types of cells and pathological states, and thus miR-modified exosome can change its functions. In addition, analyses have shown the high conservation of MiR-146a-5p amongst the mice, rats and humans. Other investigations also referred to the abundance of miR-146a-5p in the MSCs, and stimulatory impact of the humans’ umbilical cord MSC-exosome on the primordial follicles is applied by carrying the functional miR-146a-5p [230,231]. Accordingly, there can be an association between the advantageous functions of exosome and the increased levels of miR-146a-5p it carries. A number of investigations revealed that miR-146a protects against diverse brain impairment [232234]. Moreover, it contributes to the regulation of microglia or macrophages in the ischemic stroke [235]. Additionally, miR-146a-5p downregulates in the ICH cases’ sera [236] and protects against ICH via suppressing TRAF6/NF-κB pathway [234]. Nonetheless, particular mechanism of miR-146a-5p in neuro-protection following the ICH should be highlighted.

Duan et al. addressed the impact of the exosome derived from the miR-146a-5p enriched bone-marrow mesenchymal stem-cells (BMSCs-miR-146a-5p-Exos) on the experimental ICH [237]. Analysis showed the induction of ICH in adult male Sprague-Dawley rats by intra-striatally injecting the collagenase type IV. Researchers verified binding miR-146a-5p and the respective target genes through the luciferase reporter assay. BMSCs-miR-146a-5p-Exos injection ameliorated the neurological functions, diminished degenerative and apoptotic neurons, and suppressed the inflammatory responses. Results also revealed the clear inhibition of M1 polarization of micro-glia after ICH in the rats by the miR-146a-5p enriched Exosome that has been followed by lower expression of the proinflammatory mediators released via M1 microglia-like monocyte chemo attractant protein-1 (MCP-1), the inducible nitric oxide synthase (iNOS), as well as cyclooxygenase-2 (COX-2). Consequently, direct targeting of interleukin-1 receptor--associated kinase1 (IRAK1) as well as nuclear factor of the activated T-cells 5 (NFAT5) has been shown by miR-146a-5p involved in inflammation responses and polarization of M1 microglia or macrophages. miR-146a-5p riched BMSCs-Exos afforded neuroprotection and functional improvement after ICH via reduction of neuronal apoptosis as well as inflammation related to suppression of the microglial M1 polarization via down-regulating IRAK1 and NFAT5expression [237].

Endogenous neural stem-cells of the adults’ brains have been largely found in the sub-ventricular zone (SVZ) area of lateral ventricle and sub-granular zone (SGZ) region of hippocampus [238,239]. Neural stem-cells are be activated following stroke and subsequently migrate into the lesioned areas and differentiate into the functional neural cells [240,241].

Exosomes from the peri-ischemic striatum and determined them through exosomal bio-markers so that the differentially expressed miRNAs have been detected with micro-array chip. In the next step, they cultured the primary stem cells and utilized OGD as well as reperfusion (OGD/R) for mimic vitro ischemic injuries. Analysis has shown the greater level of the exosomal bio-markers of CD81 and TSG101 in the peri-ischemic striatum following the EA treatment that displayed 25 differentially expressed miRNAs in the isolated exosomes, of which miR-146b has been chosen for additional analyses. Furthermore, EA enhanced the miR-146b expression and its suppressors have been capable of blocking the impacts. Researchers also approved that EA up-regulated miR-146b expression for promoting the differentiation of the neural stem cells into the neurons in the peri-ischemic striatum. According to the results, OGD/R delayed differentiating the neural stem-cells and thus miR-146b suppressors repressed its differentiation. Moreover, NeuroD1 showed its contribution to the differentiation of the neural stem cells into neurons. Additionally, in vivo experiments showed that EA enhanced differentiating the NeuroD1-mediated neural stem cells through miR-146b and ability of EA for improving the neurological deficiencies via miR-146b following the ischemic stroke. Finally, EA promoted differentiating the endogenous neural stem-cells through the exosomal miR-146b for improving the neurological injuries following the ischemic stroke [242].

miRNA-126 affords its neuro-protective effects against the ischemia injuries by regulating the genes expression like phosphor-inositide-3-kinase regulatory sub-unit 2 (PIK3R2). Moreover, the vascular cell adhesion molecule 1 (VCAM-1) in ECs [243,244]. VCAM-1 as well as PIK3R2 have a relationship to the resistance against endothelial dysfunction and vascular inflammation that have been proposed as the 2 prominent procedures correlated to the neuronal damages of reperfusion or ischemia [245]. Lower activation of the inflammatory pathways and endothelial dysfunctions must contribute to the RIPC-mediated neuro-protection through the exosomal miRNA-126 [246]. Chemical reagents such as DNMT inhibitors involve in the neuro-protection in the rodents that suffer from mild ischemia [247,248]. Pandi et al. [249] revealed that small interfering RNA (siRNA)-mediated DNMT3A knockdown decreased infarction in vivo as well as PC12-cell death in vitro. It is noteworthy that there is no information on the mechanisms involved in neuro-protection. Hu et al.[250]found that lower expression of DNMT may result in the greater expression level of a number of genes such as metallothionein because of hypo-methylation, protecting neuronal cells from the hypoxia damages or ischemia. Notably, DNMT3A andDNMT1 partially increased in SH-SY5Y cells that over-expressed miRNA-126 in our research. Consequently, a number of compensatory mechanisms possibly exist amongst DNMT3A, DNMT3B and DNMT1 so that one of them is lost or remarkably diminished [251].

Another investigation explored if exosomal miRNA-126 from RIPC serum may have neuro-protective role [252]. Researchers isolated the exosomes from the venous serum of 4 healthy-young male cases prior to and following RIPC. Consequently, the level of DNMT and DNMT3B activities has been down-regulated in the SH-SY5Y cells that have been incubated with the RIPC exosomes. Following the miRNA-126 over-expression in the SH-SY5Y cells, the overall methylation level as well as DNMT3B gene expression have been down-regulated in the above cells that has been compatible with the bio-informatics prediction. Finally, RIPC exosomes are capable of influencing the cell-cycle and increasing the tolerance of OGD in the SH-SY5Y cells and apparently RIPC has neuro-protective effects by down-regulating DNMTs expression in the neural cells via upregulating the serum exosomal miRNA-126 [252]. Table 3 lists exosomal miRNAs that are involved in stroke.

Table 3.

Exosomal miRNAs in stroke.

Cargo (s) Detection methods Effect (s) References
miR-17–92 Increases neural plasticity and functional recovery after stroke, possibly via targeting phosphatase and tensin homolog. [253]
MiR-133b promotes neural plasticity and functional recovery after treatment of stroke [254]
miR-133b improve neural plasticity and functional recovery after stroke [255]
MiR-30d-5p Prevent cerebral injury by inhibiting autophagy-mediated microglial polarization to M1. [256]
MiR-126 Promotes neurorestorative effects in T2DM mice. [257]
miR-181c-3p Inhibits Neuroinflammation by Downregulating CXCL1 in Astrocytes. [258]
miR-138-5p Reduces neurological impairment by promoting proliferation and inhibiting inflammatory responses of astrocytes following IS by targeting LCN2. [259]
miR-124 Promotes Neurogenesis [260]
microRNA-26a Enhanced neurogenesis after ischemic stroke. [261]
miR-146b Improve neurological injury after ischemic stroke. [242]
MicroRNA-17-92 Enhance Neuroplasticity and Functional Recovery After Stroke. [262]
miR-146a-5p Inhibiting Neuronal Apoptosis and Microglial M1 Polarization. [237]
miR-124 Cortical neurogenesis is increased. [263]
miR-134 It enhanced expressing LIM domain kinase to increase plasticity of synaptic-dendritic after Ischemic stroke. [264]

8. Exosomal long non-coding RNAs and stroke

Researchers showed the lncRNAs in exosomes [265]. Exosomal lncRNAs have shown as the attractive bio-markers for stroke and there is insufficient information on the contribution of exosomal lncRNAs to stroke pathogenesis.

Acute minor stroke (AMS) has been introduced as a sort of hypoxic ischemic necrosis with at least four National Institutes of Health Stroke Scale (NIHSS) score. Nonetheless, initial diagnosis of the AMS is difficult due to the absence of efficient molecular markers. Thus, experts in the field tended to reveal several long non-coding RNAs (lncRNAs) related to AMS.

Xu et al. examined potent bio-markers of lncRNAs in the exosomes isolated from the blood serum of AMS cases for initial detection of the disease [266]. For this reason, they utilized RNA-seq, GO enrichment and KEGG pathway analyses as well as RT-qPCR for validating the level of expression of 4 of 11 differentially expressed lncRNAs (lnc-NTRK3–4, lnc-CRKL-2, lnc-CALM1–7& RPS6KA2-AS1) that contribute to neuro-trophin signaling pathways. Moreover, expression level of lnc-NTRK3–4 and lnc-CRKL-2 considerably enhanced the cases suffering from AMS whereas level of expression of RPS6KA2-AS1 and lnc-CALM1–7 remarkably diminished. In conclusion, these newly revealed lncRNAs may be used as novel joint biomarkers for the early detection of AMS [266].

Chen et al. addressed modifications in the miRNAs and lncRNAs expression patterns in exosomes derived from the vascular endothelial cells upon heat stroke [267]. Analyses identified 10 considerably up-regulated and 10 down-regulated lncRNAs in exosomes derived from the heat stroke temperature treated cells. Consequently, KEGG (Kyoto Encyclopedia of Genes & Genomes) and GO (Gene Ontology) have been utilized for evaluating the signaling pathways of differential expression in lncRNAs. Interaction network of the lncRNAs-miRNAs-mRNA with ceRNA (competing endogenous RNA) principle revealed that the identified miRNAs and lncRNAs in the endothelial cell exosomes may be utilized as the noninvasive bio-markers for the heat strokes [267].

9. Conclusion

A variety of internal and external factors are associated with initiation and progression of stroke. Epigenetic regulations play essential tasks in the stroke pathogenesis. LncRNAs and miRNAs act as epigenetic regulators which are involved in the modulation of several biological mechanisms such as angiogenesis, growth, and differentiation. Several studies revealed that these molecules have critical roles in various stages of stroke. LncRNAs and miRNAs, as main regulators, have therapeutic, diagnostic and prognostic potential in the brain diseases including stroke. Circulating ncRNAs which have cell type- and tissue-specific expression patterns and remarkable stability in peripheral blood have also been studied to detect patients at risk for incident or recurrent stroke, and to predict stroke outcome. Moreover, exosomes have been regarded as the new materials with the prominent inter-cellular roles in the mediation of neuro-restorative events following strokes and neural injuries. Advancement in exosome therapy would benefit from, 1. Identifying cellular signals through which ischemic brain can influence quantity as well as volume of exosomes released by the brain parenchymal cells and remote organs, 2. Getting information of the influence of exosomal cargo on expressing the endogenous genes and proteins in the recipient cells of the damaged brains, 3. Delineating particular kinds of cells targeted by the brain parenchymal cell–derived exosomes, and 4. Deciphering the impacts of gender, comorbidity and age on cellular production of exosomes and the respective cargo as well as the impact of comorbidity, age and gender in responding to the exosome treatments after strokes. However, other investigations on exosomes as tools of inter-cellular communication in ischemic brain should provide in valuable information on the contribution of exosomes to stroke pathogenesis and exosomal therapy.

Funding

The present study was funded by a grant from the Vice Chancellor for Research, Kashan University of Medical Sciences, in Iran. MA was supported in part by grants from the National Institute of Environmental Health Sciences (NIEHS) R01ES07331 and R01ES10563.

Abbreviations:

AGO

Argonaute

AIF

apoptosis-inducing factor

ANRIL

antisense non-coding RNA in the INK4 locus

APAF-1

Apoptotic protease activating factor-1

Arg-1

Arginase 1

Arl2

ADP Ribosylation Factor Like GTPase 2

BBB

blood-brain barrier

Bcl-2

B-cell lymphoma 2

BDNF

brain-derived neurotrophic factor

BMECs

brain microvascular endothelial cells

BMF

BCL-2 modifying factor

CaMKIIδ

Ca(2+)/calmodulin-dependent protein kinase II delta

CARD

Caspase recruitment domain family member

CD

Cluster of Differentiation

C2dat1

CaMK2D-associated transcript 1

CDKN2A

Cyclin Dependent Kinase Inhibitor 2A

CDKN2B

Cyclin Dependent Kinase Inhibitor 2B

C/EBP-α

CCAAT/enhancer-binding protein alpha

ChIRP-Seq

Chromatin Isolation by RNA purification

CLIP

crosslinking immunoprecipitation

CMPs

chromatin-modifying proteins

coREST

co-repressors of the transcription factor REST

CRISPR

clustered regularly interspaced short palindromic repeats

Cytc

cytochrome c

DGCR8

DiGeorge syndrome critical region 8

dsRNA

Double-strand RNA

DUSP5

Dual specificity phosphatase 5

EAE

experimental autoimmune encephalomyelitis

ERK

extracellular signal-regulated kinase

FasL

Fas ligand

FLT-1

Fms-like tyrosine kinase 1

FosDT

Fos downstream transcript

GluR2

glutamate receptor 2

GPx1

Glutathione Peroxidase 1

HIF-1α

Hypoxia-inducible factor 1-alpha

HMGB1

high mobility group box 1 protein

HO-1

hemeoxygenase-1

H2O2

hydrogen peroxide

ICH

Intracerebral hemorrhage

IGF-1

insulin-like growth factor-1

Igf2

Insulin Like Growth Factor 2

IL13Rα1

Interleukin 13 receptor, alpha 1

I/R

Ischemia/Reperfusion

IL

interleukin

IRAK4

interleukin-1 receptor-associated kinase 4

Limk1

LIM Domain Kinase 1

lncRNAs

long non-coding RNAs

MALAT1

Metastasis Associated Lung Adenocarcinoma Transcript 1

MCAO

middle cerebral artery occlusion

MCP-1

monocyte chemotactic protein-1

MEG3

maternally expressed gene 3

miRISC

miRNA-induced silencing complex

miRNAs

microRNAs

MnSOD

manganese SOD

Nck1

non-catalytic region of tyrosine kinase adaptor protein 1

ncRNAs

Non-coding RNAs

NCX

sodium-calcium exchanger

N2a

Neuro2a

NF-κB

nuclear factor kappa-light-chain-enhancer of activated B cells

NLRP3

NOD-, LRR- and pyrin domain-containing protein 3

NMDA

N-methyl-D-aspartic acid

nNOS

nitric oxide synthase

NOX4

NADPH Oxidase 4

NPCs

neural precursor cells

Nqo1

NAD(P)H Quinone Dehydrogenase 1

Nrf2

nuclear factor namely erythroid-2-related factor-2

NSCs

neural stem cells

OGD

oxygen-glucose deprivation

OGD/R

Oxygen-glucose deprivation and reperfusion

P-bodies

processing bodies

PI3K

Phosphoinositide 3-kinases

PKN2

Protein Kinase N2

Pol II

polymerase II

PRC1/2

polycomb repressive complex 1 and 2

pre-miRNAs

precursor miRNAs

pri-miRNAs

primary miRNAs

RACE

Rapid Amplification of cDNA Ends

RAGE

Receptor for advanced glycation end products

REST

repressor element-1 silencing transcription factor

RhoB

Ras Homolog Family Member B

RNAi

RNA interference

RNase P

ribonuclease P

RNA-seq

RNA sequencing

RNS

Reactive nitrogen species

ROS

reactive oxygen species

RUNX-1

Runt-related transcription factor 1

SHIP1

SH-2 containing inositol 5′ polyphosphatase 1

SNHG14

small nucleolar RNA host gene 14

snoRNA

small nucleolar RNA

snoRNP

small nucleolar ribonucleoprotein

SNP

single nucleotide polymorphism

SOD

superoxide dismutase

ssRNA

single-stranded RNA

TGF

transforming growth factor

TLRs

Toll-like receptors

TNF-α

Tumor necrosis factor alpha

TRBP

transactivation-responsive RNA-binding protein

TUG1

Taurine Up-Regulated 1

Txnrd2

Thioredoxin Reductase 2

VCAM-1

vascular cell adhesion molecule 1

VEGF

vascular endothelial growth factor

VILIP-1

Visinin like protein 1

WHO

World Health Organization

XIAP

X-linked inhibitor of apoptosis protein

XPO5

exportin 5

Footnotes

Conflict of interest statement

The other authors declare no conflicts of interest.

Availability of data and material

The primary data for this study is available from the authors on request.

References

  • [1].W.H. Organization, The World Health Report: 2004: Changing History, 2004.
  • [2].Banerjee TK, Roy MK, Bhoi KK, Is stroke increasing in India–preventive measures that need to be implemented, J. Indian Med. Assoc 103 (3) (2005) 162, 164, 166 passim. [PubMed] [Google Scholar]
  • [3].He W, Wei D, Cai S. Chen, Li S, Chen W, Altered long non-coding RNA transcriptomic profiles in ischemic stroke, Hum. Gene Ther 29 (6) (2018) 719–732. [DOI] [PubMed] [Google Scholar]
  • [4].Jolana L, Kamil D, The role of microRNA in ischemic and hemorrhagic stroke, Curr. Drug Deliv 14 (6) (2017) 816–831. [DOI] [PubMed] [Google Scholar]
  • [5].Mirzaei H, Momeni F, Saadatpour L, Sahebkar A, Goodarzi M, Masoudifar A, Kouhpayeh S, Salehi H, Mirzaei HR, Jaafari MR, MicroRNA: relevance to stroke diagnosis, prognosis, and therapy, J. Cell Physiol 233 (2) (2018) 856–865. [DOI] [PubMed] [Google Scholar]
  • [6].Mirzaei H, Stroke in women: risk factors and clinical biomarkers, J. Cell Biochem 118 (12) (2017) 4191–4202. [DOI] [PubMed] [Google Scholar]
  • [7].Borges FT, Reis LA, Schor N, Extracellular vesicles: structure, function, and potential clinical uses in renal diseases, Braz. J. Med. Biol. Res 46 (10) (2013) 824–830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Blandford SN, Galloway DA, Moore CS, The roles of extracellular vesicle microRNAs in the central nervous system, Glia 66 (11) (2018) 2267–2278. [DOI] [PubMed] [Google Scholar]
  • [9].Barile L, Vassalli G, Exosomes: Therapy delivery tools and biomarkers of diseases, Pharmacol. Ther 174 (2017) 63–78. [DOI] [PubMed] [Google Scholar]
  • [10].Soria FN, Pampliega O, Bourdenx M, Meissner WG, Bezard E, Dehay B, Exosomes, an unmasked culprit in neurodegenerative diseases, Front. Neurosci 11 (2017) 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Schmitz SU, Grote P, Herrmann BG, Mechanisms of long noncoding RNA function in development and disease, Cell. Mol. Life Sci 73 (13) (2016) 2491–2509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Delas MJ, Hannon GJ, lncRNAs in development and disease: from functions to mechanisms. 2017. 7(7). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Akerman I, Tu Z, Beucher A, Rolando D, Sauty-Colace C, Benazra M, Nakic N, Yang J, Wang H, Pasquali L, Moran I, Garcia-Hurtado J, Castro N, Gonzalez-Franco R, Stewart AF, Bonner C, Piemonti L, Berney T, Groop L, Kerr-Conte J, Pattou F, Argmann C, Schadt E, Ravassard P, Ferrer J, Human pancreatic β cell lncRNAs control cell-specific regulatory networks, Cell Metab. 25 (2) (2017) 400–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Morán I, Akerman I, van de Bunt M, Xie R, Benazra M, Nammo T, Arnes L, Nakić N, García-Hurtado J, Rodríguez-Seguí S, Pasquali L, Sauty-Colace C, Beucher A, Scharfmann R, van Arensbergen J, Johnson PR, Berry A, Lee C, Harkins T, Gmyr V, Pattou F, Kerr-Conte J, Piemonti L, Berney T, Hanley N, Gloyn AL, Sussel L, Langman L, Brayman KL, Sander M, McCarthy MI, Ravassard P, Ferrer J, Human β cell transcriptome analysis uncovers lncRNAs that are tissue-specific, dynamically regulated, and abnormally expressed in type 2 diabetes, Cell Metab. 16 (4) (2012) 435–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Vafadar A, Shabaninejad Z, Movahedpour A, Mohammadi S, Fathullahzadeh S, Mirzaei HR, Namdar A, Savardashtaki A, Mirzaei H, Long non-coding RNAs as epigenetic regulators in cancer, Curr. Pharm. Des 25 (33) (2019) 3563–3577. [DOI] [PubMed] [Google Scholar]
  • [16].Hashemipour M, Boroumand H, Mollazadeh S, Tajiknia V, Nourollahzadeh Z, Rohani Borj M, Pourghadamyari H, Rahimian N, Hamblin MR, Mirzaei H, Exosomal microRNAs and exosomal long non-coding RNAs in gynecologic cancers, Gynecol. Oncol 161 (1) (2021) 314–327. [DOI] [PubMed] [Google Scholar]
  • [17].Rahimian N, Razavi ZS, Aslanbeigi F, Mirkhabbaz AM, Piroozmand H, Shahrzad MK, Hamblin MR, Mirzaei H, Non-coding RNAs related to angiogenesis in gynecological cancer, Gynecol. Oncol (2021). [DOI] [PubMed] [Google Scholar]
  • [18].Wu H, Yang L, Chen L-L, The diversity of long noncoding RNAs and their generation, Trends Genet. 33 (8) (2017) 540–552. [DOI] [PubMed] [Google Scholar]
  • [19].Vicens Q, Westhof E, Biogenesis of circular RNAs, Cell 159 (1) (2014) 13–14. [DOI] [PubMed] [Google Scholar]
  • [20].Dahariya S, Paddibhatla I, Kumar S, Raghuwanshi S, Pallepati A, Gutti RK, Long non-coding RNA: Classification, biogenesis and functions in blood cells, Mol. Immunol 112 (2019) 82–92. [DOI] [PubMed] [Google Scholar]
  • [21].Naganuma T, Hirose T, Paraspeckle formation during the biogenesis of long non-coding RNAs, RNA Biol. 10 (3) (2013) 456–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Yamazaki T, Hirose T, The building process of the functional paraspeckle with long non-coding RNAs, Front. Biosci. (Elite Ed.) 7 (2015) 1–41. [DOI] [PubMed] [Google Scholar]
  • [23].Yoon JH, Gorospe M, Cross-linking immunoprecipitation and qPCR (CLIP-qPCR) analysis to map interactions between long noncoding RNAs and RNA-binding proteins, Methods Mol. Biol 1402 (2016) 11–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Salehi S, Taheri MN, Azarpira N, Zare A, Behzad-Behbahani A, State of the art technologies to explore long non-coding RNAs in cancer, J. Cell. Mol. Med 21 (12) (2017) 3120–3140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Li JH, Liu S, Zheng LL, Wu J, Sun WJ, Wang ZL, Zhou H, Qu LH, Yang JH, Discovery of protein-lncRNA interactions by integrating large-scale CLIP-Seq and RNA-Seq datasets, Front. Bioeng. Biotechnol 2 (2014) 88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Zhang J, Yuan L, Zhang X, Hamblin MH, Zhu T, Meng F, Li Y, Chen YE, Yin KJ, Altered long non-coding RNA transcriptomic profiles in brain microvascular endothelium after cerebral ischemia, Exp. Neurol 277 (2016) 162–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Dykstra-Aiello C, Jickling GC, Ander BP, Shroff N, Zhan X, Liu D, Hull H, Orantia M, Stamova BS, Sharp FR, Altered expression of long noncoding RNAs in blood after ischemic stroke and proximity to putative stroke risk loci, Stroke 47 (12) (2016) 2896–2903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Ji P, Diederichs S, Wang W, Böing S, Metzger R, Schneider PM, Tidow N, Brandt B, Buerger H, Bulk E, Thomas M, Berdel WE, Serve H, Müller-Tidow C, MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer, Oncogene 22 (39) (2003) 8031–8041. [DOI] [PubMed] [Google Scholar]
  • [29].Yoshimoto R, Mayeda A, Yoshida M, Nakagawa S, MALAT1 long non-coding RNA in cancer, Biochim. Biophys. Acta 1859 (1) (2016) 192–199. [DOI] [PubMed] [Google Scholar]
  • [30].Guffanti A, Iacono M, Pelucchi P, Kim N, Soldà G, Croft LJ, Taft RJ, Rizzi E, Askarian-Amiri M, Bonnal RJ, Callari M, Mignone F, Pesole G, Bertalot G, Bernardi LR, Albertini A, Lee C, Mattick JS, Zucchi I, De Bellis G,A transcriptional sketch of a primary human breast cancer by 454 deep sequencing, BMC Genom. 10 (2009) 163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Chou J, Wang B, Zheng T, Li X, Zheng L, Hu J, Zhang Y, Xing Y, Xi T, MALAT1 induced migration and invasion of human breast cancer cells by competitively binding miR-1 with cdc42, Biochem. Biophys. Res. Commun 472 (1) (2016) 262–269. [DOI] [PubMed] [Google Scholar]
  • [32].Ip JY, Nakagawa S, Long non-coding RNAs in nuclear bodies, Dev. Growth Differ 54 (1) (2012) 44–54. [DOI] [PubMed] [Google Scholar]
  • [33].Zhao J, Li L, Peng L, MAPK1 up-regulates the expression of MALAT1 to promote the proliferation of cardiomyocytes through PI3K/AKT signaling pathway, Int. J. Clin. Exp. Pathol 8 (12) (2015) 15947–15953. [PMC free article] [PubMed] [Google Scholar]
  • [34].Watts R, Johnsen VL, Shearer J, Hittel DS, Myostatin-induced inhibition of the long noncoding RNA Malat1 is associated with decreased myogenesis, Am. J. Physiol. Cell Physiol 304 (10) (2013) C995–C1001. [DOI] [PubMed] [Google Scholar]
  • [35].Tang Y, Jin X, Xiang Y, Chen Y, Shen CX, Zhang YC, Li YG, The lncRNA MALAT1 protects the endothelium against ox-LDL-induced dysfunction via upregulating the expression of the miR-22-3p target genes CXCR2 and AKT, FEBS Lett. 589 (20 Pt B) (2015) 3189–3196. [DOI] [PubMed] [Google Scholar]
  • [36].Michalik KM, You X, Manavski Y, Doddaballapur A, Zörnig M, Braun T, John D, Ponomareva Y, Chen W, Uchida S, Boon RA, Dimmeler S, Long noncoding RNA MALAT1 regulates endothelial cell function and vessel growth, Circ. Res 114 (9) (2014) 1389–1397. [DOI] [PubMed] [Google Scholar]
  • [37].Hutchinson JN, Ensminger AW, Clemson CM, Lynch CR, Lawrence JB, Chess A, A screen for nuclear transcripts identifies two linked noncoding RNAs associated with SC35 splicing domains, BMC Genom. 8 (2007) 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Sun W, Yang Y, Xu C, Guo J, Regulatory mechanisms of long noncoding RNAs on gene expression in cancers, Cancer Genet. 216–217 (2017) 105–110. [DOI] [PubMed] [Google Scholar]
  • [39].Yuan P, Cao W, Zang Q, Li G, Guo X, Fan J, The HIF-2alpha-MALAT1-miR-216b axis regulates multi-drug resistance of hepatocellular carcinoma cells via modulating autophagy, Biochem. Biophys. Res. Commun 478 (3) (2016) 1067–1073. [DOI] [PubMed] [Google Scholar]
  • [40].Sallé-Lefort S, Miard S, Nolin MA, Boivin L, Paré MÈ, Debigaré R, Picard F, Hypoxia upregulates Malat1 expression through a CaMKK/AMPK/HIF-1alpha axis, Int. J. Oncol 49 (4) (2016) 1731–1736. [DOI] [PubMed] [Google Scholar]
  • [41].Zhang X, Tang X, Liu K, Hamblin MH, Yin KJ, Long noncoding RNA malat1 regulates cerebrovascular pathologies in ischemic, Stroke 37 (7) (2017) 1797–1806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Michalik KM, You X, Manavski Y, Doddaballapur A, Zörnig M, Braun T, John D, Ponomareva Y, Chen W, Uchida S, Boon RA, Dimmeler S, Long noncoding RNA MALAT1 regulates endothelial cell function and vessel growth, Circ. Res 114 (9) (2014) 1389–1397. [DOI] [PubMed] [Google Scholar]
  • [43].Tee AE, Liu B, Song R, Li J, Pasquier E, Cheung BB, Jiang C, Marshall GM, Haber M, Norris MD, Fletcher JI, Dinger ME, Liu T, The long noncoding RNA MALAT1 promotes tumor-driven angiogenesis by up-regulating pro-angiogenic gene expression, Oncotarget 7 (8) (2016) 8663–8675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Nduom EK, Wei J, Yaghi NK, Huang N, Kong LY, Gabrusiewicz K, Ling X, Zhou S, Ivan C, Chen JQ, Burks JK, Fuller GN, Calin GA, Conrad CA, Creasy C, Ritthipichai K, Radvanyi L, Heimberger AB, PD-L1 expression and prognostic impact in glioblastoma, Neuro Oncol. 18 (2) (2016) 195–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Shete S, Hosking FJ, Robertson LB, Dobbins SE, Sanson M, Malmer B, Simon M, Marie Y, Boisselier B, Delattre JY, Hoang-Xuan K, El Hallani S, Idbaih A, Zelenika D, Andersson U, Henriksson R, Bergenheim AT, Feychting M, Lönn S, Ahlbom A, Schramm J, Linnebank M, Hemminki K, Kumar R, Hepworth SJ, Price A, Armstrong G, Liu Y, Gu X, Yu R, Lau C, Schoemaker M, Muir K, Swerdlow A, Lathrop M, Bondy M, Houlston RS, Genome-wide association study identifies five susceptibility loci for glioma, Nat. Genet 41 (8) (2009) 899–904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].McPherson R, Pertsemlidis A, Kavaslar N, Stewart A, Roberts R, Cox DR, Hinds DA, Pennacchio LA, Tybjaerg-Hansen A, Folsom AR, Boerwinkle E, Hobbs HH, Cohen JC, A common allele on chromosome 9 associated with coronary heart disease, Science 316 (5830) (2007) 1488–1491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Pasmant E, Laurendeau I, Héron D, Vidaud M, Vidaud D, Bièche I, Characterization of a germ-line deletion, including the entire INK4/ARF locus, in a melanoma-neural system tumor family: identification of ANRIL, an antisense noncoding RNA whose expression coclusters with ARF, Cancer Res. 67 (8) (2007) 3963–3969. [DOI] [PubMed] [Google Scholar]
  • [48].Folkersen L, Kyriakou T, Goel A, Peden J, Mälarstig A, Paulsson-Berne G, Hamsten A, Hugh W, Franco-Cereceda A, Gabrielsen A, Eriksson P, c PROCARDIS, Relationship between CAD risk genotype in the chromosome 9p21 locus and gene expression. Identification of eight new ANRIL splice variants, PLoS One 4 (11) (2009) 7677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Lu Y, Zhou X, Xu L, Rong C, Shen C, Bian W, Long noncoding RNA ANRIL could be transactivated by c-Myc and promote tumor progression of non-small-cell lung cancer, Onco Targets Ther. 9 (2016) 3077–3084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Wei X, Wang C, Ma C, Sun W, Li H, Cai Z, Long noncoding RNA ANRIL is activated by hypoxia-inducible factor-1α and promotes osteosarcoma cell invasion and suppresses cell apoptosis upon hypoxia, Cancer Cell Int. 16 (2016) 73. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [51].Aguilo F, Zhou MM, Walsh MJ, Long noncoding RNA, polycomb, and the ghosts haunting INK4b-ARF-INK4a expression, Cancer Res. 71 (16) (2011) 5365–5369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Holdt LM, Beutner F, Scholz M, Gielen S, Gäbel G, Bergert H, Schuler G, Thiery J, Teupser D, ANRIL expression is associated with atherosclerosis risk at chromosome 9p21, Arterioscler. Thromb. Vasc. Biol 30 (3) (2010) 620–627. [DOI] [PubMed] [Google Scholar]
  • [53].Consortium P, Deloukas P, Kanoni S, Willenborg C, Farrall M, Assimes TL, Thompson JR, Ingelsson E, Saleheen D, Erdmann J, Goldstein BA, Stirrups K, König IR, Cazier JB, Johansson A, Hall AS, Lee JY, Willer CJ, Chambers JC, Esko T, Folkersen L, Goel A, Grundberg E, Havulinna AS, Ho WK, Hopewell JC, Eriksson N, Kleber ME, Kristiansson K, Lundmark P, Lyytikäinen LP, Rafelt S, Shungin D, Strawbridge RJ, Thorleifsson G, Tikkanen E, Van Zuydam N, Voight BF, Waite LL, Zhang W, Ziegler A, Absher D, Altshuler D, Balmforth AJ, Barroso I, Braund PS, Burgdorf C, Claudi-Boehm S, Cox D, Dimitriou M, Do R, DIAGRAM C, CARDIOGENICS C, Doney AS, El Mokhtari N, Eriksson P, Fischer K, Fontanillas P, Franco-Cereceda A, Gigante B, Groop L, Gustafsson S, Hager J, Hallmans G, Han BG, Hunt SE, Kang HM, Illig T, Kessler T, Knowles JW, Kolovou G, Kuusisto J, Langenberg C, Langford C, Leander K, Lokki ML, Lundmark A, McCarthy MI, Meisinger C, Melander O, Mihailov E, Maouche S, Morris AD, Müller-Nurasyid M, MuTHER C, Nikus K, Peden JF, Rayner NW, Rasheed A, Rosinger S, Rubin D, Rumpf MP, Schäfer A, Sivananthan M, Song C, Stewart AF, Tan ST, Thorgeirsson G, van der Schoot CE, Wagner PJ, Wellcome C Trust Case Control, Wells GA, Wild PS, Yang TP, Amouyel P, Arveiler D, Basart H, Boehnke M, Boerwinkle E, Brambilla P, Cambien F, Cupples AL, de Faire U, Dehghan A, Diemert P, Epstein SE, Evans A, Ferrario MM, Ferrières J, Gauguier D, Go AS, Goodall AH, Gudnason V, Hazen SL, Holm H, Iribarren C, Jang Y, Kähönen M, Kee F, Kim HS, Klopp N, Koenig W, Kratzer W, Kuulasmaa K, Laakso M, Laaksonen R, Lee JY, Lind L, Ouwehand WH, Parish S, Park JE, Pedersen NL, Peters A, Quertermous T, Rader DJ, Salomaa V, Schadt E, Shah SH, Sinisalo J, Stark K, Large-scale association analysis identifies new risk loci for coronary artery disease, Nat. Genet 45 (1) (2013) 25–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Cunnington MS, Santibanez Koref M, Mayosi BM, Burn J, Keavney B, Chromosome 9p21 SNPs associated with multiple disease phenotypes correlate with ANRIL expression, PLoS Genet. 6 (4) (2010), 1000899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Amouyel P, From genes to stroke subtypes, Lancet Neurol. 11 (11) (2012) 931–933. [DOI] [PubMed] [Google Scholar]
  • [56].Zhang W, Chen Y, Liu P, Chen J, Song L, Tang Y, Wang Y, Liu J, Hu FB, Hui R, Variants on chromosome 9p21.3 correlated with ANRIL expression contribute to stroke risk and recurrence in a large prospective stroke population, Stroke 43 (1) (2012) 14–21. [DOI] [PubMed] [Google Scholar]
  • [57].Zhang B, Wang D, Ji TF, Shi L, Yu JL, Overexpression of lncRNA ANRIL up-regulates VEGF expression and promotes angiogenesis of diabetes mellitus combined with cerebral infarction by activating NF-kappaB signaling pathway in a rat model, Oncotarget 8 (10) (2017) 17347–17359. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [58].Cao Y, Positive and negative modulation of angiogenesis by VEGFR1 ligands, Sci. Signal 2 (59) (2009) 1. [DOI] [PubMed] [Google Scholar]
  • [59].Smith EM, Gregg M, Hashemi F, Schott L, Hughes TK, Corticotropin Releasing Factor (CRF) activation of NF-kappaB-directed transcription in leukocytes, Cell Mol. Neurobiol 26 (4–6) (2006) 1021–1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Stilo R, Leonardi A, Formisano L, Di Jeso B, Vito P, Liguoro D, TUCAN/CARDINAL and DRAL participate in a common pathway for modulation of NF-kappaB activation, FEBS Lett. 521 (1–3) (2002) 165–169. [DOI] [PubMed] [Google Scholar]
  • [61].Ben Hamad M, Cornelis F, Marzouk S, Chabchoub G, Bahloul Z, Rebai A, Fakhfakh F, Ayadi H, Petit-Teixeira E, Maalej A, Association study of CARD8 (p.C10X) and NLRP3 (p.Q705K) variants with rheumatoid arthritis in French and Tunisian populations, Int. J. Immunogenet 39 (2) (2012) 131–136. [DOI] [PubMed] [Google Scholar]
  • [62].Bai Y, Nie S, Jiang G, Zhou Y, Zhou M, Zhao Y, Li S, Wang F, Lv Q, Huang Y, Yang Q, Li Q, Li Y, Xia Y, Liu Y, Liu J, Qian J, Li B, Wu G, Wu Y, Wang B, Cheng X, Yang Y, Ke T, Li H, Ren X, Ma X, Liao Y, Xu C, Tu X, Wang QK, Regulation of CARD8 expression by ANRIL and association of CARD8 single nucleotide polymorphism rs2043211 (p.C10X) with ischemic stroke, Stroke 45 (2) (2014) 383–388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Brunkow ME, Tilghman SM, Ectopic expression of the H19 gene in mice causes prenatal lethality, Genes Dev. 5 (6) (1991) 1092–1101. [DOI] [PubMed] [Google Scholar]
  • [64].Gabory A, Jammes H, Dandolo L, The H19 locus: role of an imprinted non-coding RNA in growth and development, Bioessays 32 (6) (2010) 473–480. [DOI] [PubMed] [Google Scholar]
  • [65].Jinno Y, Ikeda Y, Yun K, Maw M, Masuzaki H, Fukuda H, Inuzuka K, Fujishita A, Ohtani Y, Okimoto T, Establishment of functional imprinting of the H19 gene in human developing placentae, Nat. Genet 10 (3) (1995) 318–324. [DOI] [PubMed] [Google Scholar]
  • [66].Wilkin F, Paquette J, Ledru E, Hamelin C, Pollak M, Deal CL, H19 sense and antisense transgenes modify insulin-like growth factor-II mRNA levels, Eur. J. Biochem 267 (13) (2000) 4020–4027. [DOI] [PubMed] [Google Scholar]
  • [67].Wang WT, Ye H, Wei PP, Han BW, He B, Chen ZH, Chen YQ, LncRNAs H19 and HULC, activated by oxidative stress, promote cell migration and invasion in cholangiocarcinoma through a ceRNA manner, J. Hematol. Oncol 9 (1) (2016) 117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Matouk IJ, Raveh E, Abu-lail R, Mezan S, Gilon M, Gershtain E, Birman T, Gallula J, Schneider T, Barkali M, Richler C, Fellig Y, Sorin V, Hubert A, Hochberg A, Czerniak A, Oncofetal H19 RNA promotes tumor metastasis, Biochim. Biophys. Acta 1843 (7) (2014) 1414–1426. [DOI] [PubMed] [Google Scholar]
  • [69].Matouk IJ, DeGroot N, Mezan S, Ayesh S, Abu-lail R, Hochberg A, Galun E, The H19 non-coding RNA is essential for human tumor growth, PLoS One 2 (9) (2007) 845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Wang J, Zhao H, Fan Z, Li G, Ma Q, Tao Z, Wang R, Feng J, Luo Y, Long noncoding RNA H19 promotes neuroinflammation in ischemic stroke by driving histone deacetylase 1-dependent M1 microglial polarization, Stroke 48 (8) (2017) 2211–2221. [DOI] [PubMed] [Google Scholar]
  • [71].Wang J, Cao B, Han D, Sun M, Feng J, Long non-coding RNA H19 induces cerebral ischemia reperfusion injury via activation of autophagy, Aging Dis. 8 (1) (2017) 71–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Han W, Fu X, Xie J, Meng Z, Gu Y, Wang X, Li L, Pan H, Huang W, MiR-26a enhances autophagy to protect against ethanol-induced acute liver injury, J. Mol. Med 93 (9) (2015) 1045–1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Tao H, Cao W, Yang JJ, Shi KH, Zhou X, Liu LP, Li J, Long noncoding RNA H19 controls DUSP5/ERK1/2 axis in cardiac fibroblast proliferation and fibrosis, Cardiovasc. Pathol 25 (5) (2016) 381–389. [DOI] [PubMed] [Google Scholar]
  • [74].Wu Z, et al. , Erratum to: LncRNA-N1LR enhances neuroprotection against ischemic stroke probably by inhibiting p53 phosphorylation, Mol. Neurobiol (2017) 7686–7688. [DOI] [PubMed] [Google Scholar]
  • [75].Wu Z, Wu P, Zuo X, Yu N, Qin Y, Xu Q, He S, Cen B, Liao W, Ji A, LncRNA-N1LR enhances neuroprotection against ischemic stroke probably by inhibiting p53 phosphorylation, Mol. Neurobiol 54 (10) (2017) 7670–7685. [DOI] [PubMed] [Google Scholar]
  • [76].Wu Z, Wu P, Zuo X, Yu N, Qin Y, Xu Q, He S, Cen B, Liao W, Ji A, LncRNA-N1LR enhances neuroprotection against ischemic stroke probably by inhibiting p53 phosphorylation, Mol. Neurobiol 54 (10) (2017) 7670–7685. [DOI] [PubMed] [Google Scholar]
  • [77].Ji Y, Guo X, Zhang Z, Huang Z, Zhu J, Chen QH, Gui L, CaMKIIδ meditates phenylephrine induced cardiomyocyte hypertrophy through store-operated Ca2+ entry, Cardiovasc. Pathol 27 (2017) 9–17. [DOI] [PubMed] [Google Scholar]
  • [78].Mehta SL, Kim T, Vemuganti R, Long noncoding RNA FosDT promotes ischemic brain injury by interacting with REST-associated chromatin-modifying proteins, J. Neurosci 35 (50) (2015) 16443–16449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Shimazu M, Mizushima H, Sasaki K, Arai Y, Matsumoto K, Shioda S, Nakai Y, Expression of c-fos in the rat cerebral cortex after focal ischemia and reperfusion, Brain Res. Bull 33 (6) (1994) 689–697. [DOI] [PubMed] [Google Scholar]
  • [80].Dharap A, Pokrzywa C, Vemuganti R, Increased binding of stroke-induced long non-coding RNAs to the transcriptional corepressors Sin3A and coREST, ASN Neuro 5 (4) (2013) 283–289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Paonessa F, Criscuolo S, Sacchetti S, Amoroso D, Scarongella H, Pecoraro Bisogni F, Carminati E, Pruzzo G, Maragliano L, Cesca F, Benfenati F, Regulation of neural gene transcription by optogenetic inhibition of the RE1-silencing transcription factor, Proc. Natl. Acad. Sci. USA 113 (1) (2016) E91–E100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Ren W, Yang X, Pathophysiology of long non-coding RNAs in ischemic stroke, Front. Mol. Neurosci 11 (2018) 96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Wang Y, Mo Y, Gong Z, Yang X, Yang M, Zhang S, Xiong F, Xiang B, Zhou M, Liao Q, Zhang W, Li X, Li X, Li Y, Li G, Zeng Z, Xiong W, Circular RNAs in human cancer, Mol. Cancer 16 (1) (2017) 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Wang J, Cao B, Han D, Sun M, Feng J, Long non-coding RNA H19 induces cerebral ischemia reperfusion injury via activation of autophagy, Aging Dis. 8 (1) (2017) 71–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Chen S, Wang M, Yang H, Mao L, He Q, Jin H, Ye ZM, Luo XY, Xia YP, Hu B, LncRNA TUG1 sponges microRNA-9 to promote neurons apoptosis by up-regulated Bcl2l11 under ischemia, Biochem. Biophys. Res. Commun 485 (1) (2017) 167–173. [DOI] [PubMed] [Google Scholar]
  • [86].Xu Q, Deng F, Xing Z, Wu Z, Cen B, Xu S, Zhao Z, Nepomuceno R, Bhuiyan MI, Sun D, Wang QJ, Ji A, Long non-coding RNA C2dat1 regulates CaMKIIδ expression to promote neuronal survival through the NF-κB signaling pathway following cerebral ischemia, Cell Death Dis. 7 (3) (2016) 2173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Zhang B, Wang D, Ji TF, Shi L, Yu JL, Overexpression of lncRNA ANRIL up-regulates VEGF expression and promotes angiogenesis of diabetes mellitus combined with cerebral infarction by activating NF-κB signaling pathway in a rat model, Oncotarget 8 (10) (2017) 17347–17359. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [88].Lee RC, Feinbaum RL, Ambros V, The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14, Cell 75 (5) (1993) 843–854. [DOI] [PubMed] [Google Scholar]
  • [89].Lee RC, Ambros V, An extensive class of small RNAs in Caenorhabditis elegans, Science 294 (5543) (2001) 862–864. [DOI] [PubMed] [Google Scholar]
  • [90].Lau NC, Lim LP, Weinstein EG, Bartel DP, An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans, Science 294 (5543) (2001) 858–862. [DOI] [PubMed] [Google Scholar]
  • [91].Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T, Identification of novel genes coding for small expressed RNAs, Science 294 (5543) (2001) 853–858. [DOI] [PubMed] [Google Scholar]
  • [92].Filipowicz W, Bhattacharyya SN, Sonenberg N, Mechanisms of posttranscriptional regulation by microRNAs: are the answers in sight? Nat. Rev. Genet 9 (2) (2008) 102–114. [DOI] [PubMed] [Google Scholar]
  • [93].Lee Y, Kim M, Han J, Yeom KH, Lee S, Baek SH, Kim VN, MicroRNA genes are transcribed by RNA polymerase II, EMBO J. 23 (20) (2004) 4051–4060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Denli AM, Tops BB, Plasterk RH, Ketting RF, Hannon GJ, Processing of primary microRNAs by the Microprocessor complex, Nature 432 (7014) (2004) 231–235. [DOI] [PubMed] [Google Scholar]
  • [95].Gregory RI, Yan KP, Amuthan G, Chendrimada T, Doratotaj B, Cooch N, Shiekhattar R, The Microprocessor complex mediates the genesis of microRNAs, Nature 432 (7014) (2004) 235–240. [DOI] [PubMed] [Google Scholar]
  • [96].Han J, Lee Y, Yeom KH, Kim YK, Jin H, Kim VN, The Drosha-DGCR8 complex in primary microRNA processing, Genes Dev. 18 (24) (2004) 3016–3027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, Lee J, Provost P, Rådmark O, Kim S, Kim VN, The nuclear RNase III Drosha initiates microRNA processing, Nature 425 (6956) (2003) 415–419. [DOI] [PubMed] [Google Scholar]
  • [98].Heo I, Ha M, Lim J, Yoon MJ, Park JE, Kwon SC, Chang H, Kim VN, Monouridylation of pre-microRNA as a key step in the biogenesis of group II let-7 microRNAs, Cell 151 (3) (2012) 521–532. [DOI] [PubMed] [Google Scholar]
  • [99].Burke JM, Kelenis DP, Kincaid RP, Sullivan CS, A central role for the primary microRNA stem in guiding the position and efficiency of Drosha processing of a viral pri-miRNA, RNA 20 (7) (2014) 1068–1077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Zeng Y, Yi R, Cullen BR, Recognition and cleavage of primary microRNA precursors by the nuclear processing enzyme Drosha, EMBO J. 24 (1) (2005) 138–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Han J, Lee Y, Yeom KH, Nam JW, Heo I, Rhee JK, Sohn SY, Cho Y, Zhang BT, Kim VN, Molecular basis for the recognition of primary microRNAs by the Drosha-DGCR8 complex, Cell 125 (5) (2006) 887–901. [DOI] [PubMed] [Google Scholar]
  • [102].Bohnsack MT, Czaplinski K, Görlich D, Exportin 5 is a RanGTP-dependent dsRNA-binding protein that mediates nuclear export of pre-miRNAs, RNA 10 (2) (2004) 185–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Lund E, Güttinger S, Calado A, Dahlberg JE, Kutay U, Nuclear export of microRNA precursors, Science 303 (5654) (2004) 95–98. [DOI] [PubMed] [Google Scholar]
  • [104].Yi R, Qin Y, Macara IG, Cullen BR, Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs, Genes Dev. 17 (24) (2003) 3011–3016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Park J-E, Heo I, Tian Y, Simanshu DK, Chang H, Jee D, Patel DJ, Kim VN, Dicer recognizes the 5′ end of RNA for efficient and accurate processing, Nature 475 (7355) (2011) 201–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Bernstein E, Caudy AA, Hammond SM, Hannon GJ, Role for a bidentate ribonuclease in the initiation step of RNA interference, Nature 409 (6818) (2001) 363–366. [DOI] [PubMed] [Google Scholar]
  • [107].Chendrimada TP, Gregory RI, Kumaraswamy E, Norman J, Cooch N, Nishikura K, Shiekhattar R, TRBP recruits the Dicer complex to Ago2 for microRNA processing and gene silencing, Nature 436 (7051) (2005) 740–744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Kim Y, Yeo J, Lee JH, Cho J, Seo D, Kim JS, Kim VN, Deletion of human tarbp2 reveals cellular microRNA targets and cell-cycle function of TRBP, Cell Rep. 9 (3) (2014) 1061–1074. [DOI] [PubMed] [Google Scholar]
  • [109].Lee HY, Doudna JA, TRBP alters human precursor microRNA processing in vitro, RNA 18 (11) (2012) 2012–2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Eulalio A, Behm-Ansmant I, Schweizer D, Izaurralde E, P-body formation is a consequence, not the cause, of RNA-mediated gene silencing, Mol. Cell. Biol 27 (11) (2007) 3970–3981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Liu J, Valencia-Sanchez MA, Hannon GJ, Parker R, MicroRNA-dependent localization of targeted mRNAs to mammalian P-bodies, Nat. Cell Biol 7 (7) (2005) 719–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].Gregory RI, Chendrimada TP, Cooch N, Shiekhattar R, Human RISC couples microRNA biogenesis and posttranscriptional gene silencing, Cell 123 (4) (2005) 631–640. [DOI] [PubMed] [Google Scholar]
  • [113].Yin K-J, Deng Z, Huang H, Hamblin M, Xie C, Zhang J, Chen YE, miR-497 regulates neuronal death in mouse brain after transient focal cerebral ischemia, Neurobiol. Dis 38 (1) (2010) 17–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Tao Z, Zhao H, Wang R, Liu P, Yan F, Zhang C, Ji X, Luo Y, Neuroprotective effect of microRNA-99a against focal cerebral ischemia–reperfusion injury in mice, J. Neurol. Sci 355 (1–2) (2015) 113–119. [DOI] [PubMed] [Google Scholar]
  • [115].Buller B, Liu X, Wang X, Zhang RL, Zhang L, Hozeska-Solgot A, Chopp M, Zhang ZG, MicroRNA-21 protects neurons from ischemic death, FEBS J. 277 (20) (2010) 4299–4307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Zhao H, Wang J, Gao L, Wang R, Liu X, Gao Z, Tao Z, Xu C, Song J, Ji X, Luo Y, MiRNA-424 protects against permanent focal cerebral ischemia injury in mice involving suppressing microglia activation, Stroke 44 (6) (2013) 1706–1713. [DOI] [PubMed] [Google Scholar]
  • [117].Yuan B, Shen H, Lin L, Su T, Zhong L, Yang Z, MicroRNA367 negatively regulates the inflammatory response of microglia by targeting IRAK4 in intracerebral hemorrhage, J. Neuroinflamm 12 (1) (2015) 206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Kong F, Zhou J, Zhou W, Guo Y, Li G, Yang L, Protective role of microRNA-126 in intracerebral hemorrhage, Mol. Med. Rep 15 (3) (2017) 1419–1425. [DOI] [PubMed] [Google Scholar]
  • [119].Zhang Y, Han B, He Y, Li D, Ma X, Liu Q, Hao J, MicroRNA-132 attenuates neurobehavioral and neuropathological changes associated with intracerebral hemorrhage in mice, Neurochem. Int 107 (2017) 182–190. [DOI] [PubMed] [Google Scholar]
  • [120].Zeng L, He X, Wang Y, Tang Y, Zheng C, Cai H, Liu J, Wang Y, Fu Y, Yang GY, MicroRNA-210 overexpression induces angiogenesis and neurogenesis in the normal adult mouse brain, Gene Ther. 21 (1) (2014) 37–43. [DOI] [PubMed] [Google Scholar]
  • [121].Liu Y, Pan Q, Zhao Y, He C, Bi K, Chen Y, Zhao B, Chen Y, Ma X, MicroRNA-155 regulates ROS production, NO generation, apoptosis and multiple functions of human brain microvessel endothelial cells under physiological and pathological conditions, J. Cell. Biochem 116 (12) (2015) 2870–2881. [DOI] [PubMed] [Google Scholar]
  • [122].Yin M, Chen Z, Ouyang Y, Zhang H, Wan Z, Wang H, Wu W, Yin X, Thrombin-induced, TNFR-dependent miR-181c downregulation promotes MLL1 and NF-κB target gene expression in human microglia, J. Neuroinflamm 14 (1) (2017) 132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Schratt GM, Tuebing F, Nigh EA, Kane CG, Sabatini ME, Kiebler M, Greenberg ME, A brain-specific microRNA regulates dendritic spine development, Nature 439 (7074) (2006) 283–289. [DOI] [PubMed] [Google Scholar]
  • [124].Xiao X, Jiang Y, Liang W, Wang Y, Cao S, Yan H, Gao L, Zhang L, miR-212-5p attenuates ferroptotic neuronal death after traumatic brain injury by targeting Ptgs2, Mol. Brain 12 (1) (2019) 78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Venkat P, Cui C, Chopp M, Zacharek A, Wang F, Landschoot-Ward J, Shen Y, Chen J, MiR-126 mediates brain endothelial cell exosome treatment–induced neurorestorative effects after stroke in type 2 diabetes mellitus mice, Stroke 50 (10) (2019) 2865–2874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Xie W, Li M, Xu N, Lv Q, Huang N, He J, Zhang Y, MiR-181a regulates inflammation responses in monocytes and macrophages, PLoS One 8 (3) (2013) 58639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Moon J.-m, Xu L, Giffard RG, Inhibition of microRNA-181 reduces forebrain ischemia-induced neuronal loss, J. Cereb. Blood Flow Metab 33 (12) (2013) 1976–1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Turrin NP, Rivest S, Molecular and cellular immune mediators of neuroprotection, Mol. Neurobiol 34 (3) (2006) 221–242. [DOI] [PubMed] [Google Scholar]
  • [129].Brown GC, Neher JJ, Inflammatory neurodegeneration and mechanisms of microglial killing of neurons, Mol. Neurobiol 41 (2–3) (2010) 242–247. [DOI] [PubMed] [Google Scholar]
  • [130].Ponomarev ED, Veremeyko T, Weiner HL, MicroRNAs are universal regulators of differentiation, activation, and polarization of microglia and macrophages in normal and diseased CNS, Glia 61 (1) (2013) 91–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Chamorro An, Hallenbeck J, The harms and benefits of inflammatory and immune responses in vascular disease, Stroke 37 (2) (2006) 291–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Moskowitz MA, Lo EH, Iadecola C, The science of stroke: mechanisms in search of treatments, Neuron 67 (2) (2010) 181–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Bazzoni F, et al. , Induction and regulatory function of miR-9 in human monocytes and neutrophils exposed to proinflammatory signals, Proc. Natl. Acad. Sci. USA 106 (13) (2009) 5282–5287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Amantea D, Nappi G, Bernardi G, Bagetta G, Corasaniti MT, Post-ischemic brain damage: pathophysiology and role of inflammatory mediators, FEBS J. 276 (1) (2009) 13–26. [DOI] [PubMed] [Google Scholar]
  • [135].Eikelenboom P, Rozemuller AJ, Hoozemans JJ, Veerhuis R, van Gool WA, Neuroinflammation and Alzheimer disease: clinical and therapeutic implications, Alzheimer Dis. Assoc. Disord 14 (1) (2000) S54–S61. [DOI] [PubMed] [Google Scholar]
  • [136].Mrak RE, Griffin WST, Glia and their cytokines in progression of neurodegeneration, Neurobiol. Aging 26 (3) (2005) 349–354. [DOI] [PubMed] [Google Scholar]
  • [137].Kong F-Y, Zhang JW, Li RF, Wang ZX, Wang WJ, Wang W, Unique roles of gold nanoparticles in drug delivery, targeting and imaging applications, Molecules 22 (9) (2017) 1445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Bache S, Rasmussen R, Rossing M, Laigaard FP, Nielsen FC, Møller K, MicroRNA changes in cerebrospinal fluid after subarachnoid hemorrhage, Stroke 48 (9) (2017) 2391–2398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Ferrarese C, Mascarucci P, Zoia C, Cavarretta R, Frigo M, Begni B, Sarinella F, Frattola L, De Simoni MG, Increased cytokine release from peripheral blood cells after acute stroke, J. Cereb. Blood Flow. Metab 19 (9) (1999) 1004–1009. [DOI] [PubMed] [Google Scholar]
  • [140].Guo D, Liu J, Wang W, Hao F, Sun X, Wu X, Bu P, Zhang Y, Liu Y, Liu F, Zhang Q, Jiang F, Alteration in abundance and compartmentalization of inflammation-related miRNAs in plasma after intracerebral hemorrhage, Stroke 44 (6) (2013) 1739–1742. [DOI] [PubMed] [Google Scholar]
  • [141].Tan JR, Kaur P, Liu F, Armugam A, Wong PT, Jeyaseelan K, Koo YX, microRNAs in stroke pathogenesis, Curr. Mol. Med 11 (2) (2011) 76–92. [DOI] [PubMed] [Google Scholar]
  • [142].Saugstad JA, MicroRNAs as effectors of brain function with roles in ischemia and injury, neuroprotection, and neurodegeneration, J. Cereb. Blood Flow. Metab 30 (9) (2010) 1564–1576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Ni J, Wang X, Chen S, Liu H, Wang Y, Xu X, Cheng J, Jia J, Zhen X, MicroRNA let-7c-5p protects against cerebral ischemia injury via mechanisms involving the inhibition of microglia activation, Brain Behav. Immun 49 (2015) 75–85. [DOI] [PubMed] [Google Scholar]
  • [144].Laterza OF, Lim L, Garrett-Engele PW, Vlasakova K, Muniappa N, Tanaka WK, Johnson JM, Sina JF, Fare TL, Sistare FD, Glaab WE, Plasma MicroRNAs as sensitive and specific biomarkers of tissue injury, Clin. Chem 55 (11) (2009) 1977–1983. [DOI] [PubMed] [Google Scholar]
  • [145].Meza-Sosa KF, Pedraza-Alva G, Pérez-Martínez L, microRNAs: key triggers of neuronal cell fate, Front. Cell. Neurosci 8 (2014) 175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Karikó K, Ni H, Capodici J, Lamphier M, Weissman D, mRNA is an endogenous ligand for Toll-like receptor 3, J. Biol. Chem 279 (13) (2004) 12542–12550. [DOI] [PubMed] [Google Scholar]
  • [147].Kawai T, Akira S, The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors, Nat. Immunol 11 (5) (2010) 373–384. [DOI] [PubMed] [Google Scholar]
  • [148].Weinstein JR, Koerner IP, Möller T, Microglia in ischemic brain injury, Fut. Neurol 5 (2) (2010) 227–246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Zhang L, Li YJ, Wu XY, Hong Z, Wei WS, Micro RNA-181c negatively regulates the inflammatory response in oxygen-glucose-deprived microglia by targeting Toll-like receptor 4, J. Neurochem 132 (6) (2015) 713–723. [DOI] [PubMed] [Google Scholar]
  • [150].Yang Z, Zhong L, Xian R, Yuan B, MicroRNA-223 regulates inflammation and brain injury via feedback to NLRP3 inflammasome after intracerebral hemorrhage, Mol. Immunol 65 (2) (2015) 267–276. [DOI] [PubMed] [Google Scholar]
  • [151].Ruffell D, et al. , A CREB-C/EBPβ cascade induces M2 macrophage-specific gene expression and promotes muscle injury repair, Proc. Natl. Acad. Sci. USA 106 (41) (2009) 17475–17480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [152].Martinez-Nunez RT, Louafi F, Sanchez-Elsner T, The interleukin 13 (IL-13) pathway in human macrophages is modulated by microRNA-155 via direct targeting of interleukin 13 receptor α1 (IL13Rα1), J. Biol. Chem 286 (3) (2011) 1786–1794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [153].Elmore S, Apoptosis: a review of programmed cell death, Toxicol. Pathol 35 (4) (2007) 495–516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Rathmell JC, Thompson CB, Pathways of apoptosis in lymphocyte development, homeostasis, and disease, Cell 109 (2) (2002) S97–S107. [DOI] [PubMed] [Google Scholar]
  • [155].Meier P, Finch A, Evan G, Apoptosis in development, Nature 407 (6805) (2000) 796–801. [DOI] [PubMed] [Google Scholar]
  • [156].Shao A, Wang Z, Wu H, Dong X, Li Y, Tu S, Tang J, Zhao M, Zhang J, Hong Y, Enhancement of autophagy by histone deacetylase inhibitor trichostatin a ameliorates neuronal apoptosis after subarachnoid hemorrhage in rats, Mol. Neurobiol 53 (1) (2016) 18–27. [DOI] [PubMed] [Google Scholar]
  • [157].Reed JC, Apoptosis-based therapies, Nat. Rev. Drug Discov 1 (2) (2002) 111–121. [DOI] [PubMed] [Google Scholar]
  • [158].Kroemer G, Galluzzi L, Brenner C, Mitochondrial membrane permeabilization in cell death, Physiol. Rev 87 (1) (2007) 99–163. [DOI] [PubMed] [Google Scholar]
  • [159].Adams JM, Ways of dying: multiple pathways to apoptosis, Genes Dev. 17 (20) (2003) 2481–2495. [DOI] [PubMed] [Google Scholar]
  • [160].Culmsee C, Zhu C, Landshamer S, Becattini B, Wagner E, Pellecchia M, Blomgren K, Plesnila N, Apoptosis-inducing factor triggered by poly (ADP-ribose) polymerase and Bid mediates neuronal cell death after oxygen-glucose deprivation and focal cerebral ischemia, J. Neurosci 25 (44) (2005) 10262–10272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [161].Broughton BR, Reutens DC, Sobey CG, Apoptotic mechanisms after cerebral ischemia, Stroke 40 (5) (2009) e331–e339. [DOI] [PubMed] [Google Scholar]
  • [162].Cardoso AL, Guedes JR, Pereira de Almeida L, Pedroso de Lima MC, miR-155 modulates microglia-mediated immune response by down-regulating SOCS-1 and promoting cytokine and nitric oxide production, Immunology 135 (1) (2012) 73–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].Peng Z, Li J, Li Y, Yang X, Feng S, Han S, Li J, Downregulation of miR-181b in mouse brain following ischemic stroke induces neuroprotection against ischemic injury through targeting heat shock protein A5 and ubiquitin carboxyl-terminal hydrolase isozyme L1, J. Neurosci. Res 91 (10) (2013) 1349–1362. [DOI] [PubMed] [Google Scholar]
  • [164].Liu D-Z, Tian Y, Ander BP, Xu H, Stamova BS, Zhan X, Turner RJ, Jickling G, Sharp FR, Brain and blood microRNA expression profiling of ischemic stroke, intracerebral hemorrhage, and kainate seizures, J. Cereb. Blood Flow Metab 30 (1) (2010) 92–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].Zhang L, Zhang S, Yao J, Lowery FJ, Zhang Q, Huang WC, Li P, Li M, Wang X, Zhang C, Wang H, Ellis K, Cheerathodi M, McCarty JH, Palmieri D, Saunus J, Lakhani S, Huang S, Sahin AA, Aldape KD, Steeg PS, Yu D, Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth, Nature 527 (7576) (2015) 100–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [166].Zhai F, Zhang X, Guan Y, Yang X, Li Y, Song G, Guan L, Expression profiles of microRNAs after focal cerebral ischemia/reperfusion injury in rats, Neural Regen. Res 7 (12) (2012) 917–923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Xu W, Gao L, Zheng J, Li T, Shao A, Reis C, Chen S, Zhang J, The roles of microRNAs in stroke: possible therapeutic targets, Cell Transplant. 27 (12) (2018) 1778–1788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [168].Valko M, Leibfritz D, Moncol J, Cronin MTD, Mazur M, Telser J, Free radicals and antioxidants in normal physiological functions and human disease, Int. J. Biochem. Cell Biol 39 (1) (2007) 44–84. [DOI] [PubMed] [Google Scholar]
  • [169].Giasson BI, Duda JE, Murray IV, Chen Q, Souza JM, Hurtig HI, Ischiropoulos H, Trojanowski JQ, Lee VM, Oxidative damage linked to neurodegeneration by selective α-synuclein nitration in synucleinopathy lesions, Science 290 (5493) (2000) 985–989. [DOI] [PubMed] [Google Scholar]
  • [170].Gasche Y, Copin JC, Sugawara T, Fujimura M, Chan PH, Matrix metalloproteinase inhibition prevents oxidative stress-associated blood-brain barrier disruption after transient focal cerebral ischemia, J. Cereb. Blood Flow. Metab 21 (12) (2001) 1393–1400. [DOI] [PubMed] [Google Scholar]
  • [171].Cherubini A, Ruggiero C, Polidori MC, Mecocci P, Potential markers of oxidative stress in stroke, Free Radic. Biol. Med 39 (7) (2005) 841–852. [DOI] [PubMed] [Google Scholar]
  • [172].Lafon-Cazal M, Pietri S, Culcasi M, Bockaert J, NMDA-dependent superoxide production and neurotoxicity, Nature 364 (6437) (1993) 535–537. [DOI] [PubMed] [Google Scholar]
  • [173].Cuzzocrea S, et al. , Antioxidant therapy: a new pharmacological approach in shock, inflammation, and ischemia/reperfusion injury, Pharmacol. Rev 53 (1) (2001) 135–159. [PubMed] [Google Scholar]
  • [174].Coyle JT, Puttfarcken P, Oxidative stress, glutamate, and neurodegenerative disorders, Science 262 (5134) (1993) 689–695. [DOI] [PubMed] [Google Scholar]
  • [175].Piantadosi CA, Zhang J, Mitochondrial generation of reactive oxygen species after brain ischemia in the rat, Stroke 27 (2) (1996) 327–332. [DOI] [PubMed] [Google Scholar]
  • [176].Spranger M, Krempien S, Schwab S, Donneberg S, Hacke W, Superoxide dismutase activity in serum of patients with acute cerebral ischemic injury: correlation with clinical course and infarct size, Stroke 28 (12) (1997) 2425–2428. [DOI] [PubMed] [Google Scholar]
  • [177].Gariballa S, Hutchin T, Sinclair A, Antioxidant capacity after acute ischaemic stroke, Qjm 95 (10) (2002) 685–690. [DOI] [PubMed] [Google Scholar]
  • [178].Johnson JA, Johnson DA, Kraft AD, Calkins MJ, Jakel RJ, Vargas MR, Chen PC, The Nrf2-ARE pathway: an indicator and modulator of oxidative stress in neurodegeneration, Ann. N. Y. Acad. Sci 1147 (2008) 61–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Papp D, Lenti K, Módos D, Fazekas D, Dúl Z, Türei D, Földvári-Nagy L, Nussinov R, Csermely P, Korcsmáros T, The NRF2–related interactome and regulome contain multifunctional proteins and fine-tuned autoregulatory loops, FEBS Lett. 586 (13) (2012) 1795–1802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [180].Wang P, Liang X, Lu Y, Zhao X, Liang J, MicroRNA-93 downregulation ameliorates cerebral ischemic injury through the Nrf2/HO-1 defense pathway, Neurochem. Res 41 (10) (2016) 2627–2635. [DOI] [PubMed] [Google Scholar]
  • [181].Liu P, Zhao H, Wang R, Wang P, Tao Z, Gao L, Yan F, Liu X, Yu S, Ji X, Luo Y, MicroRNA-424 protects against focal cerebral ischemia and reperfusion injury in mice by suppressing oxidative stress, Stroke 46 (2) (2015) 513–519. [DOI] [PubMed] [Google Scholar]
  • [182].Zhao H, Tao Z, Wang R, Liu P, Yan F, Li J, Zhang C, Ji X, Luo Y, MicroRNA-23a-3p attenuates oxidative stress injury in a mouse model of focal cerebral ischemia-reperfusion, Brain Res. 1592 (2014) 65–72. [DOI] [PubMed] [Google Scholar]
  • [183].Li P, Shen M, Gao F, Wu J, Zhang J, Teng F, Zhang C, An antagomir to microRNA-106b-5p ameliorates cerebral ischemia and reperfusion injury in rats via inhibiting apoptosis and oxidative stress, Mol. Neurobiol 54 (4) (2017) 2901–2921. [DOI] [PubMed] [Google Scholar]
  • [184].Dharap A, Bowen K, Place R, Li LC, Vemuganti R, Transient focal ischemia induces extensive temporal changes in rat cerebral microRNAome, J. Cereb. Blood Flow Metab 29 (4) (2009) 675–687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Xu W, Li F, Liu Z, Xu Z, Sun B, Cao J, Liu Y, MicroRNA-27b inhibition promotes Nrf2/ARE pathway activation and alleviates intracerebral hemorrhage-induced brain injury, Oncotarget 8 (41) (2017) 70669–70684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Zhang RL, Chopp M, Roberts C, Liu X, Wei M, Nejad-Davarani SP, Wang X, Zhang ZG, Stroke increases neural stem cells and angiogenesis in the neurogenic niche of the adult mouse, PLoS One 9 (12) (2014), 113972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [187].Wu F, Yang Z, Li G, Role of specific microRNAs for endothelial function and angiogenesis, Biochem. Biophys. Res. Commun 386 (4) (2009) 549–553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [188].Ma X-L, Li S-Y, Shang F, Effect of microRNA-129-5p targeting HMGB1-RAGE signaling pathway on revascularization in a collagenase-induced intracerebral hemorrhage rat model, Biomed. Pharmacother 93 (2017) 238–244. [DOI] [PubMed] [Google Scholar]
  • [189].Li M, Zhang J, Circulating MicroRNAs: potential and emerging biomarkers for diagnosis of cardiovascular and cerebrovascular diseases, BioMed Res. Int 2015 (2015) 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [190].Cheng YD, Al-Khoury L, Zivin JA, Neuroprotection for ischemic stroke: two decades of success and failure, NeuroRx 1 (1) (2004) 36–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [191].Mellios N, Huang HS, Grigorenko A, Rogaev E, Akbarian S, A set of differentially expressed miRNAs, including miR-30a-5p, act as posttranscriptional inhibitors of BDNF in prefrontal cortex, Hum. Mol. Genet 17 (19) (2008) 3030–3042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [192].Zhang R, Zhang Z, Wang L, Wang Y, Gousev A, Zhang L, Ho KL, Morshead C, Chopp M, Activated neural stem cells contribute to stroke-induced neurogenesis and neuroblast migration toward the infarct boundary in adult rats, J. Cereb. Blood Flow Metab 24 (4) (2004) 441–448. [DOI] [PubMed] [Google Scholar]
  • [193].Liu FJ, Lim KY, Kaur P, Sepramaniam S, Armugam A, Wong PT, Jeyaseelan K, microRNAs involved in regulating spontaneous recovery in embolic stroke model, PLoS One 8 (6) (2013) 66393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [194].Chen J, Ning R, Zacharek A, Cui C, Cui X, Yan T, Venkat P, Zhang Y, Chopp M, M i R-126 contributes to human umbilical cord blood cell-induced neurorestorative effects after stroke in type-2 diabetic mice, Stem Cells 34 (1) (2016) 102–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [195].Lusardi TA, Murphy SJ, Phillips JI, Chen Y, Davis CM, Young JM, Thompson SJ, Saugstad JA, MicroRNA responses to focal cerebral ischemia in male and female mouse brain, Front. Mol. Neurosci 7 (2014) 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [196].Tan JR, Tan KS, Koo YX, Yong FL, Wang CW, Armugam A, Jeyaseelan K, Blood microRNAs in low or no risk ischemic stroke patients, Int. J. Mol. Sci 14 (1) (2013) 2072–2084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [197].Wu P, Zuo X, Ji A, Stroke-induced microRNAs: The potential therapeutic role for stroke, Exp. Ther. Med 3 (4) (2012) 571–576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [198].Long G, Wang F, Li H, Yin Z, Sandip C, Lou Y, Wang Y, Chen C, Wang DW, Circulating miR-30a, miR-126 and let-7b as biomarker for ischemic stroke in humans, BMC Neurol. 13 (2013) 178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [199].Tan KS, Armugam A, Sepramaniam S, Lim KY, Setyowati KD, Wang CW, Jeyaseelan K, Expression profile of MicroRNAs in young stroke patients, PLoS One 4 (11) (2009) 7689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [200].Jeyaseelan K, Lim KY, Armugam A, MicroRNA expression in the blood and brain of rats subjected to transient focal ischemia by middle cerebral artery occlusion, Stroke 39 (3) (2008) 959–966. [DOI] [PubMed] [Google Scholar]
  • [201].Selvamani A, Williams MH, Miranda RC, Sohrabji F, Circulating miRNA profiles provide a biomarker for severity of stroke outcomes associated with age and sex in a rat model, Clin. Sci 127 (2) (2014) 77–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [202].Dharap A, Bowen K, Place R, Li LC, Vemuganti R, Transient focal ischemia induces extensive temporal changes in rat cerebral microRNAome, J. Cereb. Blood Flow Metab 29 (4) (2009) 675–687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [203].Liu DZ, Tian Y, Ander BP, Xu H, Stamova BS, Zhan X, Turner RJ, Jickling G, Sharp FR, Brain and blood microRNA expression profiling of ischemic stroke, intracerebral hemorrhage, and kainate seizures, J. Cereb. Blood Flow Metab 30 (1) (2010) 92–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [204].Ghosh G, Subramanian IV, Adhikari N, Zhang X, Joshi HP, Basi D, Chandrashekhar YS, Hall JL, Roy S, Zeng Y, Ramakrishnan S, Hypoxia-induced microRNA-424 expression in human endothelial cells regulates HIF-alpha isoforms and promotes angiogenesis, J. Clin. Investig 120 (11) (2010) 4141–4154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [205].Laterza OF, Lim L, Garrett-Engele PW, Vlasakova K, Muniappa N, Tanaka WK, Johnson JM, Sina JF, Fare TL, Sistare FD, Glaab WE, Plasma MicroRNAs as sensitive and specific biomarkers of tissue injury, Clin. Chem 55 (11) (2009) 1977–1983. [DOI] [PubMed] [Google Scholar]
  • [206].Weng H, Shen C, Hirokawa G, Ji X, Takahashi R, Shimada K, Kishimoto C, Iwai N, Plasma miR-124 as a biomarker for cerebral infarction, Biomed. Res 32 (2) (2011) 135–141. [DOI] [PubMed] [Google Scholar]
  • [207].Lopez-Verrilli MA, Exosomes: mediators of communication in eukaryotes, Biol. Res 46 (1) (2013) 5–11. [DOI] [PubMed] [Google Scholar]
  • [208].Simons M, Raposo G, Exosomes–vesicular carriers for intercellular communication, Curr. Opin. Cell Biol 21 (4) (2009) 575–581. [DOI] [PubMed] [Google Scholar]
  • [209].Mianehsaz E, Mirzaei HR, Mahjoubin-Tehran M, Rezaee A, Sahebnasagh R, Pourhanifeh MH, Mirzaei H, Hamblin MR, Mesenchymal stem cell-derived exosomes: a new therapeutic approach to osteoarthritis? Stem Cell Res. Ther 10 (1) (2019) 340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [210].Luarte A, Bátiz LF, Wyneken U, Lafourcade C, Potential therapies by stem cell-derived exosomes in CNS diseases: focusing on the neurogenic niche, Stem Cells Int. 2016 (2016), 5736059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [211].Xin H, Li Y, Chopp M, Exosomes/miRNAs as mediating cell-based therapy of stroke, Front. Cell. Neurosci 8 (2014) 377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [212].Théry C, et al. , Isolation and characterization of Exosomes from cell culture supernatants and biological fluids. Curr Curr Protoc Cell Biol Chapter 3: Unit 3.22. 2006. [DOI] [PubMed] [Google Scholar]
  • [213].Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO, Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells, Nat. Cell Biol 9 (6) (2007) 654–659. [DOI] [PubMed] [Google Scholar]
  • [214].Ghoreishy A, Khosravi A, Ghaemmaghami A, Exosomal microRNA and stroke: a review, J. Cell Biochem 120 (10) (2019) 16352–16361. [DOI] [PubMed] [Google Scholar]
  • [215].Emanueli C, Shearn AI, Angelini GD, Sahoo S, Exosomes and exosomal miRNAs in cardiovascular protection and repair, Vascul. Pharmacol 71 (2015) 24–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [216].Sun Z, Shi K, Yang S, Liu J, Zhou Q, Wang G, Song J, Li Z, Zhang Z, Yuan W, Effect of exosomal miRNA on cancer biology and clinical applications, Mol. Cancer 17 (1) (2018) 147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [217].Choi DS, Kim DK, Kim YK, Gho YS, Proteomics of extracellular vesicles: exosomes and ectosomes, Mass Spectrom. Rev 34 (4) (2015) 474–490. [DOI] [PubMed] [Google Scholar]
  • [218].Théry C, Ostrowski M, Segura E, Membrane vesicles as conveyors of immune responses, Nat. Rev. Immunol 9 (8) (2009) 581–593. [DOI] [PubMed] [Google Scholar]
  • [219].Lakkaraju A, Rodriguez-Boulan E, Itinerant exosomes: emerging roles in cell and tissue polarity, Trends Cell Biol. 18 (5) (2008) 199–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [220].Ling H, Fabbri M, Calin GA, MicroRNAs and other non-coding RNAs as targets for anticancer drug development, Nat. Rev. Drug Discov 12 (11) (2013) 847–865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [221].Lai CP-K, Breakefield XO, Role of exosomes/microvesicles in the nervous system and use in emerging therapies, Front. Physiol 3 (2012) 228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [222].Fruhbeis C, et al. , Extracellular vesicles as mediators of neuron-glia communication. Front. Cell Neurosci, 2013; 7: 182; 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [223].György B, Hung ME, Breakefield XO, Leonard JN, Therapeutic applications of extracellular vesicles: clinical promise and open questions, Annu. Rev. Pharmacol. Toxicol 55 (2015) 439–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [224].Perez-Gonzalez R, Gauthier SA, Kumar A, Levy E, The exosome secretory pathway transports amyloid precursor protein carboxyl-terminal fragments from the cell into the brain extracellular space, J. Biol. Chem 287 (51) (2012) 43108–43115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [225].Banigan MG, Kao PF, Kozubek JA, Winslow AR, Medina J, Costa J, Schmitt A, Schneider A, Cabral H, Cagsal-Getkin O, Vanderburg CR, Delalle I, Differential expression of exosomal microRNAs in prefrontal cortices of schizophrenia and bipolar disorder patients, PLoS One 8 (1) (2013) 48814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [226].Rajendran L, et al. , Alzheimer’s disease β-amyloid peptides are released in association with exosomes, Proc. Natl. Acad. Sci. USA 103 (30) (2006) 11172–11177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [227].Dinkins MB, Dasgupta S, Wang G, Zhu G, Bieberich E, Exosome reduction in vivo is associated with lower amyloid plaque load in the 5XFAD mouse model of Alzheimer’s disease, Neurobiol. Aging 35 (8) (2014) 1792–1800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [228].Gareev I, Yang G, Sun J, Beylerli O, Chen X, Zhang D, Zhao B, Zhang R, Sun Z, Yang Q, Li L, Pavlov V, Safin S, Zhao S, Circulating MicroRNAs as potential noninvasive biomarkers of spontaneous intracerebral hemorrhage, World Neurosurg. 133 (2020) e369–e375. [DOI] [PubMed] [Google Scholar]
  • [229].Ouyang Y, Li D, Wang H, Wan Z, Luo Q, Zhong Y, Yin M, Qing Z, Li Z, Bao B, Chen Z, Yin X, Zhu LQ, MiR-21–5p/dual-specificity phosphatase 8 signalling mediates the anti-inflammatory effect of haem oxygenase-1 in aged intracerebral haemorrhage rats, Aging Cell 18 (6) (2019) 13022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [230].Yang W, Zhang J, Xu B, He Y, Liu W, Li J, Zhang S, Lin X, Su D, Wu T, Li J, HucMSC-derived exosomes mitigate the age-related retardation of fertility in female mice, Mol. Ther. J. Am. Soc. Gene Ther 28 (2020) 1200–1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [231].Baglio SR, Rooijers K, Koppers-Lalic D, Verweij FJ, Pérez Lanzón M, Zini N, Naaijkens B, Perut F, Niessen HW, Baldini N, Pegtel DM, Human bone marrow-and adipose-mesenchymal stem cells secrete exosomes enriched in distinctive miRNA and tRNA species, Stem Cell Res. Ther 6 (1) (2015) 127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [232].Xie Y, Chu A, Feng Y, Chen L, Shao Y, Luo Q, Deng X, Wu M, Shi X, Chen Y, MicroRNA-146a: a comprehensive indicator of inflammation and oxidative stress status induced in the brain of chronic T2DM rats, Front. Pharmacol 9 (2018) 478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [233].Chu B, Zhou Y, Zhai H, Li L, Sun L, Li Y, The role of microRNA-146a in regulating the expression of IRAK1 in cerebral ischemia–reperfusion injury, Can. J. Physiol. Pharmacol 96 (6) (2018) 611–617. [DOI] [PubMed] [Google Scholar]
  • [234].Qu X, Wang N, Cheng W, Xue Y, Chen W, Qi M, MicroRNA-146a protects against intracerebral hemorrhage by inhibiting inflammation and oxidative stress, Exp. Ther. Med 18 (5) (2019) 3920–3928. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [235].Zhang H, Lu M, Zhang X, Kuai Y, Mei Y, Tan Q, Zhong K, Sun X, Tan W, Isosteviol Sodium Protects against Ischemic Stroke by Modulating Microglia/Macrophage Polarization via Disruption of GAS5/miR-146a-5p sponge, Sci. Rep 9 (1) (2019) 12221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [236].Hu YL, Wang H, Huang Q, Wang G, Zhang HB, MicroRNA-23a-3p promotes the perihematomal edema formation after intracerebral hemorrhage via ZO-1, Eur. Rev. Med Pharmacol. Sci 22 (9) (2018) 2809–2816. [DOI] [PubMed] [Google Scholar]
  • [237].Duan S, Wang F, Cao J, Wang C, Exosomes derived from microRNA-146a-5p-enriched bone marrow mesenchymal stem cells alleviate intracerebral hemorrhage by inhibiting neuronal apoptosis and microglial M1 polarization, Drug Des. Dev. Ther 14 (2020) 3143–3158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [238].Péron S, Berninger B, Imported stem cells strike against stroke, Cell Stem Cell 17 (5) (2015) 501–502. [DOI] [PubMed] [Google Scholar]
  • [239].Boldrini M, Fulmore CA, Tartt AN, Simeon LR, Pavlova I, Poposka V, Rosoklija GB, Stankov A, Arango V, Dwork AJ, Hen R, Mann JJ, Human hippocampal neurogenesis persists throughout aging, Cell Stem Cell 22 (4) (2018) 589–599. e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [240].Yamashita T, Ninomiya M, Hernández Acosta P, García-Verdugo JM, Sunabori T, Sakaguchi M, Adachi K, Kojima T, Hirota Y, Kawase T, Araki N, Abe K, Okano H, Sawamoto K, Subventricular zone-derived neuroblasts migrate and differentiate into mature neurons in the post-stroke adult striatum, J. Neurosci 26 (24) (2006) 6627–6636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [241].Ngalula KP, Cramer N, Schell MJ, Juliano SL, Transplanted neural progenitor cells from distinct sources migrate differentially in an organotypic model of brain injury, Front. Neurol 6 (2015) 212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [242].Zhang S, Jin T, Wang L, Liu W, Zhang Y, Zheng Y, Lin Y, Yang M, He X, Lin H, Chen L, Tao J, Electro-acupuncture promotes the differentiation of endogenous neural stem cells via exosomal microRNA 146b after ischemic stroke, Front. Cell Neurosci 14 (2020) 223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [243].Xi T, Jin F, Zhu Y, Wang J, Tang L, Wang Y, Liebeskind DS, He Z, MicroRNA-126-3p attenuates blood-brain barrier disruption, cerebral edema and neuronal injury following intracerebral hemorrhage by regulating PIK3R2 and Akt, Biochem. Biophys. Res. Commun 494 (1–2) (2017) 144–151. [DOI] [PubMed] [Google Scholar]
  • [244].Harris TA, et al. , MicroRNA-126 regulates endothelial expression of vascular cell adhesion molecule 1, Proc. Natl. Acad. Sci. USA 105 (5) (2008) 1516–1521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [245].Lasek-Bal A, et al. , The importance of selected markers of inflammation and blood-brain barrier damage for short-term ischemic stroke prognosis, J. Physiol. Pharmacol 70 (2019) 2. [DOI] [PubMed] [Google Scholar]
  • [246].Garcia-Bonilla L, Benakis C, Moore J, Iadecola C, Anrather J, Immune mechanisms in cerebral ischemic tolerance, Front. Neurosci 8 (2014) 44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [247].Dock H, Theodorsson A, Theodorsson E, DNA methylation inhibitor zebularine confers stroke protection in ischemic rats, Transl. Stroke Res 6 (4) (2015) 296–300. [DOI] [PubMed] [Google Scholar]
  • [248].Endres M, Meisel A, Biniszkiewicz D, Namura S, Prass K, Ruscher K, Lipski A, Jaenisch R, Moskowitz MA, Dirnagl U, DNA methyltransferase contributes to delayed ischemic brain injury, J. Neurosci 20 (9) (2000) 3175–3181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [249].Pandi G, Nakka VP, Dharap A, Roopra A, Vemuganti R, MicroRNA miR-29c down-regulation leading to de-repression of its target DNA methyltransferase 3a promotes ischemic brain damage, PLoS One 8 (3) (2013) 58039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [250].Hu Z, Zhong B, Tan J, Chen C, Lei Q, Zeng L, The emerging role of epigenetics in cerebral ischemia, Mol. Neurobiol 54 (3) (2017) 1887–1905. [DOI] [PubMed] [Google Scholar]
  • [251].Elliott EN, Sheaffer KL, Kaestner KH, The ‘de novo’ DNA methyltransferase Dnmt3b compensates the Dnmt1-deficient intestinal epithelium, Elife 5 (2016), e12975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [252].Cui J, Liu N, Chang Z, Gao Y, Bao M, Xie Y, Xu W, Liu X, Jiang S, Liu Y, Shi R, Xie W, Jia X, Shi J, Ren C, Gong K, Zhang C, Bade R, Shao G, Ji X, Exosomal MicroRNA-126 from RIPC serum is involved in hypoxia tolerance in SH-SY5Y cells by downregulating DNMT3B, Mol. Ther. Nucleic Acids 20 (2020) 649–660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [253].Xin H, Katakowski M, Wang F, Qian JY, Liu XS, Ali MM, Buller B, Zhang ZG, Chopp M, MicroRNA cluster miR-17–92 cluster in exosomes enhance neuroplasticity and functional recovery after stroke in rats, Stroke 48 (3) (2017) 747–753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [254].Xin H, Li Y, Liu Z, Wang X, Shang X, Cui Y, Zhang ZG, Chopp M, MiR-133b promotes neural plasticity and functional recovery after treatment of stroke with multipotent mesenchymal stromal cells in rats via transfer of exosome-enriched extracellular particles, Stem Cells 31 (12) (2013) 2737–2746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [255].Xin H, Wang F, Li Y, Lu QE, Cheung WL, Zhang Y, Zhang ZG, Chopp M, Secondary release of exosomes from astrocytes contributes to the increase in neural plasticity and improvement of functional recovery after stroke in rats treated with exosomes harvested from microRNA 133b-overexpressing multipotent mesenchymal stromal cells, Cell Transpl. 26 (2) (2017) 243–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [256].Jiang M, Wang H, Jin M, Yang X, Ji H, Jiang Y, Zhang H, Wu F, Wu G, Lai X, Cai L, Hu R, Xu L, Li L, Exosomes from MiR-30d-5p-ADSCs reverse acute ischemic stroke-induced, autophagy-mediated brain injury by promoting M2 microglial/macrophage polarization, Cell Physiol. Biochem 47 (2) (2018) 864–878. [DOI] [PubMed] [Google Scholar]
  • [257].Venkat P, Cui C, Chopp M, Zacharek A, Wang F, Landschoot-Ward J, Shen Y, Chen J, MiR-126 mediates brain endothelial cell exosome treatment-induced neurorestorative effects after stroke in type 2 diabetes mellitus mice, Stroke 50 (10) (2019) 2865–2874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [258].Song H, Zhang X, Chen R, Miao J, Wang L, Cui L, Ji H, Liu Y, Cortical neuron-derived exosomal microRNA-181c-3p inhibits neuroinflammation by downregulating CXCL1 in astrocytes of a rat model with ischemic brain injury, Neuroimmunomodulation 26 (5) (2019) 217–233. [DOI] [PubMed] [Google Scholar]
  • [259].Deng Y, Chen D, Gao F, Lv H, Zhang G, Sun X, Liu L, Mo D, Ma N, Song L, Huo X, Yan T, Zhang J, Miao Z, Exosomes derived from microRNA-138-5p-overexpressing bone marrow-derived mesenchymal stem cells confer neuroprotection to astrocytes following ischemic stroke via inhibition of LCN2, J. Biol. Eng 13 (2019) 71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [260].Yang J, Zhang X, Chen X, Wang L, Yang G, Exosome mediated delivery of miR-124 promotes neurogenesis after ischemia, Mol. Ther. Nucleic Acids 7 (2017) 278–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [261].Ling X, Zhang G, Xia Y, Zhu Q, Zhang J, Li Q, Niu X, Hu G, Yang Y, Wang Y, Deng Z, Exosomes from human urine-derived stem cells enhanced neurogenesis via miR-26a/HDAC6 axis after ischaemic stroke, J. Cell. Mol. Med 24 (1) (2020) 640–654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [262].Correction to: MicroRNA-17–92 Cluster in Exosomes Enhance Neuroplasticity and Functional Recovery After Stroke in Rats. Stroke, 2017. 48(5): p. e137. [DOI] [PubMed] [Google Scholar]
  • [263].Yang J, Zhang X, Chen X, Wang L, Yang G, Exosome mediated delivery of miR-124 promotes neurogenesis after ischemia, Mol. Ther.-Nucleic Acids 7 (2017) 278–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [264].Liu W, Wu J, Huang J, Zhuo P, Lin Y, Wang L, Lin R, Chen L, Tao J, Electroacupuncture regulates hippocampal synaptic plasticity via miR-134-mediated LIMK1 function in rats with ischemic stroke, Neural Plast. 2017 (2017), 9545646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [265].Huang X, Yuan T, Tschannen M, Sun Z, Jacob H, Du M, Liang M, Dittmar RL, Liu Y, Liang M, Kohli M, Thibodeau SN, Boardman L, Wang L, Characterization of human plasma-derived exosomal RNAs by deep sequencing, BMC Genom. 14 (2013) 319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [266].Xu X, Zhuang C, Chen L, Exosomal long non-coding RNA expression from serum of patients with acute minor stroke, Neuropsychiatr. Dis. Treat 16 (2020) 153–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [267].Chen HS, Tong HS, Zhao Y, Hong CY, Bin JP, Su L, Differential expression pattern of exosome long non-coding RNAs (lncRNAs) and MicroRNAs (miRNAs) in vascular endothelial cells under heat stroke, Med. Sci. Monit 24 (2018) 7965–7974. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES