Next Article in Journal
Spatial Omics in Clinical Research: A Comprehensive Review of Technologies and Guidelines for Applications
Previous Article in Journal
Acute Stress and Autoimmune Markers: Evaluating the Psychoneuroimmunology Axis in Firefighter Recruits
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advancing Ischemic Stroke Prognosis: Key Role of MiR-155 Non-Coding RNA

by
Catherine Hering
and
Gloria M. Conover
*
Department of Medical Education, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(9), 3947; https://doi.org/10.3390/ijms26093947
Submission received: 19 February 2025 / Revised: 11 April 2025 / Accepted: 17 April 2025 / Published: 22 April 2025
(This article belongs to the Section Molecular Neurobiology)

Abstract

:
Ischemic stroke (IS) is the leading cause of long-term disability and the second leading cause of death worldwide. It remains a significant clinical problem because only supportive therapies exist, such as thrombolytic agents and surgical thrombectomy, which do not restore function. Understanding the molecular pathogenesis of IS, including dysfunction in oxidative homeostasis, apoptosis, neuroinflammation and neuroprotection, is crucial to developing therapies. Non-coding RNAs (ncRNAs) are master regulators, and one ncRNA that stands out is miR-155, a pro-inflammatory micro-RNA elevated in stroke. This review addresses the biological mechanisms reported in the literature that support using miR-155 as a biomarker and therapeutic agent to treat IS in patients.

Graphical Abstract

1. Introduction

Stroke is a serious cardiovascular cause of morbidity and mortality [1,2,3,4,5,6]. In 2022, stroke was the third leading cause of disability worldwide and the second leading cause of death, with 7.5 million people diagnosed with acute ischemic stroke (IS) [6,7,8]. In the US, that same year, 1 in 6 people with cardiovascular disease died of stroke [8]. Globally, the incidence of stroke is 15 million people per year, and of these, 5 million people die from stroke and another 5 million will become disabled [4,5,6,9,10]. Risk factors that contribute to poor stroke prognosis span from lifestyle and environmental toxins to various accompanying comorbidities. Major modifiable risk factors for stroke are hypertension, diabetes mellitus, smoking, hyperlipidemia, obesity, poor diet/nutrition, and lack of exercise [11,12,13]. Of the modifiable risk factors, the highest is hypertension (54%) [14,15] while non-modifiable stroke risk factors are age, sex, and ethnicity [16].
Over 70% of strokes occur in patients over 65 years old, while ~75% of strokes occur in patients over 64 with males overall exhibiting a higher incidence (~33%) and prevalence (~41%) as compared to females [17,18]. Remarkably, non-Hispanic African Americans have ~2.2 times higher incidence of stroke as compared to Caucasian patients [19]. However, women over 75 years old have more strokes compared to men of the same age. After menopause, women have elevated FSH which promotes lipogenesis and fat storage, increasing androgen levels [20]. High-risk pregnancy, contraceptive use and substance abuse increase the risk for stroke in younger patients [21].
Non-coding RNAs (ncRNAs) are RNAs that do not encode functional proteins and were originally thought to regulate post-transcriptional gene expression [22]. Interestingly, many reports have shown that ncRNAs expression are detected when disease symptoms occur, making ncRNAs attractive disease biomarkers [23]. For example, deletions of miR-15a and miR-16 were found in 68% of chronic lymphocytic leukemia patients, and 296 differentially expressed miRNAs were identified in a rat middle cerebral artery occlusion (MCAO) stroke model. These studies revealed a promising clinical use of micro-RNAs (miRNAs) as plausible markers monitoring different stages of stroke within miRNA-containing exosomes or extracellular vesicles (EVs) [24,25,26].
miRNAs are conserved non-coding single-stranded RNAs that temporally regulate tissue-specific gene expression [27,28]. As powerful regulators of cellular activity, miRNAs drive cell differentiation and developmental programs [29,30]. Today, several ncRNA therapeutics are in clinical trials for cancer (liver, lymphoma and colorectal), endometriosis, and Alzheimer’s disease [27,30]. Active processed miRNAs range between 19 and 25 base pairs in length while long non-coding RNAs (lncRNAs) can reach ~200 base pairs [31]. Although both ncRNAs are prone to degradation by RNAases, circular RNAs (circRNAs) are less so due to their stability [32]. In this review, we and others propose that stroke-associated ncRNAs modulate stroke disease stage and progression. Our discussion is focused on miR-155, a proinflammatory miRNA in strokes, with pivotal roles in oxidative stress, apoptosis, neuroinflammation, and microglia polarization. Here, we compiled compelling experimental evidence that profiles miR-155 as a promising prognosis biomarker and therapeutic inhibitor.

2. Overview of Stroke Pathophysiology

Stroke is typically defined as an acute neurological deficit caused by vascular damage to the CNS [33,34,35]. The majority of IS cases reported in the United States (87%) occur when a clot forms in a blood vessel in the brain [36]. In contrast, hemorrhagic stroke occurs (13%) when blood pools in the cerebral parenchyma that compresses adjacent brain structures [37,38]. Routine tools in the clinical settings are unreliable at predicting stroke outcomes [39]. It is well-documented that insufficient cerebral blood flow in IS triggers neurological dysfunction by disrupting the brain–blood barrier and altering oxidative stress, apoptosis, and neuroinflammation [40]. The initial ischemia in a stroke triggers multiple signaling cascades that perturb homeostasis leading to neuron and vascular dysfunction (Figure 1) [41,42,43,44]. Apoptosis from ischemia causes membrane leakage of cell contents and pump malfunction, causing build-up of excitatory glutamate, activation of NMDA and AMPA glutamate receptors, and influx of calcium (Figure 1B) [45,46,47]. The influx of calcium causes mitochondria membrane swelling, exacerbating oxidative stress and elevating production of reactive oxygen species (ROS) (Figure 1B) [48,49,50,51,52]. This damage leads to necrosis, increased neuroinflammation caused by activated glial cells and invasion of leukocytes, and monocytes in the brain (Figure 1B) [53,54,55,56,57]. Activated microglia secrete pro-inflammatory cytokines, while hypoxia increases NF-κB levels and associated hypoxia-inducible proteins (Figure 1B) [58,59,60,61].

3. Stroke Management Timing Considerations

Despite the high disease burden of stroke worldwide, therapeutic options are limited [62]. The first line of treatment for IS in adults is alteplase (tPA), a thrombolytic agent that breaks down a thromboembolic clot [63]. However, tPA has a very narrow time window to be safely prescribed. tPA cannot be administered safely to stroke patients ~4.5 h after initial symptom onset, or from the time of the last known normal state of the patient [62,63]. In infants, hypoxic–ischemic brain damage (HIBD), a brain injury caused by lack of adequate oxygen and blood flow, is a major cause of long-term neurological deficits, often resulting in cerebral palsy, mental retardation and epilepsy. Use of alteplase (tPA) beyond this 4.5-h window increases the risk of a re-bleed and can cause a new hemorrhagic stroke [64]. Moreover, tPA therapy is contraindicated for patients with high blood pressure (>185 mmHg systolic blood pressure or >110 mmHg diastolic blood pressure), a diagnosis of prior stroke in the last 3 months, or recent use of anticoagulants (e.g., Warfarin or factor Xa inhibitor Eliquis) [65]. Surgical mechanical thrombectomy is the other standard treatment for IS caused by an occluding thromboembolism. However, like with tPA, mechanical thrombectomy also has a narrow window of therapeutic use and can only be used within 6 h of symptom onset [66]. With such a narrow window of opportunity for standard of care, there is an urgent clinical need to develop adjuvant therapies to treat IS patients (Figure 1A).

4. Non-Coding RNAs: Promising Biomarkers for Stroke Progression

As mentioned above, non-coding RNAs, are a class of conserved RNAs that are not translated into proteins, which surprisingly constitute up to 97% of the entire human genome [67]. These RNAs regulate protein expression by degrading or inhibiting mRNA translation or by directly silencing transcription. For example, ncRNAs can (1) target the 3′ or 5′ untranslated region of specific mRNAs to cause mRNA degradation, repression, or translation, (2) act as mediators of intracellular communication by serving as hormones or being secreted in EVs, (3) directly targeting toll-like receptors (TLRs) to activate signaling pathways and immune responses, (4) cooperatively/competitively interact to enhance or silence targets, and (5) act as pri-miRNAs, encoding peptides that can regulate mature miRNA expression [27,68,69,70,71,72,73,74,75,76,77,78].
Ongoing research shows that ncRNA dysregulation underlies the biology of many chronic diseases [79,80,81,82,83,84,85,86,87,88,89,90,91,92,93]. Notably, single ncRNAs act to restore or degrade neural plasticity, axonal regeneration, vascular reorganization and stem cell activation [92]. Hence, it is important to understand the relationship of molecular targets of specific types of ncRNAs. Remarkably, miR-155 is an excellent candidate given its central role in neuroinflammation and neurodegeneration [68,92]. miR-155, originally discovered in 1997 as a conserved ncRNA in chicken, mice and humans [94], plays a key role in B-cell/macrophage inflammation [94]. miR-155 was reported to also be involved in vascular remodeling as it was found to be abundant in the cerebrovascular endothelium, astrocytes and microglia [95]. Early uncontrolled activation of neuroinflammation during stroke leads to rapid brain dysfunction [96,97,98,99]. We and others propose, given its profile, that miR-155 is a potent regulator that could modulate cellular damage and limit the rapid progression of disability during stroke. Here we present experimental evidence on how miR-155 constitutes an attractive, reliable time-sensitive biomarker, and that inhibition could potentially mitigate oxidative stress, apoptosis, and neuroinflammation, while simultaneously increasing neuroprotection (Figure 2, Figure 3, Figure 4 and Figure 5, Table 1).

4.1. Role of MiR-155 in Oxidative Stress

The therapeutic potential for miR-155 to modulate stroke-induced oxidative stress (Figure 2, Table 2) was established when it was realized that miR-155-5p was highly expressed during ischemia/reperfusion injuries, and that miR-155-5p suppression improved stroke outcomes [116]. Consistent with these findings, lncRNA OIP5-AS1 was recognized as a beneficial molecule, since when it was upregulated it suppressed oxidative stress. In comparison, the interferon regulatory factor 2 binding protein 2 (IRF2BP2) is a transcriptional corepressor which acts as a protective factor in oxidative stress and inflammation in ischemic stroke [127,128]. Based on these findings, a bioinformatic prediction study identified binding sites between lncRNA OIP5-AS1, miR-155-5p and IRF2BP2. This suggests a functional relationship between lncRNA OIP5-AS1, miR-155-5p, IRF2BP2 (Figure 2A, Table 2) [129].
Studies using the lncRNA OIP5-AS1 in a cell model of IS using HMC3 cells (microglia) and SH-SY5Y cells (human neuroblastoma) exposed to oxygen-glucose deprivation/reoxygenation (OGD/R) mimic stroke conditions by stimulating restricted blood flow while depriving cells of oxygen and glucose, and subsequently restoring them to simulate return of blood flow. This is achieved by placing cells in a sugar-free medium in a gas chamber with low oxygen for 2 h, followed by a return to a normal sugar medium and oxygen levels for 12 h [129,136]. In this situation, the elevated reactive oxygen species (ROS), malondialdehyde (MDA) markers, and reduced superoxide dismutase (SOD) (antioxidant) showed that stroke-injured brains had decreased lncRNA OIP5-AS1 levels and increased oxidative stress (Figure 2A, Table 2). Stroke injury increases miR-155-5p levels with associated increases in oxidative stress, but overexpression of lncRNA OIP5-AS1 directly binds and inhibits miR-155-5p, reducing oxidative stress and injury [129,137]. These findings confirm previous studies where lncRNA OIP5AS1 upregulation suppressed oxidative stress in ischemic/reperfusion injuries in cardiomyocytes [138]. IRF2BP2, which is thought to be a protective protein against oxidative stress, is a downstream target of miR-155-5p. When miR-155-5p can bind to its target IRF2BP2, it results in increased oxidative stress. However, when upregulation of IRF2BP2, in the OGD/R-induced HMC3 and SH-SY5Y cell model, overcomes miR-155-5p’s inhibition, this leads to reduction in oxidative stress (Figure 2A, Table 2) [129].
Examining the effects of inhibiting miR-155-5p expression in neural stem cell (NSC) transplants the connection between miR-155 and oxidative stress during stroke (Figure 2B, Table 2) [132]. To investigate how HIF-1α is regulated by miR-155-5p, researchers focused on hypoxia-inducible factor 1α (HIF-1α), a transcription factor and master regulator of genes involved in maintaining oxygen homeostasis [139], by using the middle cerebral artery occlusion model (MCAO). This well-characterized model for stroke injury [140] simulates reperfusion similar to those found with tissue plasminogen activator (tPA) therapy, creating a setting for testing new stroke therapies [140].
When the miR-155-5p expression was inhibited in neural stem cells (NSCs) by using an miR-155-5p inhibitor (inhibitor nucleotide sequence 5′-ACCCCTATCACAATTAGCATTAA-3′ that was cloned into GV280 lentiviral vector) [132] the data showed that miR-155-5p directly targets HIF-1α and negatively regulates its expression by promoting oxidative stress. In contrast, when investigators used the miR-155-5p inhibitor in NSCs, this reversed miR-155-5p’s inhibition of HIF-1α, resulting in significantly reduced oxidative stress and inflammation, decreasing infarct volume and improving rat neurobehavioral outcomes (Figure 2B, Table 2). Taken together, these data indicate that a miR-155 inhibitor might be a promising neuroprotective agent against stroke.
The central pro-inflammatory cytokine (PIC) is a known marker used to track neurological deficits after transient global ischemia induced by cardiac arrest (Figure 2C, Table 2) [133]. In a MCAO study, transient global ischemia in the brain was created by inducing cardiac arrest in mice followed by cardiopulmonary resuscitation. To investigate whether miR-155 inhibition could improve oxidative stress and neuroinflammation in the hippocampus, the levels of oxidative stress markers 8-isoprostaglandin F2α (8-iso PGF2α, an indicator of oxidative stress), and 8-hydroxy-2′-deoxyguanosine (8-OHdG, indicator of protein oxidation), and neuroinflammatory markers IL-1β, IL-6 and TNF-α were measured (Figure 2C, Table 2). Results showed that the miR-155 inhibitor significantly reduced upregulation of hippocampus pro-inflammatory cytokines (PICs), decreased oxidative stress, improved neurological severity score and reduced brain edema, making miR-155 inhibitors a possible therapeutic for IS (Figure 2C, Table 2).

4.2. Role of MiR-155 in Stroke-Related Apoptosis

Neuronal apoptosis is another feature for cerebral ischemic injury that spans ischemia-induced ROS elevation and mitochondria dysfunction through a canonical mitochondria permeability transition pore (MPTP) [141]. Importantly, it has been recognized that the inhibition of miR-155 may be leveraged to treat IS (Figure 3C, Table 2). In vitro oxygen-glucose deprivation/reperfusion (OGD/R) cell models and in vivo hypoxic–ischemic brain damage (HIBD) animal models have showed that miR-155 inhibits SIRT1 (sirtuin1), a nicotinamide adenosine dinucleotide (NAD)-dependent protein deacetylase, resulting in increased apoptosis and inflammation (Figure 3B, Table 1) [115]. In this cell model of OGD/R-treated PC12 cells and microglia (BV2) cells, miR-155 inhibitors decreased apoptosis. However, when SIRT1 was silenced, miR-155 inhibitors were dysfunctional, indicating that SIRT1 was required to reduce apoptosis. These findings were confirmed in the in vivo rat HIBD model. Together, this indicates that miR-155 effectively targets and inhibits SIRT1 to promote apoptosis in IS (Figure 3B, Table 1).
The therapeutic potential of miR-155-5p inhibition is supported by additional evidence (Figure 3A). Results in a study showed that miR-155-5p was upregulated in a rat middle cerebral artery occlusion/reperfusion (MCAO/R) and a OGD/R SH-SY5Y cell model [116]. In both models, miR-155-5p worsened ischemic neuronal apoptosis while inhibition of miR-155-5p improved it. miR-155-5p was shown to target and inhibit DUSP14 by binding to its 3′UTR, thereby preventing DUSP14 from inhibiting the NF-κB and MAPKs signaling pathways, resulting in increased apoptosis (Figure 3A, Table 1). Neuronal apoptosis of VECs plays a vital role in the pathophysiology of stroke through the toll-like receptor on microglia (TLR4) and its adaptor protein (MyD88) [117]. miR-155 regulates the apoptosis of VECs having an impact on cerebral IS apoptosis [142,143]. A relation between miR-155, apoptosis of VECs, and the TLR4/MyD88 signaling pathway was found when increased miR-155 levels activated the TLR4/MyD88, worsening the inflammation (Figure 3B, Table 1).
A cadre of miRNAs cooperate to synchronously integrate various regulatory pathways in the brain [144] as described. The apoptosis-associated miRNAs in a rat cerebellar ischemic alcoholism model found that miR-126 and miR-155 were both increased with apoptosis [144]. In an oxygen-glucose deprivation cell model using N2a cells, researchers investigated the mechanism of action for lncRNA Oprm1 following IS [134]. Researchers found that overexpression of lncRNA Oprm1 ameliorated apoptosis, through a lncRNA Oprm1/miR-155/GATA3 axis, by significantly decreasing infarct size and improving neurological score (Figure 3C, Table 2).
In line with these studies, EVs derived from choroid plexus epithelial (CPE) cells exhibited high levels of miR-155-5p [118]. This indicates that the neuron CPE derived EVs were likely able to deliver miR-155-5p to stroke lesions in the brain. In an OGD/R cell model, miR-155-5p overexpression from the EVs resulted in increased apoptosis, autophagic protein expression, reduced neuron viability, and activation of NLRP3 inflammasomes to worsen cell injury (Figure 3A, Table 1). miR-155-5p promoted apoptosis by suppressing Rheb expression, an important component of the mTOR pathway that promotes cell growth, and by promoting NLRP3-mediated inflammasomes to worsen ischemic damage.
The relationship between miR-155, apoptosis and the Rheb/mTOR was recognized in a MCAO rats and OGD/R cell culture models, when it was reported that miR-155 binds to the 3′-UTR of Rheb to inhibit its expression (Figure 3A, Table 1) [135]. High cerebral infarct volumes and apoptosis were associated with increased miR-155 and decreased Rheb, mTOR, and pS6K, while treatment with miR-155 inhibitors were protected with reduced apoptosis and increased Rheb, mTOR, and p-S6K expression (Figure 3A, C, Table 1). These data indicate that miR-155 inhibitors prevent neuronal apoptosis through the Rheb/mTOR pathway.

4.3. Role of MiR-155 in Neuroinflammation

After stroke onset, the pro-inflammatory master regulator nuclear factor-kappa B (NFkB) triggers heterogenous brain tissue damage that can lead to substantial disability. miR-155 induces neuroinflammation by blocking anti-inflammatory processes through multiple targets (Figure 4B, C, Table 1) [68,100,101,102]. For instance, miR-155 upregulation and targeting of the KRAS gene lead to inhibition of the Ras/NFkB and reduction of proinflammatory cytokines (IL-17, IL-22, IFN-γ, TNF-α, IL-6 and 2) (Figure 4B, Table 1) [100,101]. Conversely, miR-155 upregulation and targeting of the CD33 gene lead to upregulation of proinflammatory cytokines (IL-1β, IL-8, TNF-α) (Figure 4B, Table 1) [100,101]. miR-155 also promotes production of inflammatory cytokines, such as miR-155’s modulation of toll-like receptors (TLRs) through type I interferons (IFNs), through the NFkB inflammatory pathway (Figure 4C, Table 1) [102,103]. This suggests that miR-155 regulates a network of genes that can either promote or suppress inflammation and cytokine production (Figure 4).
Following microglia activation, miR-155 serves as a pro-inflammatory mediator through the TGFβ pathway by stimulating SMAD4, IL10 and IFNγ (Figure 4C) [95,104,105,106]. Bioinformatics studies revealed that Smad2, a mediator of TGF-β signaling involved in stroke recovery, is a target for miR-155 (Figure 4C) [107]. Interestingly, after stroke, Smad2 and Smad3 are involved in reactive astrogliosis and glial scar formation [145]. Furthermore, miR-155 overexpression decreased SMAD2 protein levels, TGF-β-induced SMAD-2 phosphorylation, and SMAD-2-dependent activation (Figure 4C, Table 1) [107].
Normally, SOCS1 and SHIP1 act as inhibitors to suppress secretion of inflammatory molecules that are activated by the TLR/MyD88, NF-κB, JNK/STAT, PI3K/Akt, and MAPK signaling pathways (Figure 4B, Table 1) [146]. Conversely, miR-155 directly targets and inhibits SOCS1 and SHIP1, promoting cytokine production. In one study, injection of an miR-155 inhibitor (anti-miR-155 miRCURY LNA™, Product# 4101082-001, Exiqon) in stroke-induced mice showed significant decrease in cytokines profiles that were correlated with increased expression of miR-155 targets SOCS1 and SHIP1 (Figure 4B) [146]. miR-155 regulates the Wnt/β-catenin signaling pathway by inhibiting HBP1 (Figure 4C) [72,108]. In IS, a hypoxia-inducible factor (HIF) regulates the transcriptional response to hypoxia [106]. Increased levels of miR-155 decreased HIF-1α mRNA, protein, and transcriptional activity in hypoxic conditions, while inhibition of miR-155 reversed these effects (Figure 4C) [106]. This suggests that miR-155 contributes to a negative feedback loop for HIF-1α activity resolution, resulting in oscillatory behavior of HIF-1α-dependent transcription (Figure 4C, Table 1) [106]. miR-155 enhanced monocyte and macrophage production of proinflammatory cytokines secreting CCL2 (Figure 4C) [104,109]. Additionally, miR-155 indirectly activates the NFkB by inhibiting BCL6, a known inhibitor of NFkB, which has important implications in the signaling of foam cells (Figure 4C) [104,105,109,110,111].
miR-155 and N6-methyladenosine (m6A) RNA methylation in a cerebral ischemia/reperfusion (I/R) injury study using a MCAO model [112] showed that m6A RNA methylation plays a role in mRNA stability, splicing, nuclear retention, and translation efficiency. This process is governed by an m6A methyltransferase complex (METTL3, METTL14, and WTAP, and demethylase FTO (fat mass and obesity-associated protein) and ALKBH5) [112]. The ischemic/reperfusion injury downregulated FTO expression, leading to increased m6A RNA modification of miR-155 which worsened the inflammatory response. This was corroborated by increased infarct volume, neurological deficit score, and IL-1b and TNF-a levels (Figure 4A, Table 1) [112]. Glucocorticoids, commonly used for treating inflammation in the clinic, appear to inhibit miR-155 expression by inhibiting the NFkB (Figure 4C) [130,131]. Notably, the overexpression of miR-155 overwhelmed and reversed the anti-inflammatory effects of glucocorticoids (Figure 4C) (Table 2) [131].
MiR-155’s role in neuroinflammation is closely tied to its interactions with toll-like receptors (TLRs) (Figure 4A, Table 1). miR-155, along with miR-146a/b and miR-21, is thought to regulate TLRs (Figure 4C) [119,147,148,149,150]. When TLRs are triggered by infection, those that recognize pathogen-associated molecular patterns (TLR2, TLR3, TLR4, and TLR9) result in miR-155 upregulation (Figure 4A) [113,119,147,148,149,150]. The TLRs require specific signaling proteins to function, such as MyD88 for TLR2 and TLR9, and TRIF for TLR3 (Figure 4A). MyD88 or TRIF signaling pathways can induce miR-155 expression (Figure 4A) [114].
TNF-alpha also activates miR-155, indicating that IFNs require TNF-alpha signaling in macrophages to increase miR-155 levels (Figure 4A) [113]. Moreover, elevation of TNF-alpha or poly(I:C) can stimulate the JNK pathway leading to further miR-155 upregulation (Figure 4A) [113]. BIC is a gene associated with pathogenesis in leukemias and lymphomas [113]. When investigating regulators of miR-155 levels, BIC mRNA that was produced by upregulation of poly(I:C) or IFN was shown to be involved in miR-155 regulation (Figure 4A) [113]. Additionally, while miR-155 is a downstream target of IFN stimulation, it is an early target gene for poly(I:C) (Figure 4A, Table 1) [113].
In summary, modulation of miR-155 through regulation of target genes can simultaneously influence multiple neuroinflammation signaling pathways. This offers a promising therapeutic strategy for IS, through modulation of uncontrolled inflammation in stroke-injured astrocytes by targeting the suppressor of cytokine signaling (SOCS-1) and M1-microglia (Table 1) [151]. Future stroke treatments should consider use of a miR-155 inhibitor as a potent anti-inflammation agent (Figure 4, Table 1).

5. Influence of MiR-155 Modulating Microglia and Astrocytes After Stroke

A critical focus for new therapeutics for stroke today is modulation of neuroprotective supportive cells (e.g., microglia and astrocytes). High levels of miR-155 expression result in activated astrocytes (Table 1). In response to changes in multiple ncRNA levels, monocytes are recruited to the damaged brain tissue and differentiate into M1 pro-inflammatory or M2 anti-inflammatory types (Figure 5) [152]. Increased levels of miR-155 in stroke pre-clinical models ramps up M1 pro-inflammatory polarization, driving up cytokine production and autophagy (Figure 5B, Table 1) [108]. High miR-155 also increased M1 and M2 microglia polarization along with elevated levels of CXCL9, IL-6 and TNF cytokines (Figure 5B, Table 1) [119,120].
Apart from miR-155, other miRNAs (miR-29b, miR-146a, miR-193b, and miR-222), are also elevated during cell differentiation of monocytes to macrophages, suggesting that these miRNAs could also trigger neurotoxic differentiation (Table 3) [120,153]. Several miRNA expression studies looked at the levels of ncRNA in different types of macrophages (M1, M2a, M2b, or M2c) [120]. One showed that the expression of M1 proinflammatory transcripts was increased in THP-1 cells transfected with miR-29b, miR-125a-5p, or miR-155 mimics (Table 3) [120]. Another study using microarray PMA-induced miRNAs (miR-155, mir-222, miR-424 and miR-503) caused cell cycle arrest and partial differentiation, and of these miRNAs two (miR-155 and miR-222) induced G2 arrest and apoptosis (Table 3) [153].
Multiple miRNAs (miR-9, miR-21, miR-24, miR-26a, miR-125a, b, miR-143, miR-145, miR-146a, miR-148, miR-187, miR-223, miR-378-3p, miR-511-3p) modulate macrophage polarization (Table 3) [104,154]. Transfection assays of macrophages with miR-155 mimic resulted in dose-dependent increases in markers CXCL9, which is normally expressed during M1 polarization, IL-6 and TNF, which are expressed during M1 and M2b polarization (Figure 5B, Table 1) [120]. M1 macrophage polarization is induced using IFNγ and TLR4 in vitro by targeting IFNγR, which recruits JAK1/2 to form STAT1/STAT2 heterodimers, which activate NOS2, MHC2 and IL12 [105]. TLR4 activates NFKB and mitogen-associated protein kinase (MAPK) pathways, activating M1-specific transcription factors, with subsequent upregulation of IL12 and downregulation of IL10 (Table 1) [105].
In addition to stimulating M1 polarization, miR-155 inhibits anti-inflammatory M2 polarization by inhibiting IL13RA and C/EBP in the IL13 and IL4 pathways (Figure 5C, Table 1) [107,122,124,125]. Similarly, MafB, a transcription factor important for M2 macrophage polarization, is strongly downregulated by miR-155 (Figure 5C) [124]. Also, IL-13, a pro-M2 cytokine, functions by binding to IL-13 receptor α1 (IL13Rα1), a part of the Type II IL-4 receptor, to activate signal transducer and activator of transcription 6 (STAT6) [122]. miR-155 directly targets and reduces IL13Rα1 protein levels, causing reduced STAT6 activation (Figure 5C, Table 1) [122]. miR-155 also affects IL-13-dependent regulation of SOCS1, DC-SIGN, CCL18, CD23, and SERPINE genes which are involved in differentiating M2 macrophages (Figure 5C, Table 1) [122].
The effects of miR-155 on astrocyte subtype polarization were investigated using DJ-1 (Parkinson disease protein 7, PARK7), an anti-oxidative stress protein with chaperone and signaling functions, and its role in regulating astrocyte neuron survival [126]. Looking for the underlying mechanisms for astrocyte DJ-1 anti-oxidation function and its effects on miR-155 and SHP-1 (Src homology 2 domain-containing phosphatase-1), investigators found that DJ-1 inhibited the transition of astrocytes to the harmful A1 phenotype and promoted A2 polarization, suppressing stroke injury (Table 1) [126]. Furthermore, DJ-1 also interacts with SHP-1 to influence downstream signaling, evidenced by increased SHP-1 elevation with DJ-1 overexpression (Table 1) [126].
In conclusion, miR-155 levels are upregulated by IFNγ and TLR4, both of which drive M1 microglia polarization (Figure 5A, Table 1) [105]. Moreover, miR-155 overexpression reprograms tumor-associated macrophages into M1 macrophages [121], and suppression of SH2-containing inositol-5-phosphatase 1, IL13Rα1 and SMAD2/3, presumed targets for miR-155, promotes M1 macrophage differentiation (Figure 5B, Table 1) [122,123]. Specifically, macrophage M1 to M2 repolarization decreased miR-155 levels while M2 to M1 repolarization increased miR-155, indicating miR-155 plays a vital role in M1 macrophage polarization (Table 1) [121]. Lastly, DJ-1 suppresses miR-155 levels, suggesting that DJ-1 may regulate astrocyte activation via a miR-155/SHP-1 signaling pathway to reduce IS injury (Table 1) [126].

6. Proposal to Use NcRNA MiR-155 as a Biomarker for Ischemic Stroke

In the last decade, ncRNAs have been gaining popularity as attractive clinical biomarkers. First, the ease of obtaining accurate miRNA expression level profiles in patient cohorts, combined with expression in pathological tissues, allows for patient risk stratification [169]. Second, quantifying miRNAs expression levels across different tissue types is feasible because biopsies of patient specimens can be processed for qPCR as fresh, frozen or fixed tissue samples which are amenable for analysis with hybridization-based methods or next-generation sequencing [170,171]. Lastly, miRNA liquid biopsies can be conveniently harvested from diseased cells and urine, saliva, cerebrospinal fluid (CSF), synovial fluid, placenta or breast milk [172]. For stroke patients, CSF or blood serum are more likely to be used as the source of miRNAs because of the difficulty associated with resecting brain tissue from living patients. When comparing the technical challenges of quantifying protein versus miRNA biomarkers in CSF or blood samples, miRNA biomarkers are generally much easier to quantify due to the availability of synthetic oligonucleotides that specifically detect PCR products [172]. Furthermore, standardized assays used to analyze miRNAs, such as qRT-PCR, antibody or DNA microarrays, and RNA-sequencing, have high reliability and resolution making them increasingly accessible tools in both academic and clinical settings [173].
There are numerous ncRNAs being investigated as biomarkers in the pipeline. For example, circRNAs are being used as clinical biomarkers for a number of neurological diseases [174,175,176]. In Alzheimer’s disease, circRNAs derived from patients’ CSF, serum, or plasma [177,178], and dysregulated circRNAs were correlated with known disease risk factors (inflammation, dysregulated metabolism, and immune response). Remarkably, for multiple sclerosis, high levels of miR-181c derived from CSF were associated with a conversion from a clinically isolated syndrome to a relapsing–remitting (recovering) multiple sclerosis, while high levels of miR-191-5p and miR-128-3p derived from serum samples were associated with progressive (non-recoverable) multiple sclerosis [179,180]. For stroke, high levels of circRNAs (circFUNDC1, circPDS5B and circCDC14A), were positively correlated with infarct volume (Table 3) [155]. These experiments give a proof-of-principle that profiling miRNA expression levels in CSF or serum could be used clinically to follow disease progression.
The use of miRNAs as clinical biomarkers is also advancing rapidly. For example, miRNAs are being investigated for use in cancer (breast, lung, gastric, pancreatic, biliary tract, neuroendocrine), cardiovascular, neurological, and infectious disease [27,181,182,183]. Investigators have shown that miRNAs also have potential to be used as biomarkers to predict stroke progression [184]. Moreover, miRNA levels in IS brain tissue were profiled (miR-25, miR-125b-2, miR-125b-627, miR-125b-27a, miR-125b-488 and miR-145) as potential biomarkers in cerebral ischemia [157,158]. As stated above, miR-155 was first recognized as a master biomarker when researchers discovered that miR-155 and miR-21 levels were consistently elevated in the serum of diffuse large B-cell lymphoma patients, suggesting that miR-155 levels could predict the progression of the disease [185,186]. Given miR-155’s central role in stroke-related oxidative stress, apoptosis, and neuroinflammation, we and others have proposed that miR-155 is a formidable master biomarker, and future research should address how its modulation can delay the pathological progression of IS [112,117,118,129]. Furthermore, high levels of circulating miR-155 are expected to correlate with ischemic stroke severity in acute settings, and to be indicative of poor long-term outcomes for patients.

6.1. Urgent Clinical Need for Time-Sensitive Biomarkers to Manage Stroke

Although miRNAs are accessible in the CSF and blood serum of stroke patients, their clinical application warrants further detailed discussion. As mentioned before, the standard IS treatment depends on a narrow, limited ~4.5-h window for tPA treatment, and a ~6-h window to administer a mechanical thrombectomy. Administration of tPA is counted from the onset of symptoms or last known normal state of the patient. Often, the initial onset of stroke is unknown, putting patients outside the window for therapeutic intervention and excluding patients that may have benefited from standard treatments. To address this challenge, a time-sensitive IS biomarker is urgently needed to reduce or prevent stroke disability.
If scientists could identify a reliable time-dependent stroke biomarker whose level correlated to time of stroke onset, physicians could use it in the clinic to rule patients who should or not receive standard of care treatments (Figure 6). As discussed above, miR-155 and accompanying ncRNAs are prime candidates to meet this need. By carefully establishing the timing, dose and administration route for miR-155-based therapeutics, future research could determine a detailed relationship between stroke onset and blood miR-155 levels in different patient stroke populations.
Indeed, such critical time-dependent biomarkers are already used clinically to treat acute heart disease. Creatine kinase (CK-MB), troponin I, and troponin T are time-dependent cardiac biomarkers that have been used for decades to assess the severity of cardiac pathology in myocardial ischemia [187]. CK-MB peaks at 24 h and stabilizes between 48 and 72 h, troponin I peaks at 24 h and stabilizes in 5 to 10 days, and troponin T peaks at 12 to 48 h and normalizes in 5 to 14 days [187]. Investigators have established that miRNAs are promising biomarkers that exhibit time-dependent courses. In a study investigating IS, researchers showed that, after inducing permanent ischemia, levels of miR-101a-3p increased at 30 min and 180 min, and later up to 9 h during transient ischemia [188]. This suggests that miRNA levels such as miR-155, a master regulation of gene expression, could be extremely useful in stroke care (Figure 6).
Finally, it remains to be established if other well-studied, time-sensitive cytokines could be concurrently used clinically to monitor the severity of IS progression. In IS rat models, levels of proinflammatory cytokines TNF-α and IL-1β rise 1 h after a stroke starts [3]. TNF-α reaches its peak at 3 h post-injury, while IL-1β reached its peak at 6 h post-injury. Both cytokines remained elevated up to 2 days after stroke injury [60]. Although TNF-α, a key regulator of neuroinflammation, rises 1–3 h after stroke onset, it may confound stroke severity with other inflammatory conditions. Therefore, a more specific approach is needed.
By carefully measuring the levels of miR-155 and other associated stroke ncRNA at different stroke stages in specific patient populations, we and others propose this could be beneficial for clinical use [189]. We propose that future stroke research profiles must be investigated following time-dependent miR-155 levels in CSF fluid and blood serum in patients between 40 and 50 years old presenting to the emergency room. Ideally, time intervals should measure miR-155 and its precursor gene (10 min, 30 min, 1 h, 2 h, 24 h) (Figure 6).
This kinetic analysis of miR-155 would have great clinical utility because it would allow physicians to determine how far along their patients are in the stroke process and what complications could ensue allowing more personalized decision making to manage stroke in clinical settings. Moreover, a reliable group of time-sensitive biomarkers would thereby help physicians rule in patients to receive treatment with tPA or a mechanical thrombectomy. Another use of miR-155 biomarkers would be to monitor improvement in stroke patients after standard of care treatment, or after taking a miR-155 inhibitor, discussed below (Section 7.2). This information would alert physicians of new strokes or ineffective treatment, allowing them to intervene early to reduce the disability and mortality of stroke patients.
Future research should systematically determine the profile levels of miR-155 in different stroke presentations, considering a patient’s clinical history and adverse effects. Clinical trials should address how stroke outcomes improve after timely administration of miR-155 antagomirs, engineered oligonucleotides designed to complementarily bind and silence miRNAs, in cohort studies. Together, these studies will help determine miR-155’s potential to be used as a biomarker in stroke clinical diagnosis, prognosis and treatment.

6.2. Artificial Intelligence and Stroke Biomarkers

In treating IS injury, the timeliness of diagnosis and intervention are crucial to preserve brain tissue function and to maximize long-term cognitive, behavioral, and motor outcomes for patients. Therefore, multiple efforts, including integrating the clinical use of artificial intelligence (AI), are underway to improve timely diagnosis of stroke and outcomes. Apart from miR-155, other adjuvant stroke-specific biomarkers could enhance the predictive value of AI. In a proteomics stroke study, researchers identified ICAM-2, STXBP5, PLGLA, C3, and IGHV3-64 as candidate biomarkers in blood, yielding a high (75% to 88%) sensitivity for identifying stroke patients [190]. Remarkably, it was found that glial fibrillary acidic protein (GFAP) was sensitive in differentiating IS from hemorrhagic stroke at 3 h and 24 h after stroke onset [191]. Antithrombin III (ATIII), fibrinogen, and ischemia-modified albumin (IMA) are being validated pre-clinically for this purpose [192].
AI is also being analyzed for its potential to improve the routine stroke treatment paradigm, by image differentiation of ischemic and hemorrhagic stroke, large vessel detection, and early CT score grading [193]. Studies are focusing on the use of AI in predicting outcomes for patients based on a combination of clinical parameters [194]. Moreover, progress in predicting reliable stroke diagnosis and treatment requires standardized and validated protocols to holistically analyze clinical history and social determinants of health [194]. Lastly, machine learning algorithms identify large vessel occlusion (LVO) on CT scans to screen patients who are eligible for endovascular interventions with thrombectomy [195].

7. Discussion

7.1. Central Role of MiR-155: Could the Pathology of Acute Ischemic Stroke Be Reversed by miRNA-155 Specific Inhibitors

miR-155 plays an intricate role in the pathophysiology of acute IS through its modulatory effects encompassing oxidative stress, apoptosis, neuroinflammation, and polarization of microglia and astrocytes (Figure 1B). In regulating oxidative stress, miR-155 affects the expression of many key pathways. For instance, in the lncRNA OIP5-AS1/miR-155-5p/IRF2BP2 axis, lncRNA OIP5-AS1 interacts and represses miR-155-5p, preventing miR-155-5p from binding its target IRF2BP2 who suppresses oxidative stress (Figure 2A) [129]. During acute stroke, higher levels of miR-155-5p overwhelm the inhibitory effects of lncRNA OIP5-AS1, allowing miR-155-5p to bind its target IRF2BP2 to enhance oxidative stress [129]. Given miR-155’s impact on oxidative stress, the use of miR-155 inhibitors and their outcomes on ischemic injury have been widely investigated. Use of miR-155-5p inhibitors in NSCs significantly reduced oxidative stress and inflammation, decreased infarct volume, and improved neurobehavioral outcomes in IS models, suggesting miR-155 inhibitors could be neuroprotective against cerebral infarction if applied to clinical settings (Figure 2B) [132]. Importantly, these findings were confirmed when miR-155 inhibitors reduced upregulation of pro-inflammatory cytokines (PICs) into the hippocampus, decreased oxidative stress, and improved neurological severity score and brain edema after ischemia, suggesting miR-155 inhibitors may have a potential therapeutic role in treating oxidative stress triggered by ischemia (Figure 2C) [133]. In another mouse MCAO model of IS, use of intravenous injections of miR-155 inhibitor resulted in reduced infarct size, decreased neuron damage, and improved functional recovery in the inhibitor-injected mice, suggesting that future miR-155 inhibitors could be therapeutically effective by intravenous routes [196].
Data from hypoxic–ischemic brain preclinical studies show that miR-155 enhances apoptosis and inflammation via inhibiting SIRT1 in neonatal rats with HIBD (Figure 3B) [115]. Also, miR-155-5p directly targets DUSP14 by regulating the NF-κB and MAPKs signaling pathways to worsen apoptosis (Figure 3A) [116]. Additionally, the increase in miR-155 levels triggered by IS activates the TLR4/MyD88 signaling pathway, worsening apoptosis of vascular endothelial cells (Figure 3B) [117]. While miR-155 promotes apoptosis, its effects can be overcome by modulation of downstream signaling pathways. For example, in the Oprm1/miR-155/GATA3 axis-regulated apoptosis, overexpression of lncRNA Oprm 1 alleviated apoptosis of IS, significantly decreasing the infarct size and improving neurological score (Figure 3C)[134]. Taken together, these findings suggest that miR-155 is a prime regulator of apoptosis and could potentially serve as a target for stroke-specific development of RNA-therapeutic inhibitors.
We describe here miR-155’s potent modulatory role in neuroinflammation through regulation of NFkB, as it promotes production of inflammatory cytokines that worsen stroke injury (Figure 4C, Table 1). The role of miR-155 and neuroinflammation is closely tied to its interactions with toll-like receptors (TLRs) (Figure 4A, Table 1). miR-155 likely collaborates with miR-146a/b and miR-21 to regulate TLR pathways to oversee inflammation (Figure 4C, Table 1) [119,147,148,149,150]. It is therefore possible that using a miR-155 inhibitor may serve as an adjuvant to reduce inflammation triggered by IS. miR-155 also interferes with the neuroprotective effects of microglia and astrocytes in the brain during ischemic injury (Figure 5, Table 1). miR-155 promotes M1 pro-inflammatory microglia polarization and inhibits M2 anti-inflammatory polarization to reverse microglia normal neuroprotection (Figure 5B,C, Table 1) [108,120,124]. Similarly, miR-155 promotes A1 pro-inflammatory astrocyte activation which is reversed by DJ-1 suppression of miR-155 through the miR-155/SHP-1 signaling pathway (Table 1). Lastly, DJ-1 suppresses miR-155 levels, suggesting that DJ-1 may regulate astrocyte activation via a miR-155/SHP-1 signaling pathway to reduce IS injury [126].

7.2. MiR-155 Inhibitors (anti-RNA) as Therapeutic for Ischemic Stroke: Antisense Oligonucleotides

As stated above, the approach of using miRNA inhibitors as therapeutics is being considered for many diseases [197,198,199]. Given miR-155’s role in regulating acute stroke pathogenesis in oxidative stress, apoptosis, neuroinflammation, and microglia/astrocyte polarization, a miR-155 inhibitor antisense oligonucleotide (ASO) could be used in combination with standard stroke therapy with tPA or thrombectomy. Cobomarsen (also called MRG-106) is an oligonucleotide miR-155 inhibitor used for treating cutaneous T-cell lymphoma [200]. Currently, a phase I clinical trial using Cobomarsen has shown that the miR-155 inhibitor was well-tolerated in patients, indicating it could be used potentially in other disease processes, such as IS [30].
MicroRNA (miRNA) inhibitors, or anti-microRNAs (anti-RNAs), are the antisense oligonucleotide (ASO) which can bind to its complementary miRNA to inhibit its function [201]. In pre-clinical research studies, there are already several miR-155 inhibitors in use [202,203,204]. miRNAs are not currently available for use clinically, but there are several clinical trials assessing their safety and effectiveness. MRX34, a mimic of the tumor suppressor miRNA miR-34a for cancer, and an anti-miR targeting miR-122, is being evaluated to treat hepatitis C [205,206]. Interestingly, miR-155 inhibitors are also being investigated in clinical trials for rheumatoid arthritis (RA), an autoimmune disease with elevated miR-155 and M1 macrophages [207]. In a clinical trial for RA, transfection of healthy monocytes with miR-155 promoted M1 microglia, while transfecting RA monocytes with antagomir miR-155 promoted anti-inflammatory M2 microglia [207].
While miR-155 and other miRNA inhibitors could have therapeutic benefit in treating ischemic stroke, it is important to discuss possible complications in using these inhibitors clinically. MiRNAs, like miR-155, are master regulators in multiple disease pathways, so miR-155 inhibitors could have potential off-target effects that need to be thoroughly investigated before they can be used in clinical settings. Additionally, miRNAs are thought to be interconnected in their regulations of pathways, therefore miR-155 inhibitors could trigger compensatory mechanisms by other miRNAs, such as miR-21 and miR-124, that could influence therapeutic outcomes. Future research should address the nuances of miR-based therapeutics for all stroke types.

7.3. MiR-155 as Biomarker for Ischemic Stroke

As mentioned above, miRNAs today are being assessed in clinical practice to monitor cancer, atrial fibrillation, endocrine disorders, heart failure, lung diseases, and neurological disorders (traumatic brain injury, Alzheimer’s Disease, and Parkinson’s disease) [187,208,209,210,211,212,213,214,215,216,217]. However, no validated clinical biomarker currently exists, to our knowledge, for IS. In the clinic, stroke diagnosis is reached by provider clinical evaluation of CT/MRI imaging [218].
A biomarker for acute IS must reliably measure levels across different stages of stroke (Figure 6). It is possible to evaluate TNF-α, an inflammatory cytokine, during IS injury as it is known to initially peak in the first 1–3 h, and again after 24–36 h [60,61]. In stroke patients, the rise in TNF-α can be measured 6–12 h from symptom onset [219]. Additionally, a decrease in TNF-α concentration within 72 to 144 h after stroke was correlated with clinical improvement [220]. IL-1β has been detected within 1 h after ischemic brain injury, and IL-6 was increased within a few hours after onset of ischemia and up to 90 days after stroke [221,222]. miRNAs can also be time-sensitive markers. miR-101a-3p, another miRNA being considered as a biomarker for IS, is time-sensitive. During IS, miR-101a-3p levels increased at 30 min and 180 min, and after inducing permanent ischemia remained elevated up to 9 h after ischemia (Figure 6) [188].
Given miR-155’s close regulatory role in the pathophysiology of a stroke, it is imperative that the levels of miR-155 be systematically profiled. Research would determine the feasibility at stroke onset, and as a time-sensitive biomarker capable of indicating clinical progression. A combination of miR-155 with other miRNAs would likely be useful as biomarkers to monitor acute IS progress. To tailor stroke treatment to a particular patient population, future research should determine the combination of miRNAs involved in IS that could potentially serve as biomarkers (Table 3). Candidates include miR-181c, miR-124, miR-126, miR-130, miR-181, miR-107, miR-15a, miR-16–1, miR-133, miR-1906, miR-99a, miR-497, miR-424, and miR-210 [3,156,157,158,159,160,161,162,163,164,165,166,167]. We refer the reader to other reviews that have discussed mechanistic details on how miR-155-associated miRNAs might also be included to investigate biomarker panels for specific patient populations [168,223,224,225].

7.4. Outstanding Challenges and Future Stroke Research Directions

While the proposal of miR-155 as a time-sensitive biomarker for ischemic stroke is promising, there remains a need to critically discuss clinical challenges. Concerns for patient safety remain in determining miR-155 as a time-sensitive biomarker for ischemic stroke in its expression across patient populations with different demographics and co-morbidities, with their expression in different number and stroke type presentation. Next, sample stability of miR-155 in CSF versus serum samples needs to be carefully addressed.
As such, future studies should investigate large-scale patient cohorts analyzing the levels of miR-155 and other miRNAs to strengthen miR-155’s validity as a time-sensitive biomarker for ischemic stroke. In the clinic, standard stroke protocols exist where once a “stroke code” has been activated, blood samples are often analyzed after performing head CT imaging and administering tPA [65]. Therefore, if a biomarker panel of miR-155 and other miRNAs were validated for clinical use, this could conveniently be added to the standard blood work for stroke patients to transform stroke outcomes. The miR-155 biomarker values could be monitored to evaluate stroke progression, similar to protocols in place for myocardial ischemia and troponins.
While Cobomarsen (miR-155 inhibitor) has the potential to be incorporated into clinical standard stroke regimens as therapeutics, there are several barriers that need to be considered. First, the ability of miR-155 inhibitors to penetrate the blood–brain barrier using EVs or carrier liposomes must be determined. Next, the level of uptake of miR-155 into neurons and glial cells, and the lifetime of their therapeutic outcomes on reducing oxidative stress, apoptosis, infarct volume and neuroinflammation, must be established. Finally, the long-term cognitive and motor outcomes must be carefully followed. Similarly, potential off-target effects, interactions with other miRNAs, and drug interactions need to be investigated before miR-155 inhibitors can be used in a clinical routine setting in conjunction with standard stroke therapies.

8. Conclusions

Stroke continues to be a significant cause of mortality and disability worldwide and is difficult to treat clinically to avoid long-term disability due to the time-restrictive nature of current standard treatments. Current clinical treatment for IS is focused on re-establishing blood flow and oxygenation to the brain using tPA and thrombectomy. However, these treatments do not address the underlying pathophysiology of stroke injury, including oxidative stress, apoptosis and neuroinflammation. Reliable stroke therapeutics that target the molecular and cellular deficits caused by stroke are needed to reverse neurotoxicity and disability.
miR-155 plays a key role in promoting the pathogenesis of IS, including oxidative stress, apoptosis, neuroinflammation, and microglia/astrocyte polarization, making miR-155 inhibitors attractive therapeutics to manage the acute phase of stroke that could be combined with current standard IS therapies (e.g., tPA and thrombectomy). Given the complex and overlapping regulation of stroke pathophysiology by ncRNAs, future research should focus on establishing the critical time frame to administer miR-155 to patients, with or without adjuvant miRs. This regimen will determine whether ncRNA-based stroke therapeutics are validated in the clinic as effective agents for monitoring stroke onset and progression (Table 4) [168].

Author Contributions

C.H. and G.M.C. conceptualized the study; C.H. wrote the original manuscript draft, including figure creation and table compilation; C.H. and G.M.C. revised the figures and tables and edited multiple drafts, including the final manuscript revisions. All authors have read and agreed to the published version of the manuscript.

Funding

This review received external funding from Texas A&M Presidential Transformational Teaching (TTLC 246063) Grant awarded by the Texas A&M Center for Teaching Excellence to G.M.C. as principal investigator.

Institutional Review Board Statement

Not applicable as this project did not involve humans or animals.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The figures were drawn using BioRender (available online: https://www.biorender.com/). The authors thank the faculty and staff of the Medical Research Pathway Program for their support of medical student academic research.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

alteplase (tPA), anti-microRNAs (anti-RNAs), antisense oligonucleotide (ASO), antithrombin III (ATIII), artificial intelligence (AI), central nervous system (CNS), cerebrospinal fluid (CSF), choroid plexus epithelial (CPE), circular RNA (circRNA), creatine kinase (CK-MB), extracellular vesicles (EVs), fat mass and obesity-associated protein (FTO), glial fibrillary acidic protein (GFAP), 8-hydroxy-2′-deoxyguanosine (8-OHdG), hypoxia-inducible factor 1α (HIF-1α), hypoxic–ischemic brain damage (HIBD), IL-13 receptor α1 (IL13Rα1), interferon regulatory factor 2 binding protein 2 (IRF2BP2), ischemia-modified albumin (IMA), ischemia/reperfusion (I/R), ischemic stroke (IS), isoprostaglandin F2α (8-iso PGF2α), large vessel occlusion (LVO), long non-coding RNA (lncRNA), malondialdehyde (MDA), microRNA (miRNA), middle cerebral artery occlusion (MCAO), middle cerebral artery occlusion/reperfusion (MCAO/R), mitochondria permeability transition pore (MPTP), mitogen associated protein kinase (MAPK), neural stem cell (NSC), N6—methyl adenosine (m6A), N-methyl-D-aspartate (NMDA), nicotinamide adenosine nucleotide (NAD), non-coding RNA (ncRNA), nuclear factor-κB (NF-κB), oxygen-glucose deprivation/reoxygenation (OGD/R), Parkinson disease protein 7, PARK7 (DJ-1), pro-inflammatory cytokine (PIC), reactive oxygen species (ROS), reduced superoxide dismutase (SOD), rheumatoid arthritis (RA), signal transducer and activator of transcription 6 (STAT6), silent information regulator, sirtuin 1 (SIRT1), Src homology 2 domain-containing phosphatase-1(SHP-1), toll-like receptors (TLRs), vascular endothelial cell (VEC).

References

  1. Haupt, M.; Gerner, S.T.; Bähr, M.; Doeppner, T.R. Neuroprotective Strategies for Ischemic Stroke-Future Perspectives. Int. J. Mol. Sci. 2023, 24, 4334. [Google Scholar] [CrossRef] [PubMed]
  2. Saini, V.; Guada, L.; Yavagal, D.R. Global Epidemiology of Stroke and Access to Acute Ischemic Stroke Interventions. Neurology 2021, 97 (Suppl. 2), S6–S16. [Google Scholar] [CrossRef]
  3. Maida, C.D.; Norrito, R.L.; Rizzica, S.; Mazzola, M.; Scarantino, E.R.; Tuttolomondo, A. Molecular Pathogenesis of Ischemic and Hemorrhagic Strokes: Background and Therapeutic Approaches. Int. J. Mol. Sci. 2024, 25, 6297. [Google Scholar] [CrossRef]
  4. Krishnamurthi, R.V.; Feigin, V.L.; Forouzanfar, M.H.; Mensah, G.A.; Connor, M.; Bennett, D.A.; Moran, A.E.; Sacco, R.L.; Anderson, L.M.; Truelsen, T.; et al. Global and regional burden of first-ever ischaemic and haemorrhagic stroke during 1990–2010: Findings from the Global Burden of Disease Study 2010. Lancet Glob. Health 2013, 1, e259–e281. [Google Scholar] [CrossRef]
  5. Feigin, V.L.; Nguyen, G.; Cercy, K.; Johnson, C.O.; Alam, T.; Parmar, P.G.; Abajobir, A.A.; Abate, K.H.; Abd-Allah, F.; Abejie, A.N.; et al. Global, Regional, and Country-Specific Lifetime Risks of Stroke, 1990 and 2016. N. Engl. J. Med. 2018, 379, 2429–2437. [Google Scholar] [CrossRef] [PubMed]
  6. Global, regional, and national burden of stroke and its risk factors, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet Neurol. 2021, 20, 795–820. [CrossRef] [PubMed]
  7. Feigin, V.L.; Brainin, M.; Norrving, B.; Martins, S.; Sacco, R.L.; Hacke, W.; Fisher, M.; Pandian, J.; Lindsay, P. World Stroke Organization (WSO): Global Stroke Fact Sheet 2022. Int. J. Stroke 2022, 17, 18–29. [Google Scholar] [CrossRef] [PubMed]
  8. Close, S.M. Stroke Facts What to know. In Stroke; CDC: Atlanta, GA, USA, 2024. [Google Scholar]
  9. World Health Organization. The Atlas of Heart Disease and Stroke/Judith Mackay and George Mensah with Shanthi Mendis and Kurt Greenland; World Health Organization: Geneva, Switzerland, 2004.
  10. Tsao, C.W.; Aday, A.W.; Almarzooq, Z.I.; Alonso, A.; Beaton, A.Z.; Bittencourt, M.S.; Boehme, A.K.; Buxton, A.E.; Carson, A.P.; Commodore-Mensah, Y.; et al. Heart Disease and Stroke Statistics-2022 Update: A Report From the American Heart Association. Circulation 2022, 145, e153–e639. [Google Scholar] [CrossRef]
  11. Guzik, A.; Bushnell, C. Stroke Epidemiology and Risk Factor Management. Continuum 2017, 23, 15–39. [Google Scholar] [CrossRef]
  12. Shi, Y.; Guo, L.; Chen, Y.; Xie, Q.; Yan, Z.; Liu, Y.; Kang, J.; Li, S. Risk factors for ischemic stroke: Differences between cerebral small vessel and large artery atherosclerosis aetiologies. Folia Neuropathol. 2021, 59, 378–385. [Google Scholar] [CrossRef]
  13. Tirschwell, D.L.; Smith, N.L.; Heckbert, S.R.; Lemaitre, R.N.; Longstreth, W.T., Jr.; Psaty, B.M. Association of cholesterol with stroke risk varies in stroke subtypes and patient subgroups. Neurology 2004, 63, 1868–1875. [Google Scholar] [CrossRef] [PubMed]
  14. Ibrahim, S.; Bielecki, J.; Kocabas, E.; Singh, S.; Senff, J.R.; Casaubon, L.K.; Rosand, J.; Rac, V.E.; Pikula, A. Lifestyle approaches to hypertension for prevention of stroke and vascular cognitive impairment: A realist review protocol. BMJ Open 2024, 14, e088631. [Google Scholar] [CrossRef] [PubMed]
  15. Wassertheil-Smoller, S. Stroke in women. Nutr. Metab. Cardiovasc. Dis. 2010, 20, 419–425. [Google Scholar] [CrossRef] [PubMed]
  16. Boehme, A.K.; Esenwa, C.; Elkind, M.S. Stroke Risk Factors, Genetics, and Prevention. Circ. Res. 2017, 120, 472–495. [Google Scholar] [CrossRef]
  17. Appelros, P.; Stegmayr, B.; Terént, A. Sex differences in stroke epidemiology: A systematic review. Stroke 2009, 40, 1082–1090. [Google Scholar] [CrossRef]
  18. Kelly-Hayes, M. Influence of age and health behaviors on stroke risk: Lessons from longitudinal studies. J. Am. Geriatr. Soc. 2010, 58 (Suppl. 2), S325–S328. [Google Scholar] [CrossRef]
  19. Stewart, J.A.; Dundas, R.; Howard, R.S.; Rudd, A.G.; Wolfe, C.D. Ethnic differences in incidence of stroke: Prospective study with stroke register. BMJ 1999, 318, 967–971. [Google Scholar] [CrossRef]
  20. Sohrabji, F.; Okoreeh, A.; Panta, A. Sex hormones and stroke: Beyond estrogens. Horm. Behav. 2019, 111, 87–95. [Google Scholar] [CrossRef]
  21. Putaala, J. Ischemic Stroke in Young Adults. Continuum 2020, 26, 386–414. [Google Scholar] [CrossRef]
  22. Wei, J.W.; Huang, K.; Yang, C.; Kang, C.S. Non-coding RNAs as regulators in epigenetics (Review). Oncol. Rep. 2017, 37, 3–9. [Google Scholar] [CrossRef]
  23. Zhang, X.; Hamblin, M.H.; Yin, K.J. Noncoding RNAs and Stroke. Neuroscientist 2019, 25, 22–26. [Google Scholar] [CrossRef]
  24. Jeyaseelan, K.; Lim, K.Y.; Armugam, A. MicroRNA expression in the blood and brain of rats subjected to transient focal ischemia by middle cerebral artery occlusion. Stroke 2008, 39, 959–966. [Google Scholar] [CrossRef] [PubMed]
  25. Mirzaei, H.; Momeni, F.; Saadatpour, L.; Sahebkar, A.; Goodarzi, M.; Masoudifar, A.; Kouhpayeh, S.; Salehi, H.; Mirzaei, H.R.; Jaafari, M.R. MicroRNA: Relevance to stroke diagnosis, prognosis, and therapy. J. Cell Physiol. 2018, 233, 856–865. [Google Scholar] [CrossRef]
  26. Calin, G.A.; Dumitru, C.D.; Shimizu, M.; Bichi, R.; Zupo, S.; Noch, E.; Aldler, H.; Rattan, S.; Keating, M.; Rai, K.; et al. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc. Natl. Acad. Sci. USA 2002, 99, 15524–15529. [Google Scholar] [CrossRef] [PubMed]
  27. Nemeth, K.; Bayraktar, R.; Ferracin, M.; Calin, G.A. Non-coding RNAs in disease: From mechanisms to therapeutics. Nat. Rev. Genet. 2024, 25, 211–232. [Google Scholar] [CrossRef] [PubMed]
  28. Yuan, X.; Liu, C.; Yang, P.; He, S.; Liao, Q.; Kang, S.; Zhao, Y. Clustered microRNAs’ coordination in regulating protein-protein interaction network. BMC Syst. Biol. 2009, 3, 65. [Google Scholar] [CrossRef]
  29. Saliminejad, K.; Khorram Khorshid, H.R.; Soleymani Fard, S.; Ghaffari, S.H. An overview of microRNAs: Biology, functions, therapeutics, and analysis methods. J. Cell Physiol. 2019, 234, 5451–5465. [Google Scholar] [CrossRef]
  30. Witten, L.; Slack, F.J. miR-155 as a novel clinical target for hematological malignancies. Carcinogenesis 2020, 41, 2–7. [Google Scholar] [CrossRef]
  31. Zheng, B.; Jeong, S.; Zhu, Y.; Chen, L.; Xia, Q. miRNA and lncRNA as biomarkers in cholangiocarcinoma(CCA). Oncotarget 2017, 8, 100819–100830. [Google Scholar] [CrossRef]
  32. Long, F.; Lin, Z.; Li, L.; Ma, M.; Lu, Z.; Jing, L.; Li, X.; Lin, C. Comprehensive landscape and future perspectives of circular RNAs in colorectal cancer. Mol. Cancer 2021, 20, 26. [Google Scholar] [CrossRef]
  33. Sacco, R.L.; Kasner, S.E.; Broderick, J.P.; Caplan, L.R.; Connors, J.J.; Culebras, A.; Elkind, M.S.; George, M.G.; Hamdan, A.D.; Higashida, R.T.; et al. An updated definition of stroke for the 21st century: A statement for healthcare professionals from the American Heart Association/American Stroke Association. Stroke 2013, 44, 2064–2089. [Google Scholar] [CrossRef] [PubMed]
  34. Coupland, A.P.; Thapar, A.; Qureshi, M.I.; Jenkins, H.; Davies, A.H. The definition of stroke. J. R. Soc. Med. 2017, 110, 9–12. [Google Scholar] [CrossRef] [PubMed]
  35. Hankey, G.J. Stroke. Lancet 2017, 389, 641–654. [Google Scholar] [CrossRef]
  36. Morotti, A.; Poli, L.; Costa, P. Acute Stroke. Semin. Neurol. 2019, 39, 61–72. [Google Scholar] [CrossRef]
  37. Ohashi, S.N.; DeLong, J.H.; Kozberg, M.G.; Mazur-Hart, D.J.; van Veluw, S.J.; Alkayed, N.J.; Sansing, L.H. Role of Inflammatory Processes in Hemorrhagic Stroke. Stroke 2023, 54, 605–619. [Google Scholar] [CrossRef]
  38. Zhao, Y.; Zhang, X.; Chen, X.; Wei, Y. Neuronal injuries in cerebral infarction and ischemic stroke: From mechanisms to treatment (Review). Int. J. Mol. Med. 2022, 49, 15. [Google Scholar] [CrossRef]
  39. Matys, P.; Mirończuk, A.; Starosz, A.; Grubczak, K.; Kochanowicz, J.; Kułakowska, A.; Kapica-Topczewska, K. Expanding Role of Interleukin-1 Family Cytokines in Acute Ischemic Stroke. Int. J. Mol. Sci. 2024, 25, 10515. [Google Scholar] [CrossRef] [PubMed]
  40. Yuan, Y.; Kang, R.; Yu, Y.; Liu, J.; Zhang, Y.; Shen, C.; Wang, J.; Wu, P.; Shen, C.; Wang, Z. Crosstalk between miRNAs and their regulated genes network in stroke. Sci. Rep. 2016, 6, 20429. [Google Scholar] [CrossRef]
  41. Qin, C.; Yang, S.; Chu, Y.H.; Zhang, H.; Pang, X.W.; Chen, L.; Zhou, L.Q.; Chen, M.; Tian, D.S.; Wang, W. Signaling pathways involved in ischemic stroke: Molecular mechanisms and therapeutic interventions. Signal Transduct. Target. Ther. 2022, 7, 215. [Google Scholar] [CrossRef] [PubMed]
  42. Hénaut, L.; Grissi, M.; Brazier, F.; Assem, M.; Poirot-Leclercq, S.; Lenglet, G.; Boudot, C.; Avondo, C.; Boullier, A.; Choukroun, G.; et al. Cellular and molecular mechanisms associated with ischemic stroke severity in female mice with chronic kidney disease. Sci. Rep. 2019, 9, 6432. [Google Scholar] [CrossRef]
  43. Nakamura, K.; Shichita, T. Cellular and molecular mechanisms of sterile inflammation in ischaemic stroke. J. Biochem. 2019, 165, 459–464. [Google Scholar] [CrossRef] [PubMed]
  44. Gasull, T.; Arboix, A. Molecular Mechanisms and Pathophysiology of Acute Stroke: Emphasis on Biomarkers in the Different Stroke Subtypes. Int. J. Mol. Sci. 2022, 23, 9476. [Google Scholar] [CrossRef]
  45. Nalbandian, A.; Sehgal, K.; Gupta, A.; Madhavan, M.V.; McGroder, C.; Stevens, J.S.; Cook, J.R.; Nordvig, A.S.; Shalev, D.; Sehrawat, T.S.; et al. Post-acute COVID-19 syndrome. Nat. Med. 2021, 27, 601–615. [Google Scholar] [CrossRef] [PubMed]
  46. Sekerdag, E.; Solaroglu, I.; Gursoy-Ozdemir, Y. Cell Death Mechanisms in Stroke and Novel Molecular and Cellular Treatment Options. Curr. Neuropharmacol. 2018, 16, 1396–1415. [Google Scholar] [CrossRef] [PubMed]
  47. Jurcau, A.; Ardelean, A.I. Oxidative Stress in Ischemia/Reperfusion Injuries following Acute Ischemic Stroke. Biomedicines 2022, 10, 574. [Google Scholar] [CrossRef]
  48. Belov Kirdajova, D.; Kriska, J.; Tureckova, J.; Anderova, M. Ischemia-Triggered Glutamate Excitotoxicity From the Perspective of Glial Cells. Front. Cell. Neurosci. 2020, 14, 51. [Google Scholar] [CrossRef]
  49. Sun, Y.; Feng, X.; Ding, Y.; Li, M.; Yao, J.; Wang, L.; Gao, Z. Phased Treatment Strategies for Cerebral Ischemia Based on Glutamate Receptors. Front. Cell. Neurosci. 2019, 13, 168. [Google Scholar] [CrossRef]
  50. Suzuki, H.; Kawakita, F.; Asada, R. Neuroelectric Mechanisms of Delayed Cerebral Ischemia after Aneurysmal Subarachnoid Hemorrhage. Int. J. Mol. Sci. 2022, 23, 3102. [Google Scholar] [CrossRef]
  51. Wen, B.; Xu, K.; Huang, R.; Jiang, T.; Wang, J.; Chen, J.; Chen, J.; He, B. Preserving mitochondrial function by inhibiting GRP75 ameliorates neuron injury under ischemic stroke. Mol. Med. Rep. 2022, 25, 165. [Google Scholar] [CrossRef]
  52. Rahi, V.; Kaundal, R.K. Exploring the intricacies of calcium dysregulation in ischemic stroke: Insights into neuronal cell death and therapeutic strategies. Life Sci. 2024, 347, 122651. [Google Scholar] [CrossRef]
  53. Ferrari, F.; Gorini, A.; Hoyer, S.; Villa, R.F. Glutamate metabolism in cerebral mitochondria after ischemia and post-ischemic recovery during aging: Relationships with brain energy metabolism. J. Neurochem. 2018, 146, 416–428. [Google Scholar] [CrossRef] [PubMed]
  54. Yilmaz, G.; Arumugam, T.V.; Stokes, K.Y.; Granger, D.N. Role of T lymphocytes and interferon-gamma in ischemic stroke. Circulation 2006, 113, 2105–2112. [Google Scholar] [CrossRef]
  55. Lambertsen, K.L.; Gregersen, R.; Meldgaard, M.; Clausen, B.H.; Heibøl, E.K.; Ladeby, R.; Knudsen, J.; Frandsen, A.; Owens, T.; Finsen, B. A role for interferon-gamma in focal cerebral ischemia in mice. J. Neuropathol. Exp. Neurol. 2004, 63, 942–955. [Google Scholar] [CrossRef]
  56. Guruswamy, R.; ElAli, A. Complex Roles of Microglial Cells in Ischemic Stroke Pathobiology: New Insights and Future Directions. Int. J. Mol. Sci. 2017, 18, 496. [Google Scholar] [CrossRef] [PubMed]
  57. Schilling, M.; Besselmann, M.; Müller, M.; Strecker, J.K.; Ringelstein, E.B.; Kiefer, R. Predominant phagocytic activity of resident microglia over hematogenous macrophages following transient focal cerebral ischemia: An investigation using green fluorescent protein transgenic bone marrow chimeric mice. Exp. Neurol. 2005, 196, 290–297. [Google Scholar] [CrossRef] [PubMed]
  58. Stone, M.J.; Hayward, J.A.; Huang, C.; Huma, Z.E.; Sanchez, J. Mechanisms of Regulation of the Chemokine-Receptor Network. Int. J. Mol. Sci. 2017, 18, 342. [Google Scholar] [CrossRef]
  59. Liu, T.; Clark, R.K.; McDonnell, P.C.; Young, P.R.; White, R.F.; Barone, F.C.; Feuerstein, G.Z. Tumor necrosis factor-alpha expression in ischemic neurons. Stroke 1994, 25, 1481–1488. [Google Scholar] [CrossRef]
  60. Wang, X.; Yue, T.L.; Barone, F.C.; White, R.F.; Gagnon, R.C.; Feuerstein, G.Z. Concomitant cortical expression of TNF-alpha and IL-1 beta mRNAs follows early response gene expression in transient focal ischemia. Mol. Chem. Neuropathol. 1994, 23, 103–114. [Google Scholar] [CrossRef]
  61. Murakami, Y.; Saito, K.; Hara, A.; Zhu, Y.; Sudo, K.; Niwa, M.; Fujii, H.; Wada, H.; Ishiguro, H.; Mori, H.; et al. Increases in tumor necrosis factor-alpha following transient global cerebral ischemia do not contribute to neuron death in mouse hippocampus. J. Neurochem. 2005, 93, 1616–1622. [Google Scholar] [CrossRef]
  62. Del Zoppo, G.J.; Saver, J.L.; Jauch, E.C.; Adams, H.P., Jr. Expansion of the time window for treatment of acute ischemic stroke with intravenous tissue plasminogen activator: A science advisory from the American Heart Association/American Stroke Association. Stroke 2009, 40, 2945–2948. [Google Scholar] [CrossRef]
  63. Hughes, R.E.; Tadi, P.; Bollu, P.C. TPA Therapy. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  64. Saver, J.L.; Fonarow, G.C.; Smith, E.E.; Reeves, M.J.; Grau-Sepulveda, M.V.; Pan, W.; Olson, D.M.; Hernandez, A.F.; Peterson, E.D.; Schwamm, L.H. Time to treatment with intravenous tissue plasminogen activator and outcome from acute ischemic stroke. JAMA 2013, 309, 2480–2488. [Google Scholar] [CrossRef] [PubMed]
  65. Powers, W.J.; Rabinstein, A.A.; Ackerson, T.; Adeoye, O.M.; Bambakidis, N.C.; Becker, K.; Biller, J.; Brown, M.; Demaerschalk, B.M.; Hoh, B.; et al. Guidelines for the Early Management of Patients With Acute Ischemic Stroke: 2019 Update to the 2018 Guidelines for the Early Management of Acute Ischemic Stroke: A Guideline for Healthcare Professionals From the American Heart Association/American Stroke Association. Stroke 2019, 50, e344–e418. [Google Scholar] [CrossRef]
  66. Kohli, V.; Koltz, M.T. Indications for Surgical Intervention in the Treatment of Ischemic Stroke. In Stroke; Dehkharghani, S., Ed.; Exon Publications: Brisbane, AU, USA, 2021. [Google Scholar]
  67. Nie, J.H.; Li, T.X.; Zhang, X.Q.; Liu, J. Roles of Non-Coding RNAs in Normal Human Brain Development, Brain Tumor, and Neuropsychiatric Disorders. Noncoding RNA 2019, 5, 36. [Google Scholar] [CrossRef]
  68. Zingale, V.D.; Gugliandolo, A.; Mazzon, E. MiR-155: An Important Regulator of Neuroinflammation. Int. J. Mol. Sci. 2021, 23, 90. [Google Scholar] [CrossRef] [PubMed]
  69. Vasudevan, S.; Tong, Y.; Steitz, J.A. Switching from repression to activation: microRNAs can up-regulate translation. Science 2007, 318, 1931–1934. [Google Scholar] [CrossRef] [PubMed]
  70. Li, G.; Wu, X.; Qian, W.; Cai, H.; Sun, X.; Zhang, W.; Tan, S.; Wu, Z.; Qian, P.; Ding, K.; et al. CCAR1 5’ UTR as a natural miRancer of miR-1254 overrides tamoxifen resistance. Cell Res. 2016, 26, 655–673. [Google Scholar] [CrossRef]
  71. Ørom, U.A.; Nielsen, F.C.; Lund, A.H. MicroRNA-10a binds the 5’UTR of ribosomal protein mRNAs and enhances their translation. Mol. Cell 2008, 30, 460–471. [Google Scholar] [CrossRef]
  72. Bayraktar, R.; Van Roosbroeck, K.; Calin, G.A. Cell-to-cell communication: microRNAs as hormones. Mol. Oncol. 2017, 11, 1673–1686. [Google Scholar] [CrossRef]
  73. Drula, R.; Pardini, B.; Fu, X.; De Los Santos, M.C.; Jurj, A.; Pang, L.; El-Daly, S.M.; Fabris, L.; Knutsen, E.; Dragomir, M.P.; et al. 17β-estradiol promotes extracellular vesicle release and selective miRNA loading in ERα-positive breast cancer. Proc. Natl. Acad. Sci. USA 2023, 120, e2122053120. [Google Scholar] [CrossRef]
  74. Fabbri, M. TLRs as miRNA receptors. Cancer Res. 2012, 72, 6333–6337. [Google Scholar] [CrossRef]
  75. Fabbri, M.; Paone, A.; Calore, F.; Galli, R.; Gaudio, E.; Santhanam, R.; Lovat, F.; Fadda, P.; Mao, C.; Nuovo, G.J.; et al. MicroRNAs bind to Toll-like receptors to induce prometastatic inflammatory response. Proc. Natl. Acad. Sci. USA 2012, 109, E2110–E2116. [Google Scholar] [CrossRef] [PubMed]
  76. Chen, X.; Liang, H.; Zhang, J.; Zen, K.; Zhang, C.Y. Secreted microRNAs: A new form of intercellular communication. Trends Cell Biol. 2012, 22, 125–132. [Google Scholar] [CrossRef] [PubMed]
  77. Lehmann, S.M.; Krüger, C.; Park, B.; Derkow, K.; Rosenberger, K.; Baumgart, J.; Trimbuch, T.; Eom, G.; Hinz, M.; Kaul, D.; et al. An unconventional role for miRNA: Let-7 activates Toll-like receptor 7 and causes neurodegeneration. Nat. Neurosci. 2012, 15, 827–835. [Google Scholar] [CrossRef]
  78. Chen, X.; Liang, H.; Zhang, J.; Zen, K.; Zhang, C.Y. microRNAs are ligands of Toll-like receptors. RNA 2013, 19, 737–739. [Google Scholar] [CrossRef]
  79. Ziats, M.N.; Rennert, O.M. Identification of differentially expressed microRNAs across the developing human brain. Mol. Psychiatry 2014, 19, 848–852. [Google Scholar] [CrossRef]
  80. Oh, Y.M.; Lee, S.W.; Kim, W.K.; Chen, S.; Church, V.A.; Cates, K.; Li, T.; Zhang, B.; Dolle, R.E.; Dahiya, S.; et al. Age-related Huntington’s disease progression modeled in directly reprogrammed patient-derived striatal neurons highlights impaired autophagy. Nat. Neurosci. 2022, 25, 1420–1433. [Google Scholar] [CrossRef] [PubMed]
  81. Reed, E.R.; Latourelle, J.C.; Bockholt, J.H.; Bregu, J.; Smock, J.; Paulsen, J.S.; Myers, R.H. MicroRNAs in CSF as prodromal biomarkers for Huntington disease in the PREDICT-HD study. Neurology 2018, 90, e264–e272. [Google Scholar] [CrossRef]
  82. Maes, O.C.; Chertkow, H.M.; Wang, E.; Schipper, H.M. MicroRNA: Implications for Alzheimer Disease and other Human CNS Disorders. Curr. Genom. 2009, 10, 154–168. [Google Scholar] [CrossRef]
  83. Scheper, M.F.; Iyer, A.; Anink, J.J.; Mesarosova, L.; Mills, J.D.; Aronica, E. Dysregulation of miR-543 in Parkinson’s disease: Impact on the neuroprotective gene SIRT1. Neuropathol. Appl. Neurobiol. 2023, 49, e12864. [Google Scholar] [CrossRef]
  84. Hezroni, H.; Perry, R.B.T.; Ulitsky, I. Long Noncoding RNAs in Development and Regeneration of the Neural Lineage. Cold Spring Harb. Symp. Quant. Biol. 2019, 84, 165–177. [Google Scholar] [CrossRef]
  85. Schneider, M.F.; Müller, V.; Müller, S.A.; Lichtenthaler, S.F.; Becker, P.B.; Scheuermann, J.C. LncRNA RUS shapes the gene expression program towards neurogenesis. Life Sci. Alliance 2022, 5, e202201504. [Google Scholar] [CrossRef] [PubMed]
  86. Lin, N.; Chang, K.Y.; Li, Z.; Gates, K.; Rana, Z.A.; Dang, J.; Zhang, D.; Han, T.; Yang, C.S.; Cunningham, T.J.; et al. An evolutionarily conserved long noncoding RNA TUNA controls pluripotency and neural lineage commitment. Mol. Cell 2014, 53, 1005–1019. [Google Scholar] [CrossRef]
  87. Wu, D.P.; Zhao, Y.D.; Yan, Q.Q.; Liu, L.L.; Wei, Y.S.; Huang, J.L. Circular RNAs: Emerging players in brain aging and neurodegenerative diseases. J. Pathol. 2023, 259, 1–9. [Google Scholar] [CrossRef]
  88. Chen, J.; Fu, B.; Bao, J.; Su, R.; Zhao, H.; Liu, Z. Novel circular RNA 2960 contributes to secondary damage of spinal cord injury by sponging miRNA-124. J. Comp. Neurol. 2021, 529, 1456–1464. [Google Scholar] [CrossRef]
  89. Doxakis, E. Insights into the multifaceted role of circular RNAs: Implications for Parkinson’s disease pathogenesis and diagnosis. npj Park. Dis. 2022, 8, 7. [Google Scholar] [CrossRef] [PubMed]
  90. Verduci, L.; Tarcitano, E.; Strano, S.; Yarden, Y.; Blandino, G. CircRNAs: Role in human diseases and potential use as biomarkers. Cell Death Dis. 2021, 12, 468. [Google Scholar] [CrossRef] [PubMed]
  91. Dube, U.; Del-Aguila, J.L.; Li, Z.; Budde, J.P.; Jiang, S.; Hsu, S.; Ibanez, L.; Fernandez, M.V.; Farias, F.; Norton, J.; et al. An atlas of cortical circular RNA expression in Alzheimer disease brains demonstrates clinical and pathological associations. Nat. Neurosci. 2019, 22, 1903–1912. [Google Scholar] [CrossRef] [PubMed]
  92. Roitbak, T. MicroRNAs and Regeneration in Animal Models of CNS Disorders. Neurochem. Res. 2020, 45, 188–203. [Google Scholar] [CrossRef]
  93. Tam, W. Identification and characterization of human BIC, a gene on chromosome 21 that encodes a noncoding RNA. Gene 2001, 274, 157–167. [Google Scholar] [CrossRef]
  94. Mashima, R. Physiological roles of miR-155. Immunology 2015, 145, 323–333. [Google Scholar] [CrossRef]
  95. Elton, T.S.; Selemon, H.; Elton, S.M.; Parinandi, N.L. Regulation of the MIR155 host gene in physiological and pathological processes. Gene 2013, 532, 1–12. [Google Scholar] [CrossRef] [PubMed]
  96. Anrather, J.; Iadecola, C. Inflammation and Stroke: An Overview. Neurotherapeutics 2016, 13, 661–670. [Google Scholar] [CrossRef]
  97. Kelly, P.J.; Lemmens, R.; Tsivgoulis, G. Inflammation and Stroke Risk: A New Target for Prevention. Stroke 2021, 52, 2697–2706. [Google Scholar] [CrossRef]
  98. Liu, Q.; Shi, K.; Wang, Y.; Shi, F.D. Neurovascular Inflammation and Complications of Thrombolysis Therapy in Stroke. Stroke 2023, 54, 2688–2697. [Google Scholar] [CrossRef] [PubMed]
  99. Candelario-Jalil, E.; Dijkhuizen, R.M.; Magnus, T. Neuroinflammation, Stroke, Blood-Brain Barrier Dysfunction, and Imaging Modalities. Stroke 2022, 53, 1473–1486. [Google Scholar] [CrossRef] [PubMed]
  100. Maranini, B.; Ciancio, G.; Ferracin, M.; Cultrera, R.; Negrini, M.; Sabbioni, S.; Govoni, M. microRNAs and Inflammatory Immune Response in SARS-CoV-2 Infection: A Narrative Review. Life 2022, 12, 288. [Google Scholar] [CrossRef]
  101. Roganović, J.R. microRNA-146a and -155, upregulated by periodontitis and type 2 diabetes in oral fluids, are predicted to regulate SARS-CoV-2 oral receptor genes. J. Periodontol. 2021, 92, 35–43. [Google Scholar] [CrossRef]
  102. Tsitsiou, E.; Lindsay, M.A. microRNAs and the immune response. Curr. Opin. Pharmacol. 2009, 9, 514–520. [Google Scholar] [CrossRef]
  103. Ceppi, M.; Pereira, P.M.; Dunand-Sauthier, I.; Barras, E.; Reith, W.; Santos, M.A.; Pierre, P. MicroRNA-155 modulates the interleukin-1 signaling pathway in activated human monocyte-derived dendritic cells. Proc. Natl. Acad. Sci. USA 2009, 106, 2735–2740. [Google Scholar] [CrossRef]
  104. Alwani, A.; Andreasik, A.; Szatanek, R.; Siedlar, M.; Baj-Krzyworzeka, M. The Role of miRNA in Regulating the Fate of Monocytes in Health and Cancer. Biomolecules 2022, 12, 100. [Google Scholar] [CrossRef]
  105. Pasca, S.; Jurj, A.; Petrushev, B.; Tomuleasa, C.; Matei, D. MicroRNA-155 Implication in M1 Polarization and the Impact in Inflammatory Diseases. Front. Immunol. 2020, 11, 625. [Google Scholar] [CrossRef]
  106. Bruning, U.; Cerone, L.; Neufeld, Z.; Fitzpatrick, S.F.; Cheong, A.; Scholz, C.C.; Simpson, D.A.; Leonard, M.O.; Tambuwala, M.M.; Cummins, E.P.; et al. MicroRNA-155 promotes resolution of hypoxia-inducible factor 1alpha activity during prolonged hypoxia. Mol. Cell. Biol. 2011, 31, 4087–4096. [Google Scholar] [CrossRef]
  107. Louafi, F.; Martinez-Nunez, R.T.; Sanchez-Elsner, T. MicroRNA-155 targets SMAD2 and modulates the response of macrophages to transforming growth factor-{beta}. J. Biol. Chem. 2010, 285, 41328–41336. [Google Scholar] [CrossRef] [PubMed]
  108. Xu, W.D.; Feng, S.Y.; Huang, A.F. Role of miR-155 in inflammatory autoimmune diseases: A comprehensive review. Inflamm. Res. 2022, 71, 1501–1517. [Google Scholar] [CrossRef] [PubMed]
  109. Nazari-Jahantigh, M.; Wei, Y.; Noels, H.; Akhtar, S.; Zhou, Z.; Koenen, R.R.; Heyll, K.; Gremse, F.; Kiessling, F.; Grommes, J.; et al. MicroRNA-155 promotes atherosclerosis by repressing Bcl6 in macrophages. J. Clin. Investig. 2012, 122, 4190–4202. [Google Scholar] [CrossRef] [PubMed]
  110. O’Connell, R.M.; Chaudhuri, A.A.; Rao, D.S.; Baltimore, D. Inositol phosphatase SHIP1 is a primary target of miR-155. Proc. Natl. Acad. Sci. USA 2009, 106, 7113–7118. [Google Scholar] [CrossRef]
  111. An, H.; Xu, H.; Zhang, M.; Zhou, J.; Feng, T.; Qian, C.; Qi, R.; Cao, X. Src homology 2 domain-containing inositol-5-phosphatase 1 (SHIP1) negatively regulates TLR4-mediated LPS response primarily through a phosphatase activity- and PI-3K-independent mechanism. Blood 2005, 105, 4685–4692. [Google Scholar] [CrossRef]
  112. Jiang, Z.; Shi, L.; Huang, H.; Lei, D.; Lou, L.; Jin, Y.; Sun, J.; Wang, L. Downregulated FTO Promotes MicroRNA-155-mediated Inflammatory Response in Cerebral Ischemia/Reperfusion Injury. Neuroscience 2023, 526, 305–313. [Google Scholar] [CrossRef]
  113. O’Connell, R.M.; Taganov, K.D.; Boldin, M.P.; Cheng, G.; Baltimore, D. MicroRNA-155 is induced during the macrophage inflammatory response. Proc. Natl. Acad. Sci. USA 2007, 104, 1604–1609. [Google Scholar] [CrossRef]
  114. Dunne, A.; O’Neill, L.A. Adaptor usage and Toll-like receptor signaling specificity. FEBS Lett. 2005, 579, 3330–3335. [Google Scholar] [CrossRef]
  115. Ke, F.; Wang, H.; Geng, J.; Jing, X.; Fang, F.; Fang, C.; Zhang, B.H. MiR-155 promotes inflammation and apoptosis via targeting SIRT1 in hypoxic-ischemic brain damage. Exp. Neurol. 2023, 362, 114317. [Google Scholar] [CrossRef] [PubMed]
  116. Shi, Y.; Li, K.; Xu, K.; Liu, Q.H. MiR-155-5p accelerates cerebral ischemia-reperfusion injury via targeting DUSP14 by regulating NF-κB and MAPKs signaling pathways. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 1408–1419. [Google Scholar]
  117. Chen, W.; Wang, L.; Liu, Z. MicroRNA-155 influences cell damage in ischemic stroke via TLR4/MYD88 signaling pathway. Bioengineered 2021, 12, 2449–2458. [Google Scholar] [CrossRef]
  118. Yang, Z.; Shi, X.; Gao, Z.; Chu, L. miR-155-5p in Extracellular Vesicles Derived from Choroid Plexus Epithelial Cells Promotes Autophagy and Inflammation to Aggravate Ischemic Brain Injury in Mice. Oxid. Med. Cell. Longev. 2022, 2022, 8603427. [Google Scholar] [CrossRef] [PubMed]
  119. Taganov, K.D.; Boldin, M.P.; Chang, K.J.; Baltimore, D. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc. Natl. Acad. Sci. USA 2006, 103, 12481–12486. [Google Scholar] [CrossRef] [PubMed]
  120. Graff, J.W.; Dickson, A.M.; Clay, G.; McCaffrey, A.P.; Wilson, M.E. Identifying functional microRNAs in macrophages with polarized phenotypes. J. Biol. Chem. 2012, 287, 21816–21825. [Google Scholar] [CrossRef]
  121. Cai, X.; Yin, Y.; Li, N.; Zhu, D.; Zhang, J.; Zhang, C.Y.; Zen, K. Re-polarization of tumor-associated macrophages to pro-inflammatory M1 macrophages by microRNA-155. J. Mol. Cell Biol. 2012, 4, 341–343. [Google Scholar] [CrossRef]
  122. Martinez-Nunez, R.T.; 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 alpha1 (IL13Ralpha1). J. Biol. Chem. 2011, 286, 1786–1794. [Google Scholar] [CrossRef]
  123. Sierra-Filardi, E.; Puig-Kröger, A.; Blanco, F.J.; Nieto, C.; Bragado, R.; Palomero, M.I.; Bernabéu, C.; Vega, M.A.; Corbí, A.L. Activin A skews macrophage polarization by promoting a proinflammatory phenotype and inhibiting the acquisition of anti-inflammatory macrophage markers. Blood 2011, 117, 5092–5101. [Google Scholar] [CrossRef]
  124. Kim, H. The transcription factor MafB promotes anti-inflammatory M2 polarization and cholesterol efflux in macrophages. Sci. Rep. 2017, 7, 7591. [Google Scholar] [CrossRef]
  125. Wahl, S.M.; Allen, J.B.; Costa, G.L.; Wong, H.L.; Dasch, J.R. Reversal of acute and chronic synovial inflammation by anti-transforming growth factor beta. J. Exp. Med. 1993, 177, 225–230. [Google Scholar] [CrossRef]
  126. Xue, Y.; Wang, Y.; Chen, T.; Peng, L.; Wang, C.; Xue, G.; Yu, S. DJ-1 regulates astrocyte activation through miR-155/SHP-1 signaling in cerebral ischemia/reperfusion injury. J. Neurochem. 2025, 169, e16230. [Google Scholar] [CrossRef]
  127. Chen, Y.; Liu, W.; Chen, M.; Sun, Q.; Chen, H.; Li, Y. Up-regulating lncRNA OIP5-AS1 protects neuron injury against cerebral hypoxia-ischemia induced inflammation and oxidative stress in microglia/macrophage through activating CTRP3 via sponging miR-186-5p. Int. Immunopharmacol. 2021, 92, 107339. [Google Scholar] [CrossRef] [PubMed]
  128. Cruz, S.A.; Hari, A.; Qin, Z.; Couture, P.; Huang, H.; Lagace, D.C.; Stewart, A.F.R.; Chen, H.H. Loss of IRF2BP2 in Microglia Increases Inflammation and Functional Deficits after Focal Ischemic Brain Injury. Front. Cell. Neurosci. 2017, 11, 201. [Google Scholar] [CrossRef]
  129. Zhang, J.K.; Li, Y.; Yu, Z.T.; Jiang, J.W.; Tang, H.; Tu, G.L.; Xia, Y. OIP5-AS1 Inhibits Oxidative Stress and Inflammation in Ischemic Stroke Through miR-155-5p/IRF2BP2 Axis. Neurochem. Res. 2023, 48, 1382–1394. [Google Scholar] [CrossRef]
  130. Chinenov, Y.; Coppo, M.; Gupte, R.; Sacta, M.A.; Rogatsky, I. Glucocorticoid receptor coordinates transcription factor-dominated regulatory network in macrophages. BMC Genom. 2014, 15, 656. [Google Scholar] [CrossRef]
  131. Zheng, Y.; Xiong, S.; Jiang, P.; Liu, R.; Liu, X.; Qian, J.; Zheng, X.; Chu, Y. Glucocorticoids inhibit lipopolysaccharide-mediated inflammatory response by downregulating microRNA-155: A novel anti-inflammation mechanism. Free Radic. Biol. Med. 2012, 52, 1307–1317. [Google Scholar] [CrossRef]
  132. Wang, D.; Wang, L.; Bai, L.; Du, Y.; Liu, L.; Chen, X. Effects of Inhibition of miR-155-5p in Neural Stem Cell Subarachnoid Transplant on Rats with Cerebral Infarction. Hum. Gene Ther. Methods 2019, 30, 184–193. [Google Scholar] [CrossRef]
  133. Sun, L.; Ji, S.; Xing, J. Inhibition of microRNA-155 Alleviates Neurological Dysfunction Following Transient Global Ischemia and Contribution of Neuroinflammation and Oxidative Stress in the Hippocampus. Curr. Pharm. Des. 2019, 25, 4310–4317. [Google Scholar] [CrossRef] [PubMed]
  134. Jing, H.; Liu, L.; Jia, Y.; Yao, H.; Ma, F. Overexpression of the long non-coding RNA Oprm1 alleviates apoptosis from cerebral ischemia-reperfusion injury through the Oprm1/miR-155/GATA3 axis. Artif. Cells Nanomed. Biotechnol. 2019, 47, 2431–2439. [Google Scholar] [CrossRef] [PubMed]
  135. Xing, G.; Luo, Z.; Zhong, C.; Pan, X.; Xu, X. Influence of miR-155 on Cell Apoptosis in Rats with Ischemic Stroke: Role of the Ras Homolog Enriched in Brain (Rheb)/mTOR Pathway. Med. Sci. Monit. 2016, 22, 5141–5153. [Google Scholar] [CrossRef] [PubMed]
  136. Lu, J.; Xu, F.; Lu, H. LncRNA PVT1 regulates ferroptosis through miR-214-mediated TFR1 and p53. Life Sci. 2020, 260, 118305. [Google Scholar] [CrossRef] [PubMed]
  137. Griffiths-Jones, S. The microRNA Registry. Nucleic Acids Res. 2004, 32, D109–D111. [Google Scholar] [CrossRef]
  138. Sun, H.; Wang, C.; Zhou, Y.; Cheng, X. Long Noncoding RNA OIP5-AS1 Overexpression Promotes Viability and Inhibits High Glucose-Induced Oxidative Stress of Cardiomyocytes by Targeting MicroRNA-34a/SIRT1 Axis in Diabetic Cardiomyopathy. Endocr. Metab. Immune Disord. Drug Targets 2021, 21, 2017–2027. [Google Scholar] [CrossRef]
  139. Yoon, D.; Pastore, Y.D.; Divoky, V.; Liu, E.; Mlodnicka, A.E.; Rainey, K.; Ponka, P.; Semenza, G.L.; Schumacher, A.; Prchal, J.T. Hypoxia-inducible factor-1 deficiency results in dysregulated erythropoiesis signaling and iron homeostasis in mouse development. J. Biol. Chem. 2006, 281, 25703–25711. [Google Scholar] [CrossRef]
  140. Shahjouei, S.; Cai, P.Y.; Ansari, S.; Sharififar, S.; Azari, H.; Ganji, S.; Zand, R. Middle Cerebral Artery Occlusion Model of Stroke in Rodents: A Step-by-Step Approach. J. Vasc. Interv. Neurol. 2016, 8, 1–8. [Google Scholar]
  141. Li, Y.; Sun, J.; Wu, R.; Bai, J.; Hou, Y.; Zeng, Y.; Zhang, Y.; Wang, X.; Wang, Z.; Meng, X. Mitochondrial MPTP: A Novel Target of Ethnomedicine for Stroke Treatment by Apoptosis Inhibition. Front. Pharmacol. 2020, 11, 352. [Google Scholar] [CrossRef]
  142. Wu, X.; Wang, Y.; Yu, T.; Nie, E.; Hu, Q.; Wu, W.; Zhi, T.; Jiang, K.; Wang, X.; Lu, X.; et al. Blocking MIR155HG/miR-155 axis inhibits mesenchymal transition in glioma. Neuro Oncol. 2017, 19, 1195–1205. [Google Scholar] [CrossRef]
  143. Liang, S.; Hu, J.; Zhang, A.; Li, F.; Li, X. miR-155 induces endothelial cell apoptosis and inflammatory response in atherosclerosis by regulating Bmal1. Exp. Ther. Med. 2020, 20, 128. [Google Scholar] [CrossRef]
  144. Silva, J.P.D.; Lizarte Neto, F.S.; Cirino, M.L.A.; Carvalho, C.A.M.; Carlotti, C.G., Jr.; Colli, B.O.; Tirapelli, D.; Tirapelli, L.F. Analysis of Caspase-9 protein and microRNAs miR-21, miR-126 and miR-155 related to the apoptosis mechanism in the cerebellum of rats submitted to focal cerebral ischemia associated with an alcoholism model. Arq. Neuropsiquiatr. 2019, 77, 689–695. [Google Scholar] [CrossRef]
  145. Zhang, R.; Wu, Y.; Xie, F.; Zhong, Y.; Wang, Y.; Xu, M.; Feng, J.; Charish, J.; Monnier, P.P.; Qin, X. RGMa mediates reactive astrogliosis and glial scar formation through TGFβ1/Smad2/3 signaling after stroke. Cell Death Differ. 2018, 25, 1503–1516. [Google Scholar] [CrossRef] [PubMed]
  146. Pena-Philippides, J.C.; Caballero-Garrido, E.; Lordkipanidze, T.; Roitbak, T. In vivo inhibition of miR-155 significantly alters post-stroke inflammatory response. J. Neuroinflammation 2016, 13, 287. [Google Scholar] [CrossRef]
  147. Hou, J.; Wang, P.; Lin, L.; Liu, X.; Ma, F.; An, H.; Wang, Z.; Cao, X. MicroRNA-146a feedback inhibits RIG-I-dependent Type I IFN production in macrophages by targeting TRAF6, IRAK1, and IRAK2. J. Immunol. 2009, 183, 2150–2158. [Google Scholar] [CrossRef]
  148. Kundu, M.; Basu, J. The Role of microRNAs and Long Non-Coding RNAs in the Regulation of the Immune Response to Mycobacterium tuberculosis Infection. Front. Immunol. 2021, 12, 687962. [Google Scholar] [CrossRef]
  149. Nejad, C.; Stunden, H.J.; Gantier, M.P. A guide to miRNAs in inflammation and innate immune responses. FEBS J. 2018, 285, 3695–3716. [Google Scholar] [CrossRef]
  150. Sheedy, F.J.; Palsson-McDermott, E.; Hennessy, E.J.; Martin, C.; O’Leary, J.J.; Ruan, Q.; Johnson, D.S.; Chen, Y.; O’Neill, L.A. Negative regulation of TLR4 via targeting of the proinflammatory tumor suppressor PDCD4 by the microRNA miR-21. Nat. Immunol. 2010, 11, 141–147. [Google Scholar] [CrossRef] [PubMed]
  151. Yang, J.; Zhu, X.; Hu, H.; Lin, X. The inhibitory effect of Gualou Guizhi Decoction on post-ischemic neuroinflammation via miR-155 in MCAO rats. Ann. Palliat. Med. 2021, 10, 1370–1379. [Google Scholar] [CrossRef]
  152. Das, A.; Sinha, M.; Datta, S.; Abas, M.; Chaffee, S.; Sen, C.K.; Roy, S. Monocyte and macrophage plasticity in tissue repair and regeneration. Am. J. Pathol. 2015, 185, 2596–2606. [Google Scholar] [CrossRef]
  153. Forrest, A.R.; Kanamori-Katayama, M.; Tomaru, Y.; Lassmann, T.; Ninomiya, N.; Takahashi, Y.; de Hoon, M.J.; Kubosaki, A.; Kaiho, A.; Suzuki, M.; et al. Induction of microRNAs, mir-155, mir-222, mir-424 and mir-503, promotes monocytic differentiation through combinatorial regulation. Leukemia 2010, 24, 460–466. [Google Scholar] [CrossRef] [PubMed]
  154. Squadrito, M.L.; Etzrodt, M.; De Palma, M.; Pittet, M.J. MicroRNA-mediated control of macrophages and its implications for cancer. Trends Immunol. 2013, 34, 350–359. [Google Scholar] [CrossRef]
  155. Zuo, L.; Zhang, L.; Zu, J.; Wang, Z.; Han, B.; Chen, B.; Cheng, M.; Ju, M.; Li, M.; Shu, G.; et al. Circulating Circular RNAs as Biomarkers for the Diagnosis and Prediction of Outcomes in Acute Ischemic Stroke. Stroke 2020, 51, 319–323. [Google Scholar] [CrossRef]
  156. Sun, P.; Zhang, K.; Hassan, S.H.; Zhang, X.; Tang, X.; Pu, H.; Stetler, R.A.; Chen, J.; Yin, K.J. Endothelium-Targeted Deletion of microRNA-15a/16-1 Promotes Poststroke Angiogenesis and Improves Long-Term Neurological Recovery. Circ. Res. 2020, 126, 1040–1057. [Google Scholar] [CrossRef] [PubMed]
  157. Zhang, L.; Dong, L.Y.; Li, Y.J.; Hong, Z.; Wei, W.S. The microRNA miR-181c controls microglia-mediated neuronal apoptosis by suppressing tumor necrosis factor. J. Neuroinflammation 2012, 9, 211. [Google Scholar] [CrossRef]
  158. Wen, Y.; Zhang, X.; Dong, L.; Zhao, J.; Zhang, C.; Zhu, C. Acetylbritannilactone Modulates MicroRNA-155-Mediated Inflammatory Response in Ischemic Cerebral Tissues. Mol. Med. 2015, 21, 197–209. [Google Scholar] [CrossRef]
  159. Tan, K.S.; Armugam, A.; Sepramaniam, S.; Lim, K.Y.; Setyowati, K.D.; Wang, C.W.; Jeyaseelan, K. Expression profile of MicroRNAs in young stroke patients. PLoS ONE 2009, 4, e7689. [Google Scholar] [CrossRef]
  160. Moon, J.M.; Xu, L.; Giffard, R.G. Inhibition of microRNA-181 reduces forebrain ischemia-induced neuronal loss. J. Cereb. Blood Flow Metab. 2013, 33, 1976–1982. [Google Scholar] [CrossRef]
  161. Li, Y.; Mao, L.; Gao, Y.; Baral, S.; Zhou, Y.; Hu, B. MicroRNA-107 contributes to post-stroke angiogenesis by targeting Dicer-1. Sci. Rep. 2015, 5, 13316. [Google Scholar] [CrossRef]
  162. Xin, H.; Wang, F.; Li, Y.; Lu, Q.E.; Cheung, W.L.; Zhang, Y.; Zhang, Z.G.; 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 Transplant. 2017, 26, 243–257. [Google Scholar]
  163. Xu, X.; Wen, Z.; Zhao, N.; Xu, X.; Wang, F.; Gao, J.; Jiang, Y.; Liu, X. MicroRNA-1906, a Novel Regulator of Toll-Like Receptor 4, Ameliorates Ischemic Injury after Experimental Stroke in Mice. J. Neurosci. 2017, 37, 10498–10515. [Google Scholar] [CrossRef]
  164. 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. 2015, 355, 113–119. [Google Scholar] [CrossRef] [PubMed]
  165. Yin, K.J.; Deng, Z.; Huang, H.; Hamblin, M.; Xie, C.; Zhang, J.; Chen, Y.E. miR-497 regulates neuronal death in mouse brain after transient focal cerebral ischemia. Neurobiol. Dis. 2010, 38, 17–26. [Google Scholar] [CrossRef]
  166. Zhao, H.; Wang, J.; Gao, L.; Wang, R.; Liu, X.; Gao, Z.; Tao, Z.; Xu, C.; Song, J.; Ji, X.; et al. MiRNA-424 protects against permanent focal cerebral ischemia injury in mice involving suppressing microglia activation. Stroke 2013, 44, 1706–1713. [Google Scholar] [CrossRef]
  167. Rahmati, M.; Ferns, G.A.; Mobarra, N. The lower expression of circulating miR-210 and elevated serum levels of HIF-1α in ischemic stroke; Possible markers for diagnosis and disease prediction. J. Clin. Lab. Anal. 2021, 35, e24073. [Google Scholar] [CrossRef]
  168. Tiedt, S.; Dichgans, M. Role of Non-Coding RNAs in Stroke. Stroke 2018, 49, 3098–3106. [Google Scholar] [CrossRef]
  169. Srinivasan, S.; Yeri, A.; Cheah, P.S.; Chung, A.; Danielson, K.; De Hoff, P.; Filant, J.; Laurent, C.D.; Laurent, L.D.; Magee, R.; et al. Small RNA Sequencing across Diverse Biofluids Identifies Optimal Methods for exRNA Isolation. Cell 2019, 177, 446–462.e416. [Google Scholar] [CrossRef]
  170. Deng, Y.; Zhu, Y.; Wang, H.; Khadka, V.S.; Hu, L.; Ai, J.; Dou, Y.; Li, Y.; Dai, S.; Mason, C.E.; et al. Ratio-Based Method To Identify True Biomarkers by Normalizing Circulating ncRNA Sequencing and Quantitative PCR Data. Anal. Chem. 2019, 91, 6746–6753. [Google Scholar] [CrossRef]
  171. Chen, G.; Qian, H.M.; Chen, J.; Wang, J.; Guan, J.T.; Chi, Z.L. Whole transcriptome sequencing identifies key circRNAs, lncRNAs, and miRNAs regulating neurogenesis in developing mouse retina. BMC Genom. 2021, 22, 779. [Google Scholar] [CrossRef]
  172. Weber, J.A.; Baxter, D.H.; Zhang, S.; Huang, D.Y.; Huang, K.H.; Lee, M.J.; Galas, D.J.; Wang, K. The microRNA spectrum in 12 body fluids. Clin. Chem. 2010, 56, 1733–1741. [Google Scholar] [CrossRef]
  173. Pfaffl, M.W. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res. 2001, 29, e45. [Google Scholar] [CrossRef] [PubMed]
  174. Yokoi, A.; Matsuzaki, J.; Yamamoto, Y.; Yoneoka, Y.; Takahashi, K.; Shimizu, H.; Uehara, T.; Ishikawa, M.; Ikeda, S.I.; Sonoda, T.; et al. Integrated extracellular microRNA profiling for ovarian cancer screening. Nat. Commun. 2018, 9, 4319. [Google Scholar] [CrossRef] [PubMed]
  175. Todo, K.; Iwata, T.; Doijiri, R.; Yamagami, H.; Morimoto, M.; Hashimoto, T.; Sonoda, K.; Yamazaki, H.; Koge, J.; Okazaki, S.; et al. Frequent Premature Atrial Contractions in Cryptogenic Stroke Predict Atrial Fibrillation Detection with Insertable Cardiac Monitoring. Cerebrovasc. Dis. 2020, 49, 144–150. [Google Scholar] [CrossRef]
  176. Zhou, W.Y.; Cai, Z.R.; Liu, J.; Wang, D.S.; Ju, H.Q.; Xu, R.H. Circular RNA: Metabolism, functions and interactions with proteins. Mol. Cancer 2020, 19, 172. [Google Scholar] [CrossRef]
  177. Denk, J.; Oberhauser, F.; Kornhuber, J.; Wiltfang, J.; Fassbender, K.; Schroeter, M.L.; Volk, A.E.; Diehl-Schmid, J.; Prudlo, J.; Danek, A.; et al. Specific serum and CSF microRNA profiles distinguish sporadic behavioural variant of frontotemporal dementia compared with Alzheimer patients and cognitively healthy controls. PLoS ONE 2018, 13, e0197329. [Google Scholar] [CrossRef]
  178. Piscopo, P.; Grasso, M.; Puopolo, M.; D’Acunto, E.; Talarico, G.; Crestini, A.; Gasparini, M.; Campopiano, R.; Gambardella, S.; Castellano, A.E.; et al. Circulating miR-127-3p as a Potential Biomarker for Differential Diagnosis in Frontotemporal Dementia. J. Alzheimers Dis. 2018, 65, 455–464. [Google Scholar] [CrossRef]
  179. Ahlbrecht, J.; Martino, F.; Pul, R.; Skripuletz, T.; Sühs, K.W.; Schauerte, C.; Yildiz, Ö.; Trebst, C.; Tasto, L.; Thum, S.; et al. Deregulation of microRNA-181c in cerebrospinal fluid of patients with clinically isolated syndrome is associated with early conversion to relapsing-remitting multiple sclerosis. Mult. Scler. 2016, 22, 1202–1214. [Google Scholar] [CrossRef]
  180. Vistbakka, J.; Elovaara, I.; Lehtimäki, T.; Hagman, S. Circulating microRNAs as biomarkers in progressive multiple sclerosis. Mult. Scler. 2017, 23, 403–412. [Google Scholar] [CrossRef]
  181. Swarbrick, S.; Wragg, N.; Ghosh, S.; Stolzing, A. Systematic Review of miRNA as Biomarkers in Alzheimer’s Disease. Mol. Neurobiol. 2019, 56, 6156–6167. [Google Scholar] [CrossRef]
  182. Takousis, P.; Sadlon, A.; Schulz, J.; Wohlers, I.; Dobricic, V.; Middleton, L.; Lill, C.M.; Perneczky, R.; Bertram, L. Differential expression of microRNAs in Alzheimer’s disease brain, blood, and cerebrospinal fluid. Alzheimers Dement. 2019, 15, 1468–1477. [Google Scholar] [CrossRef]
  183. Zailaie, S.A.; Siddiqui, J.J.; Al Saadi, R.M.; Anbari, D.M.; Alomari, A.S.; Cupler, E.J. Serum Based miRNA as a Diagnostic Biomarker for Multiple Sclerosis: A Systematic Review and Meta-Analysis. Immunol. Investig. 2022, 51, 947–962. [Google Scholar] [CrossRef] [PubMed]
  184. Sonoda, T.; Matsuzaki, J.; Yamamoto, Y.; Sakurai, T.; Aoki, Y.; Takizawa, S.; Niida, S.; Ochiya, T. Serum MicroRNA-Based Risk Prediction for Stroke. Stroke 2019, 50, 1510–1518. [Google Scholar] [CrossRef]
  185. Lawrie, C.H.; Gal, S.; Dunlop, H.M.; Pushkaran, B.; Liggins, A.P.; Pulford, K.; Banham, A.H.; Pezzella, F.; Boultwood, J.; Wainscoat, J.S.; et al. Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. Br. J. Haematol. 2008, 141, 672–675. [Google Scholar] [CrossRef]
  186. Larrabeiti-Etxebarria, A.; Lopez-Santillan, M.; Santos-Zorrozua, B.; Lopez-Lopez, E.; Garcia-Orad, A. Systematic Review of the Potential of MicroRNAs in Diffuse Large B Cell Lymphoma. Cancers 2019, 11, 144. [Google Scholar] [CrossRef]
  187. Ghafoor, M.; Kamal, M.; Nadeem, U.; Husain, A.N. Educational Case: Myocardial Infarction: Histopathology and Timing of Changes. Acad. Pathol. 2020, 7, 2374289520976639. [Google Scholar] [CrossRef]
  188. Bejleri, J.; Jirström, E.; Donovan, P.; Williams, D.J.; Pfeiffer, S. Diagnostic and Prognostic Circulating MicroRNA in Acute Stroke: A Systematic and Bioinformatic Analysis of Current Evidence. J. Stroke 2021, 23, 162–182. [Google Scholar] [CrossRef]
  189. Zhang, L.; Wang, H. Long Non-coding RNA in CNS Injuries: A New Target for Therapeutic Intervention. Mol. Ther. Nucleic Acids 2019, 17, 754–766. [Google Scholar] [CrossRef]
  190. Marto, J.P.; Carvalho, A.S.; Mollet, I.G.; Mendonça, M.; Salavisa, M.; Meira, B.; Fernandes, M.; Serrazina, F.; Cabral, G.; Ventura, R.; et al. Proteomics to Identify New Blood Biomarkers for Diagnosing Patients With Acute Stroke. J. Am. Heart Assoc. 2023, 12, e030021. [Google Scholar] [CrossRef]
  191. Li, Q.; Zhao, L.; Chan, C.L.; Zhang, Y.; Tong, S.W.; Zhang, X.; Ho, J.W.K.; Jiao, Y.; Rainer, T.H. Multi-Level Biomarkers for Early Diagnosis of Ischaemic Stroke: A Systematic Review and Meta-Analysis. Int. J. Mol. Sci. 2023, 24, 13821. [Google Scholar] [CrossRef]
  192. di Biase, L.; Bonura, A.; Pecoraro, P.M.; Carbone, S.P.; Di Lazzaro, V. Unlocking the Potential of Stroke Blood Biomarkers: Early Diagnosis, Ischemic vs. Haemorrhagic Differentiation and Haemorrhagic Transformation Risk: A Comprehensive Review. Int. J. Mol. Sci. 2023, 24, 11545. [Google Scholar] [CrossRef]
  193. Soun, J.E.; Chow, D.S.; Nagamine, M.; Takhtawala, R.S.; Filippi, C.G.; Yu, W.; Chang, P.D. Artificial Intelligence and Acute Stroke Imaging. AJNR Am. J. Neuroradiol. 2021, 42, 2–11. [Google Scholar] [CrossRef] [PubMed]
  194. Bonkhoff, A.K.; Grefkes, C. Precision medicine in stroke: Towards personalized outcome predictions using artificial intelligence. Brain 2022, 145, 457–475. [Google Scholar] [CrossRef] [PubMed]
  195. Shlobin, N.A.; Baig, A.A.; Waqas, M.; Patel, T.R.; Dossani, R.H.; Wilson, M.; Cappuzzo, J.M.; Siddiqui, A.H.; Tutino, V.M.; Levy, E.I. Artificial Intelligence for Large-Vessel Occlusion Stroke: A Systematic Review. World Neurosurg. 2022, 159, 207–220.e201. [Google Scholar] [CrossRef]
  196. Caballero-Garrido, E.; Pena-Philippides, J.C.; Lordkipanidze, T.; Bragin, D.; Yang, Y.; Erhardt, E.B.; Roitbak, T. In Vivo Inhibition of miR-155 Promotes Recovery after Experimental Mouse Stroke. J. Neurosci. 2015, 35, 12446–12464. [Google Scholar] [CrossRef]
  197. Brillante, S.; Volpe, M.; Indrieri, A. Advances in MicroRNA Therapeutics: From Preclinical to Clinical Studies. Hum. Gene Ther. 2024, 35, 628–648. [Google Scholar] [CrossRef]
  198. Diener, C.; Keller, A.; Meese, E. Emerging concepts of miRNA therapeutics: From cells to clinic. Trends Genet. 2022, 38, 613–626. [Google Scholar] [CrossRef]
  199. Anthiya, S.; Griveau, A.; Loussouarn, C.; Baril, P.; Garnett, M.; Issartel, J.P.; Garcion, E. MicroRNA-Based Drugs for Brain Tumors. Trends Cancer 2018, 4, 222–238. [Google Scholar] [CrossRef]
  200. Seto, A.G.; Beatty, X.; Lynch, J.M.; Hermreck, M.; Tetzlaff, M.; Duvic, M.; Jackson, A.L. Cobomarsen, an oligonucleotide inhibitor of miR-155, co-ordinately regulates multiple survival pathways to reduce cellular proliferation and survival in cutaneous T-cell lymphoma. Br. J. Haematol. 2018, 183, 428–444. [Google Scholar] [CrossRef]
  201. Tang, L.; Chen, H.Y.; Hao, N.B.; Tang, B.; Guo, H.; Yong, X.; Dong, H.; Yang, S.M. microRNA inhibitors: Natural and artificial sequestration of microRNA. Cancer Lett. 2017, 407, 139–147. [Google Scholar] [CrossRef]
  202. Jiang, F.; Du, L.; Chen, Z.J.; Wang, X.; Ge, D.; Liu, N. LNP-miR-155 cy5 Inhibitor Regulates the Copper Transporter via the β-Catenin/TCF4/SLC31A1 Signal for Colorectal Cancer Therapy. Mol. Pharm. 2023, 20, 4138–4152. [Google Scholar] [CrossRef]
  203. Sayyed, A.A.; Gondaliya, P.; Mali, M.; Pawar, A.; Bhat, P.; Khairnar, A.; Arya, N.; Kalia, K. MiR-155 Inhibitor-Laden Exosomes Reverse Resistance to Cisplatin in a 3D Tumor Spheroid and Xenograft Model of Oral Cancer. Mol. Pharm. 2021, 18, 3010–3025. [Google Scholar] [CrossRef]
  204. Lv, H.; Guo, J.; Li, S.; Jiang, D. miR-155 inhibitor reduces the proliferation and migration in osteosarcoma MG-63 cells. Exp. Ther. Med. 2014, 8, 1575–1580. [Google Scholar] [CrossRef] [PubMed]
  205. van der Ree, M.H.; van der Meer, A.J.; van Nuenen, A.C.; de Bruijne, J.; Ottosen, S.; Janssen, H.L.; Kootstra, N.A.; Reesink, H.W. Miravirsen dosing in chronic hepatitis C patients results in decreased microRNA-122 levels without affecting other microRNAs in plasma. Aliment. Pharmacol. Ther. 2016, 43, 102–113. [Google Scholar] [CrossRef] [PubMed]
  206. Hong, D.S.; Kang, Y.K.; Borad, M.; Sachdev, J.; Ejadi, S.; Lim, H.Y.; Brenner, A.J.; Park, K.; Lee, J.L.; Kim, T.Y.; et al. Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumours. Br. J. Cancer 2020, 122, 1630–1637. [Google Scholar] [CrossRef]
  207. Paoletti, A.; Rohmer, J.; Ly, B.; Pascaud, J.; Rivière, E.; Seror, R.; Le Goff, B.; Nocturne, G.; Mariette, X. Monocyte/Macrophage Abnormalities Specific to Rheumatoid Arthritis Are Linked to miR-155 and Are Differentially Modulated by Different TNF Inhibitors. J. Immunol. 2019, 203, 1766–1775. [Google Scholar] [CrossRef]
  208. Condrat, C.E.; Thompson, D.C.; Barbu, M.G.; Bugnar, O.L.; Boboc, A.; Cretoiu, D.; Suciu, N.; Cretoiu, S.M.; Voinea, S.C. miRNAs as Biomarkers in Disease: Latest Findings Regarding Their Role in Diagnosis and Prognosis. Cells 2020, 9, 276. [Google Scholar] [CrossRef]
  209. Chakrabortty, A.; Patton, D.J.; Smith, B.F.; Agarwal, P. miRNAs: Potential as Biomarkers and Therapeutic Targets for Cancer. Genes 2023, 14, 1375. [Google Scholar] [CrossRef] [PubMed]
  210. Mori, M.A.; Ludwig, R.G.; Garcia-Martin, R.; Brandão, B.B.; Kahn, C.R. Extracellular miRNAs: From Biomarkers to Mediators of Physiology and Disease. Cell Metab. 2019, 30, 656–673. [Google Scholar] [CrossRef]
  211. Gomes da Silva, A.M.; Silbiger, V.N. miRNAs as biomarkers of atrial fibrillation. Biomarkers 2014, 19, 631–636. [Google Scholar] [CrossRef] [PubMed]
  212. Butz, H.; Kinga, N.; Racz, K.; Patocs, A. Circulating miRNAs as biomarkers for endocrine disorders. J. Endocrinol. Investig. 2016, 39, 1–10. [Google Scholar] [CrossRef]
  213. Yan, H.; Ma, F.; Zhang, Y.; Wang, C.; Qiu, D.; Zhou, K.; Hua, Y.; Li, Y. miRNAs as biomarkers for diagnosis of heart failure: A systematic review and meta-analysis. Medicine 2017, 96, e6825. [Google Scholar] [CrossRef]
  214. Alipoor, S.D.; Adcock, I.M.; Garssen, J.; Mortaz, E.; Varahram, M.; Mirsaeidi, M.; Velayati, A. The roles of miRNAs as potential biomarkers in lung diseases. Eur. J. Pharmacol. 2016, 791, 395–404. [Google Scholar] [CrossRef]
  215. Herrold, A.A.; Kletzel, S.L.; Foecking, E.M.; Saban, K.L.; Przybycien-Szymanska, M.M.; Zilliox, M.; Bhaumik, D.; Lange, D.; Radke, J.R.; Salinas, I.; et al. miRNAs as Potential Biomarkers for Traumatic Brain Injury: Pathway From Diagnosis to Neurorehabilitation. J. Head Trauma Rehabil. 2021, 36, E155–E169. [Google Scholar] [CrossRef] [PubMed]
  216. Toffolo, K.; Osei, J.; Kelly, W.; Poulsen, A.; Donahue, K.; Wang, J.; Hunter, M.; Bard, J.; Wang, J.; Poulsen, D. Circulating microRNAs as biomarkers in traumatic brain injury. Neuropharmacology 2019, 145, 199–208. [Google Scholar] [CrossRef]
  217. Mushtaq, G.; Greig, N.H.; Anwar, F.; Zamzami, M.A.; Choudhry, H.; Shaik, M.M.; Tamargo, I.A.; Kamal, M.A. miRNAs as Circulating Biomarkers for Alzheimer’s Disease and Parkinson’s Disease. Med. Chem. 2016, 12, 217–225. [Google Scholar] [CrossRef]
  218. El-Koussy, M.; Schroth, G.; Brekenfeld, C.; Arnold, M. Imaging of acute ischemic stroke. Eur. Neurol. 2014, 72, 309–316. [Google Scholar] [CrossRef] [PubMed]
  219. Huţanu, A.; Iancu, M.; Bălaşa, R.; Maier, S.; Dobreanu, M. Predicting functional outcome of ischemic stroke patients in Romania based on plasma CRP, sTNFR-1, D-Dimers, NGAL and NSE measured using a biochip array. Acta Pharmacol. Sin. 2018, 39, 1228–1236. [Google Scholar] [CrossRef]
  220. Sotgiu, S.; Zanda, B.; Marchetti, B.; Fois, M.L.; Arru, G.; Pes, G.M.; Salaris, F.S.; Arru, A.; Pirisi, A.; Rosati, G. Inflammatory biomarkers in blood of patients with acute brain ischemia. Eur. J. Neurol. 2006, 13, 505–513. [Google Scholar] [CrossRef]
  221. Satoh, T.; Otsuka, A.; Contassot, E.; French, L.E. The inflammasome and IL-1β: Implications for the treatment of inflammatory diseases. Immunotherapy 2015, 7, 243–254. [Google Scholar] [CrossRef]
  222. Loddick, S.A.; Turnbull, A.V.; Rothwell, N.J. Cerebral interleukin-6 is neuroprotective during permanent focal cerebral ischemia in the rat. J. Cereb. Blood Flow Metab. 1998, 18, 176–179. [Google Scholar] [CrossRef]
  223. Nappi, F. Non-Coding RNA-Targeted Therapy: A State-of-the-Art Review. Int. J. Mol. Sci. 2024, 25, 3630. [Google Scholar] [CrossRef]
  224. Cai, Z.; Li, S.; Yu, T.; Deng, J.; Li, X.; Jin, J. Non-Coding RNA Regulatory Network in Ischemic Stroke. Front. Neurol. 2022, 13, 820858. [Google Scholar] [CrossRef] [PubMed]
  225. Wang, S.W.; Liu, Z.; Shi, Z.S. Non-Coding RNA in Acute Ischemic Stroke: Mechanisms, Biomarkers and Therapeutic Targets. Cell Transplant. 2018, 27, 1763–1777. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Timeline of Therapeutic Intervention in Treating Ischemic Stroke. (A) At the onset of stroke symptoms, there is a 4.5-h window to use alteplase (tPA), a powerful thrombolytic that works by converting plasminogen to plasmin to dissolve fibrin clots, and a 6-h window to use mechanical thrombectomy, a procedure where a catheter is physically inserted into a blood vessel to remove clots. Therapeutic options beyond these time windows are limited. (B) The overview of stroke pathology shows the ischemia triggers activation of simultaneous molecular cascades, (a) excitotoxicity and calcium overload, (b) oxidative stress, (c) apoptosis, and (d) neuroinflammation.
Figure 1. Timeline of Therapeutic Intervention in Treating Ischemic Stroke. (A) At the onset of stroke symptoms, there is a 4.5-h window to use alteplase (tPA), a powerful thrombolytic that works by converting plasminogen to plasmin to dissolve fibrin clots, and a 6-h window to use mechanical thrombectomy, a procedure where a catheter is physically inserted into a blood vessel to remove clots. Therapeutic options beyond these time windows are limited. (B) The overview of stroke pathology shows the ischemia triggers activation of simultaneous molecular cascades, (a) excitotoxicity and calcium overload, (b) oxidative stress, (c) apoptosis, and (d) neuroinflammation.
Ijms 26 03947 g001
Figure 2. Beneficial Effects of MiR-155 Inhibitors on Oxidative Stress. miR-155 inhibitors reduce oxidative stress. (A) LncRNA OIP5-AS1 inhibits miR-155-5p, preventing miR-155-5p from binding its target gene, IRF2B2, permitting IRF2B2 to reduce oxidative stress and inflammation. Note that “miR-155” refers to the whole miRNA molecule, “miR-155-5p” specifically denotes the mature form of miR-155 derived from the 5′ arm precursor molecule. (B) Neural stem cells (NSCs) treated with miR-155-5p inhibitor frees HIF-1α from miR-155, resulting in decreased oxidative stress, inflammation, infarct volume, and improved neurobehavioral outcomes. (C) miR-155 inhibitors decreased PICs in the hippocampus, resulting in decreased oxidative stress (8-iso PGF2α, 8-OHdG), neuroinflammation (IL-1β, IL-6 and TNF-α), improved neurological severity score and brain edema. miR-155 inhibitors have potential to decrease oxidative stress, apoptosis, neuroinflammation, and increase neuroprotection in ischemic stroke patients. It is predicted that miR-155 inhibitors will have some effect in restoring blood flow in the infarct area of the stroke, delaying neuronal death. Green arrows indicate beneficial outcomes in ischemic stroke.
Figure 2. Beneficial Effects of MiR-155 Inhibitors on Oxidative Stress. miR-155 inhibitors reduce oxidative stress. (A) LncRNA OIP5-AS1 inhibits miR-155-5p, preventing miR-155-5p from binding its target gene, IRF2B2, permitting IRF2B2 to reduce oxidative stress and inflammation. Note that “miR-155” refers to the whole miRNA molecule, “miR-155-5p” specifically denotes the mature form of miR-155 derived from the 5′ arm precursor molecule. (B) Neural stem cells (NSCs) treated with miR-155-5p inhibitor frees HIF-1α from miR-155, resulting in decreased oxidative stress, inflammation, infarct volume, and improved neurobehavioral outcomes. (C) miR-155 inhibitors decreased PICs in the hippocampus, resulting in decreased oxidative stress (8-iso PGF2α, 8-OHdG), neuroinflammation (IL-1β, IL-6 and TNF-α), improved neurological severity score and brain edema. miR-155 inhibitors have potential to decrease oxidative stress, apoptosis, neuroinflammation, and increase neuroprotection in ischemic stroke patients. It is predicted that miR-155 inhibitors will have some effect in restoring blood flow in the infarct area of the stroke, delaying neuronal death. Green arrows indicate beneficial outcomes in ischemic stroke.
Ijms 26 03947 g002
Figure 3. Effect of MiR-155 in Stroke Apoptosis. miR-155 promotes apoptosis in ischemic stroke. (A) Detrimental effects (direct)—miR-155 targets DUSP14, thereby regulating the NF-κB and MAPKs signaling pathways to worsen apoptosis. miR-155-5p decreases Rheb in the Rheb/mTOR pathway, resulting in increased NLRP3 inflammasomes and apoptosis. miR-155 binds the 3′-UTR of Rheb, inhibiting Rheb to promote increased apoptosis, infarct volume, and decreasing Rheb. (B) Detrimental effects (indirect)—miR-155 targets and inhibits SIRT1 to promote apoptosis. Ischemic stroke increases miR-155 which activates the TLR4/MyD88 signaling pathways, increasing apoptosis. (C) Beneficial Effects—Increased levels of lncRNA Oprm1 regulates the Oprm1/miR-155/GATA3 axis, reducing apoptosis and injury. miR-155 inhibitors reduce apoptosis, infarct volume, and increase Rheb, mTOR, and pS6K expression. Green arrows indicate beneficial outcomes, and red arrows indicate harmful outcomes in ischemic stroke.
Figure 3. Effect of MiR-155 in Stroke Apoptosis. miR-155 promotes apoptosis in ischemic stroke. (A) Detrimental effects (direct)—miR-155 targets DUSP14, thereby regulating the NF-κB and MAPKs signaling pathways to worsen apoptosis. miR-155-5p decreases Rheb in the Rheb/mTOR pathway, resulting in increased NLRP3 inflammasomes and apoptosis. miR-155 binds the 3′-UTR of Rheb, inhibiting Rheb to promote increased apoptosis, infarct volume, and decreasing Rheb. (B) Detrimental effects (indirect)—miR-155 targets and inhibits SIRT1 to promote apoptosis. Ischemic stroke increases miR-155 which activates the TLR4/MyD88 signaling pathways, increasing apoptosis. (C) Beneficial Effects—Increased levels of lncRNA Oprm1 regulates the Oprm1/miR-155/GATA3 axis, reducing apoptosis and injury. miR-155 inhibitors reduce apoptosis, infarct volume, and increase Rheb, mTOR, and pS6K expression. Green arrows indicate beneficial outcomes, and red arrows indicate harmful outcomes in ischemic stroke.
Ijms 26 03947 g003
Figure 4. Detrimental effects of miR-155 in neuroinflammation. miR-155 plays several roles in neuroinflammation. (A) miR-155 can be regulated through RNA modification, TLR signaling, and through inflammatory pathways through the MIRHA155 gene that controls miR-155 expression (green text). (B) miR-155 can activate or inhibit specific target genes, including KRAS, CD33, SOCS1 and SHIP1, to further promote inflammation (blue text). (C) miR-155 directly targets inflammatory pathways, including NF-kB, to promote inflammation (purple text). Red arrows indicate harmful outcomes in ischemic stroke.
Figure 4. Detrimental effects of miR-155 in neuroinflammation. miR-155 plays several roles in neuroinflammation. (A) miR-155 can be regulated through RNA modification, TLR signaling, and through inflammatory pathways through the MIRHA155 gene that controls miR-155 expression (green text). (B) miR-155 can activate or inhibit specific target genes, including KRAS, CD33, SOCS1 and SHIP1, to further promote inflammation (blue text). (C) miR-155 directly targets inflammatory pathways, including NF-kB, to promote inflammation (purple text). Red arrows indicate harmful outcomes in ischemic stroke.
Ijms 26 03947 g004
Figure 5. Detrimental Effects of MiR-155 in Stroke-related Microglia and Macrophage Polarization. (A) TLR4 and IFNγ, drivers of M1 microglia polarization, both increase miR-155 levels (green text). (B) miR-155 acts to promote M1 pro-inflammatory microglia polarization through multiple pathways. Increased miR-155 enhances cytokine production, autophagy, and M1 polarization, detected by CXCL9 (M1 marker) and IL-6 and TNF (M1/M2b markers). miR-155 levels are elevated during monocyte to macrophage differentiation and are increased when tumor-associated macrophages are reprogrammed to M1 macrophages. This is potentially through miR-155 targeting and suppressing SH2-containing inositol-5-phosphatase 1, IL13Rα1 and SMAD2/3, which promotes M1 macrophages (blue text). (C) miR-155 inhibits M2 anti-inflammatory microglia polarization through multiple pathways. miR-155 inhibits M2 polarization by inhibiting IL13RA and C/EBP in the IL13 and IL4 pathways, and by suppressing MafB, a transcription factor for M2 macrophage polarization. miR-155 also directly targets IL13Rα1 (a pro-M2 cytokine), causing reduced STAT6 activation, and affects IL-13-dependent regulation of SOCS1, DC-SIGN, CCL18, CD23, and SERPINE genes involved in differentiating M2 macrophages (purple text). Red arrows indicate harmful effects/outcomes in ischemic stroke.
Figure 5. Detrimental Effects of MiR-155 in Stroke-related Microglia and Macrophage Polarization. (A) TLR4 and IFNγ, drivers of M1 microglia polarization, both increase miR-155 levels (green text). (B) miR-155 acts to promote M1 pro-inflammatory microglia polarization through multiple pathways. Increased miR-155 enhances cytokine production, autophagy, and M1 polarization, detected by CXCL9 (M1 marker) and IL-6 and TNF (M1/M2b markers). miR-155 levels are elevated during monocyte to macrophage differentiation and are increased when tumor-associated macrophages are reprogrammed to M1 macrophages. This is potentially through miR-155 targeting and suppressing SH2-containing inositol-5-phosphatase 1, IL13Rα1 and SMAD2/3, which promotes M1 macrophages (blue text). (C) miR-155 inhibits M2 anti-inflammatory microglia polarization through multiple pathways. miR-155 inhibits M2 polarization by inhibiting IL13RA and C/EBP in the IL13 and IL4 pathways, and by suppressing MafB, a transcription factor for M2 macrophage polarization. miR-155 also directly targets IL13Rα1 (a pro-M2 cytokine), causing reduced STAT6 activation, and affects IL-13-dependent regulation of SOCS1, DC-SIGN, CCL18, CD23, and SERPINE genes involved in differentiating M2 macrophages (purple text). Red arrows indicate harmful effects/outcomes in ischemic stroke.
Ijms 26 03947 g005
Figure 6. Use of MiR-155 as early biomarker for stroke. (A) Use of miR-155 as biomarker of stroke timing. If charted out in a graph, clinicians could use the miR-155 level, or levels of time-sensitive markers to determine the length of time the patient has had a stroke to see if they are within the window for tPA therapy. (B) Use of miR-155 as biomarker of clinical improvement from stroke. If miR-155 or other markers were to be monitored in the hospital, physicians could use elevations of markers as indicators for new stroke developments that would require intervention. For a more comprehensive list of miRNA biomarkers involved in stroke pathophysiology, please see Tiedt 2018, [168]. The asterisk denotes the master role of miRNA-155 in stroke pathophysiology.
Figure 6. Use of MiR-155 as early biomarker for stroke. (A) Use of miR-155 as biomarker of stroke timing. If charted out in a graph, clinicians could use the miR-155 level, or levels of time-sensitive markers to determine the length of time the patient has had a stroke to see if they are within the window for tPA therapy. (B) Use of miR-155 as biomarker of clinical improvement from stroke. If miR-155 or other markers were to be monitored in the hospital, physicians could use elevations of markers as indicators for new stroke developments that would require intervention. For a more comprehensive list of miRNA biomarkers involved in stroke pathophysiology, please see Tiedt 2018, [168]. The asterisk denotes the master role of miRNA-155 in stroke pathophysiology.
Ijms 26 03947 g006
Table 1. MiR-155 Roles in Stroke Pathology.
Table 1. MiR-155 Roles in Stroke Pathology.
Signaling PathwaymiR-155 LevelsKey Target GeneFunctionReference
Neuroinflammation
Ras/NFkBUpregulatedKRASUpregulation of miR-155 target the KRAS gene inhibiting Ras/NFkB signaling and reducing proinflammatory cytokines (IL-17, IL-22, IFN-γ, TNF-α, IL-6 and 2).[100,101]
NFkBUpregulatedCD33miR-155 upregulation targets the CD33 gene leading to increased levels of proinflammatory cytokines (IL-1β, IL-8, TNF-α).[100,101]
NFkBUpregulatedN/AmiR-155 upregulation leads to increased inflammation through NFkB pathway activation, causing high levels of TLR-signaling inflammatory cytokines and type I interferons (IFNs).[102,103]
NFkB and TGFβN/AMIRHA155 GeneRegulatory elements in the MIRHA155 gene are stimulated by IL10 and IFNγ; and modulated by the NFkB pathway and the TGFβ pathway through SMAD4.[95,104,105,106]
TGFβUpregulatedSMAD2miR-155 targets SMAD2, a TGFβ signaling mediator, to decrease SMAD2 protein levels, TGF-β-induced SMAD-2 phosphorylation, and SMAD-2-dependent activation.[107]
TLR/MyD88, NF-κB, JNK/STAT, PI3K/Akt, and MAPKUpregulatedSOCS1 and SHIP1miR-155 targeting of SOCS1 and SHIP1 activates the TLR/MyD88, NF-κB, JNK/STAT, PI3K/Akt, and MAPK signaling pathways to promote secretion of inflammatory cytokines and chemokines.[72,108]
Wnt/β-cateninUpregulatedHBP1miR-155 regulates the Wnt/β-catenin signaling pathway by inhibiting HBP1.[72,108]
Ischemic strokeUpregulatedhypoxia-inducible factor (HIF)Increased miR-155 levels decrease HIF-1α mRNA, protein, and transcriptional activity in hypoxic conditions.[106]
Monocyte/macrophageUpregulatedN/AmiR-155 enhanced monocyte and macrophage production of proinflammatory cytokine CCL2.[104,109]
NFkBUpregulatedN/AmiR-155 inhibits BCL6, an inhibitor of NFkB.[104,105,109,110,111]
FTO/m6A RNA methylation, IL-1b/TNF-aUpregulatedN/ADownregulation of FTO expression increases N6—methyladenosine (m6 A) RNA modification of miR-155, which worsens infarct volume, neurological deficit, and inflammatory IL-1b and TNF-a levels.[112]
TLRUpregulatedN/ATLR2, TLR3, TLR4, and TLR9 induce miR-155 elevation.[113]
TLRUpregulatedN/AMyD88 and TRIF signaling pathways induce miR-155 expression. MyD88 is necessary for TLR2 and TLR9 signaling, and TRIF for TLR3 signaling.[114]
TNF-alphaUpregulatedN/ATNF-alpha is an miR-155 inducer. IFNs require TNF-alpha in macrophages to increase miR-155 levels.[113]
JNK PathwayUpregulatedN/ATNF-alpha or poly(I:C) stimulation of the JNK pathway increases miR-155 upregulation.[113]
BICUpregulatedBIC geneUpregulation of poly(I:C) or IFN- increases BIC mRNA involved in miR-155 upregulation. miR-155 is a downstream target of IFN- and an early gene target for poly(I:C).[113]
Apoptosis
ApoptosisUpregulatedSIRT1miR-155 targets and inhibits SIRT1, promoting apoptosis.[115]
ApoptosisUpregulated miR-155-5p directly targets and inhibits DUSP14 by binding the 3′UTR, thereby preventing DUSP14 from inhibiting the NF-κB and MAPKs signaling pathways, resulting in increased apoptosis.[116]
Apoptosis (TLR4/MyD88)Upregulated Increased miR-155 levels activate the TLR4/MyD88 signaling pathway, worsening the inflammatory response following stroke.[117]
Apoptosis (Rheb/mTOR)Upregulated miR-155-5p promotes apoptosis by suppressing Rheb expression and by promoting NLRP3-mediated inflammasomes.[118]
Microglia
M1 microglia polarizationUpregulated High miR-155 levels increase cytokine production, autophagy, and M1 pro-inflammatory polarization.[108]
M1 and M2 microglia polarizationUpregulated High levels of passenger strand miR-155 were detected in M1 and M2 microglia polarization.[119]
M1 and M2b polarizationUpregulated Transfection of macrophages with miR-155 increased CXCL9 (M1 marker), and IL-6 and TNF (M1 and M2b markers).[120]
M1 macrophage polarizationUpregulated IFNγ and TLR4 induce M1 macrophage polarization in vitro, and miR-155 level are upregulated by IFNγ and TLR4.[105]
M1 macrophage polarizationUpregulated miR-155 overexpression reprogrammed tumor-associated macrophages into M1 macrophages.[121]
M1 macrophage polarizationUpregulated miR-155 targets and suppresses SH2-containing inositol-5-phosphatase 1, IL13Rα1 and SMAD2/3, promoting M1 macrophages.[122,123]
M2 macrophage polarizationUpregulated miR-155 inhibits the anti-inflammatory M2 polarization by inhibiting IL13RA and C/EBP in the IL13 and IL4 pathways. [107,122,124,125]
M2 macrophage polarizationUpregulated miR-155 downregulated MafB, a transcription factor important for M2 macrophage polarization.[124]
M2 macrophage polarizationUpregulated IL-13, a pro-M2 cytokine, functions by binding to IL-13 receptor α1 (IL13Rα1), a part of the Type II IL-4 receptor, to activate signal transducer and activator of transcription 6 (STAT6). miR-155 directly targets and reduces IL13Rα1 protein levels, causing reduced STAT6 activation. [122]
M2 macrophage polarizationUpregulated miR-155 impacts IL-13-dependent regulation of SOCS1, DC-SIGN, CCL18, CD23, and SERPINE genes involved in differentiating M2 macrophages.[122]
Astrocyte polarizationUpregulated DJ-1 suppresses miR-155, regulating astrocyte activation via miR-155/SHP-1 signaling pathway. DJ-1 inhibits transition of astrocytes to harmful A1 subtypes, promoting beneficial A2 polarization.[126]
Table 2. MiR-155 Inhibitors in Management of Ischemic Stroke.
Table 2. MiR-155 Inhibitors in Management of Ischemic Stroke.
miR-155 LevelsPrincipal Target GeneFunctionReference
NFkBDownregulatedN/AmiR-155 overexpression reversed the anti-inflammatory effects of glucocorticoids, while miR-155 inhibition restored them.[130,131]
Oxidative StressDownregulatedN/AlncRNA OIP5-AS1 interacts and represses miR-155-5p, preventing miR-155-5p from binding its target IRF2BP2, which then suppresses oxidative stress.[129]
Oxidative StressDownregulatedN/AmiR-155-5p directly targets HIF-1α and negatively regulates its expression, promoting oxidative stress. miR-155-5p inhibitor in NSCs reversed miR-155-5p’s inhibition of HIF-1α, resulting in reduced oxidative stress and inflammation, decreased infarct volume and improved neurobehavioral outcomes.[132]
Oxidative StressDownregulatedN/AmiR-155 inhibitor significantly reduced upregulation of hippocampus pro-inflammatory cytokines (PICs), decreased oxidative stress, and improved neurological severity score and reduced brain edema.[133]
ApoptosisDownregulated Overexpression of lncRNA Oprm1 decreases apoptosis, through a lncRNA Oprm1/miR-155/GATA3 axis, by significantly decreasing infarct size and improving neurological score.[134]
ApoptosisDownregulated High cerebral infarct volumes and apoptosis were associated with increased miR-155 and decreased Rheb, mTOR, and pS6K, while treatment with miR-155 inhibitors were protected with reduced apoptosis and increased Rheb, mTOR, and p-S6K expression[135]
Table 3. MiR-155 vs Other NcRNAs involved in Cellular Processes of Ischemic Stroke.
Table 3. MiR-155 vs Other NcRNAs involved in Cellular Processes of Ischemic Stroke.
NcRNAFunction in Ischemic StrokeReference
miR-155
miR-146a/b
miR-21
miR-155, miR-146a/b and miR-21 create a trinity that regulate TLR pathways to control inflammation in stroke[119,147,148,149,150,151]
miR-155
miR-29b
miR-146a
miR-193b
miR-222
miR-155, in addition to miR-29b, miR-146a, miR-193b, and miR-222, are elevated in monocyte to macrophage differentiation.[120,153]
miR-9
miR-21
miR-24
miR-26a
miR-125a, b
miR-143
miR-145
miR-146a
miR-148
miR-187 miR-223 miR-378-3p miR-511-3p
miR-9, miR-21, miR-24, miR-26a, miR-125a, b, miR-143, miR-145, miR-146a, miR-148, miR-187, miR-223, miR-378-3p, miR-511-3p have been reported to play a role in macrophage polarization.[104,154]
circFUNDC1
circPDS5B
circCDC14A
High levels of circFUNDC1, circPDS5B and circCDC14A were found to be positively correlated with infarct volume in acute ischemic stroke.[155]
miR-15a/16-1 clusterEndothelium-targeted deletion of the miR-15a/16-1 cluster ameliorates blood–brain barrier dysfunction in ischemic stroke and poststroke angiogenesis[156]
miR-181cmiR-181 suppresses TNF-α expression in post-ischemic neuronal damage.[157]
miR-155miR-155 exerts both pro- and
anti-inflammatory effects by targeting
mediators of inflammatory signaling—
SHIP1, SOCS1, SMAD2 and TAB2.
[158]
miR-126Increased in endothelial cell or CV functions in ischemic stroke.[159]
miR-130Increased in angiogenesis in ischemic stroke.[159]
miR-181Increased in infarct core and decreased in penumbra after focal ischemia. miR-181 was shown to sensitize cells to apoptosis by reducing Bcl-2.[160]
miR-107Increased miR-107 levels may regulate post-ischemic stroke angiogenesis.[161]
miR-15a/16–1miR-15a/16–1 repress pro-angiogenic factors VEGFA and
FGF2 and their receptors VEGFR2 and FGFR1..
[156]
miR-133Overexpressing MSCs further stimulates and increases exosomes’ release from astrocytes, possibly by downregulating the RABEPK expression..[162]
miR-1906Is increased in glial cells and decreased in neurons. miR-1906 is involved in abolishment of TLR4 protein expression and could ameliorate brain injury in stroke.[163]
miR-99amiR-99a prevented apoptosis and blocked cell cycle progression in neuro-2a cells[164]
miR-497miR-497 promotes ischemic neuronal death by negatively regulating anti-apoptotic proteins bcl-2 and bcl-w.[165]
miR-424miR-424 prevents ischemic brain injury by suppressing microglia activation.[166]
miRNA-210 + HIF-1αHIF-1α induces miR-210 which could prevent apoptosis and induce angiogenesis.[167]
miR-124, miR-223, miR-107, miR-181aInvolved in regulating excitotoxicity in stroke.[168]
miR-21, miR-210, miR-424, miR-29b, miR-124, miR-15a, miR-181aInvolved in regulating apoptosis and programmed cell death in stroke.[168]
miR-29b, miR-124, miR-150, miR-155Involved in promoting blood–brain barrier breakdown after stroke.[168]
miR-21, miR-124, miR-223, miR-424, miR-15a, miR-155, miR-181a, miR-210, miR-377Involved in promoting inflammation in stroke.[168]
miR-124Involved in protective effects against stroke including angiogenesis and neurogenesis, and damaging effects of stroke including excitotoxicity, programmed cell death, blood–brain barrier breakdown, and inflammation.[168]
Table 4. Future Directions for MiR-155 in Diagnosis of Ischemic Stroke.
Table 4. Future Directions for MiR-155 in Diagnosis of Ischemic Stroke.
miR-155 Research Future Directions Recommendations
Profiling miR-155 stability in patientsTest the stability and abundance of miR-155 in CSF and blood serum IS patient samples as compared to healthy controls at different stages of the disease.
Measuring expression miR-155 levels variability in different patient populations with severity of stroke typesEstablishing miR-155 differential expression levels in different patient populations.
Need to evaluate efficacy in large scale cohort studiesAnalyze beneficial/adverse effects of miR-155 administration in large IS patient cohorts.
Refine integration of miR-155 with IS standard of care protocolsAnalysis of existing stroke diagnostic protocols and potential incorporation of miR-155 into diagnosis algorithm
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hering, C.; Conover, G.M. Advancing Ischemic Stroke Prognosis: Key Role of MiR-155 Non-Coding RNA. Int. J. Mol. Sci. 2025, 26, 3947. https://doi.org/10.3390/ijms26093947

AMA Style

Hering C, Conover GM. Advancing Ischemic Stroke Prognosis: Key Role of MiR-155 Non-Coding RNA. International Journal of Molecular Sciences. 2025; 26(9):3947. https://doi.org/10.3390/ijms26093947

Chicago/Turabian Style

Hering, Catherine, and Gloria M. Conover. 2025. "Advancing Ischemic Stroke Prognosis: Key Role of MiR-155 Non-Coding RNA" International Journal of Molecular Sciences 26, no. 9: 3947. https://doi.org/10.3390/ijms26093947

APA Style

Hering, C., & Conover, G. M. (2025). Advancing Ischemic Stroke Prognosis: Key Role of MiR-155 Non-Coding RNA. International Journal of Molecular Sciences, 26(9), 3947. https://doi.org/10.3390/ijms26093947

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop