Translate this page into:
Circulating microRNAs and ischemic stroke: From blood biomarkers to pathogenesis: A systematic review
*Corresponding author: Alina Koshelyuk, Department of Neurology, Psychiatry and Rehabilitation, Karaganda Medical University, Karaganda, Kazakhstan. seryoginaa@qmu.kz
-
Received: ,
Accepted: ,
How to cite this article: Grigolashvili M, Kadyrova I, Shayakhmetova Y, Beisembayeva M, Muratbekova S, Koshelyuk A. Circulating microRNAs and ischemic stroke: From blood biomarkers to pathogenesis: A systematic review. J Neurosci Rural Pract. doi: 10.25259/JNRP_106_2025
Abstract
Objectives:
Stroke is one of the leading causes of death and disability worldwide. Many studies have highlighted the potential of circulating microRNAs as important biomarkers for predicting and diagnosing acute cerebrovascular diseases. They demonstrate high sensitivity and specificity, and often correlate with the severity of stroke and its consequences. The level of some microRNAs increases in the blood, while the regulation of other microRNAs tends to decrease.
Materials and Methods:
A targeted PubMed/Medline search has critically analyzed the relevant literature. The following keywords were used in the search: “ischemic stroke”, “microRNAs”, “biomarkers”, “plasma”, “diagnostics”, “pathogenesis”. The search was limited to original articles published in peer-reviewed scientific journals.
Results:
Changes in the microRNA level affect various pathological processes such as inflammation, cell apoptosis, oxidative stress, and endothelial dysfunction, which can eventually lead to the development of acute cerebral circulatory disorders. microRNAs are also involved in such pathogenesis links as atherosclerosis, regulation of angiogenesis, vascular remodeling, neuroprotection, proliferation and differentiation of neurons, as well as in maintaining the properties of endothelial cells.
Conclusion:
The mechanisms by which microRNAs are involved in the pathogenesis of neurological disorders may provide new targets for further innovative diagnostic and therapeutic strategies. In this paper, we conducted a systematic review of the literature on circulating microRNAs in the pathogenetic mechanisms of acute ischemic stroke, the expression of which changes in blood plasma.
Keywords
Acute cerebrovascular diseases
Biomarkers
Ischemic stroke
microRNA
Pathogenesis
INTRODUCTION
Stroke is the second leading cause of death and the third leading cause of disability worldwide.[1] The majority of stroke survivors experience irreversible neurological impairments that substantially impact their ability to work and overall quality of life. As such, acute cerebrovascular events require prompt diagnosis and immediate medical intervention.
Computed tomography (CT) and magnetic resonance imaging (MRI) remain the primary diagnostic tools for acute cerebrovascular diseases. However, neuroimaging techniques may not reliably detect early signs of stroke. CT is a rapid and reliable modality for identifying hemorrhagic stroke, but it lacks sufficient sensitivity for early ischemic changes, detecting only 40–60% of acute ischemic strokes within the first 6 h. This limitation can significantly affect patient eligibility for thrombolytic therapy. MRI offers greater sensitivity and specificity for diagnosing acute ischemic stroke, identifying approximately 80% of cerebral infarctions within the first 24 h. Nevertheless, MRI is often unavailable in rural or resource-limited settings and requires more time than CT to perform.[2]
At present, there is an active search for an investigation of blood-based biomarkers that could complement neuroimaging techniques and facilitate more rapid diagnosis and treatment of acute cerebrovascular diseases. Among these biomarkers are proteins, lipids, and metabolites; however, to date, none have demonstrated adequate sensitivity, specificity, speed, or accuracy to be reliably used in the clinical diagnosis and management of stroke.[3]
In recent years, growing attention has been paid to genetic mechanisms in acute cerebrovascular diseases. Among potential biomarkers, RNA-based molecules show promise for early stroke diagnosis, offering insights into pathogenesis and new treatment approaches. Circulating microRNAs (miRNAs) are particularly notable due to their high sensitivity, specificity, and correlation with stroke severity. These small non-coding RNAs (19–25 nucleotides) regulate gene expression by binding to 3' untranslated regions (3'UTRs) of target mRNAs, modulating post-transcriptional processes [Figure 1].[4] The involvement of miRNAs in the regulation of such important cellular processes as differentiation, proliferation, apoptosis, and stress response has been shown.

- MicroRNA (miRNA) biogenesis. In the cell nucleus, miRNA genes are transcribed into a precursor miRNA (pre-miRNA), which forms a characteristic hairpin structure. Pre- miRNA is exported to the cytoplasm, where it is processed into mature double-stranded miRNA (~22 nucleotides). miRNAs are directed and complementarily bind to the protein-coding region of target mRNA, and inhibit its translation or promote mRNA degradation.
Dysregulation of specific miRNAs is linked to various neurological disorders, with circulating miRNAs showing differential expression in the blood of stroke patients and across its subtypes.[5] The purpose of this article is to review current evidence on the role of circulating miRNAs in the pathogenetic mechanisms of acute ischemic stroke, emphasizing their altered expression patterns in blood plasma and their potential utility as biomarkers for diagnosis, prognosis, and therapeutic targeting.
MATERIALS AND METHODS
This systematic review was performed in line with the reporting guidelines of the systematic review and meta-analysis (PRISMA-2020) statement.
Eligibility criteria
Inclusion/exclusion criteria
The following requirements were met by articles before they were included in our review: (1) The cases with ischemic stroke; (2) the diagnosis of ischemic stroke was conducted based on neuroimaging; (3) there is a control group; (4) plasma samples were analyzed; and (5) published in peer-reviewed English-language journals.
Studies were excluded if: (1) miRNA levels were studied exclusively in animal models; (2) other types of non-coding RNA molecules have been studied; and (3) the article was not available in English.
Search strategy
A literature search was conducted in February 2025 on the Web of Science, PubMed, and Scopus. The search was performed using a combination of MeSH terms (medical subject headings) and keyword terms. Search groups of words representing the concepts of “ischemic stroke,” “microRNA,” “plasma,” “biomarker,” and “pathogenesis” were combined into queries using logical operators. The search was limited to original papers published in peer-reviewed scientific journals during or after 1993 until March 2025, inclusively; 1993 was set as a cutoff point as miRNAs were first described in this year.
Study selection and data extraction
To identify eligible studies, titles and abstracts obtained from the search strategy were screened by two independent researchers (AS: Alina Koshelyuk, ShM: Shynar Muratbekova) using a web-based tool for systematic review screening, Rayyan (https://www.rayyan.ai). Following this, eligible studies were then selected for inclusion after full-text analysis by three independent researchers (AS, MB: Mira Beisembayeva, YSh: Yelena Shayakhmetova). A consensus was met between reviewers to resolve any inclusion/exclusion differences.
Quality assessment
The quality of the included studies was assessed using the Newcastle-Ottawa Scale (NOS) [see Supplementary file]. The scale evaluates three domains: Selection of participants (up to 4 points), comparability of study groups (up to 2 points), and ascertainment of exposure (up to 3 points). The maximum score is 9. Studies scoring 7–9 points were considered high quality, 4–6 points as moderate quality, and <4 points as low quality. Quality assessment was conducted independently by two reviewers (AS and YSh), and disagreements were resolved through discussion or with the involvement of a third reviewer (MB).
RESULTS
Literature search
The PRISMA 2020 flow diagram is shown in Figure 2, which shows the studies that were used in this systematic review. A total of 1,617 articles were identified through electronic searches, including 531 from Web of Science, 482 from PubMed, and 604 from Scopus. After removing the duplicates, the titles and abstracts of the remaining articles (n = 1088) were used to determine their eligibility. Of the 118 articles assessed in full text, 109 were excluded for not meeting the eligibility criteria. Forty-four were excluded due to an inappropriate study design, and in 36 studies, blood samples other than plasma were used. Ultimately, nine articles were included in the review.
Quality assessment
The quality and validity assessment of the NOS checklist suggested that six studies were of high quality and three studies were of moderate quality. In all nine studies, the definition of “case” groups was adequate. In one study, there was no data on the comparison of case and control groups by age and gender. In six studies, information was provided on the comparison of the case and control groups according to additional criteria.

- Flow diagram of search and selection of studies in the systematic review.
Study design and methods
The design and methods of the included studies[6-14] are summarized in Table 1. Eight studies were conducted in China, and the remaining one in Germany. In six studies, a single miRNA was investigated in both the case and control groups. In the remaining studies, the expression levels of two, three, and five miRNAs were examined. In all studies, case groups were diagnosed using neuroimaging methods: MRI, CT, or magnetic resonance angiography. The number of patients with ischemic stroke ranges from 30 to 200, whereas the number of controls ranges from 21 to a maximum of 112. In seven studies, the number of male patients with stroke predominated over females, while two studies did not report sex characteristics. Plasma was used as the blood sample in all studies. Peripheral blood was collected from patients within the first 24 h after the onset of neurological symptoms in five studies, within the first 6 h and within 72 h in only one study. In one study, blood collection was performed during the acute phase without specifying the exact timing. Multiple post-stroke time points (24 h, 1 week, 4 weeks, 28 weeks, and 48 weeks) for blood sampling were reported in only one study.
| Study | Country | microRNA | Definition of ischemic stroke | Acute ischemic stroke patients | Control | ||
|---|---|---|---|---|---|---|---|
| Total (n) | Age | M (n, %) | Total | ||||
| Feng et al. 2015[6] | China | miR-487b | MRI | 30 | from 38 to 73 years | 19 (63%) | 30 |
| Yang et al. 2016[7] | China | miR-153 | Neurological examination and CT or MRI | 114 | 61±11.3 | 78 (68.4%) | 58 |
| Tiedt et al. 2017[8] | Germany | miR-125a-5p | Neurological examination and MRI (DWI) or CT | 40 (for validation) 200 (for replication) |
74.7±13.8 (for validation) 74.1±13.4 (for replication) |
22 (55%), 113 (56.5%) |
40 (for validation) 100 (for replication) |
| Jin and Xing 2017[9] | China | miR-222 | Neurological examination, CT, MRI, and/or MRA | 106 | 60.8±9.7 | 48 (45%) |
110 |
| Liu et al. 2019[10] | China | miR-128 | Neurological examination and MRI | 40 | 55–65 | N/A | 25 |
| Long et al. 2013[11] | China | miR-126 | Imaging studies** | 38 (24 h), 42 (1 w), 40 (4 w), 38 (24 w), 39 (48 w) |
19 (50%, 24 h), 20 (48%, 1 w), 21 (52.5, 4 w), 20 (52.6%, 24 w), 20 (51.3%, 48 w) |
62,5±6 (24 h), 64.0±6 (1 w), 64.0±6,3 (4 w), 65.0±6 (24 w), 64.8±6,8 (48 w) |
50 |
| Wu et al. 2020[12] | China | miR-99b | CT or MRI | 112 | 64.56±6.03 | 68 (60.7%) | 112 |
| Zhou and Zhang 2014[13] | China | miR-21 | Neurological examination, MRI and MRA | 68 | 64 (55.76) | 45 (66.2%) | 21 |
| Zhao et al. 2016[14] | China | miR-335 | Neurological examination, MRI and MRA | 168 | 70±8 | 88 (52.4%) | 104 |
| Study | Country | Control | Sample type | Sampling time point from stroke onset | Regulation | miRNA quantification | |
| Age | M (%) | ||||||
| Feng et al. 2015[6] | China | from 35 to 64 years | 17 (57%) | Plasma | Acute stage* | Up (P<0.05) | qRT-PCR |
| Yang et al. 2016[7] | China | 56±3.9 | 35 (60.3%) | Plasma | Within 24 h | Up (2.13-fold (2.13±0.10) |
qRT-PCR |
| Tiedt et al. 2017[8] | Germany | 69.7±8.8 (for validation) 65.6±13.4 (for replication) |
16 (40%), 35 (35%) | Plasma | Within 24 h | Up (1.8-fold;P=1.5×10−6) |
qRT-PCR |
| Up (2.5-fold;P=5.6×10−6) |
|||||||
| Up (4.8-fold;P=7.8×10−9) |
|||||||
| Jin and Xing 2017[9] | China | 58.6±15.2 | 59 (54%) | Plasma | Within 24 h | Up (10.62, P=0.032) |
qRT-PCR |
| Up (9.10, P=0.002) |
|||||||
| Up (6.88, P=0.011) |
|||||||
| Down (5.58, P<0.001) |
|||||||
| Down (7.28, P=0.033) |
|||||||
| Liu et al. 2019[10] | China | age-matched | N/A | Plasma | Within 72 h | Up (P=0,0288) | qRT-PCR |
| Long et al. 2013[11] | China | 64±6 | 24 (48%) | Plasma | At 24 h, 1 w, 4 w, 24 w and 48 w | Down (P<0.05) | qRT-PCR |
| Wu et al. 2020[12] | China | 63.42±5.71 | 70 (62,5%) | Plasma | Within 6 h | Down (−0.56, P<0.00) |
qRT-PCR |
| Zhou and Zhang 2014[13] | China | (54,67) | 10 (47,6%) | Plasma | Within 24 h | Down (P<0.05) |
qRT-PCR |
| Down (P<0.05) |
|||||||
| Zhao et al. 2016[14] | China | 69±9 | 55 (52,9%) | Plasma | Within 24 h | Down (P<0.001) | qRT-PCR |
Plasma circulating miRNAs
In all included studies, miRNA expression in plasma was analyzed using quantitative real-time polymerase chain reaction. A total of 16 miRNAs were investigated. The regulation of nine miRNAs was upregulated in the blood plasma (miR-487b, miR-153, miR-125a-5p, miR-125b-5p, miR-143-3p, miR-222, miR-218, miR-185, and miR-128), while the expression of the remaining seven miRNAs (miR-130a, miR-378, miR-126, miR-99b, miR-21, miR-24, and miR-335) was downregulated compared to the control group.
DISCUSSION
Circulating miRNAs contained in blood plasma are of particular interest because they are relatively stable and readily available.[15] The stability of circulating miRNAs is a clinically relevant characteristic. Alterations in miRNA expression profiles emerge immediately following focal cerebral ischemia, highlighting the involvement of miRNAs in the pathophysiological processes of ischemic stroke.
Upregulated Plasma miRNAs and Their Potential Roles in Stroke Pathophysiology
A key mechanism by which miR-487b may exert its effects is through the modulation of angiogenesis, a critical process in tissue repair and vascular remodeling following ischemic injury. In vitro studies using human umbilical vein endothelial cells (HUVECs) have demonstrated that miR-487b overexpression significantly promotes endothelial cell proliferation, migration, invasion, and capillary-like tube formation, all of which are essential steps in the angiogenic cascade.[6] These findings suggest that miR-487b may play a pro-angiogenic role, potentially enhancing vascular regeneration in ischemic tissues. Furthermore, miR-487b is thought to influence the expression of angiogenesis-related genes and signaling pathways, such as vascular endothelial growth factor (VEGF) and components of the PI3K/Akt or mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathways. Understanding the precise molecular targets and regulatory networks of miR-487b could provide valuable insights into its therapeutic potential in promoting angiogenesis and improving outcomes after acute ischemic stroke. In addition to its proangiogenic role, miR-487b has been implicated in promoting vascular injury under pathological conditions. It has been reported to contribute to cell death and loss of vascular wall integrity through suppression of insulin receptor substrate 1, an anti-apoptotic signaling molecule, particularly in the context of hypertension-related vascular disease.[16] miR-153 has been identified as a potential regulator of neuronal survival mechanisms following ischemic injury. Specifically, overexpression of miR-153 in cortical neurons leads to a significant increase in the activation of mTOR downstream effectors, suggesting that miR-153 may function as an activator of the mTOR signaling pathway. Given that the mTOR complexes (mTORC1 and mTORC2) are central mediators of cell metabolism, growth, and survival through integration of intracellular energy status, oxygen availability, amino acid levels, and extracellular growth factor signals, the modulation of mTOR activity by miR-153 may have important implications in the cellular response to ischemic stress. By enhancing mTOR signaling, miR-153 could contribute to neuroprotective processes, including inhibition of apoptosis, promotion of neuronal recovery, and metabolic adaptation during cerebral ischemia.[17] miR-143-3p plays a crucial role in maintaining vascular homeostasis through intercellular communication between vascular smooth muscle cells (VSMCs) and endothelial cells (ECs). It is a key regulatory molecule that contributes to the coordination of cellular responses required for vascular function and adaptation. In ECs, miR-143 contributes to vessel stabilization by repressing important target genes such as hexokinase II and integrin b8, thereby modulating endothelial metabolism and angiogenic capacity. By downregulating these proangiogenic and metabolic factors, miR-143 promotes a quiescent endothelial state, which is important for vessel maturation and stability. Simultaneously, in VSMCs, miR-143 supports a contractile and differentiated phenotype, which is essential for maintaining vascular tone and structural integrity. It achieves this by targeting signaling molecules involved in VSMC dedifferentiation and proliferation, such as ELK-1 (ETS Like-1 protein) and KLF4 (Kruppel-like factor 4), thus preventing phenotypic switching toward a synthetic, proliferative state often associated with vascular pathology. Given the importance of vascular remodeling, stabilization, and regeneration following ischemic injury, elevated levels of miR-143 after stroke may significantly influence endothelial function, angiogenesis, and structural repair of the vasculature in the post-stroke period.[18,19] Furthermore, dysregulation of miR-143 may contribute to impaired revascularization or pathological remodeling, making it a potential biomarker and therapeutic target for enhancing vascular recovery and improving neurological outcomes in patients with acute ischemic stroke. miR-125b has emerged as a potential regulator of neuronal function in the context of cerebral ischemia. A significant downregulation of miR-125b was observed during the early reperfusion period following ischemic stroke, mirroring changes in motor function assessments over the first 48 h.[20] This temporal association suggests that miR-125b may be involved in post-ischemic neuroplasticity and functional recovery, highlighting its potential as a target for miRNA-based therapies. Moreover, miR-125b-5p has been shown to regulate synaptic morphology and function, contribute to neuronal differentiation, and influence cytoskeletal organization – processes that are critically disrupted during and after ischemic injury.[21] miR-125a-5p has emerged as a key regulator of microglial cell fate in the context of cerebral ischemia. Its expression is significantly upregulated in microglial cells subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) conditions in vitro, as well as in the brains of rats following middle cerebral artery occlusion (MCAO). This elevated expression is associated with increased microglial apoptosis, suggesting a detrimental role for miR-125a-5p during ischemic injury. Experimental evidence indicates that downregulation of miR-125a-5p reduces microglial cell death, thereby exerting a protective effect against ischemia-induced injury. The pro-apoptotic function of miR-125a-5p is mediated through its direct targeting of insulin-like growth factor binding protein 3 (IGFBP3), a protein known for its role in promoting cell survival. Notably, silencing IGFBP3 abolishes the anti-apoptotic effect observed with miR-125a-5p inhibition, confirming the functional significance of the miR-125a-5p–IGFBP3 axis. Beyond its influence on apoptosis, miR-125a-5p may also contribute to the neuroinflammatory response by modulating microglial activity, including the release of pro-inflammatory cytokines and regulation of phagocytic behavior. These actions may amplify neuronal damage and hinder post-ischemic recovery. Collectively, these findings suggest that miR-125a-5p plays a critical role in ischemia-induced microglial dysfunction and represents a potential therapeutic target for attenuating neuroinflammation and limiting secondary brain injury following stroke.[22] miR-222 is involved in various physiological and pathological processes within the cardiovascular system, including vascular remodeling and atherosclerosis. Notably, reduced miR-222 expression has been observed at sites of atherosclerotic plaque rupture, suggesting its association with plaque stability.[23] In experimental models, suppression of miR-222 in the carotid arteries inhibited VSMC proliferation and reduced neointimal lesion formation following angioplasty. These findings indicate that miR-222 promotes VSMC proliferation and neointimal hyperplasia, processes that contribute to vascular injury and may influence stroke pathogenesis by affecting plaque vulnerability and post-ischemic vascular repair.[24] miR-218-5p has been implicated in the regulation of endothelial cell survival and vascular inflammation. A reduction in miR-218-5p levels exacerbates injury to HUVECs, particularly in the context of downregulated small nucleolar RNA SNHG12. This downregulation suppresses endothelial proliferation, promotes apoptosis, and enhances the inflammatory response, thereby contributing to the development of atherosclerosis.[25] Given the critical role of endothelial dysfunction and inflammation in stroke pathogenesis, miR-218-5p may serve as a protective factor in ischemic cerebrovascular injury. miR-185 has been shown to play a regulatory role in vascular homeostasis and the development of atherosclerosis, a major risk factor for ischemic stroke. One of its key molecular targets is stromal interaction molecule 1 (STIM1), a calcium sensor within the endoplasmic reticulum. Downregulation of miR-185 leads to increased STIM1 expression, which promotes vascular cell proliferation, migration, and invasion, thereby accelerating atherosclerotic plaque progression. The miR-185/STIM1 axis thus represents a potential mechanism linking vascular remodeling with ischemic stroke pathogenesis.[26] miR-128-3p has been identified as a critical regulator of neuronal survival in the context of cerebral ischemia. It exerts its effects primarily by targeting p38a (Mapk14), a well-known pro-apoptotic protein kinase involved in cellular stress responses, inflammation, and programmed cell death. Although p38a mRNA levels remain unchanged following ischemic injury, its protein expression is significantly reduced, indicating that miR-128-3p mediates a post-transcriptional regulatory mechanism rather than transcriptional silencing. Mechanistically, miR-128-3p binds directly to the 3'UTR of p38a mRNA, thereby repressing its translation into protein. This targeted suppression results in decreased p38a-mediated activation of downstream apoptotic pathways, including caspase activation and mitochondrial dysfunction, both of which are central to ischemia-induced neuronal death. Experimental studies have demonstrated that overexpression of miR-128-3p significantly reduces infarct volume, attenuates neuronal apoptosis, and improves functional neurological outcomes in animal models of acute ischemic stroke.[27] Conversely, inhibition of miR-128-3p leads to increased p38a protein levels, greater neuronal loss, and exacerbation of ischemic damage, further supporting its neuroprotective role. These findings highlight miR-128-3p as a promising therapeutic target for reducing neuronal injury and improving outcomes in patients with ischemic stroke, particularly through its selective suppression of stress-related pro-apoptotic pathways.
Downregulated Plasma miRNAs and Their Potential Roles in Stroke Pathophysiology
Besides increased regulation, reduced miRNA levels in blood plasma have been identified as potential biomarkers for early acute stroke detection [Table 1].
miR-126 is a key regulator of endothelial cell responses to growth factors such as VEGF and fibroblast growth factor, promoting angiogenesis primarily through activation of the MAPK signaling pathway. It exerts its pro-angiogenic effects by targeting Spred-1, a negative regulator of Raf phosphorylation and ERK activation. Suppression of Spred-1 by miR-126 enhances endothelial cell proliferation, migration, and cytoskeletal reorganization, all of which are essential for vascular repair following ischemic injury. Deficiency of miR-126 leads to impaired endothelial function and vascular abnormalities that closely resemble phenotypes observed with disrupted MAPK signaling, underscoring its critical role in post-stroke vascular remodeling.[28] Moreover, experimental models demonstrate that miR-126 restoration promotes revascularization and supports neurovascular integrity, highlighting its potential as a therapeutic target in ischemic stroke. miR-130a has been shown to exert neuroprotective effects in ischemic stroke models by modulating the PTEN/PI3K/AKT (Phosphatase and Tensin Homolog/Phosphatidylinositol 3-Kinase/Protein Kinase B) signaling pathway signaling pathway, which plays a central role in regulating cell survival, apoptosis, and oxidative stress responses. Its expression is significantly downregulated following oxygen-glucose deprivation/reperfusion (OGD/R) in PC12 cells and in the MCAO model in rats. Restoration of miR-130a levels improves cell viability, reduces apoptosis and oxidative damage, and leads to a marked decrease in infarct volume and neurological deficits. Mechanistically, miR-130a exerts its effects by directly targeting PTEN, a negative regulator of the PI3K/AKT pathway, thereby promoting AKT activation and enhancing neuronal survival after ischemic injury.[29] miR-99b has been shown to play a protective role in ischemic brain injury by enhancing neuronal cell survival and reducing apoptosis. In an in vitro OGD/R model, expression of miR-99b was significantly upregulated in SH-SY5Y neuroblastoma cells, suggesting a potential adaptive response to ischemic stress. Functional studies demonstrated that overexpression of miR-99b promotes cell viability, inhibits apoptotic cell death, and contributes to the preservation of neuronal integrity under ischemic conditions. Mechanistically, miR-99b directly targets the insulin-like growth factor 1 receptor (IGF1R), a critical mediator of neuronal survival and anti-apoptotic signaling. This interaction was confirmed using a luciferase reporter assay, supporting the specificity of miR-99b binding to the 3'UTR of IGF1R mRNA and its role in regulating downstream signaling pathways. Suppression of IGF1R expression by miR-99b may help to fine-tune IGF1-mediated responses, preventing excessive or maladaptive activation of survival signaling cascades during reperfusion.[12] These findings suggest that miR-99b contributes to neuroprotection by modulating IGF1R-dependent signaling, and its upregulation may represent an endogenous mechanism aimed at limiting ischemia-induced neuronal injury. Further investigation into the role of miR-99b in vivo may clarify its potential as a therapeutic target for stroke intervention. miR-21 plays a critical role in regulating the phenotype of VSMCs, which is essential for vascular remodeling and stabilization following ischemic stroke. Its expression is rapidly induced by transforming growth factor-beta and bone morphogenetic protein signaling through a Sma and Mad related proteins (SMAD)-dependent, post-transcriptional mechanism, which facilitates the processing of primary miR-21 transcripts (pri-miR-21) through enhanced interaction with the DROSHA (Drosha Ribonuclease III) microprocessor complex. Functionally, miR-21 downregulates programmed cell death protein 4, a known inhibitor of contractile gene expression, thereby promoting a contractile, differentiated VSMC phenotype and suppressing proliferation and phenotypic switching associated with vascular pathology.[30] This shift toward a contractile state contributes to vessel wall stabilization and may prevent maladaptive remodeling processes such as neointima formation or vascular stiffening. These findings suggest that miR-21 not only supports structural repair after stroke but may also serve as a potential target for therapeutic modulation of VSMC behavior in cerebrovascular disease. miR-24 plays a neuroprotective role in ischemic brain injury by modulating microglia/macrophage polarization. In a rat model of MCAO, overexpression of miR-24 reduced infarct volume and neurological deficits, while its inhibition exacerbated ischemic damage. miR-24 suppresses pro-inflammatory M1 polarization and promotes anti-inflammatory M2 polarization in both in vivo brain tissue and BV-2 microglial cells. Mechanistically, miR-24 targets Clcn3, a chloride channel protein, to regulate microglial phenotype.[31] These findings suggest that miR-24 contributes to post-stroke recovery by shifting microglial activation toward a protective, anti-inflammatory state. miR-335 is downregulated in the acute phase of ischemic stroke and plays a key role in regulating neuronal survival and stress granule (SG) formation. In a rat MCAO model, restoration of miR-335 expression promoted SG assembly, reduced infarct volume, and suppressed apoptosis. Mechanistically, miR-335 directly targets Rho-associated protein kinase-2 (ROCK2), whose inhibition contributes to enhanced SG formation and decreased neuronal death.[32] These findings suggest that miR-335 exerts neuroprotective effects during ischemic injury by modulating the ROCK2 pathway and cellular stress responses. miR-378 has been identified as a neuroprotective factor in ischemic stroke, with its expression markedly reduced in both the peri-infarct region of MCAO mice and OGD-treated neuronal cells. Overexpression of miR-378 enhances cell viability and reduces apoptosis by directly targeting the 3'-UTR of Caspase-3 mRNA, thereby suppressing its expression and activity. In vivo, miR-378 agomir treatment led to decreased cleaved-caspase-3 levels, reduced infarct volume, and mitigated neuronal death following MCAO.[33] These findings suggest that miR-378 protects against ischemic injury by inhibiting caspase-3–mediated apoptosis, making it a promising therapeutic target.
Comparison of the diagnostic potential of circulating miRNAs versus neuroimaging techniques
Neuroimaging remains a cornerstone in the diagnosis of acute ischemic stroke, enabling rapid assessment of infarct size, vascular occlusion, and tissue viability. Advanced techniques such as diffusion-weighted imaging and perfusion-weighted imaging provide critical information for therapeutic decision-making within narrow time windows.[34] Circulating miRNAs have emerged as promising minimally invasive biomarkers for the early diagnosis of acute ischemic stroke, offering the potential for rapid detection through blood-based assays. Unlike neuroimaging techniques such as MRI, which require specialized equipment and may be limited by contraindications or delayed availability in emergency settings, circulating miRNAs can be measured quickly and repeatedly, including in pre-hospital or resource-limited environments. While MRI remains the gold standard for anatomical localization and confirmation of infarction – with high sensitivity and specificity, particularly in the acute phase – it may not detect very early or small ischemic lesions. In contrast, circulating miRNAs may reflect molecular changes that occur before structural abnormalities visible on imaging. Therefore, although circulating miRNAs cannot replace neuroimaging, they may serve as a valuable complementary tool, particularly for early triage and diagnosis when imaging is unavailable or inconclusive.
Diagnostic cost comparison
MRI remains the gold standard for anatomical and diagnostic assessment in acute ischemic stroke. However, direct cost– utility analysis in a Spanish hospital revealed that MRI yields an average direct cost of €5,693 per patient – comparable to CT scan (mean cost €5,831) – with only marginal quality-adjusted life-year (QALY) improvement, resulting in an incremental cost-effectiveness ratio of €11,869/QALY.[35] By contrast, assays for circulating miRNAs are substantially lower-cost: Reverse transcription quantitative polymerase chain reaction profiling through commercial platforms incurs approximately $0.53–$0.82 per miRNA per sample, with individual panels costing $250–400.[36] Moreover, such assays can be performed in standard molecular laboratories without the need for specialized imaging infrastructure, offering faster turnaround times and wider accessibility – particularly in settings where MRI availability is limited. These data support the cost-effective and practical potential of circulating miRNA assessment as a complement to, or triage tool for, MRI in stroke care. The development and implementation of strategies aimed at removing barriers to accessibility and improving the quality of services are crucial for ensuring the health and well-being of the population.[37] Despite their promise as diagnostic and prognostic biomarkers for ischemic stroke, the clinical utility of miRNAs requires further investigation.
Thus, we reviewed data from nine independent studies. This study is subject to several limitations. The included studies varied in sample sizes, which may affect the comparability of findings. In many cases, the number of patients analyzed was relatively small, limiting the statistical power and generalizability of the results.
CONCLUSION
This systematic review describes 16 circulating miRNAs that are expressed differently in plasma in patients with ischemic stroke. miRNAs play an important role in processes occurring after injury, such as pathological changes in neurons, restoration of the penumbra zone, and subsequent regeneration of nervous tissue.
miRNAs may have good diagnostic potential. However, studies involving larger samples with the most similar miRNA expression assessment system are needed to determine the clinical value of miRNAs as biomarkers of ischemic stroke. Further study and deeper understanding of the mechanisms by which miRNAs are involved in the pathogenesis of neurological disorders may provide new targets for further innovative diagnostic and therapeutic strategies.
Ethical approval:
The research/study was approved by the Institutional Review Board at Karaganda Medical University, number 20, dated December 03, 2024.
Declaration of patient consent:
Patient’s consent was not required as there are no patients in this study.
Conflicts of interest:
There are no conflicts of interest.
Use of artificial intelligence (AI)-assisted technology for manuscript preparation:
The authors confirm that there was no use of artificial intelligence (AI)-assisted technology for assisting in the writing or editing of the manuscript and no images were manipulated using AI.
Financial support and sponsorship: This work was supported by the Ministry of Science and Higher Education of the Republic of Kazakhstan (individual registration number AP23490807, Protocol version Contract № 331/GF 24-26 January 10, 2024 year); the Karaganda Medical University.
References
- Addressing disparities in the global epidemiology of stroke. Nat Rev Neurol. 2024;20:207-21.
- [CrossRef] [PubMed] [Google Scholar]
- Magnetic resonance imaging in acute ischemic stroke. Cureus. 2022;14:e27224.
- [CrossRef] [PubMed] [Google Scholar]
- Blood biomarkers for the differentiation of cardiac ischemic stroke subtypes: A systematic review. Cardiovasc Hematol Disord Drug Targets. 2019;19:215-27.
- [CrossRef] [PubMed] [Google Scholar]
- Expression profile of MicroRNAs in young stroke patients. PLoS One. 2009;4:e7689.
- [CrossRef] [PubMed] [Google Scholar]
- miR-487b promotes human umbilical vein endothelial cell proliferation, migration, invasion and tube formation through regulating THBS1. Neurosci Lett. 2015;591:1-7.
- [CrossRef] [PubMed] [Google Scholar]
- The diagnostic value of circulating brain-specific MicroRNAs for ischemic stroke. Intern Med. 2016;55:1279-86.
- [CrossRef] [PubMed] [Google Scholar]
- RNA-seq identifies circulating miR-125a-5p, miR-125b-5p, and miR-143-3p as potential biomarkers for acute ischemic stroke. Circ Res. 2017;121:970-80.
- [CrossRef] [PubMed] [Google Scholar]
- Circulating pro-angiogenic and anti-angiogenic microRNA expressions in patients with acute ischemic stroke and their association with disease severity. Neurol Sci. 2017;38:2015-23.
- [CrossRef] [PubMed] [Google Scholar]
- Upregulation of microRNA-128 in the peripheral blood of acute ischemic stroke patients is correlated with stroke severity partially through inhibition of neuronal cell cycle reentry. Cell Transplant. 2019;28:839-50.
- [CrossRef] [PubMed] [Google Scholar]
- Circulating miR-30a, miR-126 and let-7b as biomarker for ischemic stroke in humans. BMC Neurol. 2013;13:178.
- [CrossRef] [PubMed] [Google Scholar]
- Plasma level of miR-99b may serve as potential diagnostic and short-term prognostic markers in patients with acute cerebral infarction. J Clin Lab Anal. 2020;34:e23093.
- [CrossRef] [PubMed] [Google Scholar]
- Identification of miRNA-21 and miRNA-24 in plasma as potential early stage markers of acute cerebral infarction. Mol Med Rep. 2014;10:971-6.
- [CrossRef] [PubMed] [Google Scholar]
- Decreased plasma miR-335 expression in patients with acute ischemic stroke and its association with calmodulin expression. J Int Med Res. 2016;44:1331-8.
- [CrossRef] [PubMed] [Google Scholar]
- Biological significance of microRNA biomarkers in ALS-innocent bystanders or disease culprits? Front Neurol. 2019;10:578.
- [CrossRef] [PubMed] [Google Scholar]
- The 14q32 microRNA-487b targets the antiapoptotic insulin receptor substrate 1 in hypertension-induced remodeling of the aorta. Ann Surg. 2013;258:743-51. discussion 752-3
- [CrossRef] [PubMed] [Google Scholar]
- miR-7 and miR-153 protect neurons against MPP(+)-induced cell death via upregulation of mTOR pathway. Front Cell Neurosci. 2014;8:182.
- [CrossRef] [PubMed] [Google Scholar]
- MicroRNAs in vascular and metabolic disease. Circ Res. 2012;110:508-22.
- [CrossRef] [PubMed] [Google Scholar]
- MicroRNA-125b mimic inhibits ischemia reperfusion-induced neuroinflammation and aberrant p53 apoptotic signalling activation through targeting TP53INP1. Brain Behav Immun. 2018;74:154-65.
- [CrossRef] [PubMed] [Google Scholar]
- Argonaute HITS-CLIP decodes microRNA-mRNA interaction maps. Nature. 2009;460:479-86.
- [CrossRef] [PubMed] [Google Scholar]
- MiR-125a-5p silencing inhibits cerebral ischemia-induced injury through targeting IGFBP3. Folia Neuropathol. 2021;59:121-30.
- [CrossRef] [PubMed] [Google Scholar]
- Acute loss of miR-221 and miR-222 in the atherosclerotic plaque shoulder accompanies plaque rupture. Stroke. 2015;46:3285-7.
- [CrossRef] [PubMed] [Google Scholar]
- A necessary role of miR-221 and miR-222 in vascular smooth muscle cell proliferation and neointimal hyperplasia. Circ Res. 2009;104:476-87.
- [CrossRef] [PubMed] [Google Scholar]
- LncRNA SNHG12 regulates ox-LDL-induced endothelial cell injury by the miR-218-5p/IGF2 axis in atherosclerosis. Cell Cycle. 2021;20:1561-77.
- [CrossRef] [PubMed] [Google Scholar]
- miR-185 silencing promotes the progression of atherosclerosis via targeting stromal interaction molecule 1. Cell Cycle. 2019;18:682-95.
- [CrossRef] [PubMed] [Google Scholar]
- MicroRNA-128-3p protects mouse against cerebral ischemia through reducing p38a mitogen-activated protein kinase activity. J Mol Neurosci. 2017;61:152-8.
- [CrossRef] [PubMed] [Google Scholar]
- The endothelial-specific microRNA miR-126 governs vascular integrity and angiogenesis. Dev Cell. 2008;15:261-71.
- [CrossRef] [PubMed] [Google Scholar]
- MiR-130a exerts neuroprotective effects against ischemic stroke through PTEN/PI3K/AKT pathway. Biomed Pharmacother. 2019;117:109117.
- [CrossRef] [PubMed] [Google Scholar]
- SMAD proteins control DROSHA-mediated microRNA maturation. Nature. 2008;454:56-61.
- [CrossRef] [PubMed] [Google Scholar]
- miR-24 protects against ischemia-induced brain damage in rats via regulating microglia polarization by targeting Clcn3. Neurosci Lett. 2021;759:135998.
- [CrossRef] [PubMed] [Google Scholar]
- miR335 promotes stress granule formation to inhibit apoptosis by targeting ROCK2 in acute ischemic stroke. Int J Mol Med. 2019;43:1452-66.
- [CrossRef] [Google Scholar]
- MicroRNA-378 alleviates cerebral ischemic injury by negatively regulating apoptosis executioner caspase-3. Int J Mol Sci. 2016;17:1427.
- [CrossRef] [PubMed] [Google Scholar]
- Neuroimaging of acute ischemic stroke: Multimodal imaging approach for acute endovascular therapy. J Stroke. 2023;25:55-71.
- [CrossRef] [PubMed] [Google Scholar]
- Cost-utility analysis of magnetic resonance imaging management of patients with acute ischemic stroke in a Spanish hospital. Neurol Ther. 2015;4:25-37.
- [CrossRef] [PubMed] [Google Scholar]
- Technology in microRNA profiling: Circulating microRNAs as noninvasive cancer biomarkers in breast cancer. J Lab Autom. 2015;20:574-88.
- [CrossRef] [PubMed] [Google Scholar]
- Approaches to improving the effectiveness of the quality of medical services. Med Ecol. 2025;1:153-63.
- [CrossRef] [Google Scholar]

