Next Article in Journal
Hepatoprotective and Antioxidant Effects of Nanopiperine against Cypermethrin via Mitigation of Oxidative Stress, Inflammations and Gene Expression Using qRT-PCR
Next Article in Special Issue
Patients with Chronic Spinal Cord Injury Display a Progressive Alteration over the Years of the Activation Stages of the T Lymphocyte Compartment
Previous Article in Journal
The Effect of Temperature on the Embryo Development of Cephalopod Sepiella japonica Suggests Crosstalk between Autophagy and Apoptosis
Previous Article in Special Issue
Histamine H4 Receptor Antagonist Ameliorates the Progression of Experimental Autoimmune Encephalomyelitis via Regulation of T-Cell Imbalance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

MicroRNAs as Potential Biomarkers of Post-Traumatic Epileptogenesis: A Systematic Review

by
Anastasia A. Vasilieva
,
Elena E. Timechko
,
Kristina D. Lysova
,
Anastasia I. Paramonova
,
Alexey M. Yakimov
,
Elena A. Kantimirova
and
Diana V. Dmitrenko
*
Department of Medical Genetics and Clinical Neurophysiology of Postgraduate Education, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(20), 15366; https://doi.org/10.3390/ijms242015366
Submission received: 27 September 2023 / Revised: 16 October 2023 / Accepted: 17 October 2023 / Published: 19 October 2023

Abstract

:
Structural or post-traumatic epilepsy often develops after brain tissue damage caused by traumatic brain injury, stroke, infectious diseases of the brain, etc. Most often, between the initiating event and epilepsy, there is a period without seizures—a latent period. At this time, the process of restructuring of neural networks begins, leading to the formation of epileptiform activity, called epileptogenesis. The prediction of the development of the epileptogenic process is currently an urgent and difficult task. MicroRNAs are inexpensive and minimally invasive biomarkers of biological and pathological processes. The aim of this study is to evaluate the predictive ability of microRNAs to detect the risk of epileptogenesis. In this study, we conducted a systematic search on the MDPI, PubMed, ScienceDirect, and Web of Science platforms. We analyzed publications that studied the aberrant expression of circulating microRNAs in epilepsy, traumatic brain injury, and ischemic stroke in order to search for microRNAs—potential biomarkers for predicting epileptogenesis. Thus, 31 manuscripts examining biomarkers of epilepsy, 19 manuscripts examining biomarkers of traumatic brain injury, and 48 manuscripts examining biomarkers of ischemic stroke based on circulating miRNAs were analyzed. Three miRNAs were studied: miR-21, miR-181a, and miR-155. The findings showed that miR-21 and miR-155 are associated with cell proliferation and apoptosis, and miR-181a is associated with protein modifications. These miRNAs are not strictly specific, but they are involved in processes that may be indirectly associated with epileptogenesis. Also, these microRNAs may be of interest when they are studied in a cohort with each other and with other microRNAs. To further study the microRNA-based biomarkers of epileptogenesis, many factors must be taken into account: the time of sampling, the type of biological fluid, and other nuances. Currently, there is a need for more in-depth and prolonged studies of epileptogenesis.

1. Introduction

Epilepsy is a neurological disease characterized by a predisposition to the occurrence of epileptic seizures, as well as neurobiological changes and cognitive, psychological, and social impairments [1]. Epilepsy is one of the most common diseases of the nervous system. In the world, according to the WHO, there are about 75 million people diagnosed with epilepsy [2].
Structural or post-traumatic epilepsy (PTE) develops as a result of damage to brain tissue caused by traumatic brain injury (TBI), stroke, infectious diseases of the brain, etc. The frequency of brain damage in people of working age is significant, so the problem of predicting the development of post-traumatic epilepsy is an urgent task for researchers [3,4].
The types of post-traumatic seizures are classified as follows:
(1)
Immediate—arising in the first 24 h after a traumatic event;
(2)
Early—from 24 h to 1 week after the traumatic event;
(3)
Late—occurring more than 1 week after the traumatic event [3].
At the same time, immediate and early seizures are not considered truly “epileptic”, since it is assumed that they are provoked by an initiating event, and are not a consequence of the molecular, biochemical, and histological rearrangements of brain tissue [4]. The latent period of post-traumatic epileptogenesis is characterized by the absence of epileptic seizures from the moment of injury to the first late seizure. This period can last up to 10 years or more [5].
Epileptogenesis is a pathological process of the reorganization of tissues and cells in the brain, leading to the formation of hyperexcitable neural networks and the subsequent appearance of epileptiform activity. It was previously believed that epileptogenesis occurs only in the latent period—between the epileptogenic event and the onset of the first clinical seizure. Currently, according to studies [6,7,8], epileptogenesis is a longer process and includes the mechanisms of disease progression, since the frequency and severity of seizures increase with time. Pathological mechanisms leading to the development of epileptogenesis and subsequent chronicization of structural epilepsy are considered to be neuroinflammation of the brain (production of specific cytokines and chemokines that activate glia, which can lead to the recruitment of macrophages and lymphocytes to a dysfunctional BBB, leading to chronic inflammation and neurodegeneration), dysfunction of the blood–brain barrier, oxidative stress (proliferation of reactive oxygen species capable of organic damage to neuronal cells), apoptosis of neurons, activation of astrocytes, dendritic growth, gliosis (increased number, altered morphology with hypertrophy of the soma and processes, spatial overlap and various functional changes in astrocytes, leading to reorganization of the neuronal network and the formation of hyperexcitable areas), etc. [4,5].
The problem of assessing the risks of developing post-traumatic epilepsy after traumatic brain injury and stroke in the early stages and during the progression of the disease is one of the most urgent topics in the study of epilepsy. Currently, it is impossible to predict the development of the epileptogenic process after the initiating event (TBI, stroke, etc.); it is only possible to diagnose if the disease already exists [4].
The standard methods for diagnosing epilepsy today are electroencephalography (EEG) and neuroimaging methods such as computed tomography (CT) and magnetic resonance imaging (MRI) [9].
The use of non-invasive and minimally invasive biomarkers for diagnosing and predicting the development of epilepsy can serve as an alternative to classical diagnostic methods. A biomarker is an indicator of biological and physiological processes occurring in the body, as well as pathogenic processes or reactions to various influences, including therapeutic intervention [10]. According to the BEST (Biomarkers, EndpointS, and other Tools) Resource, there are the following types of biomarkers: susceptibility (risk) biomarkers; diagnostic biomarkers; biomarkers for disease monitoring; prognostic biomarkers; response biomarkers; and biomarkers of safety/toxicity [10].
A potential prognostic biomarker of epileptogenesis is microRNAs (miRNAs), endogenous non-coding RNA molecules that can bind to one or more mRNAs, mainly inducing their degradation and, consequently, repression of translation [11]. MicroRNA processing and the mechanism of action are summarized in Figure 1.
The advantages of using microRNAs are the minimally invasive methods for determination, relative stability in biological fluids, and the simplicity and low cost of the methods used [12].
This review analyzed manuscripts related to biomarkers for epilepsy, TBI, and ischemic stroke. The purpose of the review was to analyze the available information and search for miRNAs that have potential as biomarkers and predictors of epileptogenesis.

2. Methods

When writing the systematic review, the following aggregators of scientific journals were used: MDPI, PubMed, ScienceDirect, and Web of Science. Scientific articles were searched using the following keywords: microRNA, biomarker, epilepsy, traumatic brain injury, ischemic stroke.
Scientific articles from 2010 to 2023 were analyzed, including original research and review articles. Publications were included in the systematic review if they were published in English and complied with the inclusion criteria defined below. Duplicates were excluded from the review. Research publications containing information on aberrant miRNA expression in plasma/serum/saliva/cerebrospinal fluid in epilepsy, TBI, and ischemic stroke in humans were included.
Only studies with statistically significant aberrant miRNA expression (according to AUC criteria and/or p-value, depending on the criteria used in the original research) were analyzed. The review excluded manuscripts that investigated miRNA expression in animal models and in brain tissues.
This review was produced in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA 2020). The block diagram is shown in Figure 2.

2.1. Inclusion Criteria

Study Material

The material included studies of miRNA expression in patients with epilepsy, traumatic brain injury, and ischemic stroke. No limitations were placed on age, ethnicity, country of origin, family history of recurrent miscarriage, or any other patient demographics. Only original studies were included in the review.
The material on patients’ investigation samples included cerebrospinal fluid, plasma, and blood.

2.2. Exclusion Criteria

The studies of patients with epilepsy were limited by type of epilepsy. Genetic forms of epilepsy were excluded. The study of brain tissue was also excluded from the analysis. Animal model studies were also excluded from the review. Reviews were also excluded.

3. Results and Discussion

The search for biomarkers of post-traumatic epilepsy is difficult. Epileptogenesis is a long, non-specific, and dynamic process. It may be necessary to investigate different panels of biomarkers depending on the stage and etiology of the initiating event. Another problem is the search for those biomarkers that are specific for epileptogenesis, and not the severity of brain damage.
When searching for biomarkers of epileptogenesis, a number of factors must be taken into account. MicroRNA expression profiles may differ depending on the type of biological fluid taken for analysis. There is evidence [13] that cerebrospinal fluid is more informative than peripheral blood. However, the sampling procedure is more invasive, so CSF is less frequently studied. It is worth paying attention to the study of exosomal miRNAs, which is independent of the total content in the biofluid. Exosomal miRNAs can also be more informative [14]. Despite this, most studies are aimed specifically at the general study of the miRNAs profile in peripheral blood due to the greater availability of the method.
The timing of biosampling also plays an important role in miRNA analysis since miRNA expression in the acute phase of an epileptic seizure/trauma/stroke and miRNA expression, which indicates molecular and metabolic changes caused by trauma, may differ. Only a few publications that have studied models of epilepsy examine the biomaterial immediately after a seizure [14,15]. Obviously, this is difficult in studying the model of epilepsy. In TBI and ischemic stroke, almost all studies indicate the time of biomaterial collection.

3.1. Circulating miRNAs in Epilepsy

The development of epilepsy occurs due to a series of pathological changes that form a repetitive cycle of excitation in the hippocampus, leading to the appearance of seizures. In patients with epilepsy, many molecular anomalies in various metabolic processes are observed in the brain [16]. It has been proven that they are associated with the impaired transcriptional and post-transcriptional regulation of genes in epileptogenesis [17].
Thirty-one manuscripts were analyzed that examined circulating miRNAs for which aberrant expression was observed in patients with epilepsy (Table 1). miRNAs, as biomarkers of epilepsy, have been extensively studied in various publications, but the association with post-traumatic epilepsy is not often made. Chun-Hong Shen et al., in their study [18], determined that miR-145-5p was reduced in patients with drug-resistant epilepsy compared to healthy volunteers. The decreased expression of these miRNAs correlated with a history of TBI or encephalitis in patients. However, in two other studies, miR-145 was overexpressed in patients with mesial temporal lobe epilepsy immediately after a seizure [15] and in patients with established hippocampal sclerosis [19].
Most often, blood plasma or serum was studied, but cerebrospinal fluid was studied in some publications [20,21]. In some cases, miRNA expression was compared in various biofluids. Ye Zhao et al. found a significant increase in miR-1275 expression in the serum of patients with epilepsy of unknown etiology, but it was reduced in the CSF [21]. Similarly, Rana Raoof et al. did not find a statistically significant correlation between miR-19b-3p, miR-21-5p, or miR-451a expression in CSF compared with plasma [22]. Therefore, it should be assumed that the information content of studies of different types of biological fluids will vary.
It is worth paying attention to studies of epilepsy in pediatric patients. It is generally accepted that epileptogenic mechanisms in children differ from those in adults [23]. However, data on the microRNA–age correlation are insufficient. Some miRNAs show similar expression patterns in both children and adults and may not be related to age. Examples include miR-106b [24,25,26] and miR-155 [27,28,29].
Although tissue expression studies were excluded from the review, some authors performed a comparative analysis between tissue and circulating miRNAs. One study found that the miR-145, miR-181c, miR199a, and miR-1183 miRNAs circulating in biological fluids were overexpressed in patients with mesial temporal lobe epilepsy, but hypoexpressed in hippocampal tissues [19]. In contrast, Yuqiang Sun et al. did not find a difference in miR-129-2-3p expression levels in plasma and cerebral cortex tissues in patients with drug-resistant epilepsy—in both cases, it was hyperexpressed.
Table 1. Circulating miRNAs in epilepsy.
Table 1. Circulating miRNAs in epilepsy.
Experimental DesignSampleSampling TimeMiRNAsSignificanceYearLink
AUCp-Value
TLE/HVCSF miR-219↓ <0.0012014[20]
DR E/HVSerum miR-194-5p↑
miR-301a-3p↑
miR-30b-5p↑
miR-342-5p↑
miR-4446-3p↑
0.740
0.893
0.684
0.721
0.703
2015[30]
DR E/HVSerum miR-4521↑0.718 2016[31]
DR E/HVPlasma miR-153↓ 0.0012016[32]
DR E/HVPlasma, brain tissue miR-129-2-3p↑0.7780.0008 [33]
E/HVSerum miR-106b↑
miR-146a↑
miR-301a↑
miR-194-5p↓
0.786
0.774
0.686
0.696
2016[24]
E/HVSerum miR-378↑
miR-30a↑ (associated with the frequency of seizures)
miR-106b↑
miR-15a↑
<0.0102016[25]
Seizure mTLE/mTLESerum30 min after seizuremiR-143↑
miR-145↑
miR-532↑
miR-365a↑
<0.0502016[15]
DR E/HVPlasma miR-323a-5p↑ 0.0142017[34]
mTLE+HS/HVPlasma miR-3613-5p↑
miR-4668-5p↓
miR-8071↓
miR-197-5p↓
miR-4322↓
miR-6781-5p↓
0.844
0.789
0.932
0.802
0.714
0.781
2017[35]
DR-mTLE/mTLE/HVPlasma miR-134↓0.671 2017[36]
TLE/SE/HVCSF TLE/SE:
miR-19b-3p↑
miR-21-5p↑
miR-204-5p↑
miR-223-3p↑
miR-451a↑
miR-886- 3p↓
TLE/HV:
miR-19b-3p↑
miR-21-5p↑
miR-204-5p↑
miR-223-3p↑
miR-451a↑
miR-886-3p↑

0.780
0.800


0.910


0.730
0.830


0.800
2017[22]
E/HVPlasma miR-153↓ <0.0102018[37]
E/HVSerum miR-155↑ <0.0102018[27]
E/E after seizure/HVPlasma24 hTLE/TLE after seizure:
miR-27a-3p↓
miR-328-3p↑
miR-335-5p↓
TLE/HV:
miR-27a-3p↓
miR-328-3p↑
miR-335-5p↓


<0.010
<0.050
<0.050

<0.010
<0.010
<0.010
2018[14]
DR E/mTLE/HV Plasma DR E/HV:
miR-145-5p↓
mTLE/HV:
miR-145-5p↓

0.632

0.829
2019[18]
mTLE+HS/HVSerum miR-145↑
miR-181c↑
miR199a↑
miR-1183↑
0.005
0.030
0.010
0.001
2019[19]
DR E/ESerumSeizures during 12 months/Without seizures during 12 monthsmiR-146a-5p↑
miR-134-5p↑
0.640
0.617
<0.001

2020[38]
ChE/HVPlasma miR-106b↑
miR-146a↑
0.885
0.763
2019[26]
GGE/HVSerum miR-146a↑
miR-155↑
miR-132↓
Average: 0.850 2020[28]
IE/HVPlasma miR-125a↓
miR-181a↓
0.815
0.704
0.001
0.001
2020[39]
mTLE+HS(Engel I, Engel III-IV)/HVSerum E I/HV:
miR-328-3p↑
miR-654-3p↑
E III-IV /HV:
miR-328-3p↑
miR-654-3p↑
E I/E III-IV:
miR-328-3p↑
miR-654-3p↑

0.001
0.004

<0.001
0.89


<0.001
0.190
[40]
UEE/HVSerum, CSF Serum:
miR-1275↑
CSF:
miR-1275↓
2020[21]
TLE/HVSerum miR-142↑
miR-146a↑
miR-223↑
<0.001
0.020
<0.001
2021[41]
Ch TLE/HVPlasma miR-194-5p↓0.896 2021[42]
TLE/HVPlasma miR-146a↑
miR-132↑
0.808
0.791
2022[43]
ChE/HVPlasma (sEVs) miR-584-5p↑
miR-342-5p↑
miR-150-5p↑
0.846
0.835
0.826
2022[44]
ChE/HVSerum miR-155 0.813 2022[29]
ChE/HVSerum miR-324-5p↑
miR-146a-5p↓
miR-138-5p↓
miR-187-3p↓
2022[45]
E/HVSerum miR-378↓
miR-575↓
<0.001
<0.001
2022[46]
mTLE+HS/HVSerum miR-629-3p↑
miR-1202↑
miR-1225-5p↑
2022[47]
TLE—temporal lobe epilepsy; HV—healthy volunteers; CSF—cerebrospinal fluid; DR—drug-resistant epilepsy; mTLE—mesial temporal lobe epilepsy; HS—hippocampal sclerosis; SE—status epilepticus; E—epilepsy; GGE—genetic generalized epilepsy; ChE—childhood epilepsy; UEE—unknown etiology epilepsy; IE—idiopathic epilepsy; Engel I—good surgical outcome; Engel III—bad surgical outcome; ↑—upregulation; ↓—downregulation; AUC—area under the curve.

3.2. Circulating miRNAs inTraumatic Brain Injury

The risk of developing post-traumatic epilepsy after TBI is quite high. In a study conducted by Meral A Tubi et al., 45.7% of a cohort of patients with TBI of varying severity acquired PTE within two years of injury [48].
The progression of injury towards epileptogenesis occurs in a state of secondary injury—a few hours or days after it. At this time, a number of complex molecular and structural transformations occur, leading to the development of epilepsy in certain circumstances [3]. Structural changes include neurodegeneration [49], gliosis [50], damage or sprouting of axons [51], dendritic plasticity [52], damage to the blood–brain barrier [53], and recruitment of inflammatory cells in brain tissue [54].
To diagnose and predict the outcome of TBI, the Glasgow scale is used, based on eye-tracking and the verbal and motor activity of the patient. Together with the presence and amount of loss of consciousness, the severity of the injury is determined [55]. This method, in combination with classical CT and MRI, does not always make it possible to identify the severity and focus of the injury [4]. It is also difficult to find the prerequisites for the development of epileptogenesis [4].
Table 2 summarizes the aberrantly expressed miRNAs in TBI patients. Studies of circulating microRNAs as biomarkers of TBI often aim to diagnose TBI after a head injury, as well as to determine the severity. For example, Manish Bhomia et al. found the aberrant expression of 18 microRNAs in moderate TBI and 20 in severe TBI. Subsequently, verification analyses of 10 miRNAs were carried out in the cerebrospinal fluid of patients with various TBIs. A correlation was also made between CT-positive and CT-negative patients. It was found that miR-328, miR-362, and miR-486 are potential biomarkers of severe versus moderate TBI. The expression of selected miRNAs differed in CT-negative patients—some of them were diagnosed with TBI, but the authors did not identify a separate cohort [56]. It is also worth paying attention to prolonged studies in which miRNA levels are measured over a long period of time [57,58,59,60].
MicroRNAs can be predictors of comorbidities even years after the initiating event. In one study, the cohort of patients included individuals with TBI-induced hypopituitarism [58]. In patients who developed hypopituitarism after TBI, there was an increase in miR-3610 by 5.0 times on the 1st day, 8.5 times on the 7th day, 2.3 times on the 28th day, and 10.0 times at year 5 compared to healthy controls. Another miRNA, miR126-3p, was reduced 11-fold on the 1st day and 12-fold on the 28th day, while at the 5th year after trauma, there was a 12-fold increase compared to the control group. In addition, it was noted that miR-126-3p showed a 15-fold increase at year 5 after trauma when patients with and without hypopituitarism after TBI were compared. The problem with such studies is that there are difficulties in the long-term monitoring of patients. Therefore, such studies involve patients with already-established diagnoses and a history of TBI of a certain prescription. Nevertheless, such a research model is optimal in the study of PTE.
Table 2. Circulating miRNAs in TBI.
Table 2. Circulating miRNAs in TBI.
Experimental DesignSampleSampling TimeMiRNAsAUCp-ValueYearLink
sTBI/HVPlasma24 h after traumamiR-16↓
miR-92a↓
miR-765↑
0.890
0.820
0.860
2010[61]
sTBI/OTmiR-16↓
miR-92a↓
miR-765↑
0.820
0.830
0.790
sTBI/HVmiR-16↓
miR-26a↑
miR-92a↓
miR-638↑
miR-765↑
0.820
0.730
0.780
0.510
0.680
TBI of varying severity/HVSerum24 h after trauma–21 days after traumamiR-93↑
miR-191↑
miR-499↑
1.000
0.727
0.801
2016[57]
sTBI (coma)/HVCSFComa during two weeks after traumamiR-141↑
miR-572↑
miR-181a-star↑
miR-27b-star↑
miR-483-5p↑
miR-30b↑
miR-431-star↑
miR-193b-star↑
miR-499-3p↑
miR-1297↓
miR-33b↓
miR-933↓
miR-449b↓
0.005
0.002
0.001
0.012
0.003
0.030
0.006
0.021
0.002
0.013
0.016
0.009
0.042
2016[62]
TBI/HVSerum24 h after traumamiR-155↓0.726 2019[63]
TBI of varying severit/OT/HVSerum24–48 h after traumamiR-151-5p↑
miR-195↑
miR-20a↑
miR-328↑
miR362-3p↑
miR-30d↑
miR-451↑
miR-486↑
miR-505*↑
miR-92a↑
0.660
0.810
0.780
0.730
0.790
0.750
0.820
0.810
0.820
0.860
2016[56]
TBI+ hypopituitarism /HVSerum1, 7, 28 days after traumamiR-3907↑
miR126-3p↓
< 0.0502016[58]
лЧМT/ЗДSerum24 h–15 days after traumamiR-425-5p↓
miR-21↑
miR-502↓
miR-335↑
1.000
0.961
1.000
0.990
2017[59]
TBI of varying severity/HVSaliva, CSF miR-182-5p↓
miR-221-3p↓
mir-26b-5p↓
miR-320c↓
miR-29c-3p↑
miR-30e-5p↑
Average
0.852
2018[64]
TBI of varying severity/HVPlasma miR-6867-5p ↑
miR-3665↑
miR-328-5p↑
miR-762↑
miR-3195↑
miR-4669↑
miR-2861↑
0.854
0.877
0.888
0.916
0.899
0.907
0.913
2018[65]
mTBI/HVSaliva, Serum miR-10b-5p↑
miR-30b-5p↑
miR-3678-3p↓
miR-455-5p↓
miR-5694↓
miR-6809-3p↓
miR-92a-3p↓
Average
0.890
2019[66]
sTBI/HVSerum<24 h after traumamiR-103a-3p↑
miR-219a-5p↑
miR-302d-3p↑
miR-422a↑
miR-518f-3p↑
miR-520d-3p↑
miR-627↑
<0.0502019[67]
mTBI/HVSerum24-48 h after trauma miR-151-5p↑
miR-362-3p↑
miR-486↑
miR-505↑
miR-499↑
miR-625↑
miR-638↑
miR-381↑
<0.0502020[68]
TBI/HVSerum<24 h after traumamiR-124-3p↑
miR-219a-5p↑
miR-9-5p↑
miR-9-3p↑
miR-137↑
miR-128-3p↑
0.710
0.890
0.890
0.780
0.810
0.740
2020[69]
Isolated TBI/HVPlasma/Serum6 h after traumamiR-423-3p↑0.790 2020[70]
TBI/HVSerum<24 h after traumamiR-93↑
miR-191↑
0.712
0.660
2020[71]
TBI/HVPlasma<24 h after traumamiR-203b-5p↓
miR-203a-3p↓
miR-206↑
miR-185-5p↑
0.840 2020[72]
modTBI/HVPlasmaless than 1 day, 7, 28 days after traumamiR-32-5p↑0.700–0.900 2022[60]
sTBI—severe traumatic brain injury; HV—healthy volunteers; CSF—cerebrospinal fluid; OT—orthopedic injury; mTBI—mild traumatic brain injury; modTBI—moderate craniocerebral injury; ↑—upregulation; ↓—downregulation; AUC—area under the curve.

3.3. Circulating miRNAs in Ischemic Stroke

Ischemic stroke, along with TBI, is one of the main causes of post-traumatic epilepsy [73]. The incidence of PTE ranges from 3% to 25% depending on various factors [74]. Neurological deficit, disability, and damage to the cerebral cortex and hippocampus are the main factors in the development of post-stroke seizures [75]. Seizures with early onset do not cause epilepsy, but are considered a risk factor. However, late seizures occurring more than a week later pose a greater risk of PTE. This is due to structural changes in the neural networks of the brain, such as dysfunction of the blood–brain barrier. Late attack causes multiple mechanisms—gliotic scarring with changes in membrane properties, albumin extravasation, changes in astrocytic proteins, and brain irritation due to hemosiderin deposits, as well as thrombin stimulation of the protease-activated receptor [76]. The pathogenesis of PTE in terms of molecular markers is associated with elevated plasma interleukin-6 (IL-6), high mobility group B1 protein, and low-serum neuropeptide Y [76]. The dysregulation of miRNA expression can also be a factor regulating the processes described above. It was previously reported [77] that miRNAs are involved in the processes of oxidative stress, inflammation, and apoptosis; in this case, either protective or pathogenic functions are manifested [77].
The publications that have studied miRNA expression in ischemic stroke are listed in Table 3. The clinical use of miRNAs as biomarkers of ischemic stroke is most often in diagnostics—in determining differences between controls and a group of patients. However, other goals may be pursued, such as establishing the etiology of a stroke. The publication of G. Long et al. [78] included patients with ischemic stroke of various etiologies: damage to small vessels, cardioembolism, and atherosclerosis of large vessels. It was found that the levels of let-7b miRNA were different in patients with atherosclerosis of large vessels and were reduced compared to the control. In other patients, the levels of this miRNA were significantly higher than in controls.
One of the goals in the study of miRNAs in ischemia is to establish a correlation with possible deterioration in the patient’s condition. The authors of one of the studies showed that miR-132 miRNA levels can be statistically correlated depending on the presence of post-stroke cognitive impairment in patients [79]. Hyperexpressed levels of miR-124-3p positively correlated with mortality within 3 months in patients with ischemic stroke [80].
Table 3. Circulating miRNAs in ischemic stroke.
Table 3. Circulating miRNAs in ischemic stroke.
Experimental DesignSampleSampling TimeMiRNAsAUCp-ValueYearLink
IS/HVSerum3, 4, 7 days after attackmiR-210↓ 0.0012011[81]
IS/A/HVSerum miR-21↑
miR-221↓
<0.0001
0.0002
2013[82]
IS/HVSerum24 h, 1 week, 4 weeks, 24 weeks and 48 weeks after attack.24 h–24 weeks:
miR-30a↓
miR-126↓
let-7b↑
(but let-7b ↓ for atherosclerosis of large vessels)

0.910–0.930
0.920–0.940
0.920–0.930
2013[78]
IS/HVSerum miR-145↑ 2014[83]
IS/HVSerum72 h after attackmiR-223↑ <0.0502014[84]
IS/HV with vascular risk factorsSerum>2 weeks after attackmiR-122↓
miR-148a↓
let-7i↓
miR-19a↓
miR-320d↓
miR-4429↓
miR-363↑
miR-487b↑
0.047
0.009
0.023
0.030
0.020
0.034
0.037
0.044
2014[85]
ACI/HVPlasma miR-21↑
miR-24↓
<0.0502014[86]
AS/HVPlasma0–24 h after attackhsa-miR106b-5p↑
hsa-miR-4306↑
hsa-miR-320e↓
hsa-miR320d↓
0.962
0.877
0.981
0.987
2014[87]
IS/HSPlasma miR-124-3p↓
miR-16↑
0.010
0.039
2014[88]
AcuteIS/HVSerum24 h after attackmiR-124↓
miR-9↓
0.011
<0.001
2015[89]
IS/HVSerum24 h after attackmiR-32-3p↑
miR-106-5p↑
miR-532-5p↓
<0.050
<0.010
<0.010
2015[90]
AS/IS/HVSerum24 h after attackmiR-145↑
miR-23a↓
miR-221↓
0.794
0.816
0.819
<0.001
<0.001
<0.001
2015[91]
IS/HVSerum miR-15a↑
miR-16↑
miR-17-5p↑
0.698
0.820
0.784
2015[92]
IS/HVSerum, CSF let-7e↑ 0.0332015[93]
IS/HVSerum miR-9,-22,-23,-27,-125,-524↑
miR-9,-30,-33,-124,-135, -181,-197,-218,-330,-452↓
<0.100


<0.150
2015[94]
IS/HVSerum miR-107↑
miR-128b↑
miR-153↑
0.970
0.903
0.893
2016[95]
PSCI/PSCN/HVSerum-miR-132↑0.961 2016[79]
Fatal post-stroke/survivorsPlasma<24 h after attackmiR-124-3p↑
miR-16↓
0.750
0.690
2016[80]
IS/HVSerum<72 h after attacklet-7i↓ <0.000012016[96]
IS/HV TIA/HVPlasma<24 h after attackmiR-125a-5p↑
miR-125b-5p↑
miR-143-3p↑
IS/HV
1.1 × 10−7
8.2 × 10−9,
2.4 × 10−9
TIS/HV
0.003
0.003
0.005
2017[97]
IS/HVPlasma1–3 days after attack


4–14 days after attack
miR-422a↑
miR-125b-2-3p↓

miR-422a↓
miR-125b-2-3p↓
0.769


0.971
0.889
2017[98]
IS/HVSerum<24 h after attackmiR-221-3p↓
miR-382-5p↓
0.8106
0.7483
2017[99]
IS/TIA/HVSerum<24 h after attackIS:
miR-23b-3p↑
miR-29b-3p↑
miR-181a-5p↑
miR-21-5p↑
TIS:
miR-23b-3p↑
miR-29b-3p↑
miR-181a-5p↑

<0.001
<0.001
0.027
<0.001

<0.001
0.001
0.007
2017[100]
IS/HVSerum1–3 days after attackmiR-146a↓0.910 2017[101]
IS/HVCSF<48 h after attackmiR-9-5p↑
miR-128-3p↑
<0.0502017[102]
IS/HVSerum<48 h after attackmiR-4656,-432, -503↑
miR-874↓
<0.0502018[103]
IS/HVSerum<24 h after attackmiR-146b↑ 0.8632018[104]
IS/HVSerum<72 h after attackmiR-15a↑ <0.0502018[105]
IS/HVSerum<24 h after attackmiR-148b-3p↓
miR-151b↑
miR-27b-3p↑
0.665
0.685
0.666
2018[106]
IS/HVSerum<24 h after attackmiR-186-5p↓
miR-19a-3p↓
miR-32-5p↓
miR-340-5p↓
miR-579-3p↓
let-7e↑
miR-362-3p↑
miR-1238-5p↓
<0.0502019[107]
Acute IS/HVSerum<24 h after attackmiR-124↓ <0.0012019[108]
IS/HVSerum<24 h after attackmiR-146a↓ <0.0012019[109]
IS/HVSerum miR-451↑0.912 2019[110]
Acute IS/HVSerum<24 h after attackmiR-124↑ 0.0072020[111]
IS/HVSerum miR-24↓
miR-29b↓
0.802
0.835
2020[112]
IS/HVSerum<24 h
14 days after attack
miR-135b↑
(24 h after attack)
miR-135b↑
(14 days after attack)
0.780 2020[113]
IS/HVSerum<24 h after attackmiR-602↓0.817 [114]
IS/HVPlasma miR-155↑0.851 2020[115]
IS/HVSerum miR-128↑ <0.0012020[116]
IS/HVSerum<72 h after attackmiR-124↓0.953 2021[117]
IS/HVSerum<72 h after attackmiR-148a↑
miR-342-3p↑
miR-19a↑
miR-320d↑
0.872
0.844
0.721
0.673
2021[118]
Acute IS/HVSerum miR-9-5p↑
miR-128-3p↑
0.946
0.929
2021[119]
IS/HVSerum miR-659-5p↑
miR-151a-3p↑
miR-29c-5p↑
0.003
0.0007
0.0007
2022[120]
IS/A/HVPlasmaIS <6 h after attackmiR-129-1-3p↑
miR-4312↑
miR-5196-3p(IS↑, A↓)
<0.0012022[121]
IS/HVSerum<24 h after attackmiR-451a↑
miR-574-5p↓
miR-142-3p↓
0.970 2022[122]
IS/HVSerum miR-15b-5p↑
miR-184↑
miR-16-5p↑
0.581
0.607
0.719
2022[123]
IS/HVSerum miR-6089↓ <0.0502023[124]
IS—ischemic stroke; HV—healthy volunteers; A—atherosclerosis; TIA—transient ischemic attack; CI—cerebral ischemia; ACI—acute cerebral infarction; AS—acute stroke; HS—hemorrhagic stroke; PSCI—post-stroke cognitive impairment; PSCN—post-stroke cognitive normality; ↑—upregulation; ↓—downregulation; AUC—area under the curve.

3.4. Discussions

According to the analyzed information, three microRNAs were found that exhibit aberrant expression in TBI, stroke, and epilepsy: miR-21 [22,59,82,86,100], miR-181a [39,62,100], and miR-155 [27,28,29,63,115]. A summary of the data is shown in Table 4 and also in Figure 3.

3.4.1. miR-21

miR-21 microRNA is a well-known and well-studied biomarker of many diseases [125], is involved in inflammatory [126] processes, and is associated with cancer [127] and cardiovascular [128] diseases. The targets of this miRNA and related processes were analyzed using the MiRTarBase database and ShinyGO v.0.77 and TissueAtlas software (https://ccb-web.cs.uni-saarland.de/tissueatlas/ (accessed on 27 September 2023)). Figure 4 shows the biological processes regulated by miR-21. These processes are not specific to epilepsy. However, the processes of cell development, proliferation, and apoptosis remain important for such neurological disorders. miR-21-3p is most actively expressed in the arachnoid mater (Figure 5), which may indicate an association with epileptogenesis. Previously, only miR-21-5p was studied in the model of epilepsy [22].
A study by R. Raoof et al. [22] used miR-21-5p, miR-451a, and miR-19b-3p expression in epilepsy as a test material for CSF samples from patients with temporal lobe epilepsy, status epilepticus, and the control group. Patients suffering from other neurological diseases (Alzheimer’s disease, multiple sclerosis) were also included in order to test the specificity of biomarkers. The miR-21-5p expression was significantly higher in patients with status epilepticus than in patients with temporal lobe epilepsy and controls [22]. Also, the values were overestimated in patients with other neurological diseases.
The miR-21 expression in patients with TBI [59] of various severity (moderate, severe), as well as in patients with extra-cranial injury without TBI and in healthy controls, was measured in blood serum at different time points (0–1 h, 12 h, 48–72 h, 15 days) from injury. It was found that the miR-21 expression level is strongly overestimated in patients with severe TBI in comparison with other groups of patients during the entire observation period. After 6 months, miR-21 showed no predictive value.
In a model of ischemic stroke, the level of miR-21 was compared in serum in patients with stroke, atherosclerosis, and in the control group [82]. Serum levels of miR-21 were elevated in patients with atherosclerosis along with IS. After adjusting for traditional risk factors, miR-21 levels remained a significant predictor of stroke [82]. In another study [86], patients with acute ischemic infarction served as an experimental group; the expression data are similar to previous studies.
As shown earlier, miR-21 is not specific. The studies listed above show that miR-21 is overexpressed in the brain lesions of various etiologies. However, this miRNA may be useful for epileptogenesis in terms of controlling the neurodegenerative regulation associated with inflammation and apoptosis.

3.4.2. miR-181a

miR-181a is a member of the miR-181 family, which consists of four microRNAs: miR181a, miR181b, miR181c, and miR181d. The impaired expression of these miRNAs is associated with the immune response, particularly with the differentiation of B- and T-lymphocytes [129]. miR-181a targets include heat shock protein GRP78 [130], transcription factor CREB1 [131], and the Bcl-2 family of proteins [132]. This microRNA is largely associated with protein modifications in the body (Figure 6), which may be of great interest for epileptogenesis. It was also found that miR-181a is predominantly expressed in the brain and spinal cord (Figure 7).
Publication [39] determined the relationship between miR-125a and miR-181a expression levels, IFN-γ and TNF-α production, and epileptogenic processes. The study involved children with an idiopathic form of epilepsy. The microRNA levels were significantly reduced in epileptic patients compared to healthy controls. However, the levels of pro-inflammatory cytokines in patients were slightly increased, and the authors indicated the absence of a correlation with miRNA expression. Based on this, it can be concluded that miR-181a is not associated with idiopathic epilepsy. It is likely that structural damage is necessary for its aberrant expression.
Another study [62] studied the expression of a spectrum of miRNAs, including miR-181a, in patients with severe TBI with coma. The sample for the study was CSF. The miR-181a expression level was 2.47 times higher in patients compared to healthy controls. In an IS model, miR-181a showed increased expression in a model of ischemic stroke and transient ischemic attack [100].
It is not known how specific miR-181a may be for epileptogenesis, but there are a number of prerequisites for this. This miRNA is of interest from the point of view of metabolic functions. Studies also confirm its importance, but they are too few for accurate judgments.

3.4.3. miR-155

miR-155 plays an important role in inflammatory processes, apoptosis, and cell proliferation [133,134]. An analysis of biological processes (Figure 8) showed that miR-155 is predominantly involved in cell migration and proliferation. The tissue expression of miR-155 is not specific; however, data are only available for miR-155-5p (Figure 9).
In epilepsy, the increased expression of miR-155 was observed in three independent studies [27,28,29]. Wei Duan et al. [27] found an overexpression of miR-155 in the blood serum of patients with temporal lobe epilepsy. It was concluded that miR-155 is associated with the PI3K/Akt/mTOR signaling pathway, inducing neuronal apoptosis. A study by R. Martins-Ferreira et al. [28] determined the expression patterns of the miR-146a, miR-155, and miR-132 microRNA panel in patients with genetic generalized epilepsy, all of which were overestimated. Exosomal miR-155 was also elevated in children with epilepsy in a study by Ya Liu et al. [29]. However, children with PTE were excluded from the cohort.
In patients with TBI, the level of miR-155 in the peripheral blood was reduced [63]. The authors associated the reduced expression of this microRNA with the formation of damage to the intestinal mucosa and further dysfunction by affecting the expression of the claudin1 protein.
In the model of acute and subacute ischemic stroke, the vesicular microRNA miR-155 was overestimated compared to the control by 1.6 and 2.5 times, respectively [115]. Higher levels were found in patients with mild to moderate stroke.
The common gene targets for these three microRNAs are BCL2, EGFR, and SOX6. According to the STRING database, the most significant pathways in which these genes are involved are intrinsic apoptotic signaling, EGFR signaling, and glial cell fate commitment with an indicated strength above 2.

3.4.4. Other Potential miRNA-Based Markers

The microRNAs associated with TBI/epilepsy as well as IS/epilepsy are listed below and are shown in Table 4.
The most important potential biomarkers are miRNAs, whose expression is associated with activity from ion channels. Among them are miR-30b [135], miR-219a [136], miR-335 [137], miR-342 [138], and miR-30a [25].
miR-328-3p is widely studied in relation to cancer [139]. Despite this, it has shown itself as a biomarker for epilepsy [14,40] and for determining the severity of TBI [56]. Also, miR-30a [140,141], miR-15a [142], and miR-125a [143] are associated with cancer.
miR-153 is associated with refractory epilepsy in terms of its effect on the hypoxia factor HIF-1α [37].
miR-106b, as an immune response factor [26], is an interesting target for epileptogenesis studies. It has been actively studied both in the model of epilepsy [24,25,26] and in ischemic stroke [87,90]. Similarly, miR-146a is one of the most frequently studied epilepsy and IS biomarkers, as shown in the respective tables. Despite this, this miRNA in ischemic stroke is dysregulated in the acute phase and goes up in the subacute phase [144]. This may help predict epileptogenesis after IS, but more long-term studies are needed.
The miRNA cluster 143-3p/145-3p is involved in vascular stabilization and smooth muscle contractility [145]. These miRNAs are more likely to serve as an indicator of the seizure state than of the metabolic processes associated with epileptogenesis.
miR-223 has been studied as a biomarker for epilepsy [22,41] and cerebral ischemia [84]. It is known that this miRNA affects the expression of GluR2 and NR2B and is neuroprotective [146].
miRNAs such as miR-451a are associated with the anti-apoptotic genes of BCL-2 and AKT1 proteins [147]. miR-451 also targets the drug resistance protein ABCB1/MDR1 [148], which links it to drug resistance.
miR-132 is of interest for studying the mechanisms of epilepsy. This miRNA is associated with the regulation of neuroplasticity [148,149].
Some of the above microRNAs may be of interest for the study of PTE, but more detailed studies are needed.

4. Conclusions

In this work, we reviewed the available information on aberrant miRNA expression in various pathological conditions. The basic problems of the available research related to miRNAs were identified. These include the timing of sampling, the type of biofluid being studied, and the individual characteristics of the diseases included in the review.
We propose a spectrum of miRNAs that should be paid attention to when studying epileptogenesis. This includes miR-21, miR-155, and miR-181a as a set of potential microRNA-based biomarkers. Also suggested is the role of miR-30b, miR-219a, miR-335, miR-342, and miR-30a. These miRNAs are not specific for epileptogenesis, but they may be of interest for further study of epileptogenic processes, both separately and in a cohort.
The prediction of epileptogenesis is of great interest to the medical community. However, the study of such processes is complex, long-term, and multifactorial. In any case, more experimental studies are needed to study miRNAs as prognostic biomarkers of epileptogenesis.

Author Contributions

Conceptualization, E.E.T., A.I.P., K.D.L. and A.M.Y.; methodology, A.A.V., E.A.K. and E.E.T.; investigation, A.A.V., D.V.D. and A.M.Y.; writing—original draft preparation, A.A.V.; writing—review and editing, E.E.T., A.M.Y., D.V.D. and A.I.P.; supervision D.V.D. and E.E.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Fisher, R.S.; Acevedo, C.; Arzimanoglou, A.; Bogacz, A.; Cross, J.H.; Elger, C.E.; Engel, J., Jr.; Forsgren, L.; French, J.A.; Glynn, M.; et al. ILAE Official Report: A practical clinical definition of epilepsy. Epilepsia 2014, 55, 475–482. [Google Scholar] [CrossRef]
  2. Epilepsy. Available online: https://www.who.int/news-room/fact-sheets/detail/epilepsy (accessed on 28 July 2023).
  3. Pitkänen, A.; Immonen, R. Epilepsy Related to Traumatic Brain Injury. Neurotherapeutics 2014, 11, 286–296. [Google Scholar] [CrossRef]
  4. Golub, V.M.; Reddy, D.S. Post-Traumatic Epilepsy and Comorbidities: Advanced Models, Molecular Mechanisms, Biomarkers, and Novel Therapeutic Interventions. Pharmacol. Rev. 2022, 74, 387–438. [Google Scholar] [CrossRef] [PubMed]
  5. Dudek, F.E.; Staley, K.J. The Time Course and Circuit Mechanisms of Acquired Epileptogenesis. In Jasper’s Basic Mechanisms of the Epilepsies [Internet]; National Center for Biotechnology Information (US): Bethesda, MD, USA, 2012; pp. 405–415. [Google Scholar] [CrossRef]
  6. Kadam, S.D.; White, A.M.; Staley, K.J.; Dudek, F.E. Continuous Electroencephalographic Monitoring with Radio-Telemetry in a Rat Model of Perinatal Hypoxia–Ischemia Reveals Progressive Post-Stroke Epilepsy. J. Neurosci. 2010, 30, 404. [Google Scholar] [CrossRef] [PubMed]
  7. Williams, P.A.; White, A.M.; Clark, S.; Ferraro, D.J.; Swiercz, W.; Staley, K.J.; Dudek, F.E. Development of spontaneous recurrent seizures after kainate-induced status epilepticus. J. Neurosci. 2009, 29, 2103–2112. [Google Scholar] [CrossRef]
  8. Pitkänen, A.; Lukasiuk, K.; Dudek, F.E.; Staley, K.J. Epileptogenesis. Cold Spring Harb. Perspect. Med. 2015, 5, a022822. [Google Scholar] [CrossRef] [PubMed]
  9. Avakyan, G.N.; Blinov, D.V.; Alikhanov, A.A.; Perepelova, E.M.; Perepelov, V.A.; Burd, S.G.; Lebedeva, A.V. Recommendations of the Russian League Against Epilepsy (RLAE) on the use of magnetic resonance imaging in the diagnosis of epilepsy. Epilepsy Paroxysmal Cond. 2019, 11, 208–232. [Google Scholar] [CrossRef]
  10. BEST (Biomarkers, EndpointS, and Other Tools) Resource; Food and Drug Administration (US): Silver Spring, MD, USA; National Institutes of Health (US): Bethesda, MD, USA, 2016. Available online: https://www.ncbi.nlm.nih.gov/books/NBK326791/ (accessed on 28 July 2023).
  11. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of microRNA biogenesis, mechanisms of actions, and circulation. Front. Endocrinol. 2018, 9, 388354. [Google Scholar] [CrossRef]
  12. Simonato, M.; Agoston, D.V.; Brooks-Kayal, A.; Dulla, C.; Fureman, B.; Henshall, D.C.; Pitkänen, A.; Theodore, W.H.; Twyman, R.E.; Kobeissy, F.H.; et al. Identification of clinically relevant biomarkers of epileptogenesis—A strategic roadmap. Nat. Rev. Neurol. 2021, 17, 231–242. [Google Scholar] [CrossRef]
  13. Gallego, J.A.; Gordon, M.L.; Claycomb, K.; Bhatt, M.; Lencz, T.; Malhotra, A.K. In Vivo microRNA detection and quantitation in cerebrospinal fluid. J. Mol. Neurosci. 2012, 47, 243–248. [Google Scholar] [CrossRef]
  14. Raoof, R.; Bauer, S.; El Naggar, H.; Connolly, N.M.; Brennan, G.P.; Brindley, E.; Hill, T.; McArdle, H.; Spain, E.; Forster, R.J.; et al. Dual-center, dual-platform microRNA profiling identifies potential plasma biomarkers of adult temporal lobe epilepsy. EBioMedicine 2018, 38, 127–141. [Google Scholar] [CrossRef] [PubMed]
  15. Surges, R.; Kretschmann, A.; Abnaof, K.; van Rikxoort, M.; Ridder, K.; Fröhlich, H.; Danis, B.; Kaminski, R.M.; Foerch, P.; Elger, C.E.; et al. Changes in serum miRNAs following generalized convulsive seizures in human mesial temporal lobe epilepsy. Biochem. Biophys. Res. Commun. 2016, 481, 13–18. [Google Scholar] [CrossRef]
  16. Wang, J.; Zhao, J. MicroRNA Dysregulation in Epilepsy: From Pathogenetic Involvement to Diagnostic Biomarker and Therapeutic Agent Development. Front. Mol. Neurosci. 2021, 14, 650372. [Google Scholar] [CrossRef]
  17. Brennan, G.P.; Henshall, D.C. microRNAs in the pathophysiology of epilepsy. Neurosci. Lett. 2018, 667, 47–52. [Google Scholar] [CrossRef] [PubMed]
  18. Shen, C.-H.; Zhang, Y.-X.; Zheng, Y.; Yang, F.; Hu, Y.; Xu, S.; Yan, S.-Q.; Ding, Y.; Guo, Y.; Ding, M.-P. Expression of plasma microRNA-145-5p and its correlation with clinical features in patients with refractory epilepsy. Epilepsy Res. 2019, 154, 21–25. [Google Scholar] [CrossRef]
  19. Antônio, L.G.L.; Freitas-Lima, P.; Pereira-Da-Silva, G.; Assirati, J.A.; Matias, C.M.; Cirino, M.L.A.; Tirapelli, L.F.; Velasco, T.R.; Sakamoto, A.C.; Carlotti, C.G.; et al. Expression of MicroRNAs miR-145, miR-181c, miR-199a and miR-1183 in the Blood and Hippocampus of Patients with Mesial Temporal Lobe Epilepsy. J. Mol. Neurosci. 2019, 69, 580–587. [Google Scholar] [CrossRef]
  20. Zheng, H.; Tang, R.; Yao, Y.; Ji, Z.; Cao, Y.; Liu, Z.; Peng, F.; Wang, W.; Can, D.; Xing, H.; et al. MiR-219 Protects Against Seizure in the Kainic Acid Model of Epilepsy. Mol. Neurobiol. 2016, 53, 1–7. [Google Scholar] [CrossRef] [PubMed]
  21. Zhao, Y.; Lu, C.; Wang, H.; Lin, Q.; Cai, L.; Meng, F.; Tesfaye, E.B.; Lai, H.-C.; Tzeng, C.-M. Identification of hsa-miR-1275 as a Novel Biomarker Targeting MECP2 for Human Epilepsy of Unknown Etiology. Mol. Ther. Methods Clin. Dev. 2020, 19, 398–410. [Google Scholar] [CrossRef]
  22. Raoof, R.; Jimenez-Mateos, E.M.; Bauer, S.; Tackenberg, B.; Rosenow, F.; Lang, J.; Onugoren, M.D.; Hamer, H.; Huchtemann, T.; Körtvélyessy, P.; et al. Cerebrospinal fluid microRNAs are potential biomarkers of temporal lobe epilepsy and status epilepticus. Sci. Rep. 2017, 7, 3328. [Google Scholar] [CrossRef]
  23. Coppola, A.; Moshé, S.L. Why is the developing brain more susceptible to status epilepticus? Epilepsia 2009, 50 (Suppl. S12), 25–26. [Google Scholar] [CrossRef]
  24. An, N.; Zhao, W.; Liu, Y.; Yang, X.; Chen, P. Elevated serum miR-106b and miR-146a in patients with focal and generalized epilepsy. Epilepsy Res. 2016, 127, 311–316. [Google Scholar] [CrossRef]
  25. Sun, J.; Cheng, W.; Liu, L.; Tao, S.; Xia, Z.; Qi, L.; Huang, M. Identification of serum miRNAs differentially expressed in human epilepsy at seizure onset and post-seizure. Mol. Med. Rep. 2016, 14, 5318–5324. [Google Scholar] [CrossRef] [PubMed]
  26. Elnady, H.G.; Abdelmoneam, N.; Eissa, E.; Hamid, E.R.A.; Abu Zeid, D.; Shanab, A.M.A.; Atta, H.; Kholoussi, N.M. MicroRNAs as Potential Biomarkers for Childhood Epilepsy. Open Access Maced. J. Med. Sci. 2019, 7, 3965–3969. [Google Scholar] [CrossRef] [PubMed]
  27. Duan, W.; Chen, Y.; Wang, X. MicroRNA-155 contributes to the occurrence of epilepsy through the PI3K/Akt/mTOR signaling pathway. Int. J. Mol. Med. 2018, 42, 1577–1584. [Google Scholar] [CrossRef]
  28. Martins-Ferreira, R.; Chaves, J.; Carvalho, C.; Bettencourt, A.; Chorão, R.; Freitas, J.; Samões, R.; Boleixa, D.; Lopes, J.; Ramalheira, J.; et al. Circulating microRNAs as potential biomarkers for genetic generalized epilepsies: A three microRNA panel. Eur. J. Neurol. 2020, 27, 660–666. [Google Scholar] [CrossRef]
  29. Liu, Y.; Yu, G.; Ding, Y.-Y.; Zhang, Y.-X. Expression of miR-155 in Serum Exosomes in Children with Epilepsy and Its Diagnostic Value. Dis. Markers 2022, 2022, 7979500. [Google Scholar] [CrossRef]
  30. Wang, J.; Tan, L.; Tan, L.; Tian, Y.; Ma, J.; Tan, C.-C.; Wang, H.-F.; Liu, Y.; Tan, M.-S.; Jiang, T.; et al. Circulating microRNAs are promising novel biomarkers for drug-resistant epilepsy. Sci. Rep. 2015, 5, 10201. [Google Scholar] [CrossRef] [PubMed]
  31. Wang, X.; Sun, Y.; Tan, Z.; Che, N.; Ji, A.; Luo, X.; Sun, X.; Li, X.; Yang, K.; Wang, G.; et al. Serum MicroRNA-4521 is a Potential Biomarker for Focal Cortical Dysplasia with Refractory Epilepsy. Neurochem. Res. 2016, 41, 905–912. [Google Scholar] [CrossRef] [PubMed]
  32. Li, Y.; Huang, C.; Feng, P.; Jiang, Y.; Wang, W.; Zhou, D.; Chen, L. Aberrant expression of miR-153 is associated with overexpression of hypoxia-inducible factor-1α in refractory epilepsy. Sci. Rep. 2016, 6, 32091. [Google Scholar] [CrossRef]
  33. Sun, Y.; Wang, X.; Wang, Z.; Zhang, Y.; Che, N.; Luo, X.; Tan, Z.; Sun, X.; Li, X.; Yang, K.; et al. Expression of microRNA-129-2-3p and microRNA-935 in plasma and brain tissue of human refractory epilepsy. Epilepsy Res. 2016, 127, 276–283. [Google Scholar] [CrossRef]
  34. Che, N.; Zu, G.; Zhou, T.; Wang, X.; Sun, Y.; Tan, Z.; Liu, Y.; Wang, D.; Luo, X.; Zhao, Z.; et al. Aberrant Expression of miR-323a-5p in Patients with Refractory Epilepsy Caused by Focal Cortical Dysplasia. Genet. Test. Mol. Biomarkers 2017, 21, 3–9. [Google Scholar] [CrossRef] [PubMed]
  35. Yan, S.; Zhang, H.; Xie, W.; Meng, F.; Zhang, K.; Jiang, Y.; Zhang, X.; Zhang, J. Altered microRNA profiles in plasma exosomes from mesial temporal lobe epilepsy with hippocampal sclerosis. Oncotarget 2017, 8, 4136–4146. [Google Scholar] [CrossRef] [PubMed]
  36. Avansini, S.H.; de Sousa Lima, B.P.; Secolin, R.; Santos, M.L.; Coan, A.C.; Vieira, A.S.; Torres, F.R.; Carvalho, B.S.; Alvim, M.K.M.; Morita, M.E.; et al. MicroRNA hsa-miR-134 is a circulating biomarker for mesial temporal lobe epilepsy. PLoS ONE 2017, 12, e0173060. [Google Scholar] [CrossRef] [PubMed]
  37. Gong, G.-H.; An, F.-M.; Wang, Y.; Bian, M.; Wang, D.; Wei, C.-X. MiR-153 regulates expression of hypoxia-inducible factor-1α in refractory epilepsy. Oncotarget 2018, 9, 8542–8547. [Google Scholar] [CrossRef]
  38. Leontariti, M.; Avgeris, M.; Katsarou, M.; Drakoulis, N.; Siatouni, A.; Verentzioti, A.; Alexoudi, A.; Fytraki, A.; Patrikelis, P.; Vassilacopoulou, D.; et al. Circulating miR-146a and miR-134 in predicting drug-resistant epilepsy in patients with focal impaired awareness seizures. Epilepsia 2020, 61, 959–970. [Google Scholar] [CrossRef]
  39. Raouf, H.A.; Kholoussi, N.M.; Eissa, E.; El Nady, H.G.; Fayed, D.B.; Abdelkawy, R.F.M. MicroRNAs as Immune Regulators of Inflammation in Children with Epilepsy. Int. J. Mol. Cell Med. 2020, 9, 188–197. [Google Scholar] [CrossRef]
  40. Ioriatti, E.S.; Cirino, M.L.A.; Neto, F.S.L.; Velasco, T.R.; Sakamoto, A.C.; Freitas-Lima, P.; Tirapelli, D.P.C.; Carlotti, C.G. Expression of circulating microRNAs as predictors of diagnosis and surgical outcome in patients with mesial temporal lobe epilepsy with hippocampal sclerosis. Epilepsy Res. 2020, 166, 106373. [Google Scholar] [CrossRef]
  41. De Benedittis, S.; Fortunato, F.; Cava, C.; Gallivanone, F.; Iaccino, E.; Caligiuri, M.E.; Castiglioni, I.; Bertoli, G.; Manna, I.; Labate, A.; et al. Circulating microRNA: The Potential Novel Diagnostic Biomarkers to Predict Drug Resistance in Temporal Lobe Epilepsy, a Pilot Study. Int. J. Mol. Sci. 2021, 22, 702. [Google Scholar] [CrossRef]
  42. Niu, X.; Zhu, H.-L.; Liu, Q.; Yan, J.-F.; Li, M.-L. MiR-194-5p serves as a potential biomarker and regulates the proliferation and apoptosis of hippocampus neuron in children with temporal lobe epilepsy. J. Chin. Med. Assoc. 2021, 84, 510–516. [Google Scholar] [CrossRef]
  43. Huang, H.; Cui, G.; Tang, H.; Kong, L.; Wang, X.; Cui, C.; Xiao, Q.; Ji, H. Relationships between plasma expression levels of microRNA-146a and microRNA-132 in epileptic patients and their cognitive, mental and psychological disorders. Bioengineered 2022, 13, 941–9492. [Google Scholar] [CrossRef]
  44. Wang, Y.; Wang, Y.; Chen, Y.; Hua, Y.; Xu, L.; Zhu, M.; Zhao, C.; Zhang, W.; Sheng, G.; Liu, L.; et al. Circulating MicroRNAs From Plasma Small Extracellular Vesicles as Potential Diagnostic Biomarkers in Pediatric Epilepsy and Drug-Resistant Epilepsy. Front. Mol. Neurosci. 2022, 15, 823802. [Google Scholar] [CrossRef] [PubMed]
  45. Ünalp, A.; Coskunpinar, E.; Gunduz, K.; Pekuz, S.; Baysal, B.T.; Edizer, S.; Hayretdag, C.; Gudeloglu, E. Detection of Deregulated miRNAs in Childhood Epileptic Encephalopathies. J. Mol. Neurosci. 2022, 72, 1234–1242. [Google Scholar] [CrossRef] [PubMed]
  46. Li, X.; Gao, Z.; Ma, M.L.; Li, L.; Guo, S. Identification of serum miR-378 and miR-575 as diagnostic indicators and predicting surgical prognosis in human epilepsy. J. Med. Biochem. 2022, 41, 184–190. [Google Scholar] [CrossRef] [PubMed]
  47. Gattás, D.; Neto, F.; Freitas-Lima, P.; Bonfim-Silva, R.; de Almeida, S.M.; Cirino, M.d.A.; Tiezzi, D.G.; Tirapelli, L.; Velasco, T.; Sakamoto, A.; et al. MicroRNAs miR-629-3p, miR-1202 and miR-1225-5p as potential diagnostic and surgery outcome biomarkers for mesial temporal lobe epilepsy with hippocampal sclerosis. Neurochirurgie 2022, 68, 583–588. [Google Scholar] [CrossRef]
  48. Tubi, M.A.; Lutkenhoff, E.; Blanco, M.B.; McArthur, D.; Villablanca, P.; Ellingson, B.; Diaz-Arrastia, R.; Van Ness, P.; Real, C.; Shrestha, V.; et al. Early seizures and temporal lobe trauma predict post-traumatic epilepsy: A longitudinal study. Neurobiol. Dis. 2019, 123, 115. [Google Scholar] [CrossRef]
  49. Silva, L.F.A.; Hoffmann, M.S.; Gerbatin, R.d.R.; Fiorin, F.d.S.; Dobrachinski, F.; Mota, B.C.; Wouters, A.T.B.; Pavarini, S.P.; Soares, F.A.A.; Fighera, M.R.; et al. Treadmill exercise protects against pentylenetetrazol-induced seizures and oxidative stress after traumatic brain injury. J. Neurotrauma 2013, 30, 1278–1287. [Google Scholar] [CrossRef]
  50. Kori, P.; Garg, R.K.; Malhotra, H.S.; Gupta, R.K.; Verma, R.; Singh, M.K.; Rathore, R.K.S.; Gupta, P.K. Evaluation of cerebral white-matter micro-structural alterations in patients with medically refractory epilepsy using diffusion tensor tractography. Epilepsy Res. 2013, 107, 82–90. [Google Scholar] [CrossRef]
  51. Babb, T.; Kupfer, W.; Pretorius, J.; Crandall, P.; Levesque, M. Synaptic reorganization by mossy fibers in human epileptic fascia dentata. Neuroscience 1991, 42, 351–363. [Google Scholar] [CrossRef] [PubMed]
  52. Cole-Edwards, K.K.; Musto, A.E.; Bazan, N.G. c-Jun N-terminal kinase activation responses induced by hippocampal kindling are mediated by reactive astrocytes. J. Neurosci. 2006, 26, 8295–8304. [Google Scholar] [CrossRef]
  53. Mishra, M.; Singh, R.; Mukherjee, S.; Sharma, D. Dehydroepiandrosterone’s antiepileptic action in FeCl3-induced epileptogenesis involves upregulation of glutamate transporters. Epilepsy Res. 2013, 106, 83–91. [Google Scholar] [CrossRef]
  54. Alyu, F.; Dikmen, M. Inflammatory aspects of epileptogenesis: Contribution of molecular inflammatory mechanisms. Acta Neuropsychiatr. 2017, 29, 1–16. [Google Scholar] [CrossRef] [PubMed]
  55. Jeter, C.B.; Hergenroeder, G.W.; Hylin, M.J.; Redell, J.B.; Moore, A.N.; Dash, P.K.; Baugh, C.M.; Kroshus, E.; Kiernan, P.T.; Mendel, D.; et al. Biomarkers for the diagnosis and prognosis of mild traumatic brain injury/concussion. J. Neurotrauma 2013, 30, 657–670. [Google Scholar] [CrossRef] [PubMed]
  56. Bhomia, M.; Balakathiresan, N.S.; Wang, K.K.; Papa, L.; Maheshwari, R.K. A Panel of Serum MiRNA Biomarkers for the Diagnosis of Severe to Mild Traumatic Brain Injury in Humans. Sci. Rep. 2016, 6, 28148. [Google Scholar] [CrossRef]
  57. Yang, T.; Song, J.; Bu, X.; Wang, C.; Wu, J.; Cai, J.; Wan, S.; Fan, C.; Zhang, C.; Wang, J. Elevated serum miR-93, miR-191, and miR-499 are noninvasive biomarkers for the presence and progression of traumatic brain injury. J. Neurochem. 2016, 137, 122–129. [Google Scholar] [CrossRef]
  58. Taheri, S.; Tanriverdi, F.; Zararsiz, G.; Elbuken, G.; Ulutabanca, H.; Karaca, Z.; Selcuklu, A.; Unluhizarci, K.; Tanriverdi, K.; Kelestimur, F.; et al. Circulating MicroRNAs as Potential Biomarkers for Traumatic Brain Injury-Induced Hypopituitarism. J. Neurotrauma 2016, 33, 1818–1825. [Google Scholar] [CrossRef]
  59. Di Pietro, V.; Ragusa, M.; Davies, D.; Su, Z.; Hazeldine, J.; Lazzarino, G.; Hill, L.J.; Crombie, N.; Foster, M.; Purrello, M.; et al. MicroRNAs as Novel Biomarkers for the Diagnosis and Prognosis of Mild and Severe Traumatic Brain Injury. J. Neurotrauma 2017, 34, 1948–1956. [Google Scholar] [CrossRef] [PubMed]
  60. Mitra, B.; Reyes, J.; O’Brien, W.T.; Surendran, N.; Carter, A.; Bain, J.; McEntaggart, L.; Sorich, E.; Shultz, S.R.; O’Brien, T.J.; et al. Micro-RNA levels and symptom profile after mild traumatic brain injury: A longitudinal cohort study. J. Clin. Neurosci. 2022, 95, 81–87. [Google Scholar] [CrossRef]
  61. Redell, J.B.; Moore, A.N.; Ward, N.H.; Hergenroeder, G.W.; Dash, P.K. Human traumatic brain injury alters plasma microRNA levels. J. Neurotrauma 2010, 27, 2147–2156. [Google Scholar] [CrossRef]
  62. You, W.-D.; Tang, Q.-L.; Wang, L.; Lei, J.; Feng, J.-F.; Mao, Q.; Gao, G.-Y.; Jiang, J.-Y. Alteration of microRNA expression in cerebrospinal fluid of unconscious patients after traumatic brain injury and a bioinformatic analysis of related single nucleotide polymorphisms. Chin. J. Traumatol. 2016, 19, 11–15. [Google Scholar] [CrossRef]
  63. Pan, P.; Bai, L.; Hua, X.; Wang, Y.; Jiang, X.; Cheng, X.; Song, Y.; Yu, X. miR-155 Regulates claudin1 Expression in Humans With Intestinal Mucosa Dysfunction After Brain Injury. Transplant. Proc. 2019, 51, 3474–3480. [Google Scholar] [CrossRef]
  64. Hicks, S.D.; Johnson, J.; Carney, M.C.; Bramley, H.; Olympia, R.P.; Loeffert, A.C.; Thomas, N.J. Overlapping MicroRNA Expression in Saliva and Cerebrospinal Fluid Accurately Identifies Pediatric Traumatic Brain Injury. J. Neurotrauma 2018, 35, 64–72. [Google Scholar] [CrossRef] [PubMed]
  65. Qin, X.; Li, L.; Lv, Q.; Shu, Q.; Zhang, Y.; Wang, Y. Expression profile of plasma microRNAs and their roles in diagnosis of mild to severe traumatic brain injury. PLoS ONE 2018, 13, e0204051. [Google Scholar] [CrossRef]
  66. LaRocca, D.; Barns, S.; Hicks, S.D.; Brindle, A.; Williams, J.; Uhlig, R.; Johnson, P.; Neville, C.; Middleton, F.A. Comparison of serum and saliva miRNAs for identification and characterization of mTBI in adult mixed martial arts fighters. PLoS ONE 2019, 14, e0207785. [Google Scholar] [CrossRef] [PubMed]
  67. Yan, J.; Bu, X.; Li, Z.; Wu, J.; Wang, C.; Li, D.; Song, J.; Wang, J. Screening the expression of several miRNAs from TaqMan Low Density Array in traumatic brain injury: miR-219a-5p regulates neuronal apoptosis by modulating CCNA2 and CACUL1. J. Neurochem. 2019, 150, 202–217. [Google Scholar] [CrossRef] [PubMed]
  68. Polito, F.; Famà, F.; Oteri, R.; Raffa, G.; Vita, G.; Conti, A.; Daniele, S.; Macaione, V.; Passalacqua, M.; Cardali, S.; et al. Circulating miRNAs expression as potential biomarkers of mild traumatic brain injury. Mol. Biol. Rep. 2020, 47, 2941–2949. [Google Scholar] [CrossRef]
  69. O’connell, G.C.; Smothers, C.G.; Winkelman, C. Bioinformatic analysis of brain-specific miRNAs for identification of candidate traumatic brain injury blood biomarkers. Brain Inj. 2020, 34, 965–974. [Google Scholar] [CrossRef]
  70. Schindler, C.R.; Woschek, M.; Vollrath, J.T.; Kontradowitz, K.; Lustenberger, T.; Störmann, P.; Marzi, I.; Henrich, D. miR-142-3p Expression Is Predictive for Severe Traumatic Brain Injury (TBI) in Trauma Patients. Int. J. Mol. Sci. 2020, 21, 5381. [Google Scholar] [CrossRef] [PubMed]
  71. Tas, D.; Kaplan, O.; Sogut, O. Validity of Serum miRNA 93 and miRNA 191 to Reduce Unnecessary Computed Tomography in Patients with Mild Head Trauma. J. Clin. Med. Res. 2020, 12, 579–589. [Google Scholar] [CrossRef] [PubMed]
  72. Ko, J.; Hemphill, M.; Yang, Z.; Beard, K.; Sewell, E.; Shallcross, J.; Schweizer, M.; Sandsmark, D.K.; Diaz-Arrastia, R.; Kim, J.; et al. Multi-Dimensional Mapping of Brain-Derived Extracellular Vesicle MicroRNA Biomarker for Traumatic Brain Injury Diagnostics. J. Neurotrauma 2020, 37, 2424–2434. [Google Scholar] [CrossRef]
  73. Pitkänen, A.; Roivainen, R.; Lukasiuk, K. Development of epilepsy after ischaemic stroke. Lancet Neurol. 2016, 15, 185–197. [Google Scholar] [CrossRef]
  74. Tröscher, A.R.; Gruber, J.; Wagner, J.N.; Böhm, V.; Wahl, A.-S.; von Oertzen, T.J. Inflammation Mediated Epileptogenesis as Possible Mechanism Underlying Ischemic Post-stroke Epilepsy. Front. Aging Neurosci. 2021, 13, 781174. [Google Scholar] [CrossRef] [PubMed]
  75. Myint, P.K.; Staufenberg, E.F.A.; Sabanathan, K. Post-stroke seizure and post-stroke epilepsy. Postgrad. Med. J. 2006, 82, 568. [Google Scholar] [CrossRef] [PubMed]
  76. Phan, J.; Ramos, M.; Soares, T.; Parmar, M.S. Poststroke Seizure and Epilepsy: A Review of Incidence, Risk Factors, Diagnosis, Pathophysiology, and Pharmacological Therapies. Oxidative Med. Cell. Longev. 2022, 2022, 7692215. [Google Scholar] [CrossRef]
  77. Khoshnam, S.E.; Winlow, W.; Farbood, Y.; Moghaddam, H.F.; Farzaneh, M. Emerging Roles of microRNAs in Ischemic Stroke: As Possible Therapeutic Agents. J. Stroke 2017, 19, 166–187. [Google Scholar] [CrossRef] [PubMed]
  78. Long, G.; Wang, F.; Li, H.; Yin, Z.; Sandip, C.; Lou, Y.; Wang, Y.; Chen, C.; Wang, D.W. Circulating miR-30a, miR-126 and let-7b as biomarker for ischemic stroke in humans. BMC Neurol. 2013, 13, 178. [Google Scholar] [CrossRef]
  79. Huang, S.; Zhao, J.; Huang, D.; Zhuo, L.; Liao, S.; Jiang, Z. Serum miR-132 is a risk marker of post-stroke cognitive impairment. Neurosci. Lett. 2016, 615, 102–106. [Google Scholar] [CrossRef]
  80. Rainer, T.H.; Leung, L.Y.; Chan, C.P.Y.; Leung, Y.K.; Abrigo, J.M.; Wang, D.; Graham, C.A. Plasma miR-124-3p and miR-16 concentrations as prognostic markers in acute stroke. Clin. Biochem. 2016, 49, 663–668. [Google Scholar] [CrossRef]
  81. Zeng, L.; Liu, J.; Wang, Y.; Wang, L.; Weng, S.; Tang, Y.; Zheng, C.; Cheng, Q.; Chen, S.; Yang, G.-Y. MicroRNA-210 as a novel blood biomarker in acute cerebral ischemia. Front. Biosci. 2011, 3, 1265–1272. [Google Scholar] [CrossRef]
  82. Tsai, P.-C.; Liao, Y.-C.; Wang, Y.-S.; Lin, H.-F.; Lin, R.-T.; Juo, S.-H.H. Serum microRNA-21 and microRNA-221 as potential biomarkers for cerebrovascular disease. J. Vasc. Res. 2013, 50, 346–354. [Google Scholar] [CrossRef]
  83. Gan, C.; Wang, C.; Tan, K. Circulatory microRNA-145 expression is increased in cerebral ischemia. Genet. Mol. Res. 2012, 11, 147–152. [Google Scholar] [CrossRef]
  84. Wang, Y.; Zhang, Y.; Huang, J.; Chen, X.; Gu, X.; Wang, Y.; Zeng, L.; Yang, G.-Y. Increase of circulating miR-223 and insulin-like growth factor-1 is associated with the pathogenesis of acute ischemic stroke in patients. BMC Neurol. 2014, 14, 77. [Google Scholar] [CrossRef] [PubMed]
  85. Jickling, G.C.; Ander, B.P.; Zhan, X.; Noblett, D.; Stamova, B.; Liu, D. microRNA expression in peripheral blood cells following acute ischemic stroke and their predicted gene targets. PLoS ONE 2014, 9, e99283. [Google Scholar] [CrossRef] [PubMed]
  86. Zhou, J.; Zhang, J. Identification of miRNA-21 and miRNA-24 in plasma as potential early stage markers of acute cerebral infarction. Mol. Med. Rep. 2014, 10, 971–976. [Google Scholar] [CrossRef] [PubMed]
  87. Wang, W.; Sun, G.; Zhang, L.; Shi, L.; Zeng, Y. Circulating microRNAs as novel potential biomarkers for early diagnosis of acute stroke in humans. J. Stroke Cerebrovasc. Dis. 2014, 23, 2607–2613. [Google Scholar] [CrossRef]
  88. Leung, L.Y.; Chan, C.P.Y.; Leung, Y.K.; Jiang, H.L.; Abrigo, J.M.; Wang, D.F.; Chung, J.S.H.; Rainer, T.H.; Graham, C.A. Comparison of miR-124-3p and miR-16 for early diagnosis of hemorrhagic and ischemic stroke. Clin. Chim. Acta 2014, 433, 139–144. [Google Scholar] [CrossRef]
  89. Liu, Y.; Zhang, J.; Han, R.; Liu, H.; Sun, D.; Liu, X. Downregulation of serum brain specific microRNA is associated with inflammation and infarct volume in acute ischemic stroke. J. Clin. Neurosci. 2015, 22, 291–295. [Google Scholar] [CrossRef] [PubMed]
  90. Li, P.; Teng, F.; Gao, F.; Zhang, M.; Wu, J.; Zhang, C. Identification of circulating microRNAs as potential biomarkers for detecting acute ischemic stroke. Cell. Mol. Neurobiol. 2015, 35, 433–447. [Google Scholar] [CrossRef]
  91. Jia, L.; Hao, F.; Wang, W.; Qu, Y. Circulating miR-145 is associated with plasma high-sensitivity C-reactive protein in acute ischemic stroke patients. Cell Biochem. Funct. 2015, 33, 314–319. [Google Scholar] [CrossRef]
  92. Wu, J.; Du, K.; Lu, X. Elevated expressions of serum miR-15a, miR-16, and miR-17-5p are associated with acute ischemic stroke. Int. J. Clin. Exp. Med. 2015, 8, 21071. [Google Scholar]
  93. Peng, G.; Yuan, Y.; Wu, S.; He, F.; Hu, Y.; Luo, B. MicroRNA let-7e Is a Potential Circulating Biomarker of Acute Stage Ischemic Stroke. Transl. Stroke Res. 2015, 6, 437–445. [Google Scholar] [CrossRef]
  94. Zeng, Y.; Liu, J.-X.; Yan, Z.-P.; Yao, X.-H.; Liu, X.-H. Potential microRNA biomarkers for acute ischemic stroke. Int. J. Mol. Med. 2015, 36, 1639–1647. [Google Scholar] [CrossRef]
  95. Yang, Z.-B.; Li, T.-B.; Zhang, Z.; Ren, K.-D.; Zheng, Z.-F.; Peng, J.; Luo, X.-J. The Diagnostic Value of Circulating Brain-specific MicroRNAs for Ischemic Stroke. Intern. Med. 2016, 55, 1279–1286. [Google Scholar] [CrossRef]
  96. Jickling, G.C.; Ander, B.P.; Shroff, N.; Orantia, M.; Stamova, B.; Dykstra-Aiello, C.; Hull, H.; Zhan, X.; Liu, D.; Sharp, F.R. Leukocyte response is regulated by microRNA let7i in patients with acute ischemic stroke. Neurology 2016, 87, 2198–2205. [Google Scholar] [CrossRef]
  97. Tiedt, S.; Prestel, M.; Malik, R.; Schieferdecker, N.; Duering, M.; Kautzky, V.; Stoycheva, I.; Böck, J.; Northoff, B.H.; Klein, M.; et al. 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, 970–980. [Google Scholar] [CrossRef]
  98. Li, D.-B.; Liu, J.-L.; Wang, W.; Li, R.-Y.; Yu, D.-J.; Lan, X.-Y.; Li, J.-P. Plasma Exosomal miR-422a and miR-125b-2-3p Serve as Biomarkers for Ischemic Stroke. Curr. Neurovascular Res. 2017, 14, 330–337. [Google Scholar] [CrossRef] [PubMed]
  99. Wang, Y.; Ma, Z.; Kan, P.; Zhang, B. The Diagnostic Value of Serum miRNA-221-3p, miRNA-382-5p, and miRNA-4271 in Ischemic Stroke. J. Stroke Cerebrovasc. Dis. 2017, 26, 1055–1060. [Google Scholar] [CrossRef] [PubMed]
  100. Wu, J.; Fan, C.L.; Ma, L.J.; Liu, T.; Wang, C.; Song, J.-X.; Lv, Q.-S.; Pan, H.; Zhang, C.; Wu, J.; et al. Distinctive expression signatures of serum microRNAs in ischaemic stroke and transient ischaemic attack patients. Thromb. Haemost. 2017, 117, 992–1001. [Google Scholar] [CrossRef] [PubMed]
  101. Li, S.-H.; Chen, L.; Pang, X.-M.; Su, S.-Y.; Zhou, X.; Chen, C.-Y.; Huang, L.-G.; Li, J.-P.; Liu, J.-L. Decreased miR-146a expression in acute ischemic stroke directly targets the Fbxl10 mRNA and is involved in modulating apoptosis. Neurochem. Int. 2017, 107, 156–167. [Google Scholar] [CrossRef] [PubMed]
  102. Sørensen, S.S.; Nygaard, A.-B.; Carlsen, A.L.; Heegaard, N.H.H.; Bak, M.; Christensen, T. Elevation of brain-enriched miRNAs in cerebrospinal fluid of patients with acute ischemic stroke. Biomark. Res. 2017, 5, 24. [Google Scholar] [CrossRef] [PubMed]
  103. Bam, M.; Yang, X.; Sen, S.; Zumbrun, E.E.; Dennis, L.; Zhang, J.; Nagarkatti, P.S.; Nagarkatti, M. Characterization of Dysregulated miRNA in Peripheral Blood Mononuclear Cells from Ischemic Stroke Patients. Mol. Neurobiol. 2018, 55, 1419–1429. [Google Scholar] [CrossRef]
  104. Chen, Z.; Wang, K.; Huang, J.; Zheng, G.; Lv, Y.; Luo, N.; Liang, M.; Huang, L. Upregulated Serum MiR-146b Serves as a Biomarker for Acute Ischemic Stroke. Cell. Physiol. Biochem. 2018, 45, 397–405. [Google Scholar] [CrossRef]
  105. Lu, W.-J.; Zeng, L.-L.; Wang, Y.; Zhang, Y.; Liang, H.-B.; Tu, X.-Q.; He, J.-R.; Yang, G.-Y. Blood microRNA-15a Correlates with IL-6, IGF-1 and Acute Cerebral Ischemia. Curr. Neurovascular Res. 2018, 15, 63–71. [Google Scholar] [CrossRef]
  106. Cheng, X.; Kan, P.; Ma, Z.; Wang, Y.; Song, W.; Huang, C.; Zhang, B. Exploring the potential value of miR-148b-3p, miR-151b and miR-27b-3p as biomarkers in acute ischemic stroke. Biosci. Rep. 2018, 38, 20181033. [Google Scholar] [CrossRef] [PubMed]
  107. Yoo, H.; Kim, J.; Lee, A.-R.; Lee, J.-M.; Kim, O.-J.; Kim, J.-K.; Oh, S.-H. Alteration of microRNA 340-5p and Arginase-1 Expression in Peripheral Blood Cells during Acute Ischemic Stroke. Mol. Neurobiol. 2019, 56, 3211–3221. [Google Scholar] [CrossRef] [PubMed]
  108. Sun, M.; Hou, X.; Ren, G.; Zhang, Y.; Cheng, H. Dynamic changes in miR-124 levels in patients with acute cerebral infarction. Int. J. Neurosci. 2019, 129, 649–653. [Google Scholar] [CrossRef] [PubMed]
  109. Kotb, H.G.; Ibrahim, A.H.; Mohamed, E.F.; Ali, O.M.; Hassanein, N.; Badawy, D.; Aly, E.A. The expression of microRNA 146a in patients with ischemic stroke: An observational study. Int. J. Gen. Med. 2019, 12, 273–278. [Google Scholar] [CrossRef]
  110. Liu, G.; Cao, C.; Zhu, M. Peripheral Blood miR-451 May Serve as a Biomarker of Ischemic Stroke. Clin. Lab. 2019, 65, 1713–1718. [Google Scholar] [CrossRef]
  111. Wang, J.; Huang, Q.; Ding, J.; Wang, X. Elevated serum levels of brain-derived neurotrophic factor and miR-124 in acute ischemic stroke patients and the molecular mechanism. 3 Biotech 2019, 9, 386. [Google Scholar] [CrossRef]
  112. Guo, C.; Zhong, C.; Li, Q.; Gao, Y.; Li, W.; Ou, Y. Expressions and neural function prognostic evaluation of serum microRNA-24 and microRNA-29b in elderly patients with acute ischemic stroke. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue 2020, 32, 78–82. [Google Scholar] [CrossRef]
  113. Yang, S.; Zhan, X.; He, M.; Wang, J.; Qiu, X. miR-135b levels in the peripheral blood serve as a marker associated with acute ischemic stroke. Exp. Ther. Med. 2020, 19, 3551–3558. [Google Scholar] [CrossRef]
  114. Rahmati, M.; Azarpazhooh, M.R.; Ehteram, H.; Ferns, G.A.; Ghayour-Mobarhan, M.; Ghannadan, H.; Mobarra, N. The elevation of S100B and downregulation of circulating miR-602 in the sera of ischemic stroke (IS) patients: The emergence of novel diagnostic and prognostic markers. Neurol. Sci. 2020, 41, 2185–2192. [Google Scholar] [CrossRef]
  115. Zhang, H.; Chen, G.; Qiu, W.; Pan, Q.; Chen, Y.; Chen, Y.; Ma, X. Plasma endothelial microvesicles and their carrying miRNA-155 serve as biomarkers for ischemic stroke. J. Neurosci. Res. 2020, 98, 2290–2301. [Google Scholar] [CrossRef] [PubMed]
  116. Li, S.; Zhao, Y.; Zhao, J.; Mou, L. Expression and clinical value of miR-128 and IGF-1 in patients with acute ischemic stroke. Minerva Medica 2020, 111, 544–550. [Google Scholar] [CrossRef] [PubMed]
  117. Zhou, X.; Qi, L. miR-124 Is Downregulated in Serum of Acute Cerebral Infarct Patients and Shows Diagnostic and Prognostic Value. Clin. Appl. Thromb./Hemost. 2021, 27, 10760296211035446. [Google Scholar] [CrossRef]
  118. Zhao, X.; Chen, X.; Wu, X.; Zhu, L.; Long, J.; Su, L.; Gu, L. Machine Learning Analysis of MicroRNA Expression Data Reveals Novel Diagnostic Biomarker for Ischemic Stroke. J. Stroke Cerebrovasc. Dis. 2021, 30, 105825. [Google Scholar] [CrossRef] [PubMed]
  119. Wang, Q.; Wang, F.; Fu, F.; Liu, J.; Sun, W.; Chen, Y. Diagnostic and prognostic value of serum miR-9-5p and miR-128-3p levels in early-stage acute ischemic stroke. Clinics 2021, 76, e2958. [Google Scholar] [CrossRef]
  120. Li, K.; Shen, L.; Zheng, P.; Wang, Y.; Wang, L.; Meng, X.; Lv, Y.; Xue, Z.; Guo, X.; Zhang, A.; et al. IIdentification of MicroRNAs as potential biomarkers for detecting ischemic stroke. Genes Genom. 2022, 44, 9–17. [Google Scholar] [CrossRef]
  121. Zhou, B.; Li, B.; Feng, P.; Wang, X.; Gao, H.; Xu, L.; Wang, T.; Guo, X. Identification of a miRNA biomarker for the large artery atherosclerosis subtype of acute ischemic stroke. Folia Neuropathol. 2022, 60, 210–220. [Google Scholar] [CrossRef]
  122. Aldous, E.K.; Toor, S.M.; Parray, A.; Al-Sarraj, Y.; Diboun, I.; Abdelalim, E.M.; Arredouani, A.; El-Agnaf, O.; Thornalley, P.J.; Akhtar, N.; et al. Identification of Novel Circulating miRNAs in Patients with Acute Ischemic Stroke. Int. J. Mol. Sci. 2022, 23, 3387. [Google Scholar] [CrossRef]
  123. SJiang, S.; Wu, J.; Geng, Y.; Zhang, Y.; Wang, Y.; Wu, J.; Lu, C.; Luo, G.; Zan, J.; Zhang, Y. Identification of Differentially Expressed microRNAs Associated with Ischemic Stroke by Integrated Bioinformatics Approaches. Int. J. Genom. 2022, 2022, 9264555. [Google Scholar] [CrossRef]
  124. Zhang, J.; Shen, Y.; Kang, K.; Lin, J.; Wang, A.; Li, S.; Wu, S.; Zhao, X.; Zhang, Q. Circulating MicroRNAs as Potential Biomarkers for Ischemic Stroke in Patients with Asymptomatic Intracranial Artery Stenosis. Cell. Mol. Neurobiol. 2023, 43, 1573–1582. [Google Scholar] [CrossRef] [PubMed]
  125. Jenike, A.E.; Halushka, M.K. miR-21: A non-specific biomarker of all maladies. Biomark. Res. 2021, 9, 18. [Google Scholar] [CrossRef]
  126. Sheedy, F.J. Turning 21: Induction of miR-21 as a Key Switch in the Inflammatory Response. Front. Immunol. 2015, 6, 19. [Google Scholar] [CrossRef] [PubMed]
  127. Pfeffer, S.R.; Yang, C.H.; Pfeffer, L.M. The Role of miR-21 in Cancer. Drug Dev. Res. 2015, 76, 270–277. [Google Scholar] [CrossRef] [PubMed]
  128. Kura, B.; Kalocayova, B.; Devaux, Y.; Bartekova, M. Potential Clinical Implications of miR-1 and miR-21 in Heart Disease and Cardioprotection. Int. J. Mol. Sci. 2020, 21, 700. [Google Scholar] [CrossRef]
  129. Chen, C.-Z.; Li, L.; Lodish, H.F.; Bartel, D.P. MicroRNAs modulate hematopoietic lineage differentiation. Science 2004, 303, 83–86. [Google Scholar] [CrossRef] [PubMed]
  130. Ouyang, Y.-B.; Lu, Y.; Yue, S.; Xu, L.-J.; Xiong, X.-X.; White, R.E.; Sun, X.; Giffard, R.G. miR-181 regulates GRP78 and influences outcome from cerebral ischemia in vitro and in vivo. Neurobiol. Dis. 2012, 45, 555–563. [Google Scholar] [CrossRef]
  131. Liu, Y.; Zhao, Z.; Yang, F.; Gao, Y.; Song, J.; Wan, Y. microRNA-181a is involved in insulin-like growth factor-1-mediated regulation of the transcription factor CREB1. J. Neurochem. 2013, 126, 771–780. [Google Scholar] [CrossRef]
  132. Ouyang, Y.-B.; Lu, Y.; Yue, S.; Giffard, R.G. miR-181 targets multiple Bcl-2 family members and influences apoptosis and mitochondrial function in astrocytes. Mitochondrion 2012, 12, 213–219. [Google Scholar] [CrossRef]
  133. Mahesh, G.; Biswas, R. MicroRNA-155: A Master Regulator of Inflammation. J. Interf. Cytokine Res. 2019, 39, 321–330. [Google Scholar] [CrossRef]
  134. Hu, J.; Huang, S.; Liu, X.; Zhang, Y.; Wei, S.; Hu, X. miR-155: An Important Role in Inflammation Response. J. Immunol. Res. 2022, 2022, 7437281. [Google Scholar] [CrossRef] [PubMed]
  135. Zheng, H.; Wu, L.; Yuan, H. miR-30b-5p targeting GRIN2A inhibits hippocampal damage in epilepsy. Open Med. 2023, 18, 20230675. [Google Scholar] [CrossRef] [PubMed]
  136. Fu, N.; Yu, J.; Zhu, L.; Yang, L.; Ma, L.; He, J.; Yu, H.; Liu, J.; Tian, Y.; Xu, J. Role of miR-219a-5p in regulating NMDAR in nonylphenol-induced synaptic plasticity damage. Ecotoxicol. Environ. Saf. 2023, 252, 114576. [Google Scholar] [CrossRef] [PubMed]
  137. Heiland, M.; Connolly, N.M.C.; Mamad, O.; Nguyen, N.T.; Kesavan, J.C.; Langa, E.; Fanning, K.; Sanfeliu, A.; Yan, Y.; Su, J.; et al. MicroRNA-335-5p suppresses voltage-gated sodium channel expression and may be a target for seizure control. Proc. Natl. Acad. Sci. USA 2023, 120, e2216658120. [Google Scholar] [CrossRef]
  138. Pala, M.; Meral, I.; Acikgoz, N.P.; Yilmaz, S.G.; Okur, S.K.; Acar, S.; Polat, Y.; Akbas, F. Downregulatory effect of miR-342-3p on epileptogenesis in the PTZ-kindling model. Mol. Biol. Rep. 2022, 49, 11997–12006. [Google Scholar] [CrossRef]
  139. Wang, Z.; Xie, W.; Guan, H. The diagnostic, prognostic role and molecular mechanism of miR-328 in human cancer. Biomed. Pharmacother. 2023, 157, 114031. [Google Scholar] [CrossRef]
  140. Jiang, L.-H.; Zhang, H.-D.; Tang, J.-H. MiR-30a: A Novel Biomarker and Potential Therapeutic Target for Cancer. J. Oncol. 2018, 2018, 5167829. [Google Scholar] [CrossRef]
  141. Wang, T.; Li, F.; Tang, S. MiR-30a upregulates BCL2A1, IER3 and cyclin D2 expression by targeting FOXL2. Oncol. Lett. 2015, 9, 967–971. [Google Scholar] [CrossRef]
  142. Liu, T.; Xu, Z.; Ou, D.; Liu, J.; Zhang, J. The miR-15a/16 gene cluster in human cancer: A systematic review. J. Cell. Physiol. 2019, 234, 5496–5506. [Google Scholar] [CrossRef]
  143. Yin, H.; Sun, Y.; Wang, X.; Park, J.; Zhang, Y.; Li, M.; Yin, J.; Liu, Q.; Wei, M. Progress on the relationship between miR-125 family and tumorigenesis. Exp. Cell Res. 2015, 339, 252–260. [Google Scholar] [CrossRef]
  144. Li, S.-H.; Su, S.-Y.; Liu, J.-L. Differential Regulation of microRNAs in Patients with Ischemic Stroke. Curr. Neurovascular Res. 2015, 12, 214–221. [Google Scholar] [CrossRef]
  145. Climent, M.; Quintavalle, M.; Miragoli, M.; Chen, J.; Condorelli, G.; Elia, L. TGFβ triggers miR-143/145 transfer from smooth muscle cells to endothelial cells, thereby modulating vessel stabilization. Circ. Res. 2015, 116, 1753–1764. [Google Scholar] [CrossRef] [PubMed]
  146. Harraz, M.M.; Eacker, S.M.; Wang, X.; Dawson, T.M.; Dawson, V.L. MicroRNA-223 is neuroprotective by targeting glutamate receptors. Proc. Natl. Acad. Sci. USA 2012, 109, 18962–18967. [Google Scholar] [CrossRef] [PubMed]
  147. Henshall, D.C.; Araki, T.; Schindler, C.K.; Lan, J.-Q.; Tiekoter, K.L.; Taki, W.; Simon, R.P. Activation of Bcl-2-Associated Death Protein and Counter-Response of Akt within Cell Populations during Seizure-Induced Neuronal Death. J. Neurosci. 2002, 22, 8458–8465. [Google Scholar] [CrossRef] [PubMed]
  148. Aronica, E.; Gorter, J.; Jansen, G.; van Veelen, C.; van Rijen, P.; Leenstra, S.; Ramkema, M.; Scheffer, G.; Scheper, R.; Troost, D. Expression and cellular distribution of multidrug transporter proteins in two major causes of medically intractable epilepsy: Focal cortical dysplasia and glioneuronal tumors. Neuroscience 2003, 118, 417–429. [Google Scholar] [CrossRef]
  149. Suzuki, A.; Fukushima, H.; Mukawa, T.; Toyoda, H.; Wu, L.-J.; Zhao, M.-G.; Xu, H.; Shang, Y.; Endoh, K.; Iwamoto, T.; et al. Upregulation of CREB-Mediated Transcription Enhances Both Short- and Long-Term Memory. J. Neurosci. 2011, 31, 8786–8802. [Google Scholar] [CrossRef]
Figure 1. miRNA processing and mechanism of action: cytoplasmic processing of pre-microRNA via Dicer protein; disintegration of a double-stranded microRNA duplex, as a result of which only one of the strands becomes a guide; targeting guide microRNA and the RISC complex (RNA-induced silencing complex) to target mRNA; degradation of target mRNA.
Figure 1. miRNA processing and mechanism of action: cytoplasmic processing of pre-microRNA via Dicer protein; disintegration of a double-stranded microRNA duplex, as a result of which only one of the strands becomes a guide; targeting guide microRNA and the RISC complex (RNA-induced silencing complex) to target mRNA; degradation of target mRNA.
Ijms 24 15366 g001
Figure 2. PRISMA chart.
Figure 2. PRISMA chart.
Ijms 24 15366 g002
Figure 3. MicroRNAs common to epilepsy, TBI, and ischemic stroke. The outer circles indicate the number of miRNAs found in these diseases, at the intersection of three circles—the names of miRNAs common to all diseases.
Figure 3. MicroRNAs common to epilepsy, TBI, and ischemic stroke. The outer circles indicate the number of miRNAs found in these diseases, at the intersection of three circles—the names of miRNAs common to all diseases.
Ijms 24 15366 g003
Figure 4. Most significant biological processes in which miR-21 is involved [22,59,82,86,100].
Figure 4. Most significant biological processes in which miR-21 is involved [22,59,82,86,100].
Ijms 24 15366 g004
Figure 5. Tissue expression of (A) miR-21-3p and (B) miR-21-5p [22,59,82,86,100].
Figure 5. Tissue expression of (A) miR-21-3p and (B) miR-21-5p [22,59,82,86,100].
Ijms 24 15366 g005
Figure 6. Most significant biological processes in which miR-181a is involved [39,62,100].
Figure 6. Most significant biological processes in which miR-181a is involved [39,62,100].
Ijms 24 15366 g006
Figure 7. Tissue expression of (A) miR-181a-3p and (B) miR-181a-5p [39,62,100].
Figure 7. Tissue expression of (A) miR-181a-3p and (B) miR-181a-5p [39,62,100].
Ijms 24 15366 g007
Figure 8. Most significant biological processes in which miR-155 is involved [27,28,29,63,115].
Figure 8. Most significant biological processes in which miR-155 is involved [27,28,29,63,115].
Ijms 24 15366 g008
Figure 9. Tissue expression of miR-155-5p [27,28,29,63,115].
Figure 9. Tissue expression of miR-155-5p [27,28,29,63,115].
Ijms 24 15366 g009
Table 4. MicroRNAs associated with epilepsy, TBI, and IS.
Table 4. MicroRNAs associated with epilepsy, TBI, and IS.
Epilepsy + TBIEpilepsy + ISEpilepsy + TBI + IS
miR-30b↑
miR-21(E↓TBI↑)
miR-328-3p↑
miR-335(E↓ TBI ↑)
miR-155 (E↑ TBI↓)
miR-181a(E↓ TBI↑)
miR-342(E—3p,IS—5p)↑
miR-153 (E↓ IS↑)
miR-106b↑
miR-146a(E↑ AIS↓)
miR-30a (E↑ IS↓)
miR-15a(E↑ IS↓)
miR-143↑
miR-145(E↑↓ IS↓)
miR-21(E↓ IS ↑↓)
miR-223↑
miR-451a ↑
miR-155↑
miR-132
miR-125a(E↓ IS↑)
miR-181a(E↓ IS↑)
miR-142 (E↑ IS↓)
miR-21
miR-181a
miR-155
E—epilepsy; TBI—traumatic brain injury; IS—ischemic stroke; AIS—acute ischemic stroke; ↑—upregulation; ↓—downregulation.
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

Vasilieva, A.A.; Timechko, E.E.; Lysova, K.D.; Paramonova, A.I.; Yakimov, A.M.; Kantimirova, E.A.; Dmitrenko, D.V. MicroRNAs as Potential Biomarkers of Post-Traumatic Epileptogenesis: A Systematic Review. Int. J. Mol. Sci. 2023, 24, 15366. https://doi.org/10.3390/ijms242015366

AMA Style

Vasilieva AA, Timechko EE, Lysova KD, Paramonova AI, Yakimov AM, Kantimirova EA, Dmitrenko DV. MicroRNAs as Potential Biomarkers of Post-Traumatic Epileptogenesis: A Systematic Review. International Journal of Molecular Sciences. 2023; 24(20):15366. https://doi.org/10.3390/ijms242015366

Chicago/Turabian Style

Vasilieva, Anastasia A., Elena E. Timechko, Kristina D. Lysova, Anastasia I. Paramonova, Alexey M. Yakimov, Elena A. Kantimirova, and Diana V. Dmitrenko. 2023. "MicroRNAs as Potential Biomarkers of Post-Traumatic Epileptogenesis: A Systematic Review" International Journal of Molecular Sciences 24, no. 20: 15366. https://doi.org/10.3390/ijms242015366

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