Next Article in Journal
CircDOCK1 Regulates miR-186/DNMT3A to Promote Osteosarcoma Progression
Next Article in Special Issue
Development of Antiepileptic Drugs throughout History: From Serendipity to Artificial Intelligence
Previous Article in Journal
Pathophysiology and Outcomes of Endothelium Function in Coronary Microvascular Diseases: A Systematic Review of Randomized Controlled Trials and Multicenter Study
Previous Article in Special Issue
Acute Hippocampal Damage as a Prognostic Biomarker for Cognitive Decline but Not for Epileptogenesis after Experimental Traumatic Brain Injury
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Stage- and Subfield-Associated Hippocampal miRNA Expression Patterns after Pilocarpine-Induced Status Epilepticus

1
Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
2
Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
3
Department of Education, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
4
Radiation Physiology Lab, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore 138602, Singapore
5
Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
*
Authors to whom correspondence should be addressed.
Biomedicines 2022, 10(12), 3012; https://doi.org/10.3390/biomedicines10123012
Submission received: 5 September 2022 / Revised: 15 October 2022 / Accepted: 16 November 2022 / Published: 23 November 2022

Abstract

:
Objective: To investigate microRNA (miRNA) expression profiles before and after pilocarpine-induced status epilepticus (SE) in the cornu ammonis (CA) and dentated gyrus (DG) areas of the mouse hippocampus, and to predict the downstream proteins and related pathways based on bioinformatic analysis. Methods: An epileptic mouse model was established using a pilocarpine injection. Brain tissues from the CA and DG were collected separately for miRNA analysis. The miRNAs were extracted using a kit, and the expression profiles were generated using the SurePrint G3 Mouse miRNA microarray and validated. The intersecting genes of TargetScan and miRanda were selected to predict the target genes of each miRNA. For gene ontology (GO) studies, the parent-child-intersection (pci) method was used for enrichment analysis, and Benjamini-Hochberg was used for multiple test correction. The Kyoto Encyclopedia of Genes and Genomes (KEGG) was used to detect disease-related pathways among the large list of miRNA-targeted genes. All analyses mentioned above were performed at the time points of control, days 3, 14, and 60 post-SE. Results: Control versus days 3, 14, and 60 post-SE: in the CA area, a total of 131 miRNAs were differentially expressed; 53, 49, and 26 miRNAs were upregulated and 54, 10, and 22 were downregulated, respectively. In the DG area, a total of 171 miRNAs were differentially expressed; furthermore, 36, 32, and 28 miRNAs were upregulated and 78, 58, and 44 were downregulated, respectively. Of these, 92 changed in both the CA and DG, 39 only in the CA, and 79 only in the DG area. The differentially expressed miRNAs target 11–1630 genes. Most of these proteins have multiple functions in epileptogenesis. There were 15 common pathways related to altered miRNAs: nine different pathways in the CA and seven in the DG area. Conclusions: Stage- and subfield-associated hippocampal miRNA expression patterns are closely related to epileptogenesis, although the detailed mechanisms need to be explored in the future.

1. Introduction

Epilepsy is associated with plasticity of synaptic transmission, axonal sprouting, and neurogenesis in the hippocampus [1,2]. The hippocampus is divided into five sub-regions; CA1, CA2, CA3, CA4 (Cornu Ammonis region, Ammon’s horn), and the dentate gyrus (DG), based on cell size, shape, connectivity, and function [3]. Granule neurones are mainly distributed in the DG, whereas pyramidal neurones are predominantly concentrated in the CA region. These regions are distinguished by their unique patterns of connectivity; layer II neurones project to the DG and CA3/CA2 of the hippocampus proper, whereas layer III neurones project to CA1 and the subiculum [4].
Animal models have been used extensively to study the mechanisms of epilepsy; these include models of pilocarpine-induced status epilepticus (SE), electrical stimulation-induced SE, or kainic acid (KA)-induced SE. The ability of pilocarpine to induce SE is due to the activation of the M1 muscarinic receptor subtype. Following pilocarpine-induced SE, the mortality rate of adult male albino mice was 25–50%, the mean latent period was 14.4 days, and spontaneous recurrent seizures (SRSs) lasted 50–60 s with a frequency of 1–5 seizures per week in the chronic stage. Injection of pilocarpine induces an SE characterised by tonic-clonic generalised seizures, which spontaneously remit several hours later; this stage was defined as the acute stage with loss of neurones (days 1 to 3 post-SE), after which the mice went into a seizure-free period called the latent period with more prominent loss of neurones (days 7 to 14 post-SE), before displaying the SRSs and entering chronic epileptic condition characterised by an increase of astrocytes in the DG known as sclerosis (day 60 post-SE) [5,6]. The key cellular components of hippocampal formation, DG granule cells, and CA neurones are strikingly different during chronic epileptic activity. The DG has a uniquely permissible environment for neurogenesis, whereas the CA does not. Multipotent neuroprogenitor cells differentiate only into neurones when grafted into the DG area [7]. These characteristics prompted us to study these two parts separately.
MicroRNAs (miRNAs) are small RNAs that regulate gene expression by binding to the 3′-UTR of mRNAs, causing translational repression and/or mRNA degradation [8,9]. The mechanisms of this involve binding to target mRNAs and reducing protein production via Argonaute (AGO)-mediated translational repression [8]. Approximately 1500 miRNAs in the human genome are predicted to regulate more than one-third of all protein-coding genes because a single miRNA can target approximately 200 mRNAs [10].
Current studies on miRNA expression in epilepsy involve the analysis of blood, cerebrospinal fluid, and brain tissues in both animal models and patients, in terms of pathogenesis, diagnosis, treatment, and prognosis [11,12,13,14,15,16,17]. However, the miRNA expression profiles in the CA and DG areas are still relatively unknown. Herein, the expression profile of miRNAs in the CA and DG were examined based on miRNA microarrays and confirmed with quantitative real-time polymerase chain reaction (qRT-PCR) and bioinformatics analysis.

2. Materials and Methods

2.1. Sample Preparation

2.1.1. Experimental Animals

This study was approved by the Institutional Animal Care and Use Committee of the National University of Singapore (IACUC approved protocol No: B27/09&098/09). Experiments were performed on 8–10-week-old non-spontaneously epileptic adult male Swiss albino mice weighing in a range from 25 g–30 g. The mice used in the study were randomly divided into two groups: the control group did not undergo pilocarpine injection but saline, whereas the pilocarpine-induced seizures group underwent pilocarpine injection. All mice in the control and pilocarpine-induced seizures group were sacrificed, and the latter group was re-divided into three subgroups according to the time points at which they were sacrificed; days 3 (acute), 14 (latency), and 60 (chronic) post-SE. Three mice were included in each group.

2.1.2. Pilocarpine-Induced SE Mouse Model Preparation

The mice were administered a single subcutaneous injection of methyl-scopolamine nitrate (1 mg/kg) 30 min before the injection of either saline in the control or pilocarpine (300 mg/kg) in the experimental groups. Treated mice experienced acute SE. If the mice did not experience continuous seizures for 4 h, they were considered non-SE mice and were excluded from our study. Some mice experienced convulsions and died; only those with continuous SE for more than 4 h were eligible for the study (classification of seizures was based on Racine, 1972). Mice were placed in disposable cages in the same environment. Continuous behavioural observations were performed from SE cessation until the intended time points.

2.1.3. Tissue Preparation for miRNA Isolation

All eligible mice were sacrificed by decapitation at different time points. Hippocampal brain tissue was collected and placed in an ice-cold phosphate-buffered saline (PBS) in a Petri dish. The DG was distinguishable from the CA by the gaps between them and was separated from the CA by inserting a sharp needle tip into each side of the DG. The needles were then slid superficially along the septotemporal axis of the hippocampus for isolation [18]. The isolated DG was picked up using a needle or forceps and placed in a sample tube containing RNAlater RNA stabilisation reagent (Qiagen, Dusseldorf, Germany). The CA and DG samples in all four groups were snap-frozen in liquid nitrogen and stored at −80 °C for further use.

2.2. Total RNA and miRNA Isolation

Each frozen sample was homogenised in 1 mL QIAzol lysis reagent (Qiagen, Dusseldorf, Germany). After the addition of 140 μL chloroform and centrifugation for 15 min, the upper phase was collected and precipitated with absolute ethanol (1.5 mL). The mixture was passed through an RNeasy Mini spin column and washed with RWT and RPE buffer, and total RNAs, including small RNAs, were eluted with RNase-free water. The quantity and quality of RNA were determined at 260/280 nm and 260/230 nm, respectively, using a NanoDrop spectrophotometer (Ocean Optics, Florida, USA) and an Agilent Bioanalyzer (Agilent, Santa Clara, CA, USA). The acceptable ratios for this study were a 260/280 ratio of >2.0 and a 230/280 ratio of >1.8. Acceptable RNA integrity numbers for this study were those greater than 7. The samples were chosen to meet these criteria for downstream assays.

2.3. SurePrint G3 Mouse miRNA Microarray

Microarray analysis was performed by a service provider (Genomax Technologies, Singapore) using the SurePrint G3 Mouse miRNA microarray kit (Agilent, Santa Clara, CA, USA). This kit was produced based on miRBase 16.0 and included 1023 mouse and 57 mouse viral G4859A miRNAs. The procedure included four steps: spiking in solution preparation, sample labelling and hybridisation, microarray washing, scanning, and feature extraction. With a sample input of 100 ng of total RNA, Agilent’s miRNA complete labelling and hybridisation kit generates fluorescently labelled miRNAs via ligation of one Cyanine 3-Cytidine biphosphate (pCp) molecule to the 3′ end of an RNA molecule with more than 90% efficiency. The miRNA microarray data were analysed using Genespring software (GX 11, Agilent, Santa Clara, CA, USA).

2.4. qRT-PCR Analysis of Chosen miRNAs

To validate the miRNA results, miRCURY LNATM universal cDNA synthesis and SYBR Green master mix kits (Exiqon, Copenhagen, Denmark) were used. First, total RNA, including small RNAs, was reverse-transcribed to cDNA. A real-time experiment was performed using 7900HT fast real-time PCR instruments (Life Technologies, Carlsbad, CA, USA). Cycle threshold (Ct) values were acquired using the software provided by the manufacturer. The reference gene of each group was RNU5G (the reference gene was chosen by pre-experimental tests; after some tests, this gene was expressed stably in each sample). The relative expression of each miRNA was calculated as ΔCt = Ct target gene − Ct internal control gene. The relative quality (RQ) of mRNA from the SE group compared to that of the control group was calculated. Fold-changes in miRNA expression in the SE group were determined relative to the control using the 2−∆∆Ct method. Statistical analyses were performed using one-way analysis of variance (ANOVA) with SPSS (version 25, IBM, Armonk, NY, USA).

2.5. Bioinformatics Analysis of the miRNA Microarray Results

Genes with significant differential expression between the three different stages of SE were first filtered using significance analysis of microarrays (SAM) software (p < 0.05). One-way ANOVA and the Benjamini-Hochberg test were used to perform multiple testing corrections.
Sequence checking of the gene expression dynamics clustering algorithm was used to analyse the gene expression time series and to determine the cluster set most likely to generate the observed time series. This method explicitly considers the dynamic nature of gene expression profiles during clustering and determines the number of different clusters.
According to the random variance model (RVM) corrective ANOVA, differentially expressed genes were selected in a logical sequence. A set of unique model expression tendencies was identified according to the different signal density change tendencies of genes under different situations. The raw expression values were converted into log2 ratios. Some unique profiles were defined using a strategy for clustering the short time-series gene expression data. The actual or expected number of genes assigned to each model profile was related to the expression model profiles. Analysed by Fisher’s exact test and multiple comparison test, the significant profiles exhibited higher probabilities than expected.
To further reduce the false positive rate (FDR), the intersection genes of TargetScan and miRanda were selected for target gene prediction. The intersecting target genes were annotated for gene function based on the gene ontology (GO) database for functional analysis. The GO terms which were structured and controlled were subdivided into three ontologies: molecular function (MF), biological process (BP), and cellular component (CC). The two-sided Fisher’s exact test and Benjamini-Honchberg test were used for multiple test correction, and FDR was used to adjust the p-values for multiple comparisons, with a p value < 0.01 and an adjusted p value < 0.01.
KEGG was used to detect disease-related pathways among the large list of miRNA-targeted genes. Pathway enrichment was calculated using hypergeometric distribution, and the p-values were adjusted for multiple comparisons by FDR.

3. Results

3.1. miRNA Microarray Analysis

Control versus days 3, 14, and 60 post-SE: in the CA area, 131 miRNAs were differently expressed, including 107 in the acute stage, 59 in the latent stage, and 48 in the chronic stage, 53, 49, and 26 miRNAs were upregulated and 54, 10, and 22 were downregulated, respectively. In the DG, 171 miRNAs were differentially expressed, including 114 in the acute stage, 90 in the latent stage, and 72 in the chronic stage, 36, 32, and 28 miRNAs were upregulated and 78, 58, and 44 were downregulated, respectively. Of these, 92 changed in both the CA and DG, 39 only in the CA, and 79 only in the DG area (cut off value: absolute value of fold change ≥ 1.5). The differentially expressed miRNAs at the three time points (days 3, 14, and 60) in the CA and DG are shown in Figure 1.

3.2. Validation of the miRNA Microarray Data by qRT-PCR

Primers of the selected miRNAs were used to validate the microarray data. The detailed results are listed in Table 1 and Table 2 and the miRNAs labelled with “*” were statistically different compared with those of control. Those labelled with “#” were statistically different compared with those of day 3 post-SE and those labelled with “^” were statistically different compared with those of day 14 post-SE. In the CA area, the validated miRNAs were 124, 137, 142-3p, 19a, 203, 27a, 494, 551b, 146a, 188-5p, and 193. In the DG area, the validated miRNAs were 124, 137, 142-3p, 19a, 203, 27a, 494, 551b, 132, 135b, 148a, 188-5p, and 672 (Table 1 and Table 2). Most qRT-PCR results were consistent with the microarray results, with the exception of those for miR-494 and 188-5p in both the CA and DG areas.

4. Bioinformatics Analysis of the miRNA Microarray Results

4.1. Gene Expression Trend Analysis

There were 26 miRNA gene expression trends on days 3, 14, and 60 post-SE in the CA and DG areas; they were randomly labelled with numbers. It can be concluded that trends of No.6 (A/D), 9 (B/E) and 19 (C/F) have significant differences in both CA (A, B, C) and DG (D, E, F) areas among the 26 trends generated (Figure 2). The miRNA expression levels were relatively similar between the two areas.

4.2. Gene Ontology Enrichment Analysis

The number of intersecting genes in the CA and DG predicted by Targetscan and miRanda is shown in Table 3.
GO enrichment analysis based on the predicted intersection target genes of differentially expressed miRNAs revealed that there are numerous functions regulated by differentially expressed miRNAs. These functions are regulated in both CA and DG areas (the larger the (−LgP) value, the smaller the p value, and the higher the significance level of GO).
Some common functions are regulated by miRNAs in the CA and DG. These are G protein-related processes (4), macromolecule-related processes (5), nitrogen compound-related processes (5), RNA-related processes (3), biosynthetic processes (3), metabolic processes (15), signalling (11), neuron differentiation (1), stimulus (11), transport processes (6), and development-related processes (37). The numbers in brackets represent the number of functions associated with the keywords. Table 4 lists the different functions regulated by the altered miRNAs in the CA and DG areas and their −LgP values.

4.3. Pathway Analysis

After deleting some obviously irrelevant pathways (those associated with cancer, infection, or other organs), the pathways regulated by the altered miRNAs in both the CA and DG areas were axon guidance; regulation of the actin cytoskeleton; focal adhesion; the Rap1, MAPK, Notch, Fc epsilon RI, TNF, and VEGF signalling pathways; dorso-ventral axis formation; glycerophospholipid metabolism; glycosaminoglycan biosynthesis-chondroitin sulphate/dermatan sulphate; the HIF-1 signalling pathway; endocrine and other factor-regulated calcium reabsorption; and the calcium signalling pathway. The different pathways regulated by the differentially expressed miRNAs in the CA and DG areas are listed in Table 5.

5. Discussion

miRNAs are small single-stranded RNAs approximately 22 nucleotides in length, which act as modulators of gene expression at the post-transcriptional level. It has been demonstrated that miRNA expression differs between brain regions [19,20], and it is believed that brain region-specific miRNA expression plays a role in regulating mRNA transcription. miRNAs have important effects on brain excitability and control several processes associated with epileptogenesis, including inflammation, synaptogenesis, and ion channel expression [8,21]. During the past decade, there have been some findings related to the specific miRNAs involved in epileptogenesis. However, seizure-evoked miRNA profiles differ depending on the species, mode of seizure induction, type of tissue analysed, and time of tissue collection after seizure [22]; hence, there might be various results among studies. We used the microarray method to screen for differentially expressed miRNAs in the pilocarpine-induced mouse epileptic model and qRT-PCR for validation. Control versus days 3, 14, and 60 post-SE: in the CA area, a total of 131 miRNAs were differentially expressed; 53, 49, and 26 miRNAs were upregulated, and 54, 10, and 22 were downregulated, respectively. In the DG area, a total of 171 miRNAs were differentially expressed; 36, 32, and 28 miRNAs were upregulated and 78, 58, and 44 were downregulated, respectively. Of these, 92 changed in both the CA and DG, 39 only in the CA, and 79 only in the DG area. The randomly selected miRNA qRT-PCR results were consistent with the microarray results, with the exception of those for miR-494 and 188-5p, which were excluded. We used bioinformatics to predict target genes, their functions, and related pathways. The differentially expressed miRNA target 11–1630 genes. Most of these proteins have multiple functions in epileptogenesis and are worthy of further study. There were 15 common pathways related to altered miRNAs; nine in the CA and seven in the DG area. To better understand the effect on epileptogenesis of these changed miRNAs, we compared our results with the previous literature; most were consistent with each other regardless of the models used, and some were consistent with the analysis of temporal lobe epilepsy (TLE) patient samples.
First, we compared our validated miRNA results with those of other studies. miR-27a-3p is overexpressed in the hippocampal cells of epileptic rats and KA-treated neurones [23]. Our results showed upregulation of miR-27a after SE in both the CA and DG areas at all three time points, consistent with a previous in vitro study. miR-132 was shown to be upregulated in a hippocampal neuronal culture and rat model of SE [24,25]. A study of the hippocampus of patients with TLE hippocampal sclerosis (TLE-HS) and the DG areas of electrodes in epileptic rats in the acute stage confirmed its upregulation [25]. Epileptic patients exhibit higher miR-132 expression in plasma, which could reflect the severity of epilepsy and predict the risks of complications [26]. miR-132 was upregulated in the DG area at the three time points after SE in this study. miR-203 was shown to be upregulated in the hippocampus of epileptic mice [27]. In addition, it was upregulated in the CA and DG areas at the three time points in this study. A previous study demonstrated upregulation of miR-146a expression in all hippocampal tissues in the latent and chronic stages of SE development in a rat model and in human TLE patients with hippocampal sclerosis [28]. Plasma miR-146a expression is increased in epileptic patients [26]. In our study, miR-146a was upregulated in the CA area at the three time points and in the DG area on days 3 and 14 post-SE. miR-142-3p is upregulated in the hippocampi of mTLE/HS patients of all age groups, including children, adolescents, and adults, and rats 3 months after SE [29]. In our study, miR-142-3p was upregulated in the CA area at the three time points and in the DG area on days 3 and 14 post-SE. Lower miR-137 expression has been associated with intellectual disability [30]. In our study, it was downregulated especially on day 3 post-SE in the CA region. Downregulated miR-551b-5p was found during long-term intermittent theta burst stimulation (iTBS) treatment of ischaemia/reperfusion (I/R), enhancing neurogenesis and migration [31]. In our study, miR-551b was downregulated on days 14 and 60 post-SE in the CA area and at the three time points in the DG area. miR-19a/b-3p, FoxO3, and SPHK1 have been shown to be upregulated, whereas SIRT1 has been shown to be downregulated in I/R [32]. We found that miR-19a was upregulated on day 3 post-SE compared to the control and downregulated on day 60 compared to on day 14 post-SE in both the CA and DG areas. miRNA-672 is downregulated and localised in neurones in the dorsal root ganglia (DRG) [33]. In our study, miR-672 was downregulated on days 14 and 60 post-SE in the DG area. Overexpression of miR-148a-3p in a febrile seizure (FS) model group affected the neuroimmune system and neuronal apoptosis in a previous study [34]. miR-148a-3p may serve as a novel diagnostic biomarker for TLE [35]. In our study, we found that miR-148a was downregulated in the DG region on days 14 and 60 post-SE. miR-135b-5p was shown to be downregulated in children with TLE and in an epileptic rat neuron model [36,37]. We found that miR-135b was upregulated on day 14 post-SE in the DG.
Second, some of our miRNA microarray results showed the same expression trends as in other studies, although they were not validated by qRT-PCR. Neuronal excitability [38]-related miRNAs include upregulated miR-23a [39] and downregulated miR-211 (DG) [40] and miR-128 (DG) [41]. Transcription factor-related miRNAs include downregulated miR-124 [42]. Apoptosis-related miRNAs include upregulated miR-21 [43], miR-34a (CA) [44], miR-129-5p [45], and downregulated miR-139-5p [46]. Ion channel-related miRNAs include downregulated miR-324-5p (DG) [47]. Oxidative stress-related miRNAs include upregulated miR-221 (DG) [48] and downregulated miR-153 [49], miR-221 (CA) [50], and miR-485 [51]. P2X7 related miRNAs include upregulated miR-22 [52], and some unclassed miRNAs such as downregulated miR-145-5p (DG) [53], miR-204 (DG), and miR-218 [17]. miRNAs without a label for CA or DG were miRNAs that changed in both regions. miRNAs changed only in the DG and had opposite expression trends compared to the whole hippocampus, which may play a protective role. For example, miR-592 was found to be upregulated in the DG area at days 14 and 60 post-SE in our study, suppressing its target p75NTR BDNF receptor, and inhibiting apoptotic signalling and neuronal death [54]. These miRNAs include miR-324-5p, 135b, 148a, and 221, etc. On the contrary, miRNAs changed only in the DG area and shared the same expression trend as those in the whole hippocampus, such as miR-128, 211, 145-5p, and 204, which may have proepileptic effects. For example, loss of miR-128 in dopamine 1 receptor (Drd1)-positive neurones leads to motor hyperactivity and lethal seizures [30].
Third, there are some miRNAs which have been reported to be related to SE, but for which we did not find statistically significant fold-changes, such as miR-134, 181a, 181a-5p, 181b, 155, 34c-5p, 935, 10a-5p, 142a-5p, 431-5p, 141, 184, 421, 25-3p, and 15a [38,46,48,55].
In the pathway-related analysis, miR-137 and 148-3p are related to the PI3K-Akt-mTOR pathway [34,56]. Mitogen-activated protein kinase 4 (MAP2K4) is a direct target of miR-27a-3p, and it is related to the MAPK signalling pathway [23]. miR-132 and miR-551b-5p are involved in the BDNF/TrkB pathway [24,31]. miR-203 directly suppresses the expression of MEF2C and promotes NF-kB, phosphorylated IkB/IKK, and inflammatory effectors through the MEF2C/NF-kB signalling pathway [57]. miR-19a/b-3p/SIRT1 promotes neuroinflammation by regulating FoxO3/SPHK1 signalling, leading to cell death [32]. miR-146a and 142-3p are involved in the TLR signalling pathway [58,59,60]. miR-672 and 135b-5p are related to the pyroptosis pathway [36,37,61]. Among those, the PI3K-Akt-mTOR and MAPK signalling pathways were validated and changed the miRNAs-related pathway, which was consistent with our bioinformatics analysis.

6. Conclusions

We studied stage and subfield-associated hippocampal miRNA expression patterns after pilocarpine-induced SE and found differences in the CA and DG areas. This is important for understanding the pathogenesis of this disease in these two areas, as the loss of neurones in the DG is less severe than that in the CA. The number of animals should be increased in in-depth studies. Some of our results were consistent with other studies, regardless of the model, and these miRNAs might be more valuable for further studies, specifically miR-146a, miR-132, and miR-137. All of them are candidate therapeutic targets.

Author Contributions

Software, Y.L. (Yue Li); investigation, Y.L. (Yue Li); writing—original draft preparation, Y.L. (Yue Li); writing—review and editing, Y.L. (Yumin Luo) and R.M.; supervision, T.S.W.S. and L.Z.; project administration, S.T.D.; funding acquisition, F.T. and L.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by BMRC Grant 08/1/33/19/570 from A*Star, Singapore, and Capital Science and Technology Leading Talent Training Project (Z191100006119017), China.

Institutional Review Board Statement

The animal study protocol was approved by the Institutional Animal Care and Use Committee of the National University of Singapore (protocol code B27/09&098/09 and date of approval was 22 September 2010).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

We are grateful to Weiping Tang (Cnkingbio Company Lte, Beijing, China) for assistance with bioinformatics.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Blumcke, I.; Thom, M.; Wiestler, O.D. Ammon’s horn sclerosis: A maldevelopmental disorder associated with temporal lobe epilepsy. Brain Pathol. 2002, 12, 199–211. [Google Scholar] [PubMed]
  2. Coulter, D.A.; Yue, C.; Ang, C.W.; Weissinger, F.; Goldberg, E.; Hsu, F.-C.; Carlson, G.C.; Takano, H. Hippocampal microcircuit dynamics probed using optical imaging approaches. J. Physiol. 2011, 589, 1893–1903. [Google Scholar] [CrossRef] [PubMed]
  3. Storm-Mathisen, J.; Ottersen, O.P. Immunocytochemistry of glutamate at the synaptic level. J. Histochem. Cytochem. 1990, 38, 1733–1743. [Google Scholar] [CrossRef] [Green Version]
  4. Caballero-Bleda, M.; Witter, M.P. Projections from the presubiculum and the parasubiculum to morphologically characterized entorhinal-hippocampal projection neurons in the rat. Exp. Brain Res. 1994, 101, 93–108. [Google Scholar] [CrossRef] [PubMed]
  5. Curia, G.; Longo, D.; Biagini, G.; Jones, R.S.; Avoli, M. The pilocarpine model of temporal lobe epilepsy. J. Neurosci. Methods 2008, 172, 143–157. [Google Scholar] [CrossRef]
  6. Kim, H.; Choi, Y.; Joung, H.Y.; Choi, Y.S.; Kim, H.J.; Joo, Y.; Oh, J.H.; Hann, H.J.; Cho, Z.H.; Lee, H.W. Structural and Functional Alterations at Pre-Epileptic Stage Are Closely Associated with Epileptogenesis in Pilocarpine-induced Epilepsy Model. Exp. Neurobiol. 2017, 26, 287–294. [Google Scholar] [CrossRef] [Green Version]
  7. Gage, F.H.; Kempermann, G.; Palmer, T.; Peterson, D.A.; Ray, J. Multipotent progenitor cells in the adult dentate gyrus. J. Neurobiol. 1998, 36, 249–266. [Google Scholar] [CrossRef]
  8. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef] [Green Version]
  9. Ambros, V. The functions of animal microRNAs. Nature 2004, 431, 350–355. [Google Scholar] [CrossRef]
  10. Henshall, D.C. Antagomirs and microRNA in status epilepticus. Epilepsia 2013, 54 (Suppl. 6), 17–19. [Google Scholar] [CrossRef]
  11. Matos, G.; Scorza, F.A.; Mazzotti, D.R.; Guindalini, C.; Cavalheiro, E.A.; Tufik, S.; Andersen, M.L. The effects of sleep deprivation on microRNA expression in rats submitted to pilocarpine-induced status epilepticus. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2014, 51, 159–165. [Google Scholar] [CrossRef]
  12. Hu, K.; Xie, Y.-Y.; Zhang, C.; Ouyang, D.-S.; Long, H.-Y.; Sun, D.-N.; Long, L.-L.; Feng, L.; Li, Y.; Xiao, B. MicroRNA expression profile of the hippocampus in a rat model of temporal lobe epilepsy and miR-34a-targeted neuroprotection against hippocampal neurone cell apoptosis post-status epilepticus. BMC Neurosci. 2012, 13, 115. [Google Scholar] [CrossRef] [Green Version]
  13. Gorter, J.A.; Iyer, A.; White, I.; Colzi, A.; van Vliet, E.A.; Sisodiya, S.; Aronica, E. Hippocampal subregion-specific microRNA expression during epileptogenesis in experimental temporal lobe epilepsy. Neurobiol. Dis. 2014, 62, 508–520. [Google Scholar] [CrossRef]
  14. Gstir, R.; Schafferer, S.; Scheideler, M.; Misslinger, M.; Griehl, M.; Daschil, N.; Humpel, C.; Obermair, G.J.; Schmuckermair, C.; Striessnig, J.; et al. Generation of a neuro-specific microarray reveals novel differentially expressed noncoding RNAs in mouse models for neurodegenerative diseases. RNA 2014, 20, 1929–1943. [Google Scholar] [CrossRef] [Green Version]
  15. Chen, C.-L.; Liu, H.; Guan, X. Changes in microRNA expression profile in hippocampus during the acquisition and extinction of cocaine-induced conditioned place preference in rats. J. Biomed. Sci. 2013, 20, 96. [Google Scholar] [CrossRef] [Green Version]
  16. Risbud, R.M.; Porter, B.E. Changes in MicroRNA Expression in the Whole Hippocampus and Hippocampal Synaptoneurosome Fraction following Pilocarpine Induced Status Epilepticus. PLoS ONE 2013, 8, e53464. [Google Scholar] [CrossRef] [Green Version]
  17. Kaalund, S.S.; Venø, M.T.; Bak, M.; Møller, R.S.; Laursen, H.; Madsen, F.; Broholm, H.; Quistorff, B.; Uldall, P.; Tommerup, N.; et al. Aberrant expression of miR-218 and miR-204 in human mesial temporal lobe epilepsy and hippocampal sclerosis-Convergence on axonal guidance. Epilepsia 2014, 55, 2017–2027. [Google Scholar] [CrossRef]
  18. Hagihara, H.; Toyama, K.; Yamasaki, N.; Miyakawa, T. Dissection of Hippocampal Dentate Gyrus from Adult Mouse. J. Vis. Exp. 2009, 33, e1543. [Google Scholar] [CrossRef] [Green Version]
  19. Bak, M.; Silahtaroglu, A.; Møller, M.; Christensen, M.; Rath, M.F.; Skryabin, B.; Tommerup, N.; Kauppinen, S. MicroRNA expression in the adult mouse central nervous system. RNA 2008, 14, 432–444. [Google Scholar] [CrossRef] [Green Version]
  20. Olsen, L.; Klausen, M.; Helboe, L.; Nielsen, F.C.; Werge, T. MicroRNAs Show Mutually Exclusive Expression Patterns in the Brain of Adult Male Rats. PLoS ONE 2009, 4, e7225. [Google Scholar] [CrossRef]
  21. Brennan, G.P.; Henshall, D.C. microRNAs in the pathophysiology of epilepsy. Neurosci. Lett. 2018, 667, 47–52. [Google Scholar] [CrossRef] [PubMed]
  22. Korotkov, A.; Mills, J.D.; Gorter, J.A.; van Vliet, E.; Aronica, E. Systematic review and meta-analysis of differentially expressed miRNAs in experimental and human temporal lobe epilepsy. Sci. Rep. 2017, 7, 11592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Lu, J.; Zhou, N.; Yang, P.; Deng, L.; Liu, G. MicroRNA-27a-3p Downregulation Inhibits Inflammatory Response and Hippocampal Neuronal Cell Apoptosis by Upregulating Mitogen-Activated Protein Kinase 4 (MAP2K4) Expression in Epilepsy: In Vivo and In Vitro Studies. Med Sci. Monit. 2019, 25, 8499–8508. [Google Scholar] [CrossRef]
  24. Xiang, L.; Ren, Y.; Cai, H.; Zhao, W.; Song, Y. MicroRNA-132 aggravates epileptiform discharges via suppression of BDNF/TrkB signaling in cultured hippocampal neurons. Brain Res. 2015, 1622, 484–495. [Google Scholar] [CrossRef]
  25. Korotkov, A.; Broekaart, D.W.M.; Banchaewa, L.; Pustjens, B.; Scheppingen, J.; Anink, J.J.; Baayen, J.C.; Idema, S.; Gorter, J.A.; Vliet, E.A.; et al. microRNA-132 is overexpressed in glia in temporal lobe epilepsy and reduces the expression of pro-epileptogenic factors in human cultured astrocytes. Glia 2019, 68, 60–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. 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 2021, 13, 941–949. [Google Scholar] [CrossRef]
  27. Lee, S.T.; Jeon, D.; Chu, K.; Jung, K.H.; Moon, J.; Sunwoo, J.; Park, D.K.; Yang, H.; Park, J.H.; Kim, M.; et al. Inhibition of miR-203 Reduces Spontaneous Recurrent Seizures in Mice. Mol. Neurobiol. 2017, 54, 3300–3308. [Google Scholar] [CrossRef]
  28. Aronica, E.; Fluiter, K.; Iyer, A.; Zurolo, E.; Vreijling, J.; Van Vliet, E.A.; Baayen, J.C.; Gorter, J.A. Expression pattern of miR-146a, an inflammation-associated microRNA, in experimental and human temporal lobe epilepsy. Eur. J. Neurosci. 2010, 31, 1100–1107. [Google Scholar] [CrossRef]
  29. Baloun, J.; Bencurova, P.; Totkova, T.; Kubova, H.; Hermanova, M.; Hendrych, M.; Pail, M.; Pospisilova, S.; Brazdil, M. Epilepsy miRNA Profile Depends on the Age of Onset in Humans and Rats. Front. Neurosci. 2020, 14, 924. [Google Scholar] [CrossRef]
  30. Thomas, K.T.; Gross, C.; Bassell, G.J. microRNAs Sculpt Neuronal Communication in a Tight Balance That Is Lost in Neurological Disease. Front. Mol. Neurosci. 2018, 11, 455. [Google Scholar] [CrossRef]
  31. Wang, L.; Zhou, Y.; Chen, X.; Liu, J.; Qin, X. Long-term iTBS promotes neural structural and functional recovery by enhancing neurogenesis and migration via miR-551b-5p/BDNF/TrkB pathway in a rat model of cerebral ischemia-reperfusion injury. Brain Res. Bull. 2022, 184, 46–55. [Google Scholar] [CrossRef]
  32. Zhou, F.; Wang, Y.K.; Zhang, C.G.; Wu, B.Y. miR-19a/b-3p promotes inflammation during cerebral ischemia/reperfusion injury via SIRT1/FoxO3/SPHK1 pathway. J. Neuroinflamm. 2021, 18, 122. [Google Scholar] [CrossRef]
  33. Chang, H.-L.; Wang, H.-C.; Chunag, Y.-T.; Chou, C.-W.; Lin, I.-L.; Lai, C.-S.; Chang, L.-L.; Cheng, K.-I. miRNA Expression Change in Dorsal Root Ganglia After Peripheral Nerve Injury. J. Mol. Neurosci. 2016, 61, 169–177. [Google Scholar] [CrossRef]
  34. Xu, J.; Sun, M.; Li, X.; Huang, L.; Gao, Z.; Gao, J.; Xie, A. MicroRNA expression profiling after recurrent febrile seizures in rat and emerging role of miR-148a-3p/SYNJ1 axis. Sci. Rep. 2021, 11, 1262. [Google Scholar] [CrossRef]
  35. Yu, Y.; Du, L.; Zhang, J. Febrile Seizure-Related miR-148a-3p Exerts Neuroprotection by Promoting the Proliferation of Hippocampal Neurons in Children with Temporal Lobe Epilepsy. Dev. Neurosci. 2021, 43, 312–320. [Google Scholar] [CrossRef]
  36. Li, R.; Hu, J.; Cao, S. The Clinical Significance of miR-135b-5p and Its Role in the Proliferation and Apoptosis of Hippocampus Neurons in Children with Temporal Lobe Epilepsy. Dev. Neurosci. 2020, 42, 187–194. [Google Scholar] [CrossRef] [PubMed]
  37. Chen, S.; Luo, Z.; Chen, X. Hsa_circ_0044235 regulates the pyroptosis of rheumatoid arthritis via MiR-135b-5p-SIRT1 axis. Cell Cycle 2021, 20, 1107–1121. [Google Scholar] [CrossRef] [PubMed]
  38. Feng, Y.; Yang, H.; Yue, Y.; Tian, F. MicroRNAs and target genes in epileptogenesis. Epilepsia 2020, 61, 2086–2096. [Google Scholar] [CrossRef]
  39. Roncon, P.; Soukupovà, M.; Binaschi, A.; Falcicchia, C.; Zucchini, S.; Ferracin, M.; Langley, S.R.; Petretto, E.; Johnson, M.R.; Marucci, G.; et al. MicroRNA profiles in hippocampal granule cells and plasma of rats with pilocarpine-induced epilepsy—Comparison with human epileptic samples. Sci. Rep. 2015, 5, 14143. [Google Scholar] [CrossRef] [Green Version]
  40. Bekenstein, U.; Mishra, N.; Milikovsky, D.Z.; Hanin, G.; Zelig, D.; Sheintuch, L.; Berson, A.; Greenberg, D.S.; Friedman, A.; Soreq, H. Dynamic changes in murine forebrain miR-211 expression associate with cholinergic imbalances and epileptiform activity. Proc. Natl. Acad. Sci. USA 2017, 114, E4996–E5005. [Google Scholar] [CrossRef] [PubMed]
  41. Tan, C.L.; Plotkin, J.L.; Venø, M.T.; von Schimmelmann, M.; Feinberg, P.; Mann, S.; Handler, A.; Kjems, J.; Surmeier, D.J.; O’Carroll, D.; et al. MicroRNA-128 Governs Neuronal Excitability and Motor Behavior in Mice. Science 2013, 342, 1254–1258. [Google Scholar] [CrossRef] [Green Version]
  42. Brennan, G.P.; Dey, D.; Chen, Y.; Patterson, K.P.; Magnetta, E.J.; Hall, A.M.; Dube, C.M.; Mei, Y.-T.; Baram, T.Z. Dual and Opposing Roles of MicroRNA-124 in Epilepsy Are Mediated through Inflammatory and NRSF-Dependent Gene Networks. Cell Rep. 2016, 14, 2402–2412. [Google Scholar] [CrossRef] [Green Version]
  43. Peng, J.; Omran, A.; Ashhab, M.U.; Kong, H.; Gan, N.; He, F.; Yin, F. Expression Patterns of miR-124, miR-134, miR-132, and miR-21 in an Immature Rat Model and Children with Mesial Temporal Lobe Epilepsy. J. Mol. Neurosci. 2013, 50, 291–297. [Google Scholar] [CrossRef]
  44. Sano, T.; Reynolds, J.P.; Jiménez-Mateos, E.; Matsushima, S.; Taki, W.; Henshall, D.C. MicroRNA-34a upregulation during seizure-induced neuronal death. Cell Death Dis. 2012, 3, e287. [Google Scholar] [CrossRef] [Green Version]
  45. Huang, L.-G.; Luo, Y.-H.; Xu, J.-W.; Lu, Q.-C. Plasma Exosomal MiRNAs Expression Profile in Mesial Temporal Lobe Epilepsy With Hippocampal Sclerosis: Case-Control Study and Analysis of Potential Functions. Front. Mol. Neurosci. 2020, 13, 584828. [Google Scholar] [CrossRef]
  46. Ghafouri-Fard, S.; Hussen, B.M.; Abak, A.; Taheri, M.; Khoshnoud, R.J. Aberrant expression of miRNAs in epilepsy. Mol. Biol. Rep. 2022, 49, 5057–5074. [Google Scholar] [CrossRef]
  47. Gross, C.; Yao, X.; Engel, T.; Tiwari, D.; Xing, L.; Rowley, S.; Danielson, S.W.; Thomas, K.T.; Jimenez-Mateos, E.M.; Schroeder, L.M.; et al. MicroRNA-Mediated Downregulation of the Potassium Channel Kv4.2 Contributes to Seizure Onset. Cell Rep. 2016, 17, 37–45. [Google Scholar] [CrossRef] [Green Version]
  48. 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]
  49. Li, Y.; Huang, C.; Feng, P.; Jiang, Y.; Wang, W.; Zhou, N.; 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] [Green Version]
  50. Fu, M.; Zhu, Y.; Zhang, J.; Wu, W.; Sun, Y.; Zhang, X.; Tao, J.; Li, Z. MicroRNA-221-3p Suppresses the Microglia Activation and Seizures by Inhibiting of HIF-1α in Valproic Acid-Resistant Epilepsy. Front. Pharmacol. 2021, 12, 714556. [Google Scholar] [CrossRef]
  51. Pan, W.; Song, X.; Hu, Q.; Zhang, Y. miR-485 inhibits histone deacetylase HDAC5, HIF1α and PFKFB3 expression to alleviate epilepsy in cellular and rodent models. Aging 2021, 13, 14416–14432. [Google Scholar] [CrossRef] [PubMed]
  52. Beamer, E.; Jurado-Arjona, J.; Jiménez-Mateos, E.; Morgan, J.; Reschke, C.R.; Kenny, A.; de Leo, G.; Olivos-Oré, L.A.; Arribas-Blázquez, M.; Madden, S.F.; et al. MicroRNA-22 Controls Aberrant Neurogenesis and Changes in Neuronal Morphology After Status Epilepticus. Front. Mol. Neurosci. 2018, 11, 442 2. [Google Scholar] [CrossRef] [PubMed]
  53. 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]
  54. Irmady, K.; Jackman, K.A.; Padow, V.A.; Shahani, N.; Martin, L.A.; Cerchietti, L.; Unsicker, K.; Iadecola, C.; Hempstead, B.L. Mir-592 regulates the induction and cell death-promoting activity of p75NTR in neuronal ischemic injury. J. Neurosci. 2014, 34, 3419–3428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Venø, M.T.; Reschke, C.R.; Morris, G.; Connolly, N.M.; Su, J.; Yan, Y.; Engel, T.; Jimenez-Mateos, E.M.; Harder, L.M.; Pultz, D.; et al. A systems approach delivers a functional microRNA catalog and expanded targets for seizure suppression in temporal lobe epilepsy. Proc. Natl. Acad. Sci. USA 2020, 117, 15977–15988. [Google Scholar] [CrossRef]
  56. Thomas, K.T.; Anderson, B.R.; Shah, N.; Zimmer, S.E.; Hawkins, D.; Valdez, A.N.; Gu, Q.; Bassell, G.J. Inhibition of the Schizophrenia-Associated MicroRNA miR-137 Disrupts Nrg1α Neurodevelopmental Signal Transduction. Cell Rep. 2017, 20, 1–12. [Google Scholar] [CrossRef] [Green Version]
  57. Yu, Q.; Zhao, M.; Yang, P. LncRNA UCA1 Suppresses the Inflammation Via Modulating miR-203-Mediated Regulation of MEF2C/NF-κB Signaling Pathway in Epilepsy. Neurochem. Res. 2020, 45, 783–795. [Google Scholar] [CrossRef]
  58. Taganov, K.D.; Boldin, M.P.; Chang, K.; Baltimore, D. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc. Natl. Acad. Sci. USA 2006, 103, 12481–12486. [Google Scholar] [CrossRef] [Green Version]
  59. Hou, J.; Wang, P.; Lin, L.; Liu, X.; Ma, F.; An, H.; Wang, Z.; Cao, X. MicroRNA-146a Feedback Inhibits RIG-I-Dependent Type I IFN Production in Macrophages by Targeting TRAF6, IRAK1, and IRAK2. J. Immunol. 2009, 183, 2150–2158. [Google Scholar] [CrossRef] [Green Version]
  60. Cukovic, D.; Bagla, S.; Ukasik, D.; Stemmer, P.; Jena, B.; Naik, A.; Sood, S.; Asano, E.; Luat, A.; Chugani, D.; et al. Exosomes in Epilepsy of Tuberous Sclerosis Complex: Carriers of Pro-Inflammatory MicroRNAs. Non-Coding RNA 2021, 7, 40. [Google Scholar] [CrossRef]
  61. Zhou, Z.; Li, C.; Bao, T.; Zhao, X.; Xiong, W.; Luo, C.; Yin, G.; Fan, J. Exosome-Shuttled miR-672-5p from Anti-Inflammatory Microglia Repair Traumatic Spinal Cord Injury by Inhibiting AIM2/ASC/Caspase-1 Signaling Pathway Mediated Neuronal Pyroptosis. J. Neurotrauma 2022, 39, 1057–1074. [Google Scholar] [CrossRef]
Figure 1. Differently expressed miRNAs in the CA (A) and DG (B) areas over three time points. The fold-change value on day 14 post-SE was used as the ordinate and the fold-change value on day 60 post-SE as the abscissa. The colour gradient (from green to red) and the size of dots were used to represent the fold-change value on day 3 post-SE. miR-494 and 188-5p were excluded according to the validation results. Tableau software was used to map the changes in the expression of different miRNAs. Note: the * in the figure like miR-29a* has no other meaning but the name of a specific miRNA.
Figure 1. Differently expressed miRNAs in the CA (A) and DG (B) areas over three time points. The fold-change value on day 14 post-SE was used as the ordinate and the fold-change value on day 60 post-SE as the abscissa. The colour gradient (from green to red) and the size of dots were used to represent the fold-change value on day 3 post-SE. miR-494 and 188-5p were excluded according to the validation results. Tableau software was used to map the changes in the expression of different miRNAs. Note: the * in the figure like miR-29a* has no other meaning but the name of a specific miRNA.
Biomedicines 10 03012 g001
Figure 2. CA (AC) and DG (DF) area of significant gene expression trends. The abscissa represents different time points, the ordinate represents the grading of gene changes, and each line represents a different miRNA. It can be concluded that trends of No.6 (A/D), 9 (B/E) and 19 (C/F) have significant differences in both CA and DG areas among the 26 trends generated.
Figure 2. CA (AC) and DG (DF) area of significant gene expression trends. The abscissa represents different time points, the ordinate represents the grading of gene changes, and each line represents a different miRNA. It can be concluded that trends of No.6 (A/D), 9 (B/E) and 19 (C/F) have significant differences in both CA and DG areas among the 26 trends generated.
Biomedicines 10 03012 g002
Table 1. Validation of microarray results in the CA area by qRT-PCR.
Table 1. Validation of microarray results in the CA area by qRT-PCR.
Primer NameGroupqRT-PCR Fold-ChangeMicroarray Fold-ChangeFP
miR-124Control1 ± 0.2511.6930.245
Day 30.64 ± 0.2−1.745579
Day 140.89 ± 0.3−1.1375024
Day 600.68 ± 0.12−1.1038424
miR-137Control1 ± 0.1512.1540.172
Day 30.62 ± 0.23 *−2.0571318
Day 140.78 ± 0.1−1.3803179
Day 600.75 ± 0.23−1.619207
miR-142-3pControl1 ± 0.11114.2890.001
Day 35.32 ± 0.81 *5.619121
Day 143.42 ± 1.64 *#2.8325222
Day 601.34 ± 0.19 #1.7457325
miR-19aControl1 ± 0.34125.3050.000
Day 32.87 ± 0.19 *2.878858
Day 141.17 ± 0.52 #1.1613489
Day 600.71 ± 0.17 #−1.1601521
miR-203Control1 ± 0.3618.605 0.007
Day 34.5 ± 1.14 *4.5549407
Day 143.44 ± 0.77 *3.667932
Day 601.89 ± 1.18 #2.421915
miR-27a Control1 ± 0.1612.4770.136
Day 31.39 ± 0.141.7447702
Day 141.57 ± 0.41 *1.6940101
Day 601.32 ± 0.251.7312951
miR-494Control1 ± 0.6210.2720.844
Day 31.02 ± 0.357.6855636
Day 141.05 ± 0.582.0513108
Day 600.73 ± 0.32−1.0420982
miR-551bControl1 ± 0.2112.8480.105
Day 30.59 ± 0.28−1.9337255
Day 140.53 ± 0.14 *−1.7970246
Day 600.54 ± 0.27 *−1.3070583
miR-146aControl1 ± 0.3412.6600.119
Day 32.99 ± 0.863.089705
Day 144.86 ± 3.23 *4.8941493
Day 601.69 ± 1.312.211484
miR-188-5pControl1 ± 0.116.1290.018
Day 31.23 ± 0.18333.90417
Day 140.89 ± 0.22 #22.7263
Day 600.69 ± 0.09 *#26.388815
miR-193Control1 ± 0.2610.0730.973
Day 31.02 ± 0.382.0996573
Day 141.13 ± 0.441.5109724
Day 601.09 ± 0.431.6478502
Note: * indicates comparison to control, p < 0.05; # indicates comparison to day 3 post-SE, p < 0.05.
Table 2. Validation of microarray results in the DG area by qRT-PCR.
Table 2. Validation of microarray results in the DG area by qRT-PCR.
Primer NameGroupqRT-PCR Fold-ChangeMicroarray Fold-ChangeFP
miR-124Control1 ± 0.1212.2250.163
Day 30.55 ± 0.2−1.5798804
Day 140.81 ± 0.16−1.3798828
Day 600.58 ± 0.4−1.1080296
miR-137Control1 ± 0.2510.6040.630
Day 30.77 ± 0.08−1.832393
Day 140.88 ± 0.22−1.588261
Day 601.03 ± 0.41−1.2935005
miR-142-3pControl1 ± 0.07119.7600.000
Day 36.4 ± 1.82 *4.912
Day 142.75 ± 0.9 #2.225
Day 600.72 ± 0.21 #,^1.392988
miR-19aControl1 ± 0.3515.5420.024
Day 32.61 ± 1.08 *2.1299305
Day 141.23 ± 0.48 #−1.2170526
Day 600.54 ± 0.43 #−1.7115029
miR-203Control1 ± 0.6712.7450.113
Day 37.57 ± 5.08 *5.519774
Day 143.82 ± 1.323.3713422
Day 602.43 ± 2.62.5886562
miR-27a Control1 ± 0.3112.3850.145
Day 31.6 ± 0.281.5262655
Day 141.61 ± 0.481.5529257
Day 600.96 ± 0.51.6639766
miR-494Control1 ± 0.2810.7640.545
Day 31.28 ± 0.434.505004
Day 141.51 ± 0.431.3652874
Day 601.17 ± 0.52−1.180082
miR-551bControl1 ± 0.1112.9880.002
Day 30.63 ± 0.23 *−1.7511983
Day 140.32 ± 0.04 *,#−3.644983
Day 600.32 ± 0.19 *,#−2.47584
miR-132Control1 ± 0.231
Day 33.38 ± 0.812.63845445.8010.021
Day 146.55 ± 1.34 *5.115643
Day 606.28 ± 3.43 *4.9513855
miR-135bControl1 ± 0.3513.7260.061
Day 31.56 ± 0.321.2437161
Day 144.86 ± 2.21 *,#3.8363545
Day 603.77 ± 2.373.7484105
miR-148aControl1 ± 0.1719.7890.005
Day 30.83 ± 0.36−1.731051
Day 140.34 ± 0.17 *,#−3.1871736
Day 600.17 ± 0.09 *,#−4.944259
miR-188-5pControl1 ± 0.1611.9710.197
Day 31.27 ± 0.0528.367397
Day 140.79 ± 0.273.5954535
Day 600.63 ± 0.62.1866686
miR-672Control1 ± 0.0713.8060.058
Day 30.81 ± 0.26−1.6016815
Day 140.59 ± 0.11 *−1.9541458
Day 600.52 ± 0.2 6 *−1.6448656
Note: * indicates comparison to control, p < 0.05; # indicates comparison to day 3 post-SE, p < 0.05; ^ indicates comparison to day 14 post-SE, p < 0.05.
Table 3. Numbers of predicted intersecting target genes of changed miRNAs.
Table 3. Numbers of predicted intersecting target genes of changed miRNAs.
Changed miRNA in Both the CA and DG AreasPredicted Intersecting Target Gene NumberChanged miRNA in DG AreaPredicted Intersecting Target Gene NumberChanged miRNA in CA AreaPredicted Intersecting Target Gene Number
miR-1241630miR-128828miR-193164
miR-12720miR-132305miR-487b11
miR-129-5p391miR-133b476miR-93932
miR-136170miR-143273
miR-137912miR-145552
miR-139-5p289miR-195940
miR-142-3p252miR-204396
miR-149288miR-211396
miR-150201miR-28192
miR-153599miR-31271
miR-186504miR-33313
miR-190119miR-34684
miR-203608miR-381548
miR-20b935miR-383115
miR-21230miR-384-3p152
miR-21027miR-43187
miR-218757miR-488210
miR-22370miR-497940
miR-221308miR-873178
miR-222308
miR-223220
miR-25725
miR-324-5p85
miR-326268
miR-328123
miR-335-5p175
miR-362-3p225
miR-370245
miR-377364
miR-37967
miR-384-5p1078
miR-410434
miR-411121
miR-433196
miR-485489
miR-494314
miR-49677
miR-874194
miR-91017
Table 4. Different functions regulated by changed miRNAs in CA and DG areas.
Table 4. Different functions regulated by changed miRNAs in CA and DG areas.
GO Name (CA Area)LgPGO Name (DG Area)LgP
myeloid cell differentiation4.79sensory perception of temperature stimulus5.17
positive regulation of cell differentiation4.24head development4.72
macromolecule de-acylation4.10response to metalation4.47
amyloid precursor protein metabolic process4.06modification-dependent macromolecule catabolic process4.26
positive regulation of multi-organism process3.87RNA localisation3.93
prostanoid metabolic process4.03stem cell division4.09
membrane biogenesis4.06ossification4.07
cellular protein complex assembly3.82adrenergic receptor signalling pathway3.88
ganglion development3.69cell aggregation3.74
membrane docking3.55detection of biotic stimulus3.72
response to peptide3.55developmental growth involved in morphogenesis3.70
suckling behaviour3.82cell-cell adhesion via plasma-membrane adhesion molecules3.58
Table 5. Different pathways regulated by the changed miRNAs in CA and DG areas.
Table 5. Different pathways regulated by the changed miRNAs in CA and DG areas.
CA Area-Specific PathwaysLgPDG Area-Specific PathwaysLgP
Ras signalling pathway10Hippo signalling pathway10
Hedgehog signalling pathway4.60PI3K-Akt signalling pathway10
Glycosaminoglycan biosynthesis—heparan sulphate/heparin3.95Tight junction4.22
Synaptic vesicle cycle3.77Cholinergic synapse3.98
Adipocytokine signalling pathway3.61N-Glycan biosynthesis3.27
Chemokine signalling pathway3.32Glycosaminoglycan biosynthesis—keratan sulphate2.51
B cell receptor signalling pathway3.14Renin secretion2.44
ECM-receptor interaction2.81
Jak-STAT signalling pathway2.43
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Li, Y.; Dheen, S.T.; Tang, F.; Luo, Y.; Meng, R.; Samuel, T.S.W.; Zhang, L. Stage- and Subfield-Associated Hippocampal miRNA Expression Patterns after Pilocarpine-Induced Status Epilepticus. Biomedicines 2022, 10, 3012. https://doi.org/10.3390/biomedicines10123012

AMA Style

Li Y, Dheen ST, Tang F, Luo Y, Meng R, Samuel TSW, Zhang L. Stage- and Subfield-Associated Hippocampal miRNA Expression Patterns after Pilocarpine-Induced Status Epilepticus. Biomedicines. 2022; 10(12):3012. https://doi.org/10.3390/biomedicines10123012

Chicago/Turabian Style

Li, Yue, S Thameem Dheen, Fengru Tang, Yumin Luo, Ran Meng, Tay Sam Wah Samuel, and Lan Zhang. 2022. "Stage- and Subfield-Associated Hippocampal miRNA Expression Patterns after Pilocarpine-Induced Status Epilepticus" Biomedicines 10, no. 12: 3012. https://doi.org/10.3390/biomedicines10123012

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