Loading [Contrib]/a11y/accessibility-menu.js
Research article

Key protein-coding genes related to microglia in immune regulation and inflammatory response induced by epilepsy


  • Received: 16 August 2021 Accepted: 24 October 2021 Published: 02 November 2021
  • Several studies have shown a link between immunity, inflammatory processes, and epilepsy. Active neuroinflammation and marked immune cell infiltration occur in epilepsy of diverse etiologies. Microglia, as the first line of defense in the central nervous system, are the main effectors of neuroinflammatory processes. Discovery of new biomarkers associated with microglia activation after epileptogenesis indicates that targeting specific molecules may help control seizures. In this research, we used a combination of several bioinformatics approaches, including RNA sequencing, to explore differentially expressed genes (DEGs) in epileptic lesions and control samples, and to construct a protein-protein interaction (PPI) network for DEGs, which was examined utilizing plug-ins in Cytoscape software. Finally, we aimed to identify 10 hub genes in immune and inflammation-related sub-networks, which were subsequently validated in real-time quantitative polymerase chain reaction analysis in a mouse model of kainic acid-induced epilepsy. The expression patterns of nine genes were consistent with sequencing outcomes. Meanwhile, several genes, including CX3CR1, CX3CL1, GPR183, FPR1, P2RY13, P2RY12 and LPAR5, were associated with microglial activation and migration, providing novel candidate targets for immunotherapy in epilepsy and laying the foundation for further research.

    Citation: Jing Cao, Hui Gan, Han Xiao, Hui Chen, Dan Jian, Ning Jiang, Xuan Zhai. Key protein-coding genes related to microglia in immune regulation and inflammatory response induced by epilepsy[J]. Mathematical Biosciences and Engineering, 2021, 18(6): 9563-9578. doi: 10.3934/mbe.2021469

    Related Papers:

    [1] Yuru Han, Shuo Shi, Shuang Liu, Xuefeng Gu . Effects of spaceflight on the spleen and thymus of mice: Gene pathway analysis and immune infiltration analysis. Mathematical Biosciences and Engineering, 2023, 20(5): 8531-8545. doi: 10.3934/mbe.2023374
    [2] Ning Li, Jining Zhou . Screening and validating the immune-related gene expression signatures in peripheral blood mononuclear cells of nonischaemic cardiomyopathy. Mathematical Biosciences and Engineering, 2020, 17(3): 2330-2347. doi: 10.3934/mbe.2020124
    [3] Jie Wang, Md. Nazim Uddin, Rehana Akter, Yun Wu . Contribution of endothelial cell-derived transcriptomes to the colon cancer based on bioinformatics analysis. Mathematical Biosciences and Engineering, 2021, 18(6): 7280-7300. doi: 10.3934/mbe.2021360
    [4] Sufang Wu, Hua He, Jingjing Huang, Shiyao Jiang, Xiyun Deng, Jun Huang, Yuanbing Chen, Yiqun Jiang . FMR1 is identified as an immune-related novel prognostic biomarker for renal clear cell carcinoma: A bioinformatics analysis of TAZ/YAP. Mathematical Biosciences and Engineering, 2022, 19(9): 9295-9320. doi: 10.3934/mbe.2022432
    [5] Ming-Xi Zhu, Tian-Yang Zhao, Yan Li . Insight into the mechanism of DNA methylation and miRNA-mRNA regulatory network in ischemic stroke. Mathematical Biosciences and Engineering, 2023, 20(6): 10264-10283. doi: 10.3934/mbe.2023450
    [6] Xiaowei Zhang, Jiayu Tan, Xinyu Zhang, Kritika Pandey, Yuqing Zhong, Guitao Wu, Kejun He . Aggrephagy-related gene signature correlates with survival and tumor-associated macrophages in glioma: Insights from single-cell and bulk RNA sequencing. Mathematical Biosciences and Engineering, 2024, 21(2): 2407-2431. doi: 10.3934/mbe.2024106
    [7] Rui-zhe Zheng, Jin Xing, Qiong Huang, Xi-tao Yang, Chang-yi Zhao, Xin-yuan Li . Integration of single-cell and bulk RNA sequencing data reveals key cell types and regulators in traumatic brain injury. Mathematical Biosciences and Engineering, 2021, 18(2): 1201-1214. doi: 10.3934/mbe.2021065
    [8] Li-Sha Pan, Zacharia Ackbarkha, Jing Zeng, Min-Li Huang, Zhen Yang, Hao Liang . Immune marker signature helps to predict survival in uveal melanoma. Mathematical Biosciences and Engineering, 2021, 18(4): 4055-4070. doi: 10.3934/mbe.2021203
    [9] Xin Lin, Xingyuan Li, Binqiang Ma, Lihua Hang . Identification of novel immunomodulators in lung squamous cell carcinoma based on transcriptomic data. Mathematical Biosciences and Engineering, 2022, 19(2): 1843-1860. doi: 10.3934/mbe.2022086
    [10] Qian Li, Minawaer Hujiaaihemaiti, Jie Wang, Md. Nazim Uddin, Ming-Yuan Li, Alidan Aierken, Yun Wu . Identifying key transcription factors and miRNAs coregulatory networks associated with immune infiltrations and drug interactions in idiopathic pulmonary arterial hypertension. Mathematical Biosciences and Engineering, 2023, 20(2): 4153-4177. doi: 10.3934/mbe.2023194
  • Several studies have shown a link between immunity, inflammatory processes, and epilepsy. Active neuroinflammation and marked immune cell infiltration occur in epilepsy of diverse etiologies. Microglia, as the first line of defense in the central nervous system, are the main effectors of neuroinflammatory processes. Discovery of new biomarkers associated with microglia activation after epileptogenesis indicates that targeting specific molecules may help control seizures. In this research, we used a combination of several bioinformatics approaches, including RNA sequencing, to explore differentially expressed genes (DEGs) in epileptic lesions and control samples, and to construct a protein-protein interaction (PPI) network for DEGs, which was examined utilizing plug-ins in Cytoscape software. Finally, we aimed to identify 10 hub genes in immune and inflammation-related sub-networks, which were subsequently validated in real-time quantitative polymerase chain reaction analysis in a mouse model of kainic acid-induced epilepsy. The expression patterns of nine genes were consistent with sequencing outcomes. Meanwhile, several genes, including CX3CR1, CX3CL1, GPR183, FPR1, P2RY13, P2RY12 and LPAR5, were associated with microglial activation and migration, providing novel candidate targets for immunotherapy in epilepsy and laying the foundation for further research.



    Epilepsy is a chronic neurological disorder that directly affects an estimated 68 million people worldwide [1]. Current clinical treatments manly involve the use of antiepileptic drugs and/or surgical resection of the seizure focus, whereas, they are associated with multiple adverse effects, and few patients are seizure-free after surgery. Thus, the intractability of epilepsy has prompted researchers to maintain a sustained interest in its biological mechanisms and the development of new clinical therapeutic targets.

    The different roles of inflammatory responses and immune signals in the development of epilepsy have been highlighted in several studies. Brain injuries associated with central nervous system infections may lead to early occurrence of seizures, which may result in epilepsy [2]. Concurrently, seizures trigger inflammatory responses. Several inflammatory factors, including IL-1β, IL-8, IL-12p70 and MIP-1β, have been detected in surgically resected epileptogenic cortex obtained from children with refractory epilepsy [3]. Animal studies have also found increased levels of pro-inflammatory cytokines in animal models of epilepsy and that seizure frequency correlates with the activation of these cytokines [46]. Thus, a number of small-molecule compounds targeting inflammatory signaling pathways, especially for COX-2/PGE2/EP2 axis, have been extensively studied in different animal models of epilepsy. Treatment with EP2 antagonists after seizures showed overall significant anti-inflammatory and neuroprotective effects in three rodent models of status epilepticus (SE) induced by pilocarpine, kainic acid, and diisopropyl fluorophosphate, respectively, resulting in improved behavioral and functional deficits caused by prolonged seizures [7]. Alternatively, Studies on the use of various COX-2 inhibitors in SE animal models have produced controversial results duo to they inhibit some beneficial effects that might be mediated by other COX-2-derived prostanoids, suggesting a possible consequence of COX-2 induction - early neuroprotection and later neurotoxicity [8].

    Lately, growing studies have shown that cytokine release is profoundly dependent on microglia activation, which is prevalent in human epilepsy and animal models, though their phenotypes have exhibited heterogeneity. Boer et al. studied the distribution of microglia in 20 specimens of focal cortical dysplasia(a major cause of intractable epilepsy) by immunocytochemistry and found a specific and sustained increase in the numerical density of HLA-DR-positive activated microglia in areas of abnormal development, significantly higher than in normal-appearing control cortex obtained at autopsy [9]. Benson et al. detected the changes of microglial pro-inflammatory M1 and anti-inflammatory M2 marker expression in different stages of different mouse models. M1 and M2 markers showed increased expression 3 days but not 21 days after pilocarpine-induced status epilepticus. Though, In the late chronic phase, some M1/M2 markers, IL-1β, TNFα, Arg1, Ym1, and CD206, resurged in the kainate, but not pilocarpine model, indicating a complex role of microglia in the epileptic brain [10]. Accordingly, discovering new biomarkers of microglia activation is essential for further studies on its relationship with neuroinflammation and epileptogenes.

    Given the popularity of high-throughput microarray and RNA-sequencing (RNA-Seq) analyses as research tools, the use of bioinformatics methods to improve the understanding of transcriptome changes in the onset and development of epilepsy has increased. Kalozoumi et al. reported that glial-mediated inflammatory responses play a key role during epileptogenesis in a model of acute epilepsy induced by kainic acid (KA), with hippocampal genome-wide expression analysis at 6, 12 and 24h post-injection [11]. Luo et al. explored the expression and correlation of long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) in a rat model of temporal lobe epilepsy (TLE). Results showed that 54 lncRNA, 36 miRNA, and 122 mRNA molecules that were dysregulated in experimental groups compared to normal controls [12]. Recently, Chen et al. performed a comprehensive analysis of six TLE microarray datasets using robust rank aggregation to identify co-expressed key genes across different epilepsy models and platforms. These authors have shown that the differentially-expressed genes (DEGs) in the acute, latent, and chronic phases of TLE were significantly enriched in inflammatory response, while the pivotal genes identified in each stage were associated with different biological processes [13]. However, these results were based on data from sequencing analysis of animal model samples, which do not correspond completely to the changes in transcriptome signature during human epileptogenesis. Identification of critical protein-protein interaction (PPI) modules and key regulatory genes within them is crucial for elucidating the pathogenesis of epilepsy.

    In the present study, we collected brain tissues from two neurologically healthy controls and five epileptic patients, screened the DEGs in both groups, and applied bioinformatics methods to identify the most important biological process and hub genes associated with microglia. In addition, real-time quantitative polymerase chain reaction (RT-qPCR) was used to validate the expression pattern of several hub genes in a mouse model of epilepsy.

    Brain tissue was isolated within 12 hours after death from two subjects without a history or signs of neurological or neuropsychiatric illness, and provided by the Department of Forensic Medicine, Chongqing Medical University. Surgical specimens were obtained from five secondary epileptic patients caused by brain maldevelopment who had undergone surgical resection at the Department of Neurosurgery, Children's Hospital of Chongqing Medical University. Details related to sample collection, transportation and preservation are described in the Supplementary Materials. Clinical characteristics of the patients and controls are summarized in Table 1. For more information, see Supplementary Materials Table S1. The Ethics Committee of the Children's Hospital Affiliated to Chongqing Medical University approved the present study protocol involving the use of human tissue. Written informed consent was signed by the participants or their legal representatives prior to the start of the project.

    Table 1.  Characteristics of controls and epileptic patients.
    Identifier Gender Age (years) Cause of death
    C1 Female 71 Renal failure
    C2 Male 38 Car accident
    Identifier Gender Age (years) Surgical approach
    E1 Male 4 Resection of frontal lobe lesions
    E2 Male 9 Resection of frontal lobe lesions
    E3 Male 5 Resection of parietal lobe lesions
    E4 Male 2 Resection of temporal lobe epilepsy lesions
    E5 Male 11 Resection of temporal lobe epilepsy lesions

     | Show Table
    DownLoad: CSV

    Adult male C57BL/6J mice were provided by the Experimental Animal Center of Chongqing Medical University and housed in specific pathogen free (SPF) environment. The KA-induced chronic epileptic model was established, as previously described [14]. Briefly, deeply anesthetized mice were injected with 1.0 nmol of KA (Sigma Aldrich, St. Louis, USA) dissolved in 50 nL saline into the right dorsal hippocampus (anteroposterior = -1.8 mm, mediolateral = -1.8 mm, dorsoventral = -1.9 mm) with bregma as reference. Control mice were injected 50 nL saline in the same manner. At least one behavioral spontaneous recurrent motor seizure observed during 4 weeks post-injection was considered a successful model of chronic epilepsy. All mice were sacrificed 4 weeks after surgery. All experimental procedures involving animals were reviewed and approved by the Laboratory Animal Ethics Committee of Chongqing Medical University.

    Total RNA was extracted using TRIzol Reagent (Invitrogene, USA), according to the manufacturer's instructions. Subsequent library construction and Illumina sequencing were performed by Shanghai Lifegenes Technology Co., Ltd. All downstream analyses were based on clean data, which were obtained from raw data by removing reads containing adapters, reads containing ploy-N and low-quality reads. FPKM, Fragments Per Kilobase of exon model per Million mapped fragments, is commonly used to estimate the abundance of gene expression [15]. FPKMs of the mRNAs in each sample were calculated by aligning paired-end clean reads to the reference genome utilizing HISAT2 v2.1.0., and the FPKM for each gene was computed by summing the FPKMs of transcripts in each genome. Genes with FPKM ≥ 1 were selected for subsequent analysis to remove low expression genes.

    The DESeq R package (1.8.3) was used to analyze the differentially expressed mRNAs between the epilepsy and control groups. Adjusted Р-values < 0.05 and |log2(foldchange)| ≥ 1 were set as the threshold forsignificantly differential expression.

    To elucidate interactions among the differentially expressed genes, a PPI network was constructed using STRING tool (https://www.string-db.org/) and visualized using Cytoscape software(http://cytoscape.org/) to reveal the molecular mechanisms underlying epilepsy. The Molecular Complex Detection (MCODE; Version:1.4.2) plugin in Cytoscapse software was used to screen out the key modules [16]. Parameters were set with a degree cutoff of 2, node score cutoff of 0.2, K-core of 4, and a maximum depth of 100.

    Gene Ontology (GO) and KEGG enrichment analyses were performed using the online tool DAVID 6.8 (https://david.ncifcrf.gov/). Genes in the key modules were analyzed using the following parameters: OFFICIAL-GENE-SYMBOL, species: Homo Sapiens. GOTERM_BP, GOTERM_CC, GOTERM_MF and KEGG_PATHWAY datasets were downloaded. BP, CC, and MF refer to biological process, cellular component, and molecular function, respectively. GO and KEGG terms with P-values < 0.05 were considered significantly enriched. The hub genes were distinguished by cytoHubba in Cytoscape [17]. The MCC algorithm was used to sort the genes in the network, and top 10 nodes were selected as hub genes, which were further annotated by ClueGO and CluePedia App [18,19].

    Total RNA was extracted from the right hippocampus tissue of mice in the control and epilepsy group induced by KA. Ten hub genes in key modules from RNA-seq were validated by RT-qPCR using the CFX96 Real-Time System (Bio-Rad, USA). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) served as an internal control. The primer sequences were designed using Primer Premier 5.0, and are listed in Table 4. Experiments were performed in triplicates. Relative gene expression was analyzed using the 2−ΔΔCt method.

    Statistical analyses were performed using GraphPad Prism 7.0 (GraphPad Software, La Jolla, CA, USA). Two-tailed unpaired t-test was used to the compare two groups. Differences were considered statistically significant at P-values < 0.05.

    The results of the DEG analysis are presented in volcano plot and heatmap (Figure 1). As shown in Figure 1a, a total of 1426 DEGs were detected, including 975 upregulated genes and 451 downregulated genes. Clustering analysis of genes with the same or similar expression patterns may help identify the functions of unknown genes or unknown functions of known genes. Thus, we conducted a hierarchical clustering analysis on the top 100 differentially expressed mRNAs based on the FPKM values in the two groups (Figure 1b). The different color areas represent different clusters of grouping information. Genes in the same group have similar expression patterns and may have similar functions or be involved in the same biological processes. In addition, we summarized ten genes with the most significant differences in Table 2.

    Figure 1.  The volcano plot and heatmap of differentially expressed genes. (a) Volcano plot of the DEGs. (b) Heat map of top 100 DEGs in the epileptic group compared with the control group.
    Table 2.  The top ten DEGs.
    Gene name Description log FC adj. P-Value Up/Down
    FOSL2 FOS like 2, AP-1 transcription factor subunit 2.930792 7.05E-56 Up
    SERPINF1 serpin family F member 1 4.146166 2.81E-33 Up
    OLFML3 olfactomedin like 3 3.385027 2.91E-24 Up
    TSPYL2 TSPY-like 2 2.173859 3.86E-24 Up
    CCDC88A coiled-coil domain containing 88A -1.887247 1.18E-23 Down
    TRABD2A TraB domain containing 2A 2.949808 1.05E-21 Up
    ALMS1 ALMS1, centrosome and basal body associated protein -1.339524 4.71E-20 Down
    PACSIN1 protein kinase C and casein kinase substrate in neurons 1 2.853895 4.01E-19 Up
    STIM1 stromal interaction molecule 1 1.857381 1.12E-18 Up
    C1QA complement component 1, q subcomponent, A chain 4.279842 1.12E-18 Up

     | Show Table
    DownLoad: CSV

    To further explore the crosstalk between the DEGs, top 500 DEGs were selected to construct a PPI network based on the STRING database. The network consists of 389 nodes and 1480 edges (Figure 2a). The size of the node represents the degree of connectivity, and the color of the node represents the gene expression pattern. The red nodes are upregulated genes, while blue nodes are downregulated genes. Multiple upregulated genes were at the center of the network. A small part of the PPI network with highly connected areas has a higher probability of participating in biological regulation, and the hub genes among the subnets are often of clinical importance [20]. To find biologically essential subnets and corresponding hub genes, the MCODE [16] plugin was used to investigate the whole network. Four subnets were eventually derived from the source network (Figure 2be). Table 3 shows the detailed information of each subnet. As the subnet with the highest score, cluster 1, which contains 34 nodes and 287 edges, was selected as the key module for further research.

    Figure 2.  The protein-protein interaction network. (a) PPI network for the top 500 DEGs. (b) subnet 1. (c) subnet 2. (d) subnet 3. (e) subnet 4. Red nodes represent upregulated genes. Blue nodes represent downregulated genes.
    Table 3.  Details of four subnets.
    Cluster Score Nodes Edges Node IDs
    1 17.394 34 287 LAPTM5, CSF1R, CD53, IL10RA, SPI1, AIF1, GPSM3, ADRA2C, PDYN, SSTR3, FPR1, GPR183, TAS2R20, CX3CR1, HRH3, LPAR5, P2RY13, P2RY12, CSF2RB, MCHR1, NCF2, CX3CL1, ADCY2, CYBB, ITGB2, LY86, TLR7, PLEK, HCK, C1QA, C1QC, FCGR1A, CTSS, CD86
    2 5 5 10 CAMK2A, SLC6A7, KCNA1, GAD2, GABRD
    3 4.5 13 27 TACR3, C3AR1, KCNAB2, HTR2C, SNAP25, FOLR2, GNA15, ATP6V1D, CCK, HTR2A, F13A1, VSIG4, HRH1
    4 4 8 14 MERTK, JUP, TMEM119, HPSE, NEU1, TOLLIP, CEP290, GPR34

     | Show Table
    DownLoad: CSV

    To clarify the function of genes in key module, GO annotation and KEGG pathway analyses were performed with DAVID 6.8 and visualized by bioinformatics online tool (http://www.bioinformatics.com.cn/). Figure 2 illustrates the top 10 significantly enriched GO and KEGG terms (P < 0.05). Items related to biological process were mainly enriched in inflammatory response, innate immune response and G-protein coupled receptor signaling pathway (Figure 2a). Meanwhile, the top three enriched terms of cellular components were plasma membrane, integral component of plasma membrane, integral component of membrane (Figure 2b). With regard to molecular functions, the most important functions were concentrated in receptor activity, superoxide-generating NADPH oxidase activity, G-protein coupled purinergic nucleotide receptor activity (Figure 2c). The most significantly enriched pathways were associated with Staphylococcus aureus infection, Osteoclast differentiation, Neuroactive ligand-receptor interaction (Figure 2d). Overall, these findings suggest that the immunoregulatory and inflammatory responses occupy a dominant position in the pathogenesis of epilepsy, which is consistent with previous studies [21].

    Figure 3.  Functional enrichment analysis of DEGs in cluster 1 based on GO and KEGG. (a) BP: biological process; (b) CC: cellular component; (c) MF: molecular function; (d) KEGG: signaling pathway.

    We obtained the hub genes in the module related to immunoregulatory and inflammatory responses using the cytoHubba plugin in Cytoscape. The MCC algorithm was used to sort the genes in the network, and top 10 nodes were selected, including CX3CR1, CX3CL1, GPR183, FPR1, P2RY13, P2RY12, LPAR5, ADRA2C, PDYN, SSTR3 (Figure 4a). A darker color indicates a higher score, signaling a potentially more important role in the network. In addition, we explored the GO term associated with immune system process of these genes using ClueGO and CluePedia application in Cytoscape, revealing that they were all related to regulation of microglial cell migration and macrophage migration (Figure 4b).

    Figure 4.  The hub genes. (a) Protein-protein interaction network consisting of 10 hub genes. The shade of the node color indicates its importance in the network. (b) GO terms associated with immune system process. Each node represents a term, and the connection between the nodes reflects the correlation between the terms.

    To verify the results of RNA-seq analysis, 10 hub genes were chosen for analysis using RT-qPCR. All genes had homologous genes in the mice. Gene name, description, expression of sequencing results, and primer information are shown in Table 4. Figure 5 shows the expression of each gene in epileptic mice. The expression of nine genes was consistent with the sequencing results, except for FPR1, which is down-regulated in epileptic mice compared with the control group, in contrast to the outcome of RNA-seq. This finding may be due to the inconsistent age in human specimens for sequencing and animal samples for verification, or due to species differences between humans and mice.

    Table 4.  The top10 hub genes and related information obtained from sequencing results.
    Gene name Description Up/Down Primer
    CX3CR1 C-X3-C motif chemokine receptor 1 Up F:TTCCCATCTGCTCAGGACCTC
    R:CAGACCGAACGTGAAGACGA
    CX3CL1 C-X3-C motif chemokine ligand 1 Up F:ACGAAATGCGAAATCATGTGC
    R:CTGTGTCGTCTCCAGGACAA
    GPR183 G protein-coupled receptor 183 Up F:GGATGACAGGAAAAAGGCACC
    R:CCAGTGGTCACAGTCAATATCAG
    FPR1 formyl peptide receptor 1 Up F:GCTGTTGGAAAGTTCAGGAGTC
    R:CCAGAACGATGTAGCCAGCA
    P2RY13 purinergic receptor P2Y13 Up F:TGGGTTGAGCTAGTAACTGCC
    R:TTGTCCCGAGCATCAGCTTT
    P2RY12 purinergic receptor P2Y12 Up F:AGGGTCACAGTGCAAGAACA
    R:TGGAACTTGCAGACTGGCAT
    LPAR5 lysophosphatidic acid receptor 5 Up F:CAGAGGTCCCAAGAAGGTCTCC
    R:CAGAAGCAAGACATGGGCAC
    ADRA2C adrenoceptor alpha 2C Up F:TGCACCTGTGTGCCATTAGT
    R:GTAGAACGAGACGAGAGGCG
    PDYN prodynorphin Up F:CTCCAGAACTGCCATAGGGG
    R:TGGGGATGAATGACCTGCTTAC
    SSTR3 somatostatin receptor 3 Up F:CCAAGTGCCAGAAAATGGCTG
    R:AACCAGCCCTAACCTGAAGGG

     | Show Table
    DownLoad: CSV
    Figure 5.  Validation of 10 hub genes by qPCR. Black bars represent gene expression in the control group and gray bars represent gene expression in the epilepsy group. * represents P < 0.05; ** represents P < 0.01; *** represents P < 0.001; **** represents P < 0.0001.

    In the present study, we sequenced the transcriptomes of seven samples derived from human brain tissue, screened differentially expressed genes and performed a series of bioinformatics analyses. We found that genes in the key networks were significantly enriched for immune and inflammatory processes. Ten hub genes were then selected from the key module and a gene ontology annotation analysis was performed based on immune system process. The present findings have shown that these genes are most closely related to the migration of microglia and macrophages, which may identify new immunomodulatory compounds in the development and progression of epilepsy.

    An updated review article has examined the diversity of microglia functions, including multiple aspects of microglial activation in the hippocampal sclerotic area, microglial role in epileptogenesis without inflammatory activation, anti-epileptic role of microglia, microglia-neuron interaction and perspectives on glial communication in the epileptic brain [22]. Several hub genes have been associated with microglia. CX3CL1 is a member of the chemokine gene family, also known as fractalkine, which is expressed abundantly in the brain and localized principally to neurons. Numerous studies have reported that CX3CL1 binds to its unique receptor CX3CR1, regulates the activation of microglia, and appears to function as a signal molecule from neurons to microglia [2327]. A few studies have shown the role of CX3CL1/CX3CR1 axis signaling in the pathogenesis of epilepsy. Xu et al. reported increased expression of CX3CL1 in patients with TLE and rat models [28]. Roseti et al. have shown that CX3CL1 is responsible for positively regulating the function of the GABA A receptor in human TLE brain tissue when it was expressed in Xenopus oocytes, indicating that the CX3CL1/CX3CR1 axis is a critical regulator of potentially neuroprotective microglia-neuron interactions [29]. Additionally, Eyo and colleagues have found that a deficiency of the CX3CL1 receptor reduced the microglial process convergence toward neuronal axons and dendrites, and blocking fractalkine signaling was associated with increased seizure phenotypes [30].

    GPR183 and FPR1are functionally similar in that they are both receptors expressed in the central nervous system, and their endogenous ligands direct cell migration. Rutkowska and colleagues found that GPR183 co-expressed with 7α, 25-OHC, which was considered the primary endogenous ligand for GPR183 [31], in human and murine astrocytes [32]. Wanke et al. reported that EBI2 is highly expressed in multiple sclerosis (MS) lesions and promotes early central nervous system migration of encephalitogenic CD4 T cells [33]. A recent study revealed high expression of EBI2 in astrocytes and microglia in MS brain plaques, indicating that EBI2 signaling is involved in the recovery of demyelination [34]. FPR1 encodes a high affinity receptor for N-formyl-methionyl peptides, which are powerful neutrophil chemotactic factors [3537]. FPR1 has also been reported to induce glial cell activation in a mouse model of MS [38] and to promote the migration and differentiation of rat neural stem cells [39]. However, these two genes have not been reported in epilepsy.

    P2RY12 and P2RY13 are members of the metabotropic purinergic P2Y receptor family. P2RY12 was confirmed by Sasaki et al. to be selectively expressed on microglia [40], and has been considered as a marker to distinguish microglia from other brain cells and peripheral immune cells [41]. Evidences shows it may be necessary for microglial translocation [42], response and migration to central nervous system injury [43], and communication with neurons after status epilepticus [44]. Recently, Mo and his colleagues demonstrated that microglia promote aberrant neurogenesis and increased immature neuronal projections following seizures through its P2Y12 receptor by using current selective microglial targeting approaches [45]. P2RY13 is one of the most recently identified P2Y receptors. Studies on P2Y13 receptor knockout mice have highlighted specific functions in nervous system, including pain transmission, neuroprotection, regulation of microglial morphology, and intercellular calcium wave communication [4649]. Lysophosphatidic acid receptor 5 (LPAR5) signaling has been mainly studied in the context of neuropathic pain [50,51], but Plastira and colleagues have recently shown that LPAR5 affects microglia biology and induces a distinct pro-inflammatory and migratory signature [52,53], and identified small-molecule antagonists that can interfere with LPA-induced pro-inflammatory signaling cascades in microglia [54].

    Although the remaining three genes lack evidence to support their connection with microglia, they remain closely related to many neurological symptoms. The adrenergic α2C receptor (ADRA2C) is an inhibitory modulator of the sympathetic nervous system, which regulates neurotransmission at lower levels of neural activity in the cerebral cortex [55]. Meanwhile, the protein encoded by PDYN is a preproprotein that is proteolytically processed to form the endogenous opioid peptide dynorphin, which is a ligand for the kappa-type of opioid receptor (KOR) [56]. Extensive evidence suggests that dynorphin plays an important role as an endogenous anticonvulsant in epileptogenesis and epilepsy via KORs [5761]. Preclinical data suggests a high potential of KOR agonists to control seizures [62]. SSTR3 encodes a member of the somatostatin receptor protein family with a wide range of physiological effects on neurotransmission, antiproliferative signaling and apoptosis [63,64]. Therefore, its related diseases mainly include pituitary adenoma and glioma [6568].

    Still, there were limitations to our research. First, we used a small number of samples to reflect the differential genes between epileptic brain and normal tissue, which may not be comprehensive. The uneven distribution of participants in terms of gender and age may also affect the accuracy of our results. Additionally, we selected KA-induced chronic epilepsy as an animal model to validate the reliability of sequencing data, which might not be entirely consistent with the process of epileptogenesis in humans. Thus, continuing to collect surgical specimens of epileptic brain and appropriate control tissues to achieve sufficient biological replication will be the focus of our next studies. We aim to obtain reliable biomarkers for in-depth mechanism exploration by combining the results of present study.

    In conclusion, by sequencing the transcriptome of normal and epileptic brains, screening for differentially expressed genes, and further analyzing and mining the corresponding PPI network, we obtained seven pivotal genes closely related to microglia activation and migration. These genes reveal the important role of microglia in the immune regulation and inflammatory response mediated by epilepsy, which may provide new candidates for the study of the molecular mechanisms of seizures and lay the theoretical foundation for further research.

    This study was supported by the National Natural Science Foundation of China [No.81971217].

    The authors declare there is no conflict of interest.



    [1] O. Nevalainen, H. Ansakorpi, M. Simola, J. Raitanen, J. Isojärvi, M. Artama, et al., Epilepsy-related clinical characteristics and mortality: a systematic review and meta-analysis, Neurology, 83 (2014), 1968–1977.
    [2] S. Popkirov, F. S. Ismail, W. Grönheit, M. Kapauer, J. Wellmer, C. G. Bien, Progressive hippocampal sclerosis after viral encephalitis: Potential role of NMDA receptor antibodies, Seizure, 51 (2017), 6–8.
    [3] J. Choi, D. R. Nordli, T. D. Alden, A. DiPatri, L. Laux, K. Kelley, et al., Cellular injury and neuroinflammation in children with chronic intractable epilepsy, J. Neuroinflammation, 6 (2009), 38.
    [4] A. Vezzani, S. Balosso, T. Ravizza, The role of cytokines in the pathophysiology of epilepsy, Brain Behav. Immun., 22 (2008), 797–803.
    [5] T. Ravizza, B. Gagliardi, F. Noé, K. Boer, E. Aronica, A. Vezzani, Innate and adaptive immunity during epileptogenesis and spontaneous seizures: evidence from experimental models and human temporal lobe epilepsy, Neurobiol. Dis., 29 (2008), 142–160.
    [6] K. Rijkers, H. J. Majoie, G. Hoogland, G. Kenis, M. D. Baets, J. S. Vles, The role of interleukin-1 in seizures and epilepsy: a critical review, Exp. Neurol., 216 (2009), 258–271.
    [7] M. M. Nagib, Y. Yu, J. Jiang, Targeting prostaglandin receptor EP2 for adjunctive treatment of status epilepticus, Pharmacol. Ther., 209 (2020), 107504.
    [8] A. Dey, X. Kang, J. Qiu, Y. Du, J. Jiang, Anti-inflammatory small molecules to treat seizures and epilepsy: from bench to bedside, Trends Pharmacol. Sci., 37 (2016), 463–484.
    [9] K. Boer, W. G. Spliet, P. C. V. Rijen, S. Redeker, D. Troost, E. Aronica, Evidence of activated microglia in focal cortical dysplasia, J. Neuroimmunol., 173 (2006), 188–195.
    [10] M. J. Benson, S. Manzanero, K. Borges, Complex alterations in microglial M1/M2 markers during the development of epilepsy in two mouse models, Epilepsia, 56 (2015), 895–905.
    [11] G. Kalozoumi, O. Kel-Margoulis, E. Vafiadaki, D. Greenberg, H. Bernard, H. Soreq, et al., Glial responses during epileptogenesis in Mus musculus point to potential therapeutic targets, PLoS One, 13 (2018), e0201742.
    [12] Z. H. Luo, A. A. Walid, Y. Xie, H. Long, W. Xiao, L. Xu, et al., Construction and analysis of a dysregulated lncRNA-associated ceRNA network in a rat model of temporal lobe epilepsy, Seizure, 69 (2019), 105–114.
    [13] Q. L. Chen, L. Xia, S. P. Zhong, Q. Wang, J. Ding, Bioinformatic analysis identifies key transcriptome signatures in temporal lobe epilepsy, Neuroscience, 26 (2020), 1266–1277.
    [14] V. Riban, V. Bouilleret, B. T. Pham-Lê, J. M. Fritschy, C. Marescaux, A. Depaulis, Evolution of hippocampal epileptic activity during the development of hippocampal sclerosis in a mouse model of temporal lobe epilepsy, Neuroscience, 112 (2002), 101–111.
    [15] A. Mortazavi, B. A. Williams, K. McCue, L. Schaeffer, B. Wold, Mapping and quantifying mammalian transcriptomes by RNA-Seq, Nat. Methods, 5 (2008), 621–628.
    [16] G. D. Bader, C. W. Hogue, An automated method for finding molecular complexes in large protein interaction networks, BMC Bioinf., 4 (2003), 2.
    [17] C. H. Chin, S. H. Chen, H. H. Wu, C. W. Ho, M. T. Ko, C. Y. Lin, cytoHubba: identifying hub objects and sub-networks from complex interactome, BMC Syst. Biol., 8 (2014), S11.
    [18] G. Bindea, B. Mlecnik, H. Hackl, P. Charoentong, M. Tosolini, A. Kirilovsky, et al., ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks, Bioinformatics, 25 (2009), 1091–1093.
    [19] G. Bindea, J. Galon, B. Mlecnik, CluePedia Cytoscape plugin: pathway insights using integrated experimental and in silico data, Bioinformatics, 29 (2013), 661–663.
    [20] X. He, J. Zhang, Why do hubs tend to be essential in protein networks?, PLoS Genet., 2 (2006), e88.
    [21] N. Yu, H. Liu, Q. Di, Modulation of Immunity and the Inflammatory Response: A New Target for Treating Drug-resistant Epilepsy, Curr. Neuropharmacol., 11 (2013), 114–127.
    [22] S. Kinoshita, R. Koyama, Pro- and anti-epileptic roles of microglia, Neural. Regen. Res., 16 (2021), 1369–1371.
    [23] A. Nishiyori, M. Minami, Y. Ohtani, S. Takami, J. Yamamoto, N. Kawaguchi, et al., Localization of fractalkine and CX3CR1 mRNAs in rat brain: does fractalkine play a role in signaling from neuron to microglia?, FEBS Lett., 429 (1998), 167–172.
    [24] J. K. Harrison, Y. Jiang, S. Chen, Y. Xia, D. Maciejewski, R. K. McNamara, et al., Role for neuronally derived fractalkine in mediating interactions between neurons and CX3CR1-expressing microglia, Proc. Nat. Acad. Sci. U. S. A., 95 (1998), 10896–10901.
    [25] O. Meucci, A. Fatatis, A. A. Simen, R. J. Miller, Expression of CX3CR1 chemokine receptors on neurons and their role in neuronal survival, Proc. Nat. Acad. Sci. U. S. A., 97 (2000), 8075–8080.
    [26] A. E. Cardona, E. P. Pioro, M. E. Sasse, V. Kostenko, S. M. Cardona, I. M. Dijkstra, et al., Control of microglial neurotoxicity by the fractalkine receptor, Nat. Neurosci., 9 (2006), 917–924.
    [27] A. D. Bachstetter, J. M. Morganti, J. Jernberg, A. Schlunk, S. H. Mitchell, K. W. Brewster, et al., Fractalkine and CX 3 CR1 regulate hippocampal neurogenesis in adult and aged rats, Neurobiol. Aging, 32 (2011), 2030–2044.
    [28] Y. Xu, K. Zeng, Y. Han, L. Wang, D. Chen, Z. Xi, et al., Altered expression of CX3CL1 in patients with epilepsy and in a rat model, Am. J. Pathol., 180 (2012), 1950–1962.
    [29] C. Roseti, S. Fucile, C. Lauro, K. Martinello, C. Bertollini, V. Esposito, et al., Fractalkine/CX3CL1 modulates GABAA currents in human temporal lobe epilepsy, Epilepsia, 54 (2013), 1834–1844.
    [30] U. B. Eyo, J. Peng, M. Murugan, M. Mo, A. Lalani, P. Xie, et al., Regulation of physical microglia-neuron interactions by fractalkine signaling after status epilepticus, eNeuro, 3 (2016).
    [31] S. Hannedouche, J. Zhang, T. Yi, W. Shen, D. Nguyen, J. P. Pereira, et al., Oxysterols direct immune cell migration via EBI2, Nature, 475 (2011), 524–527.
    [32] A. Rutkowska, I. Preuss, F. Gessier, A. W. Sailer, K. K. Dev, EBI2 regulates intracellular signaling and migration in human astrocyte, Glia, 63 (2015), 341–351.
    [33] F. Wanke, S. Moos, A. L. Croxford, A. P. Heinen, S. Gräf, B. Kalt, et al., EBI2 is highly expressed in multiple sclerosis lesions and promotes early CNS migration of encephalitogenic CD4 T cells, Cell Rep., 18 (2017), 1270–1284.
    [34] I. Klejbor, D. R. Shimshek, J. Klimaszewska-Łata, M. Velasco-Estevez, J. Moryś, B. Karaszewski, et al., EBI2 is expressed in glial cells in multiple sclerosis lesions, and its knock-out modulates remyelination in the cuprizone model, Eur. J. Neurosci., (2021).
    [35] F. Boulay, M. Tardif, L. Brouchon, P. Vignais, Synthesis and use of a novel N-formyl peptide derivative to isolate a human N-formyl peptide receptor cDNA, Biochem. Biophys. Res. Commun., 168 (1990), 1103–1109.
    [36] D. C. Maestes, R. M. Potter, E. R. Prossnitz, Differential phosphorylation paradigms dictate desensitization and internalization of the N-formyl peptide receptor, J. Biol. Chem., 274 (1999), 29791–29795.
    [37] B. Postma, M. J. Poppelier, J. C. V. Galen, E. R. Prossnitz, J. A. V. Strijp, C. J. de Haas, et al., Chemotaxis inhibitory protein of Staphylococcus aureus binds specifically to the C5a and formylated peptide receptor, J. Immunol., 172 (2004), 6994–7001.
    [38] K. Bihler, E. Kress, S. Esser, S. Nyamoya, S. C. Tauber, T. Clarner, et al., Formyl peptide receptor 1-mediated glial cell activation in a mouse model of cuprizone-induced demyelination, J. Mol. Neurosci., 62 (2017), 232–243.
    [39] G. Wang, L. Zhang, X. Chen, X. Xue, Q. Guo, M. Liu, et al., Formylpeptide receptors promote the migration and differentiation of rat neural stem cells, Sci. Rep., 6 (2016), 25946.
    [40] Y. Sasaki, M. Hoshi, C. Akazawa, Y. Nakamura, H. Tsuzuki, K. Inoue, et al., Selective expression of Gi/o-coupled ATP receptor P2Y12 in microglia in rat brain, Glia, 44 (2003), 242–250.
    [41] T. R. Hammond, C. Dufort, L. Dissing-Olesen, S. Giera, A. Young, A. Wysoker, et al., Single-cell RNA sequencing of microglia throughout the mouse lifespan and in the injured brain reveals complex cell-state changes, Immunity, 50 (2019), 253–271.
    [42] U. B. Eyo, M. Mo, M. H. Yi, M. Murugan, J. Liu, R. Yarlagadda, et al., P2Y12R-dependent translocation mechanisms gate the changing microglial landscape, Cell Rep., 23 (2018), 959–966.
    [43] S. E. Haynes, G. Hollopeter, G. Yang, D. Kurpius, M. E. Dailey, W. B. Gan, et al., The P2Y12 receptor regulates microglial activation by extracellular nucleotides, Nat. Neurosci., 9 (2006), 1512–1519.
    [44] U. B. Eyo, J. Peng, P. Swiatkowski, A. Mukherjee, A. Bispo, L. J. Wu, Neuronal hyperactivity recruits microglial processes via neuronal NMDA receptors and microglial P2Y12 receptors after status epilepticus, J. Neurosci., 34 (2014), 10528–10540.
    [45] M. Mo, U. B. Eyo, M. Xie, J. Peng, D. B. Bosco, A. D. Umpierre, et al., Microglial P2Y12 receptor regulates seizure-induced neurogenesis and immature neuronal projections, J. Neurosci., 39 (2019), 9453–9464.
    [46] K. Kobayashi, H. Yamanaka, F. Yanamoto, M. Okubo, K. Noguchi, Multiple P2Y subtypes in spinal microglia are involved in neuropathic pain after peripheral nerve injury, Glia, 60 (2012), 1529–1539.
    [47] V. Morente, R. Pérez-Sen, F. Ortega, J. Huerta-Cepas, E. G. Delicado, M. T. Miras-Portugal, Neuroprotection elicited by P2Y13 receptors against genotoxic stress by inducing DUSP2 expression and MAPK signaling recovery, Biochem. Biophys. Acta, 1843 (2014), 1886–1898.
    [48] V. Kyrargyri, C. Madry, A. Rifat, I. L. Arancibia-Carcamo, S. P. Jones, V. T. T. Chan, et al., P2Y(13) receptors regulate microglial morphology, surveillance, and resting levels of interleukin 1β release, Glia, 68 (2020), 328–344.
    [49] P. Jiang, F. Xing, B. Guo, J. Yang, Z. Li, W. Wei, et al., Nucleotide transmitters ATP and ADP mediate intercellular calcium wave communication via P2Y12/13 receptors among BV-2 microglia, PLoS One, 12 (2017), e0183114.
    [50] M. E. Lin, R. R. Rivera, J. Chun, Targeted deletion of LPA5 identifies novel roles for lysophosphatidic acid signaling in development of neuropathic pain, J. Biol. Chem., 287 (2012), 17608–17617.
    [51] R. Tsukahara, S. Yamamoto, K. Yoshikawa, M. Gotoh, T. Tsukahara, H. Neyama, et al., LPA5 signaling is involved in multiple sclerosis-mediated neuropathic pain in the cuprizone mouse model, J. Pharmacol. Sci., 136 (2018), 93–96.
    [52] I. Plastira, E. Bernhart, M. Goeritzer, H. Reicher, V. B. Kumble, N. Kogelnik, et al., 1-Oleyl-lysophosphatidic acid (LPA) promotes polarization of BV-2 and primary murine microglia towards an M1-like phenotype, J. Neuroinflammation, 13 (2016), 205.
    [53] I. Plastira, E. Bernhart, M. Goeritzer, T. DeVaney, H. Reicher, A. Hammer, et al., Lysophosphatidic acid via LPA-receptor 5/protein kinase D-dependent pathways induces a motile and pro-inflammatory microglial phenotype, J. Neuroinflammation, 14 (2017), 253.
    [54] I. Plastira, L. Joshi, E. Bernhart, J. Schoene, E. Specker, M. Nazare, et al., Small-molecule lysophosphatidic acid receptor 5 (LPAR5) antagonists: versatile pharmacological tools to regulate inflammatory signaling in BV-2 microglia cells, Front. Cell Neurosci., 13 (2019), 531.
    [55] L. Hein, J. D. Altman, B. K. Kobilka, Two functionally distinct alpha2-adrenergic receptors regulate sympathetic neurotransmission, Nature, 402 (1999), 181–184.
    [56] C. Chavkin, Dynorphin--still an extraordinarily potent opioid peptide, Mol. Pharmacol., 83 (2013), 729–736.
    [57] M. Simonato, P. Romualdi, Dynorphin and epilepsy, Prog. Neurobiol., 50 (1996), 557–583.
    [58] S. Loacker, M. Sayyah, W. Wittmann, H. Herzog, C. Schwarzer, Endogenous dynorphin in epileptogenesis and epilepsy: anticonvulsant net effect via kappa opioid receptors, Brain, 130 (2007), 1017–1028.
    [59] B. Queenan, R. Dunn, V. Santos, Y. Feng, M. Huizenga, R. Hammack, et al., Kappa opioid receptors regulate hippocampal synaptic homeostasis and epileptogenesis, Epilepsia, 59 (2018), 106–122.
    [60] H. Dai, P. Wang, H. Mao, X. Mao, S. Tan, Z. Chen, Dynorphin activation of kappa opioid receptor protects against epilepsy and seizure-induced brain injury via PI3K/Akt/Nrf2/HO-1 pathway, Cell Cycle, 18 (2019), 226–237.
    [61] L. Liu, Y. Xu, H. Dai, S. Tan, X. Mao, Z. Chen, Dynorphin activation of kappa opioid receptor promotes microglial polarization toward M2 phenotype via TLR4/NF-κB pathway, Cell Biosci., 10 (2020), 42.
    [62] L. Zangrandi, C. Schwarzer, The kappa opioid receptor system in temporal lobe epilepsy, Handb. Exp. Pharmacol., 2021.
    [63] D. Stanić, H. Malmgren, H. He, L. Scott, A. Aperia, T. Hökfelt, Developmental changes in frequency of the ciliary somatostatin receptor 3 protein, Brain Res., 1249 (2009), 101–112.
    [64] Q. Yao, Q. Liu, H. Xu, Z. Wu, L. Zhou, Z. Gu, et al., Upregulated expression of SSTR3 is involved in neuronal apoptosis after intracerebral hemorrhage in adult rats, Cell Mol. Neurobiol., 37 (2017), 1407–1416.
    [65] S. Nielsen, S. Mellemkjaer, L. M. Rasmussen, T. Ledet, N. Olsen, M. Bojsen-Møller, et al., Expression of somatostatin receptors on human pituitary adenomas in vivo and ex vivo, J. Endocrinol. Invest., 24 (2001), 430–437.
    [66] R. Panetta, Y. C. Patel, Expression of mRNA for all five human somatostatin receptors (hSSTR1-5) in pituitary tumors, Life Sci., 56 (1995), 333–342.
    [67] K. Lamszus, W. Meyerhof, M. Westphal, Somatostatin and somatostatin receptors in the diagnosis and treatment of gliomas, J. Neurooncol., 35 (1997), 353–364.
    [68] H. Luo, C. Tao, X. Long, K. Huang, X. Zhu, A risk signature of four aging-related genes has clinical prognostic value and is associated with a tumor immune microenvironment in glioma, Aging (Albany NY) 13 (2021), 16198–16218.
  • This article has been cited by:

    1. Fuxin Zhong, Yunhao Gan, Jiaqi Song, Wenbo Zhang, Shiyun Yuan, Zhangjin Qin, Jiani Wu, Yang Lü, Weihua Yu, The inhibition of PGAM5 suppresses seizures in a kainate-induced epilepsy model via mitophagy reduction, 2022, 15, 1662-5099, 10.3389/fnmol.2022.1047801
    2. N. P. Camacho-Hernández, F. Peña-Ortega, Long-Jun Wu, Fractalkine/CX3CR1-Dependent Modulation of Synaptic and Network Plasticity in Health and Disease, 2023, 2023, 1687-5443, 1, 10.1155/2023/4637073
    3. Ulia G. Odnoshivkina, Eva A. Kuznetsova, Alexey M. Petrov, 25-Hydroxycholesterol as a Signaling Molecule of the Nervous System, 2022, 87, 0006-2979, 524, 10.1134/S0006297922060049
    4. Shiqi Lin, Ziyuan Lin, Huaqin Sun, Effect of Levetiracetam Treatment on the Transcriptome of Pentylenetetrazole-Induced Zebrafish Epilepsy Model, 2023, 19, 18117775, 206, 10.3923/ijp.2023.206.211
  • Reader Comments
  • © 2021 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(4158) PDF downloads(205) Cited by(4)

Figures and Tables

Figures(5)  /  Tables(4)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog