Open Access

RNA‑seq analysis of blood of valproic acid‑responsive and non‑responsive pediatric patients with epilepsy

  • Authors:
    • Yan Wang
    • Zhiping Li
  • View Affiliations

  • Published online on: April 30, 2019     https://doi.org/10.3892/etm.2019.7538
  • Pages: 373-383
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Epilepsy is the most common chronic neurological disorder, affecting ~70 million individuals worldwide. However, approximately one‑third of the patients are refractory to epilepsy medication. Of note, 100% of patients with genetic epilepsy who are resistant to the traditional drug, valproic acid (VPA), are also refractory to the other anti‑epileptic drugs. The aim of the present study was to compare the transcriptomes in VPA responders and non‑responders, to explore the mechanism of action of VPA and identify possible biomarkers to predict VPA resistance. Thus, RNA‑seq was employed for transcriptomic analysis, differentially expressed genes (DEGs) were analyzed using Cuffdiff software and the DAVID database was used to infer the functions of the DEGs. A protein‑protein interaction network was obtained using STRING and visualized with Cytoscape. A total of 389 DEGs between VPA‑responsive and non‑responsive pediatric patients were identified. Of these genes, 227 were upregulated and 162 were downregulated. The upregulated DEGs were largely associated with cytokines, chemokines and chemokine receptor‑binding factors, whereas the downregulated DEGs were associated with cation channels, iron ion binding proteins, and immunoglobulin E receptors. In the pathway analysis, the toll‑like receptor signaling pathway, pathways in cancer, and cytokine‑cytokine receptor interaction were mostly enriched by the DEGs. Furthermore, three modules were identified by protein‑protein interaction analysis, and the potential hub genes, chemokine (C‑C motif) ligand 3 and 4, chemokine (C‑X‑C motif) ligand 9, tumor necrosis factor‑α and interleukin‑1β, which are known to be closely associated with epilepsy, were identified. These specific chemokines may participate in processes associated with VPA resistance and may be potential biomarkers for monitoring the efficacy of VPA.

Introduction

Epilepsy is the most common chronic neurological disorders, affecting ~70 million individuals worldwide (1). Furthermore, ~0.5–1% of the pediatric population suffer from epilepsy (2) and approximately one-third of these patients are refractory to epilepsy medication (3). Valproic acid (VPA) is an anti-epileptic drug recommended by the National Institute for Health and Care Excellence guidelines as the first-line therapy for absence epilepsy (4,5), and has been used for 50 years due to its efficacy and high tolerability (6,7). Previous studies have indicated that approximately one-third of patients are non-responsive to VPA (8,9), and the reason for this phenomenon remains elusive. Recently, Gesche et al (10) have demonstrated that resistance to VPA has a specificity of 100% regarding the identification of genetic generalized epileptic patients. Consequently, it is important to elucidate the mechanism underlying VPA efficacy and identify biomarkers predictive of VPA responses.

At present, two hypotheses for pharmaco-resistant epilepsy are commonly accepted, namely the multi-transporter hypothesis and the drug targets hypothesis. However, the mechanism of VPA resistance is distinct from that of other anti-epileptic drugs (AEDs). In fact, VPA is neither the substrate of multi-transporters, including P-glycoprotein, multi-drug resistant protein and breast cancer resistance protein, nor does it induce the expression of the multi-transporters in the brain (1114). However, reported targets of VPA, including γ-aminobutyric acid receptor (15,16), sodium channels and calcium channels, appear to not be involved in VPA resistance (1719). These results suggest that the aforementioned hypotheses hardly explain the mechanisms of VPA resistance or efficacy.

Genome-wide gene expression profiling has been increasingly used to investigate pathogenetic mechanism and identify potential biomarkers for various human diseases (20,21). RNA sequencing (RNA-seq) is a widely used method to study overall transcriptional activity and has a broad coverage. Previous studies employing the RNA-seq method led to the discovery of potential biomarkers for Alzheimer's disease and malignant glioma, including NeuroD6 and F11R, in brain tissues (22,23). Fibronectin 1 and 12 other genes implicated in oxidative phosphorylation and glycolysis/gluconeogenesis pathways in the brain were identified as candidate critical factors for temporal lobe epilepsy, with and without hippocampal sclerosis (24,25). However, few studies have addressed VPA efficacy and resistance, also due to the limited accessibility of brain tissue from VPA-treated patients.

Peripheral blood may be obtained non-invasively and is commonly used for studying biomarkers. Liew et al (26) have indicated that ~81.9% of the genes expressed in the brain were also expressed in the whole-blood microarray dataset. Borovecki et al (27) reported that the blood mRNA levels of specific target genes are associated with Huntington's disease severity and response to a histone deacetylase inhibitor. VPA is a histone deacetylase inhibitor (28,29), potentially affecting, directly or indirectly, between 2 and 5% of all genes (30). A previous study reported that 11 genes were differentially expressed after a 3-month VPA treatment (31). In the present study, the mRNA expression profile in the blood of VPA responders and non-responders was analyzed after a treatment period of ≥1 year, to identify possible biomarkers for the prediction of VPA efficacy.

Materials and methods

Patients

Subjects aged from 0 to 18 years were enrolled at the Children's Hospital of Fudan University (Shanghai, China) between July 2016 and May 2018. Each patient was evaluated according to the inclusion and exclusion criteria (32). The inclusion criteria were as follows: Pediatric patients diagnosed with epilepsy or an epileptic syndrome and administration of VPA for at least one year. The exclusion criteria were as follows: Patients with abnormal liver and kidney function, and patients who had developed infectious diseases, including upper respiratory infection and urinary tract infection, in the last three months.

Patients with a complete disappearance of seizures and a normal electroencephalogram were considered as VPA responders, while patients who continued to experience seizures were assigned to the non-responsive groups. Seizure types were identified according to the International League Against Epilepsy definition (33). Focal epileptic seizures were defined as events originating within networks limited to one hemisphere. Generalized epileptic seizures were conceptualized as originating at a certain point within, and rapidly engaging, bilaterally distributed networks (34).

RNA preparation

Blood from three VPA responders and five non-responders without seizure for 12 h was collected in PAXgene blood RNA tubes and stored at −80°C until use. PAXgene tubes were stabilized for 2 h at room temperature. After centrifugation for 10 min at 3,000–5,000 × g at 4°C by using a swing-out rotor, the supernatant was removed by pipetting. Subsequently, 4 ml of RNase-free water were added to the pellet. Total RNA was extracted according to the instructions of the PAXgene Blood RNA kit (Qiagen), and the quality and quantity of total RNA were determined using a Qubit 2.0 (Thermo Fisher Scientific, Inc.) and a Bioanalyzer 2100 (Agilent Technologies).

RNA-seq

Transcriptome sequencing libraries were prepared by using the TruSeq RNA LT V2 Sample Prep lit (Illumina, Inc.) and were qualified using the Qubit 2.0. Paired-end sequencing for 150 base pair was performed by an Illumina HiSeq 2500 instrument (Illumina, Inc.). All of the paired-end raw reads were quality-checked for low-quality bases and adapter sequences.

Analysis of differentially expressed genes (DEGs)

Low-quality fractions and Illumina universal adapters were deleted by using Trim Galore v0.4.2 with the threshold of Q<30. Fasta QC (version 0.11.5; Illumina, Inc.) was employed to assess the quality of the data. The paired-end sequencing reads were aligned to the reference genome (hg19), downloaded from the University of California Santa Cruz (UCSC) website (http://hgdownload.soe.ucsc.edu/downloads.html). Statistically significant expression changes between responders and non-responders were estimated using the Cuffilinks 2.2.1 software (http://cole-trapnell-lab.github.io/cufflinks/install/) with a threshold of 10 for NO TEST. Student's t-test was performed to calculate the P-value, while the false discovery rate controlled by the Benjamini-Hochberg procedure was used for determining the Q-value. Genes with a fold change of >1 and P<0.05 were defined as DEGs.

Validation by reverse transcription-quantitative (RT-qPCR)

Changes in the mRNA expression of specific DEGs associated with epilepsy [chemokine (C-C motif) ligand 3 and FOS], exhibiting high fold changes, were validated by qPCR in 17 samples (including 6 VPA responders and 11 non-responders). Specific primers for selected genes were designed using Primer-BLAST (http://www.ncbi.nlm.nih.gov/tools/primer-blast). All samples were amplified in triplicate. qPCR amplifications were performed with the following cycling parameters: An initial hot start at 95°C for 5 min followed by 45 cycles of 95°C for 15 sec and 60°C for 30 sec. In order to normalize the qPCR results, GAPDH was included as the reference gene. The relative expression of genes was calculated based on the average quantification cycle (Cq) values across samples. Relative expression=2{-[(Cq gene of interest-Cq GAPDH of interest) non-responders]-(Cq gene of interest-Cq GAPDH of interest) responders}} (35).

Gene functions and pathways

Gene Ontology (GO) gene functions and biochemical pathways enriched by the DEGs were determined by using the web-based annotation tool DAVID v6.7 (https://david-d.ncifcrf.gov/summary.jsp) (36), providing GO terms in the categories biological process (BP), cellular component (CC), and molecular function (MF) and Kyoto Encyclopedia of Genes and Genomes pathways. P<0.05 was used as the significance threshold.

Construction and visualization of the protein-protein interaction (PPI) network

The PPI network based on the DEGs identified were constructed by using the STRING database (https://string-db.org), a pre-computed database wherein associations between proteins are assigned on the basis of high-throughput experiments, literature mining, gene fusion, co-occurrence, co-expression analysis, and also computational predictions, e.g. genomic meta-analysis. Interactions with a confidence score of 0.7 were considered for visualization by Cytoscape v3.4.0 (https://cytoscape.org/).

Network module analysis

The Molecular Complex Deletion (MCODE) plugin for Cytoscape was used to analyze the network modules (37). Densely connected regions or clusters in the co-expression network were identified using the following parameters: Degree cut-off=2, k-core=2 and max. depth=100.

Statistical analysis

DEGs analysis was performed by using Cuffilinks 2.2.1 software (cuffdiff, ttp://cole-trapnell-lab.github.io/cufflinks/install/). RT-qPCR data were analyzed using GraphPad Prism version 7.0 software (GraphPad, Inc.). Values were expressed as the mean ± standard error. Student's t-test was performed to analyze differences between VPA-responders and non-responders.

Results

Demographic data

A total of 8 pediatric patients with epilepsy were recruited, of which 3 were responders, while 5 were non-responders. All responders were males, while 3 of the non-responders were males and 2 females. The average age of the responders was 7.0±2.2 years and was not significantly different from that of the non-responsive group (4.8±1.8 years, P=0.47). Furthermore, no significant difference in the plasma VPA concentration was identified between the responsive and non-responsive groups (107.2±17.7 vs. 80.4±17.4, P=0.35; Table I).

Table I.

Demographic data.

Table I.

Demographic data.

DemographicsRespondersNon-responders


ID123Median (min, max)12345Median (min, max)P-value
Age (Years)10.737.47.4 (3, 10.7)9.64.880.31.34.8 (0.3, 9.6)0.47
SexMaleMaleMaleMaleFemaleMaleFemaleMale
Seizure typeUnknownGeneralizedGeneralizedFocalGeneralizedGeneralizedGeneralizedGeneralized
AEDsVPA+TPMVPAVPAVPAVPA+LTG+OXCVPAVPAVPA+LEV
Dose of VPA(mg/kg)33.321.518.121.529.728.631.229.136.429.70.13
(18.1, 33.3) (28.6, 36.4)
Plasma concentration139.177.9104.6104.656.762.0148.257.377.762 (56.7, 148.2)0.35
(µg/ml) (77.9, 139.1)

[i] AEDs, antiepileptic drugs; TPM, topiramate; LTG, lamotrigine; OXC, oxcarbazepine; LEV, levetiracetam; VPA, valproic acid.

Comparative transcriptome profiling

A total of 1,153 genes were differentially expressed between responders and non-responders (P<0.05; Fig. 1). Of these DEGs, 389 had a |Log 2 fold change|≥1 and comprised of 227 upregulated and 162 downregulated genes. Among the 389 DEGs, 121 variations had a Q-value of <0.05 and included 84 upregulated and 37 downregulated genes (Fig. 2). The 20 most significantly upregulated and downregulated genes are listed in Tables II and III, respectively.

Table II.

Top 20 upregulated differentially expressed genes.

Table II.

Top 20 upregulated differentially expressed genes.

GeneDefinitionLog2 fold changeP-valueQ-value
TSIXTSIX transcript, XIST antisense RNA7.59 5.00×10−5 9.00×10−3
CXCL10Chemokine (C-X-C motif) ligand 105.99 5.00×10−5 9.00×10−3
LILRA3Leukocyte immunoglobulin-like receptor subfamily a (without tm domain) member 35.96 5.00×10−5 9.00×10−3
FN1Fibronectin 13.47 5.00×10−5 9.00×10−3
GPR84G protein-coupled receptor 843.25 5.00×10−5 9.00×10−3
PDK4Pyruvate dehydrogenase kinase, isozyme 42.99 5.00×10−5 9.00×10−3
SEMA6BSema domain. Transmembrane domain (tm) and cytoplasmic domain (semaphorin) 6b2.74 5.00×10−5 9.00×10−3
HLA-DRB5Major histocompatibility complex class ii dr β 52.73 5.00×10−5 9.00×10−3
PTGESProstaglandin E synthase2.73 5.00×10−5 9.00×10−3
MYOM2Myomesin 22.68 5.00×10−5 9.00×10−3
CCL3Chemokine (C-C motif) ligand 32.60 5.00×10−5 9.00×10−3
IL1BInterleukin 1β2.55 5.00×10−5 9.00×10−3
IFI27Interferon α-inducible protein 272.32 5.00×10−5 9.00×10−3
HJURPHolliday junction recognition protein2.17 5.00×10−5 9.00×10−3
RRM2Ribonucleotide reductase M22.06 5.00×10−5 9.00×10−3
CDCA5Cell division cycle associated 52.04 5.00×10−5 9.00×10−3
FOLR3Folate receptor 3 (γ)1.97 5.00×10−5 9.00×10−3
TNFTumor necrosis factor1.89 5.00×10−5 9.00×10−3
PLK1Polo-like kinase 11.83 5.00×10−5 9.00×10−3
SEC14L2SEC14-like 2 (S. cerevisiae)1.79 5.00×10−5 9.00×10−3

Table III.

Top 20 downregulated differentially expressed genes.

Table III.

Top 20 downregulated differentially expressed genes.

GeneDefinitionLog2 fold changeP-valueQ-value
ARHGEF10Rho guanine nucleotide exchange factor 10−3.33 5.00×10−5 9.00×10−3
KCNG1Potassium voltage-gated channel subfamily g member 1−2.63 5.00×10−5 9.00×10−3
FOSFBJ murine osteosarcoma viral oncogene homolog−2.20 5.00×10−5 9.00×10−3
PAQR8Progestin and adipoq receptor family member VIII−2.06 5.00×10−5 9.00×10−3
C21orf15Chromosome 21 open reading frame 15−2.06 5.00×10−5 9.00×10−3
PTGS2 Prostaglandin-endoperoxide synthase 2 (prostaglandin G/H synthase and cyclooxygenase)−1.72 5.00×10−5 9.00×10−3
HCAR2Hydroxycarboxylic acid receptor 2−1.71 5.00×10−5 9.00×10−3
IL8Interleukin 8−1.67 5.00×10−5 9.00×10−3
TGFATransforming growth factor α−1.57 5.00×10−5 9.00×10−3
HLA-DQA2Major histocompatibility complex class II DQ α 2−1.49 5.00×10−5 9.00×10−3
FOSBFBJ murine osteosarcoma viral oncogene homolog B−1.47 5.00×10−5 9.00×10−3
TNFRSF10CTumor necrosis factor receptor superfamily member 10c decoy without an intracellular domain−1.46 5.00×10−5 9.00×10−3
DUSP1Dual specificity phosphatase 1−1.43 5.00×10−5 9.00×10−3
RTN1Reticulon 1−1.40 5.00×10−5 9.00×10−3
TLR10Toll-like receptor 10−1.25 5.00×10−5 9.00×10−3
KCNE3Potassium voltage-gated channel Isk-related family member 3−1.20 5.00×10−5 9.00×10−3
THBDThrombomodulin−1.19 5.00×10−5 9.00×10−3
MYBL1v-myb myeloblastosis viral oncogene homolog (avian)-like 1−1.09 5.00×10−5 9.00×10−3
ARFIP1ADP-ribosylation factor interacting protein 1−1.38 1.00×10−4 1.60×10−2
FCRL5Fc receptor-like 5−1.27 1.00×10−4 1.60×10−2

Two genes (CCL3 and FOS), closely associated with epilepsy, were selected for validation by RT-qPCR, revealing significant differences in expression between VPA responders and non-responders, in accordance with the results of the RNA-seq. This indicated that the results obtained by the RNA-seq analysis were reliable (Fig. 3).

GO and pathway analysis

The GO and pathway enrichment analyses were performed for the 121 final DEGs, including 84 upregulated and 37 downregulated genes. In the GO category BP, the upregulated DEGs were significantly enriched in the GO terms ‘cell cycle’, ‘immune response’ and ‘cell cycle process’ (Fig. 4A). In the category CC, the upregulated DEGs were enriched in the GO terms ‘membrane-enclosed lumen’, ‘organelle lumen’ and ‘organelle lumen’ (Fig 4B), and in the category MF, they were enriched in the GO terms ‘cytokine activity’, ‘chemokine activity’ and ‘chemokine receptor binding’ (Fig 4C). On the other hand, the group of downregulated DEGs was highly enriched in the GO terms ‘response to wounding’, ‘defense response’ and ‘inflammatory response’ in the category BP (Fig 4D). In the category CC, the downregulated DEGs were highly enriched in the GO terms ‘integral to membrane’, ‘intrinsic to membrane’ and ‘plasma membrane’ (Fig 4E), and in the category MF, they accumulated in the GO terms ‘cation channel’, ‘iron ion binding’ and ‘IgE receptor activity’ (Fig. 4F).

The 121 DEGs were enriched in 22 pathways (P<0.05), of which the Toll-like receptor signaling pathway, cancer pathways and cytokine-cytokine receptor interactions were the most represented (Fig. 5).

PPI network analysis and module identification

Analysis of the 121 DEGs by STRING and visualization with the Cytoscape plugin MCODE revealed 197 interactions, covering three modules (Fig. 6). Of these, module 1 contained 73 interactions with 13 nodes (CDT1, PLK1, NUSAP1, RRM2, MKI67, CCNB2, HJURP, TPX2, CDCA8, TOP2A, BIRC5, KIF11, CDC45), module 2 included 21 interactions with 7 nodes (IFIT3, IFI44L, ISG15, RSAD2, IFI27, OASL, IFI44L), and module 3 contained 32 interactions with 12 nodes [chemokine (C-X-C motif) ligand 9 (CXCL9), CCL3, CCL4, tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IRF1, HLA-DRB1, HLA-DRB5, IL8, PTGS2, FOS, HLA-DQA2)] (Fig. 7).

A literature review in PubMed confirmed that the function of CCL3, CCL4, CXCL9, TNF-α, IL-1β, and FOS is associated with epilepsy (Table IV) (3844).

Table IV.

Functions of chemokine genes in epilepsy.

Table IV.

Functions of chemokine genes in epilepsy.

GeneFunction(Refs.)
CCL3Inhibition of systemic receptor leads to decrease in seizure activity(38,39)
CCL4Inhibition of systemic receptor leads to decrease in seizure activity(38,39)
CXCL9Immune-cell recruitment across the BBB(38)
TNFActivation of NF-κB and regulation of the process of post-seizure neurogenesis(40,41)
IL-1βInduction of spontaneously recurring seizures(42)
FOSRegulation of CA3 neuronal excitability and survival(43,44)

[i] BBB, blood-brain barrier; IL, interleukin; TNF, tumor necrosis factor; CXCL9, chemokine (C-X-C motif) ligand 9; CCL3, chemokine (C-C motif) ligand 3.

Discussion

Pharmaco-resistant epilepsy remains a major clinical issue with elusive underlying mechanisms. In the present study, a total of 1,153 DEGs between VPA responders and non-responders (P<0.05) were initially identified. Of these genes, 123 upregulated and 60 downregulated genes fulfilled the criterion of P<0.001. This number was higher than that in the study of Tang et al (45), which may be accounted for by the different methods used. In the latter study, oligonucleotide microarrays were employed, containing probe sets for more than 12,000 genes and ESTs. However, previous studies have concluded that microarray platforms suffer from technical issues including cross-hybridization, nonspecific hybridization and limited range of detection of individual probes (46,47). As a result, genes with expression below or near the background level may exhibit increased variability and as such, calculated fold-changes for these genes may be difficult to detect with statistical significance. RNA-seq avoids such technical issues and exhibits a broader dynamic range. In the analysis of the current study, specific DEGs (dual specificity phosphatase 1, ribosomal protein S6 kinase A1 and aldehyde dehydrogenase 2 family member) reported by Tang et al (45) were also identified.

In the present study, most of the DEGs were implicated in the immune and inflammatory response, and associated with cytokine-cytokine receptor interactions, as also identified in epileptic patients by Floriano-Sánchez et al (32). Inflammation is increasingly recognized as an important pathogenetic factor in epilepsy. Evidence suggests the presence of all of the hallmarks of a chronic inflammatory state, i.e., infiltration of leukocytes, reactive gliosis, as well as overexpression of cytokines and their target proteins, in the brain of pharmaco-resistant epileptic patients and animal models (48). CCL3, CCL4 and CXCL9 are the chemokines that guide directional migration of leukocytes and have an important role in the inflammation of the central nervous system. Several studies have demonstrated increased mRNA and protein expression of CCL3, CCL4 and CXCL9 in the cortex and hippocampus of epileptic rats and drug-refractory patients (4951). The present study revealed that the expression of CCL3, CCL4 and CXCL9 was higher in the blood of VPA non-responsive vs. responsive pediatric patients, which was consistent with the results obtained by Srivastava et al (52). The reason for the overexpression of CCL3, CCL4 and CXCL9 in the brain and blood of drug-refractory patients remains elusive. It has been reported that TNF-α and IL-1β induce the expression of CCL3 and CCL4 through NF-κB and activate inflammation via the mTOR signaling pathway (41,53). Of note, VPA was demonstrated to reduce the amount of leukocytes and inhibit the expression of TNF-α (54,55). The present study indicated that the mRNA levels of TNF-α, IL-1β, NF-κB and IL-1 receptor-associated kinase 2 (a regulator of TNF-α), were significantly higher in VPA non-responders than in responders, which suggested that CCL3 and CCL4 overexpression was associated with the high expression of TNF-α and IL-1β, and indicated that the transcriptional states of CCL3, CCL4, CXCL9, TNF-α and IL-1β are potential markers for monitoring the patients' resistance to VPA.

Previous reports have revealed that increased TNF-α may result in the inhibition of cytochrome P450 family 2 subfamily D member 6 (CYP2D6) and CYP2C19 expression, and may results in the enhanced expression of CYP3A4 and CYP2C9 (the enzymes responsible for VPA metabolism in the brain) (56). Furthermore, TNF-α has been reported to induce the overexpression of transporter (P-gp), which is associated with AED efficacy (57). However, Feng et al (58) indicated that there was no difference in the plasma VPA concentration between responders and non-responders, suggesting that the role of TNF-α in the efficacy of VPA may be independent of its effects on drug metabolism and P-gp.

CCL3, CCL4, IL-1β and TNF-α are able to increase the permeability of the blood-brain barrier (59,60), possibly resulting in their passage into the brain and cerebrospinal fluid (61). Furthermore, overexpression of CCL3 and TNF-α was identified to induce the influx of Ca2+ and to enhance the expression of N-methyl-D-aspartate receptor (NMDAR), leading to increased excitatory neurotransmission and contributing to the development of epileptic seizures and excitotoxicity (62,63). Previous studies have demonstrated an association between NMDAR and the efficacy of AEDs. Zellinger et al (64) indicated that blocking the glycine-binding site of the NR2B subunit of NMDAR may increase the sensitivity to AEDs. Hung et al (65) confirmed that a specific mutation (−200 T>G) of NR2B is associated with s sustained dosage of VPA. Therefore, it may be speculated that NMDAR functionally links CCL3, CCL4, IL-1β and TNF-α with VPA efficacy. However, further study is required for verification.

The protein expression of FOS, an immediate early gene and recognized biomarker of neuronal activity, is activated during spontaneous seizure (6668). Previous studies have indicated that the expression of FOS rapidly increases 1.5 h after seizure stimulation by pentylenetetrazol and in amygdala-kindled seizures (69). However, the expression profile of FOS during seizures is complex and varies depending on the seizure type and status. FOS expression is increased in generalized seizure but not change in focal seizures. With respect to seizure status, high FOS mRNA expression was detected in rats at 1 h after the injection of kainic acid, while it tended to be low after 6 h (70). Furthermore, Madsen et al (71) identified a large increase in FOS expression at 2 h after a kindling stimulus, while after 18 h, the expression was lower than that observed upon a sham stimulation, and reached the control levels after 3 weeks. Of note, all plasma samples in the present study were collected during a non-seizure period which may account for the slightly decreased FOS expression.

In conclusion, the chemokines CCL3, CCL4, CXCL9, TNF-α and IL-1β may participate in processes associated with VPA resistance and serve as potential biomarkers for monitoring the efficacy of VPA. The study also revealed numerous critical pathways and sub-modules of potential pathogenetic relevance, deserving further investigation.

Acknowledgements

Not applicable.

Funding

The current study was supported by Important Discipline of Shanghai (grant no. 20162B0305) and the National Natural Science Foundation of China (grant nos. 81370776 and 81874325).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contribution

YW analyzed the data and drafted the manuscript. ZL designed the study and revised the manuscript.

Ethics approval and consent to participate

The study protocol was approved by the Ethics Committee of the Children's Hospital of Fudan University (Shanghai, China) in 2016 (no. 136). Written informed consent was obtained from the guardians of patients prior to enrolment.

Patient consent for publication

Not applicable.

Competing interest

The authors declare that they have no competing interests.

References

1 

Singh A and Trevick S: The epidemiology of global epilepsy. Neurol Clin. 34:837–847. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Lv RJ, Shao XQ, Cui T and Wang Q: Significance of MDR1 gene C3435T polymorphism in predicting childhood refractory epilepsy. Epilepsy Res. 132:21–28. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Voll A, Hernández-Ronquillo L, Buckley S and Téllez-Zenteno JF: Predicting drug resistance in adult patients with generalized epilepsy: A case-control study. Epilepsy Behav. 53:126–130. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Rakitin A, Kõks S and Haldre S: Valproate modulates glucose metabolism in patients with epilepsy after first exposure. Epilepsia. 56:e172–e175. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Fernando-Dongas MC, Radtke RA, Vanlandingham KE and Husain AM: Characteristics of valproic acid resistant juvenile myoclonic epilepsy. Seizure. 9:385–388. 2000. View Article : Google Scholar : PubMed/NCBI

6 

Nevitt ST, Sudell M, Weston J, Tudur Smith C and Marson AG: Antiepileptic drug monotherapy for epilepsy: A network meta-analysis of individual participant data. Cochrane Database Syst Rev. 6:CD01141212017.

7 

Brigo F, Igwe SC and Lattanzi S: Ethosuximide, sodium valproate or lamotrigine for absence seizure in children and adolescents. Cochrane Database Syst Rev. 2:CD0030322019.PubMed/NCBI

8 

Yasiry Z and Shorvon SD: The relative effectiveness of five antiepileptic drugs in treatment of benzodiazepine-resistant convulsive status epilepticus: A meta-analysis of published studies. Seizure. 23:167–174. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Trinka E, Höfler J, Zerbs A and Brigo F: Efficacy and safety of intravenous valproate for status epilepticus: A systematic review. CNS Drugs. 28:623–639. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Gesche J, Khanevski M, Solberg C and Beier CP: Resistance to valproic acid as predictor of treatment resistance in genetic generalized epilepsies. Epilepsia. 58:E64–E69. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Luna-Tortós C, Fedrowitz M and Löscher W: Evaluation of transport of common antiepileptic drugs by human multidrug resistance-associated proteins (MRP1, 2 and 5) that are overexpressed in pharmacoresistant epilepsy. Neuropharmacology. 58:1019–1032. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Baltes S, Fedrowitz M, Tortos CL, Potschka H and Löscher W: Valproic acid is not a substrate for P-glycoprotein or multidrug resistance proteins 1 and 2 in a number of in vitro and in vivo transport assays. J Pharmacol Exp Ther. 320:331–343. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Moerman L, Wyffels L, Slaets D, Raedt R, Boon P and De Vos F: Antiepileptic drugs modulate P-glycoproteins in the brain: A mice study with (11)C-desmethylloperamide. Epilepsy Res. 94:18–25. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Römermann K, Helmer R and Löscher W: The antiepileptic drug lamotrigine is a substrate of mouse and human breast cancer resistance protein (ABCG2). Neuropharmacology. 93:7–14. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Zhou L, Cao Y, Long H, Long L, Xu L, Liu Z, Zhang Y and Xiao B: ABCB1, ABCC2, SCN1A, SCN2A, GABRA1 gene polymorphisms and drug resistant epilepsy in the Chinese Han population. Pharmazie. 70:416–420. 2015.PubMed/NCBI

16 

Balan S, Sathyan S, Radha SK, Joseph V, Radhakrishnan K and Banerjee M: GABRG2, rs211037 is associated with epilepsy susceptibility, but not with antiepileptic drug resistance and febrile seizures. Pharmacogenet Genomics. 23:605–610. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Singh E, Pillai KK and Mehndiratta M: Characterization of a lamotrigine-resistant kindled model of epilepsy in mice: Evaluation of drug resistance mechanisms. Basic Clin Pharmacol Toxicol. 115:373–378. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Glauser TA, Holland K, O'Brien VP, Keddache M, Martin L, Clark PO, Cnaan A, Dlugos D, Hirtz DG6 Shinnar S, et al: Pharmacogenetics of antiepileptic drug efficacy in childhood absence epilepsy. Ann Neurol. 81:444–453. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Lv N, Qu J, Long H, Zhou L, Cao Y, Long L, Liu Z and Xiao B: Association study between polymorphisms in the CACNA1A, CACNA1C, and CACNA1H genes and drug-resistant epilepsy in the Chinese Han population. Seizure. 30:64–69. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Mou P, Chen Z, Jiang L, Cheng J and Wei R: PTX3: A potential biomarker in thyroid associated ophthalmopathy. Biomed Res Int. 2018:59619742018. View Article : Google Scholar : PubMed/NCBI

21 

Waters WR, Maggioli MF, Palmer MV, Thacker TC, McGill JL, Vordermeier HM, Berney-Meyer L, Jacobs WR Jr and Larsen MH: Interleukin-17A as a Biomarker for bovine tuberculosis. Clin Vaccine Immunol. 23:168–180. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Satoh J, Yamamoto Y, Asahina N, Kitano S and Kino Y: RNA-Seq data mining: Downregulation of NeuroD6 serves as a possible biomarker for alzheimer's disease brains. Dis Markers. 2014:1231652014. View Article : Google Scholar : PubMed/NCBI

23 

Pong WW, Walker J, Wylie T, Magrini V, Luo J, Emnett RJ, Choi J, Cooper ML, Griffith M, Griffith OL, et al: F11R is a novel monocyte prognostic biomarker for malignant glioma. PLoS One. 8:e775712013. View Article : Google Scholar : PubMed/NCBI

24 

Dixit AB, Banerjee J, Srivastava A, Tripathi M, Sarkar C, Kakkar A, Jain M and Chandra PS: RNA-seq analysis of hippocampal tissues reveals novel candidate genes for drug refractory epilepsy in patients with MTLE-HS. Genomics. 107:178–188. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Griffin NG, Wang Y, Hulette CM, Halvorsen M, Cronin KD, Walley NM, Haglund MM, Radtke RA, Skene JH, Sinha SR and Heinzen EL: Differential gene expression in dentate granule cells in mesial temporal lobe epilepsy with and without hippocampal sclerosis. Epilepsia. 57:376–385. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Liew CC, Ma J, Tang HC, Zheng R and Dempsey AA: The peripheral blood transcriptome dynamically reflects system wide biology: A potential diagnostic tool. J Lab Clin Med. 147:126–132. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Borovecki F, Lovrecic L, Zhou J, Jeong H, Then F, Rosas HD, Hersch SM, Hogarth P, Bouzou B, Jensen RV and Krainc D: Genome-wide expression profiling of human blood reveals biomarkers for Huntington's disease. Proc Natl Acad Sci USA. 102:11023–11028. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Jergil M, Forsberg M, Salter H, Stockling K, Gustafson AL, Dencker L and Stigson M: Short-time gene expression response to valproic acid and valproic acid analogs in mouse embryonic stem cells. Toxicol Sci. 121:328–342. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Dozawa M, Kono H, Sato Y, Ito Y, Tanaka H and Ohshima T: Valproic acid, a histone deacetylase inhibitor, regulates cell proliferation in the adult zebrafish optic tectum. Dev Dyn. 243:1401–1415. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Van Lint C, Emiliani S and Verdin E: The expression of a small fraction of cellular genes is changed in response to histone hyperacetylation. Gene Expr. 5:245–253. 1996.PubMed/NCBI

31 

Rakitin A, Kõks S, Reimann E, Prans E and Haldre S: Changes in the peripheral blood gene expression profile induced by 3 months of valproate treatment in patients with newly diagnosed epilepsy. Front Neurol. 6:1882015. View Article : Google Scholar : PubMed/NCBI

32 

Floriano-Sánchez E, Brindis F, Ortega-Cuellar D, Ignacio-Mejía I, Moreno-Arriola E, Romero-Morelos P, Ceballos-Vasquez E, Córdova-Espinoza MG, Arregoitia-Sarabia CK, Sandoval-Pacheco R, et al: Differential gene expression profile induced by valproic acid (VPA) in pediatric epileptic patients. Genes (Basel). 9(pii): E3282018. View Article : Google Scholar : PubMed/NCBI

33 

Kwan P, Arzimanoglou A, Berg AT, Brodie MJ, Allen Hauser W, Mathern G, Moshé SL, Perucca E, Wiebe S and French J: Definition of drug resistant epilepsy: Consensus proposal by the ad hoc task force of the ILAE commission on therapeutic Strategies. Epilepsia. 51:1069–1077. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Berg AT, Berkovic SF, Brodie MJ, Buchhalter J, Cross JH, van Emde Boas W, Engel J, French J, Glauser TA, Mathern GW, et al: Revised terminology and concepts for organization of seizures and epilepsies: Report of the ILAE commission on classification and terminology, 2005–2009. Epilepsia. 51:676–685. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

36 

Huang DW, Sherman BT, Tan Q, Collins JR, Alvord WG, Roayaei J, Stephens R, Baseler MW, Lane HC and Lempicki RA: The DAVID gene functional classification tool: A novel biological module-centric algorithm to functionally analyze large gene lists. Genome Biol. 8:R1832007. View Article : Google Scholar : PubMed/NCBI

37 

Brahma R, Gurumayum S, Naorem LD, Muthaiyan M, Gopal J and Venkatesan A: Identification of hub genes and pathways in Zika Virus infection using RNA-Seq Data: A network-based computational approach. Viral Immunol. 31:321–332. 2018. View Article : Google Scholar : PubMed/NCBI

38 

Kan AA, de Jager W, de Wit M, Heijnen C, van Zuiden M, Ferrier C, van Rijen P, Gosselaar P, Hessel E, van Nieuwenhuizen O and de Graan PN: Protein expression profiling of inflammatory mediators in human temporal lobe epilepsy reveals co-activation of multiple chemokines and cytokines. J Neuroinflammation. 9:2072012. View Article : Google Scholar : PubMed/NCBI

39 

Louboutin JP, Chekmasova A, Marusich E, Agrawal L and Strayer DS: Role of CCR5 and its ligands in the control of vascular inflammation and leukocyte recruitment required for acute excitotoxic seizure induction and neural damage. FASEB J. 25:737–753. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Pocock JM and Liddle AC: Microglial signalling cascades in neurodegenerative disease. Prog Brain Res. 132:555–565. 2001. View Article : Google Scholar : PubMed/NCBI

41 

Chui R and Dorovini-Zis K: Regulation of CCL2 and CCL3 expression in human brain endothelial cells by cytokines and lipopolysaccharide. J Neuroinflammation. 7:12010. View Article : Google Scholar : PubMed/NCBI

42 

De Simoni MG, Perego C, Ravizza T, Moneta D, Conti M, Marchesi F, De Luigi A, Garattini S and Vezzani A: Inflammatory cytokines and related genes are induced in the rat hippocampus by limbic status epilepticus. Eur J Neurosci. 12:2623–2633. 2000. View Article : Google Scholar : PubMed/NCBI

43 

Jin W, Zhang J, Lou D, Chavkin C and Xu M: C-fos-deficient mouse hippocampal CA3 pyramidal neurons exhibit both enhanced basal and kainic acid-induced excitability. Neurosci Lett. 331:151–154. 2002. View Article : Google Scholar : PubMed/NCBI

44 

Zhang J, Zhang D, McQuade JS, Behbehani M, Tsien JZ and Xu M: c-fos regulates neuronal excitability and survival. Nat Genet. 30:416–420. 2002. View Article : Google Scholar : PubMed/NCBI

45 

Tang Y, Glauser TA, Gilbert DL, Hershey AD, Privitera MD, Ficker DM, Szaflarski JP and Sharp FR: Valproic acid blood genomic expression patterns in children with epilepsy-a pilot study. Acta Neurol Scand. 109:159–168. 2004. View Article : Google Scholar : PubMed/NCBI

46 

Minnier J, Pennock ND, Guo Q, Schedin P and abd Harrington CA: RNA-Seq and expression arrays: Selection guidelines for genome-wide expression profiling. Methods Mol Bio. 1783:7–33. 2018. View Article : Google Scholar

47 

Zhao S, Fung-Leung WP, Bittner A, Ngo K and Liu X: Comparison of RNA-Seq and microarray in transcriptome profiling of activated T cells. PLoS One. 9:e786442014. View Article : Google Scholar : PubMed/NCBI

48 

Walker L and Sills GJ: Inflammation and epilepsy: The foundations for a new therapeutic approach in epilepsy? Epilepsy Curr. 12:8–12. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Guzik-Kornacka A, Sliwa A, Plucinska G and Lukasiuk K: Status epilepticus evokes prolonged increase in the expression of CCL3 and CCL4 mRNA and protein in the rat brain. Acta Neurobiol Exp (Wars). 71:193–207. 2011.PubMed/NCBI

50 

Owens GC, Huynh MN, Chang JW, McArthur DL, Hickey MJ, Vinters HV, Mathern GW and Kruse CA: Differential expression of interferon-γ and chemokine genes distinguishes Rasmussen encephalitis from cortical dysplasia and provides evidence for an early Th1 immune response. J Neuroinflammation. 10:562013. View Article : Google Scholar : PubMed/NCBI

51 

Arisi GM, Foresti ML, Katki K and Shapiro LA: Increased CCL2, CCL3, CCL5, and IL-1β cytokine concentration in piriform cortex, hippocampus, and neocortex after pilocarpine-induced seizures. J Neuroinflammation. 12:1292015. View Article : Google Scholar : PubMed/NCBI

52 

Srivastava A, Dixit AB, Paul D, Tripathi M, Sarkar C, Chandra PS and Banerjee J: Comparative analysis of cytokine/chemokine regulatory networks in patients with hippocampal sclerosis (HS) and focal cortical dysplasia (FCD). Sci Rep. 7:159042017. View Article : Google Scholar : PubMed/NCBI

53 

Saber S, Mahmoud AAA, Helal NS, El-Ahwany E and Abdelghany RH: Renin-angiotensin system inhibition ameliorates CCl4-induced liver fibrosis in mice through the inactivation of nuclear transcription factor kappa B. Can J Physiol Pharmacol. 96:569–576. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Guenther S, Bauer S, Hagge M, Knake S, Olmes DG, Tackenberg B, Rosenow F and Hamer HM: Chronic valproate or levetiracetam treatment does not influence cytokine levels in humans. Seizure. 23:666–669. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Ichiyama T, Okada K, Lipton JM, Matsubara T, Hayashi T and Furukawa S: Sodium valproate inhibits production of TNF-alpha and IL-6 and activation of NF-kappaB. Brain Res. 857:246–251. 2000. View Article : Google Scholar : PubMed/NCBI

56 

Ghosh C, Hossain M, Solanki J, Najm IM, Marchi N and Janigro D: Overexpression of pregnane X and glucocorticoid receptors and the regulation of cytochrome P450 in human epileptic brain endothelial cells. Epilepsia. 58:576–585. 2017. View Article : Google Scholar : PubMed/NCBI

57 

Lee NY, Riechmann P and Kang YS: The changes of P-glycoprotein activity by interferon-γ and tumor necrosis factor-α in primary and immortalized human brain microvascular endothelial cells. Blomol Ther (Seoul). 20:293–298. 2012. View Article : Google Scholar

58 

Feng W, Mei S, Zhu L, Yu Y, Yang W, Gao B, Wu X, Zhao Z and Feng F: Effects of UGT2B7, SCN1A and CYP3A4 on the therapeutic response of sodium valproate treatment in children with generalized seizures. Seizure. 58:96–100. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Mantle JL and Lee KH: A differentiating neural stem cell-derived astrocytic population mitigates the inflammatory effects of TNF-α and IL-6 in an iPSC-based blood-brain barrier model. Neurobiol Dis. 119:113–120. 2018. View Article : Google Scholar : PubMed/NCBI

60 

Alluri H, Grimsley M, Anasooya Shaji C, Varghese KP, Zhang SL, Peddaboina C, Robinson B, Beeram MR, Huang JH and Tharakan B: Attenuation of blood-brain barrier breakdown and hyperpermeability by calpain inhibition. J Biol Chem. 291:26958–26969. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Sakuma H, Tanuma N, Kuki I, Takahashi Y, Shiomi M and Hayashi M: Intrathecal overproduction of proinflammatory cytokines and chemokines in febrile infection-related refractory status epilepticus. J Neurol Neurosurg Psychiatry. 86:820–822. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Kuijpers M, van Gassen KL, de Graan PN and Gruol D: Chronic exposure to the chemokine CCL3 enhances neuronal network activity in rat hippocampal cultures. J Neuroimmunol. 229:73–80. 2010. View Article : Google Scholar : PubMed/NCBI

63 

Anaparti V, Pascoe CD, Jha A, Mahood TH, Ilarraza 3, Unruh H, Moqbel R and Halayko AJ: Tumor necrosis factor regulates NMDA receptor-mediated airway smooth muscle contractile function and airway responsiveness. Am J Physiol Lung Cell Mol Physiol. 311:L467–480. 2016. View Article : Google Scholar : PubMed/NCBI

64 

Zellinger C, Salvamoser JD, Soerensen J, van Vliet EA, Aronica E, Gorter J and Potschka H: Pre-treatment with the NMDA receptor glycine-binding site antagonist L-701,324 improves pharmacosensitivity in a mouse kindling model. Epilepsy Res. 108:634–643. 2014. View Article : Google Scholar : PubMed/NCBI

65 

Hung CC, Ho JL, Chang WL, Tai JJ, Hsieh TJ, Hsieh YW and Liou HH: Association of genetic variants in six candidate genes with valproic acid therapy optimization. Pharmacogenomics. 12:1107–1117. 2011. View Article : Google Scholar : PubMed/NCBI

66 

Gautier NM and Glasscock E: Spontaneous seizures in Kcna1-null mice lacking voltage-gated Kv1.1 channels activate Fos expression in select limbic circuits. J Neurochem. 135:157–164. 2015. View Article : Google Scholar : PubMed/NCBI

67 

Szyndler J, Maciejak P, Turzyńska D, Sobolewska A, Taracha E, Skórzewska A, Lehner M, Bidziński A, Hamed A, Wisłowska-Stanek A, et al: Mapping of c-Fos expression in the rat brain during the evolution of pentylenetetrazol-kindled seizures. Epilepsy Behav. 16:216–224. 2009. View Article : Google Scholar : PubMed/NCBI

68 

Cunningham JT, Mifflin SW, Gould GG and Frazer A: Induction of c-Fos and DeltaFosB immunoreactivity in rat brain by Vagal nerve stimulation. Neuropsychopharmacology. 33:1884–1895. 2008. View Article : Google Scholar : PubMed/NCBI

69 

Deransart C Lê BT, Marescaux C and Depaulis A: Role of the subthalamo-nigral input in the control of amygdala-kindled seizures in the rat. Brain Res. 807:78–83. 1998. View Article : Google Scholar : PubMed/NCBI

70 

Bozzi Y, Vallone D and Borrelli E: Neuroprotective role of dopamine against hippocampal cell death. J Neurosci. 20:8643–8649. 2000. View Article : Google Scholar : PubMed/NCBI

71 

Madsen TM, Bolwig TG and Mikkelsen JD: Differential regulation of c-Fos and FosB in the rat brain after amygdala kindling. Cell Mol Neurobiol. 26:87–100. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2019
Volume 18 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang Y and Wang Y: RNA‑seq analysis of blood of valproic acid‑responsive and non‑responsive pediatric patients with epilepsy. Exp Ther Med 18: 373-383, 2019
APA
Wang, Y., & Wang, Y. (2019). RNA‑seq analysis of blood of valproic acid‑responsive and non‑responsive pediatric patients with epilepsy. Experimental and Therapeutic Medicine, 18, 373-383. https://doi.org/10.3892/etm.2019.7538
MLA
Wang, Y., Li, Z."RNA‑seq analysis of blood of valproic acid‑responsive and non‑responsive pediatric patients with epilepsy". Experimental and Therapeutic Medicine 18.1 (2019): 373-383.
Chicago
Wang, Y., Li, Z."RNA‑seq analysis of blood of valproic acid‑responsive and non‑responsive pediatric patients with epilepsy". Experimental and Therapeutic Medicine 18, no. 1 (2019): 373-383. https://doi.org/10.3892/etm.2019.7538