1. Introduction
Primary central nervous system (CNS) and brain tumors are abnormal masses and tissue growth that have no biological function and disturb the neural processes. They are well known for their high mortality rate and increasing global cases each year [
1,
2,
3]. They cause serious health problems such as severe headaches, neurocognitive degeneration, loss of vision and hearing, seizures, and mental disorders, in some cases they trigger autoimmunity, and cancer [
4,
5,
6,
7]. Tumors can either be benign or malignant, benign tumors are less likely to spread and can be cured by surgery (WHO Grade I) while malignant tumors grow rapidly and cause serious health problems or worst, death (WHO Grade IV) [
8]. Recently, the fifth edition of the World Health Organization Classification of Tumors of the Central Nervous System (WHO CNS5) introduced twenty-two new tumor types and subtypes [
9]. CNS tumors may arise from different factors which may be sex, age, genetics, ethnicity/race, diet, socioeconomic status, environment (radiation, oncogenic pathogens, stress), and smoking [
10,
11,
12]. Malignant CNS tumors are fatal cancers that remain a major global health burden, it significantly increased up to 94.35% from 1999 to 2019 [
3,
13]. Currently, In the U.S., CNS cancer represents 1.3% of all new cancer cases with a 33.8% survival rate and ranked 16
th common type of cancer according to the National Cancer Institute’s (NCI) Surveillance, Epidemiology, and End Results (SEER) Program August 2023. Cancer has been one of the global public health burdens of the century, the increasing cases of CNS tumors require further knowledge in prognostic, imaging, therapeutics, surgery, alternative treatment, and drug discovery [
13,
14,
15,
16]. Understanding their molecular behavior based on gene expression prompts an opportunity for other treatment options.
Pediatric brain tumors are the most common brain cancer-related deaths [
17]. It ranges from rare to common to most malignant. There are numerous CNS tumors, and most of them are named after their tissue localization. Glioblastoma multiforme (WHO Grade IV) is the most common brain cancer, it forms from three different cell types, parenchyma neurons, astrocytes, and oligodendrocytes [
18]. Astrocytomas are tumors derived from astrocytes, according to the WHO grading system they can be grade II or III. Anaplastic astrocytomas (WHO Grade III) are diffusely infiltrating forming anaplasia following aggressive manifestation to higher cancer stage [
19]. On the other hand, Pilocytic astrocytomas (WHO Grade I) are slow-growing tumors that often can be treated with surgery, chemotherapy, and drugs [
20,
21]. Oligodendrogliomas are tumors that originated from oligodendrocytes, they are graded as WHO II, III, or IV, and their molecular hallmark is the codeletion of 1p and 19q chromosome arms [
22,
23,
24]. Ependymal cells derived from tumors are called ependymomas (WHO Grade II or III), they are found in the posterior fossa or supratentorial. Subgroups arising from posterior fossa are identified by Groups A and B. Group A tumors require more treatment with a low survival rate while Group B can survive through surgery [
25,
26,
27]. The following mentioned tumors fall under the umbrella of gliomas. There are more specific and rare types of CNS tumors such as Meningioma, Medulloblastoma, and Atypical teratoid/rhabdoid tumors. Medulloblastoma is the most common malignant tumor among children, formed at the lower back brain called the cerebellum. It has four distinct groups the Wingless Type (WNT), Sonic Hedgehog (SHH), Group 3, and Group 4 [
28,
29,
30,
31]. Meningiomas, on the other hand, are tumors originating from the meninges, they can be resolved by surgery as they’re benign, however, tumor relapse is frequently observed [
32]. Atypical teratoid/rhabdoid tumors are aggressive tumors that have distinguished loss function of SMARCB1 protein [
33,
34]. Central nervous system primitive neuroectodermal tumors grow in the extra cerebellar site, they can also derive from the pineal gland forming pineoblastoma, this tumor has been correlated to the WNT/β-catenin pathway disrupting normal CNS development [
35]. A comparative study of human and mouse gene expression profile revealed that experimental PNETs corresponds to AT/TR [
36]. Numerous CNS tumors have not been classified or are still unknown due to the continuous increase in new cases of tumors that are entirely unfamiliar to cancer physicians and biologists. Access to early diagnosis and screening and a poor understanding of cancer biology may contribute to this factor. The molecular characteristics of the tumor may serve as a key to further classifying tumors, particularly tumors with the same morphological characteristics but behave differently.
It is important to identify genetic markers for specific tumors as they help to identify, classify, and grade tumors [
23,
24,
37]. These genetic markers may serve as a potential drug target, biomarker, distinguish gene regulator, and biological insight into the function and mechanism of tumorigenesis. There are numerous options for treating tumors. Commonly, surgery is the primary option to remove tumors, however, the high-risk failure of cranioplasty implant surgery after resection of the malignant infiltrating tumor in the skull may not be suitable, especially for pediatric patients [
38]. Radiotherapy may be suitable for people with financial stability, however, an increased risk of secondary tumors in adults after radiotherapy was reported in the literature [
39]. Clinical benefits of immunotherapy against brain metastases are promising, however, their response to primary tumors such as glioblastoma is poor [
40]. Chemotherapy aided by nanomicelles as a tumor-targeting drug delivery complex proposed an effective strategy for treating glioma [
41]. There are several options available for treating tumors such as hormone therapy, bone marrow transplant, oncolytic virus therapy, targeted therapy, and combinatory treatment and/or therapy [
41,
42,
43,
44,
45,
46,
47]. Medical interventions can often extend the lifespan of a tumor patient, but it is not entirely cured. The need for more tumor treatment options is a leading concern in mitigating CNS tumors as a global burden.
Recently, computational approaches in biology provided avenues for studying gene expression profiles, advancement in the screening, detection, potential biomarkers, new drug candidates, and candidate repurposed drugs [
8,
37,
48,
49]. The drug repurposing approach employed known drugs for new therapeutic purposes. In cancer, the search for interesting new drugs is rapidly increasing due to the rise of new types and subtypes. Existing anticancer drugs have several adverse side effects, drug repurposing of a non-cancer drug may pave the way for better tumor treatment. Therefore, drug repurposing is a novel approach to reduce the time in new drug discovery utilizing approved drugs for new use [
50]. Translational bioinformatics and computational oncology proposed that mutation-specific therapy may serve as a key pathway in treating tumors [
50]. In gliomas, a molecular subtype of mouse glioblastomas predicted candidate drugs through gene-drug interaction using a computational drug repurposing approach [
51]. A deep learning approach to expression profiles of tumors and cheminformatics may reveal opportunities in drug repurposing [
52,
53]. Understanding the expression profile of CNS tumors may contribute insights into tumor biology and new drug discovery [
54,
55]. In the case of CNS tumors, the use of drug repurposing provides a rapid, cost-efficient, and lower risk of drug side effects.
In this study, differentially expressed genes from expression profiles of different CNS tumors (from DNA microarray) provides a hallmark to identify gene target [
37,
55]. The current study searched for the shared differentially expressed genes from the four expression arrays of various CNS tumors and reconstructed co-expression networks. Here, we utilized computational web tools and databases, such as GEO2R, Network Analyst, STRING, DAVID, and DGIdb for co-expression network analysis. Co-expression networks revealed the correlation of hub genes, transcription factors, and miRNAs in finding candidate repurposed drugs that may treat CNS tumors [
56,
57]. Our study may serve as a significant strategy in the future of drug development against CNS tumors.
4. Discussion
Microarray expression profile is a technique to identify the behavior of genes. It has been used to compare diseased tissue samples to normal. Tumor samples are commonly used to extract RNAs, through reverse transcription RNAs are converted to DNA and used in DNA microarray chips. Expression profiles provide insights into the behavior of the tumor based on clinical samples. Here, we used DNA microarray datasets with surgically removed tumor samples to identify genes that are behaving differently when compared to normal or non-tumor samples. The datasets were GSE66354, GSE68848, GSE74195, and GSE43290 which are publicly available in the NCBI GEO database. To identify genes behaving differently, also known as, differentially expressed genes, we used an online web tool, such as GEO2R. GEO2R uses R packages from Bioconductor projects based on the R computing language. Based on the clinical classification of the tumor samples we were able to compare the gene expressed on each tumor type. However, in this study we did not perform the sampling collection, RNA extraction, and microarray expression, instead, we utilized openly available microarray datasets.
The use of the computational approach has been growing in popularity due to the shorter time frame it requires to understand diseases in terms of prognosis, diagnosis, screening, therapeutics, and treatment. Gene co-expression network analysis has been a growing trend for understanding molecular biology and the characteristics of certain diseases. It has been used to identify important genes that may serve as biomarkers and specific molecular targets for treatment [
37,
88]. Further, CNS tumors have become the primary tumor emerging in pediatric patients [
17,
27,
60,
89]. CNS tumors are often associated with debilitating diseases or conditions, such as seizure, stroke, brain edema, severe headaches, mental disorders, neurocognitive impairment, and ultimately cancer [
7,
90,
91]. Glioblastoma multiforme is a terminal brain cancer that is often the result of the metastasis and malignancy of high-grade CNS tumors such as astrocytoma and oligodendroglioma [
18]. Other malignant brain tumors include CNS lymphoma, ependymoma, medulloblastoma, and meningioma. Molecular biology in understanding CNS tumors is still infancy due to the continuous surface of new types and subtypes with increasing cases each year [
3,
13]. In this present study, gene co-expression analysis of PA, ACM, ODG, GBM, EPN, MED, MEN, ATRT, and PNET tumors, using four NCBI GEO expression profiles to investigate common differentially expressed genes between the tumors and identify candidate repurposed drug for the preserved genes and their co-regulating transcription factors. In this study, we highly utilized web tools and online databases to ease the analysis and provide a unique computational approach for drug repurposing strategy against CNS tumors.
Table 4 displays the fourteen hub genes identified from common differentially expressed genes in GSE66354, GSE68848, GSE74195, and GSE43290 when we compare the tumor group and normal tissue samples. The shared differentially expressed genes were utilized for co-expression network, we've identified 10 upregulated (CACNA1A, DNM1, GABRA1, GRIA2, MAPT, SLC17A7, SNAP25, SNAP91, STXBP1, and SYT1) and 4 downregulated (COL1A1, COL6A2, FBN2, and FN1) hub DEGs. Voltage-gated calcium ion channels, such as CACNA1A, have been associated with cell proliferation, differentiation, apoptosis, and metastasis. CACNA1A was reported to be significantly downregulated in brain tumors such as GBM, AA, ATRT, PNET, MED, ODG, and ACM, where it served as a tumor suppressor gene. Moreover, CACNA1A was reported to be downregulated in colorectal, esophagus, gastric, and breast cancer [
92]. DNM1 has been associated with epileptic encephalopathy patients, particularly the Arg237Trp variant [
93]. It was also validated for its liked to several cancer-related pathways and positive correlation with Neutrophils, Tregs, NK cells, and macrophage infiltration in colon cancer [
94]. Neurotransmitters have been linked to cancer cell proliferation. The downregulation of the neurotransmitter receptor gene, GABRA1, was reported to be associated with GBM and a positive prognosis of low-grade glioma with a negative correlation on genes responsible for immune response and inflammation [
95]. In the coculture of neurons and brain metastases tumors, neurotransmitter receptor genes, such as GRIA2, were observed in early expression in breast and lung tumors [
96]. GRIA2 was also reported to be downregulated in chemosensitive and chemoresistant advanced serous papillary ovarian adenocarcinoma and a promising prognosis of patients [
97]. In MAPT gene expression, it was determined that upregulation increases the survival rate of patients with low-grade glioma [
98]. Therefore, the downregulation of MAPT contributes to the malignancy of the tumor. In experimental validation on protein and RNA levels, SLC17A7 was reported to act as a bivalent tumor suppressor gene in GBM when compared to normal brain tissues. SLC17A7 also inhibits cell proliferation, migration, and invasion of GBM [
99]. Likewise, SNAP25 inhibits carcinogenesis of glioma by fostering control in glutamine metabolism and regulating glutaminase expression [
100]. Downregulation of SH3GL2 and SNAP91 in GBM was reported and inversely correlated with glioma grades [
101]. Like DNM1, STXBP1 was reported an association with encephalopathies, however, studies involving CNS tumors and cancer were lacking [
102]. Dysregulation of the SYT1 leads to severe neurodegenerative impairment as reported in the literature [
103]. COL1A1 and COL6A2 have been reported to cause infiltration of CD4 + T and CD8 + T cells, neutrophils, macrophages, and dendritic cells in low-grade brain tumors and gliomas [
104,
105]. FBN2 is known to contribute to connective tissue disorders due to matrix sequestering of the transforming growth factor-β (TGF-β) family. TGF-β activation contributes to tumor angiogenesis and carcinogenesis. FBN1 was reported to have higher binding affinity compared to FBN2, therefore, investigating FBN1 on brain tumors in future studies is promising [
106]. FN1 was reported expressed in glioblastoma and low-grade glioma through the bioinformatics approach, it is important in metastasis and malignancy of tumors [
107]. Based on the literature, the downregulated genes that we have identified have tumor-suppressing behavior while the upregulated genes are involved in pathways that contribute to tumor progression. It is interesting to speculate that switching the regulation of these genes may change the tumor behavior to further suppression.
In each nine tumors, we compared them against normal brain tissues to identify the differentially expressed genes.
Table 12 displays the five most upregulated and downregulated upon comparison while in
Figure A4 are the volcanic plot on each tumor. GABRA1, SLC12A5, and SNAP25 hub genes that were downregulated based on the co-expression network, appeared to be associated with certain CNS tumors. The GABRA1 and MFSD4A genes displayed a downregulation for ACM, ODG, and GBM or the gliomas while TOP2A was observed to be upregulated. Astrocytoma and oligodendroglioma shared upregulation of SOX4 and HES6 while RGS4 was downregulated. Oligodendroglioma and glioblastoma were observed to share the NDC80 (upregulated) and GJB6 (downregulated) genes. In astrocytoma and glioblastoma, both were observed to have CACNG3 downregulation. Interestingly, medulloblastoma, primitive neuroectodermal tumor, and meningioma shared the downregulation of the MBP gene. PNET shared upregulation of COL1A1 with medulloblastoma while downregulation of SNAP25 with meningioma. Surprisingly, ependymoma was observed to share downregulation of SL12A5 with pilocytic astrocytoma and SV2B with ATRT. PA and ATRT shared two downregulated genes, PACSIN1 and GJB6. VSNL1 appears to be suppressed in ATRT, medulloblastoma, and oligodendroglioma. The TOP2A gene was observed to be active in four CNS tumors, such as ATRT, ACM, ODG, and GBM. In literature, TOP2A over-expression was observed to contribute to radioresistance in medulloblastoma patients [
108]. Likewise, in glioma patients, TOP2A is highly expressed [
109]. Our study suggests that TOP2A may have an oncogenic role in CNS tumors. GJB6 was found to be suppressed in ATRT, PA, ODG, and GBM. It is known to produce connexin 30 mainly plays a role in nervous system cells, which might suggest the correlation of CNS tumors with neurodegenerative diseases and their debilitating effects [
110]. The DEGs for each tumor considered can be accessed in Supplementary Data 6. Our study may contribute to future understanding of CNS tumors and the similarity in the behavior of the tumors. This study provides insight into the shared genes that were behaving differently in CNS tumors. They may serve as prognostic biomarkers or future drug targets.
The SYT1, DNM1, and STXBP1 are HDEGs that have been associated with debilitating neurological diseases while reports on their role in CNS tumors were still lacking. Here, we identified ten downregulated genes that may serve as tumor-suppressing genes with inverse correlations with CNS tumor proliferation, invasion, metastasis, malignancy, and differentiation. It is interesting to investigate further the role of immune response and neurotransmitters in contributing to tumorigenesis. The four upregulated contribute to the infiltration of neurotransmitters contributing to tumor proliferation. In this present study, fourteen identified HDEGs as promising therapeutic targets and biomarkers in CNS tumors. Differential expression of genes provides insight into tumor behavior based on clinical characteristics and samples. To further validate the tumor-suppressing behavior of the downregulated genes and tumor-contributing behavior of upregulated genes we highly encourage experts to perform In vivo and In vitro experimental studies.
Three clusters were identified as preserved in the co-expression network, two represent the downregulated HDEGs, and one for the upregulated HDEGs. In this study, enrichment analysis in each cluster was achieved (
Figure 7). In the enrichment analysis, GO BP, synaptic communication was significant in downregulated groups. This biological process provides insight into glutamatergic neuron-to-brain tumor synaptic communication [
111]. For the upregulated group cell adhesion, bone trabecula formation, and wound healing are enriched. It is interesting to speculate that cell adhesion contributes to tumor proliferation and the trabeculae process responsible for aggressive size growth. In GO CC cluster 1, the plasma membrane, neuronal cell body, and synapse while in cluster 2, the cytoplasm, perinuclear region of cytoplasm, and microtubule are the enriched cellular components. Cellular responses are highly influenced by microtubule alteration which may lead to tumor development and chemoresistance [
112]. Cluster 1 genes are mainly involved in neurons for the transmission of nervous impulses while cluster 2 facilitates cellular structure. The downregulation of cluster 1 genes provided insight into the neurological impairment in tumor patients while cluster 2 genes support the abnormal formation of tumor cells. Furthermore, enriched GO CC of cluster 3 are mainly extracellular matrix, extracellular space, and extracellular region. The extracellular matrix serves as the microenvironment for the cells, dysregulation may lead to the development of cancer. that was mainly responsible for tumor progression [
113]. The GO MF highly preserved the protein kinase binding and identical protein binding functions. It has been reported that the protein kinase C functions as a tumor suppressor while its isoforms contribute to tumor progression [
114]. In KEGG pathways, cluster 1 associated its genes with Nicotine addiction, Synaptic vesicle cycle, GABAergic synapse, and Retrograde endocannabinoid signaling. Mostly the pathways involve the disturbance of neurotransmitters in the brain [
111,
115,
116]. Cluster 2 KEGG preserved pathways were Pathways of neurodegeneration - multiple diseases and Parkinson's disease both relative in neurocognitive impairment that hijacks glutamate signaling [
117]. In cluster 3, KEGG pathways were ECM-receptor interaction and Focal adhesion, which is essential for cell anchoring and migration, these pathways were also reported in gastric cancer [
118]. The GO and KEGG enrichment analysis provided insight into the molecular and biological functions of the cluster genes. Enrichment data of the top 5 DEGs in each tumor can be accessed in Supplementary Data 5. These may serve as key insights for the neurologist, oncologist, and biologist in assessing CNS tumors.
In this study, we explored the five most upregulated and downregulated genes in each of the nine CNS tumors for their KEGG and GO pathways. Observed in
Figure 12 is the chord diagram of the following genes considered. We discovered that the upregulated genes mostly contribute to the extracellular region, extracellular space, and extracellular exosome. COL3A1 and COL1A1 appear to be involved in most of the pathways and their active expression is interesting to explore towards the behavior of different CNS tumors. Meanwhile, in the downregulated group, plasma membrane, cytosol, and neuron projection were highly enriched GO pathways. Among the suppress genes, GABRA1, GABRB2, and GABRBG2 are well involved in the GO and KEGG pathways. It is interesting to speculate that the suppression of these genes contributes to tumor progression. Their possible tumor-suppressing characteristics may open a door to drug development. This study opens a prospect for understanding CNS tumors for the future of drug development for anti-tumor.
Transcription factors and miRNA play a role in the coregulation of genes in post-transcription. Through the co-expression network of HDEGs, TFs, and miRNAs significant co-regulating TFs and miRNAs were identified. In this study, we have identified 33 transcription factors (
Table 5) and 46 miRNAs (
Table 6). Among the miRNAs, we considered five significant miRNA families, the
let-7 family
, the mir-124 family, the
mir-1 family, the
mir-103 family, and the
mir-27 family. The
let-7 mRNA family was reported as a promising biomarker and modulated cancer stemlike cells [
119,
120]. Expression of
mir-124 in neurons contributes to suppressing tumor malignancy through coregulation of STAT3 and EZH2 [
121]. Suppressive activity of
mir-1 against GBM has been reported through FN1 targeting [
122]. Likewise,
mir-103 suppresses glioma cell proliferation and invasion by interacting with BDNF [
123]. Lastly, the
mir-27 family was reported to exhibit tumor suppression in colon cancer, pancreatic cancer, breast cancer, bladder cancer, and hepatocellular carcinoma [
124]. It is interesting to examine and clinically validate these miRNA families in CNS tumor treatment. They might as well serve as biomarkers in examining tumor patients.
Table 13.
Proposed candidate repurposed drugs and their regulatory effect on gene target, indication, and mechanism.
Table 13.
Proposed candidate repurposed drugs and their regulatory effect on gene target, indication, and mechanism.
Drug |
HDEG |
TF |
Upregulated gene |
Class |
Quercetin |
MAPT |
NFKB1, PRKCA, RELA |
SNCA |
Kinase inhibitors |
Vorinostat |
TUBB2A, TUBB4A |
EZH2, MYC, TP53 |
DNAJC6, DNM3, KIF5C, SLC17A7, STXBP1, SYT1, TUBBA2A |
Antineoplastic agents |
Moving on, the candidate repurposed drugs were identified by the DGIdb online database. Here, we considered the HDEGs and the TFs as drug targets, resulting in 32 candidate drugs. The Drug-TFs network and Drug-HDEGs network were reconstructed (
Figure 9 and
Figure 10) to identify drugs with a high degree of interaction. Curcumin and ocriplasmin were the considered candidate drugs in drug-HDEGs. Curcumin interacts with downregulated genes MAPT, TUBB2A, and TUBB4A while ocriplasmin interacts with upregulated genes COL1A1, COL6A2, and FN1. However, the two-lead drug failed CMAP analysis for drug validation. The Drug-TFs network showed cisplatin and daunorubicin as lead candidate repurposed drugs, both interacting with MAPT. Both have five TF interactions, cisplatin has GABPA, MYC, SMARCA4, STAT1, and TP53 while daunorubicin has CBFB, GATA1, IKZF1, THRB, and WT1. In the CMAP analysis, thirteen drugs were validated for their regulatory effect on HDEGs, namely, colchicine, doxorubicin, indoprofen, levothyroxine, methotrexate, nifedipine, paclitaxel, quercetin, raloxifene, resveratrol, trifluoperazine, vinblastine, and vorinostat. Based on the regulatory effect of the candidate drug, colchicine, and indoprofen are not suggestible. Colchicine will further downregulate the downregulated genes which were DNM3, SLC12A5, and TUBB2A while indoprofen will upregulate the upregulated gene COL1A1. Moreover, doxorubicin, levothyroxine, methotrexate, nifedipine, paclitaxel, raloxifene, resveratrol, trifluoperazine, and vinblastine were observed to have gene regulations that contradict the HDEGs. In considering drug contradictions with gene targets, drugs that upregulate the downregulated genes or vice versa are promising drug candidates for repurposing. Here, we proposed two major candidate repurposed drugs, quercetin and vorinostat. Furthermore, quercetin and resveratrol have been reported in the literature on their inhibitory effect on tumor proliferation. Quercetin is a polyphenol with antioxidant properties, its anticancer mechanism focuses on apoptosis through PI3K/Akt/mTOR, Wnt/β-catenin, and RAS/MAPK/ERK1/2 pathways, Upregulation of P53 protein through ROS generation and ER stress was also illustrated in the literature. Tumor invasion, proliferation, and migration are suppressed by MMP-9, PLD-1, ecto-5’-NT/CD73 inhibition, decrease in FN1, and cell arrest by G2 immune checkpoint [
125] In rodent models, vorinostat has prevented brain metastases by 62% [
126]. Vorinostat was also reported in peripheral mononuclear cells and tumor tissue acting as histone deacetylase (HDAC) inhibitors as an antitumor agent [
127]. In rodent models, vorinostat was reported to hinder brain metastasis of triple-negative breast cancer due to its permeability in the brain. Identified drugs based on the drug-HDEGs and drug-TFs networks may pose a future use in treating CNS tumors. Currently, surgery is the most promising tumor removal or treatment. However, due to the reoccurrence of secondary tumors, metastases on different tissues or organs, and localization of tumors in areas of the body where surgery is not possible, drug treatment may be useful. Further validation on the candidate drugs might be needed such as their toxicity, administration, absorption, distribution, and excretion. The drug must also pass the blood-brain barrier if it will be used in treating CNS tumors.
In this present study, we have reported 17/74 (upregulated/downregulated) common differentially expressed genes and fourteen hub genes that are significant in CNS tumors. The 33 transcription factors may lead to further drug discovery and repurposing studies such as molecular docking, target binding sites, and molecular dynamics simulations. We reported five miRNA families that play a major role in CNS tumors. Their potential as biomarkers and therapeutic targets for tumor mitigation is promising in the landscape of cancer treatment. Furthermore, 32 speculated antitumor drugs have been identified and thirteen were validated. Among the validated candidate repurposed drugs only two fit the regulatory requirement for gene-targeted drugs, quercetin and vorinostat. Future experimental studies may be required to further validate the candidate repurposed drugs. Current challenges in CNS tumors are mostly associated with their debilitating effect on patients, neurocognitive impairment, and localization and infiltration in areas of the body making surgery removal of tumors difficult. In this present study, we utilized the DNA microarray of various CNS tumors to provide insights into their molecular biology. Our study contributes to society by providing prospects that may be useful in the future of drug treatment to mitigate CNS tumors as a global burden.
Figure 1.
Volcano plot of differentially expressed genes for (a) GSE66354, (b) GSE68848, (c) GSE74195, and (d) GSE43290. Highlighted genes are differentially expressed genes following the adjusted p-value cutoff of ≤ 0.05 and log2FC threshold of ≥ 1 (upregulated) and ≤ -1 (downregulated). Red represents upregulated genes while blue represents downregulated genes.
Figure 1.
Volcano plot of differentially expressed genes for (a) GSE66354, (b) GSE68848, (c) GSE74195, and (d) GSE43290. Highlighted genes are differentially expressed genes following the adjusted p-value cutoff of ≤ 0.05 and log2FC threshold of ≥ 1 (upregulated) and ≤ -1 (downregulated). Red represents upregulated genes while blue represents downregulated genes.
Figure 2.
Venn diagram of intersected differentially expressed genes from GSE66354 (blue), GSE68848 (red), GSE74195 (green), and GSE43290 (yellow) datasets. Common DEGs are (a) upregulated and (b) downregulated.
Figure 2.
Venn diagram of intersected differentially expressed genes from GSE66354 (blue), GSE68848 (red), GSE74195 (green), and GSE43290 (yellow) datasets. Common DEGs are (a) upregulated and (b) downregulated.
Figure 3.
Protein-protein interaction network for the common differentially expressed genes of different CNS tumors obtained from the STRING database. The nodes are 90 and the number of edges is 185. Disconnected nodes were removed.
Figure 3.
Protein-protein interaction network for the common differentially expressed genes of different CNS tumors obtained from the STRING database. The nodes are 90 and the number of edges is 185. Disconnected nodes were removed.
Figure 4.
Co-expression network of the DEGs. Green nodes are upregulated, and blue nodes are downregulated.
Figure 4.
Co-expression network of the DEGs. Green nodes are upregulated, and blue nodes are downregulated.
Figure 5.
Top 10 hub genes identified by Cytoscape-cytoHubba plugin through MCC algorithm. Rank is based on the intensity of color. Red is the highest rank to yellow is the lowest rank.
Figure 5.
Top 10 hub genes identified by Cytoscape-cytoHubba plugin through MCC algorithm. Rank is based on the intensity of color. Red is the highest rank to yellow is the lowest rank.
Figure 6.
Sub-networks based on clusters identified by the ClusterViz in the co-expression network. (a) cluster 1, (b) cluster 2, and (c) cluster 3. Highlighted nodes (yellow) are the highest edges (red) on their sub-network.
Figure 6.
Sub-networks based on clusters identified by the ClusterViz in the co-expression network. (a) cluster 1, (b) cluster 2, and (c) cluster 3. Highlighted nodes (yellow) are the highest edges (red) on their sub-network.
Figure 7.
Bubble plot of the KEGG and GO enrichment pathways of DEGs. (a) Cluster 1, (b) Cluster 2, and (c) Cluster 3.
Figure 7.
Bubble plot of the KEGG and GO enrichment pathways of DEGs. (a) Cluster 1, (b) Cluster 2, and (c) Cluster 3.
Figure 8.
TFs-HDEGs-miRNAs coregulatory network. (a) Cluster 1, (b) Cluster 2, and (c) Cluster 3. Blue circle nodes are the hub genes, green square nodes are the transcription factors, and red triangle nodes are the miRNAs.
Figure 8.
TFs-HDEGs-miRNAs coregulatory network. (a) Cluster 1, (b) Cluster 2, and (c) Cluster 3. Blue circle nodes are the hub genes, green square nodes are the transcription factors, and red triangle nodes are the miRNAs.
Figure 9.
Drug-Hub DEGs network. Blue circle nodes are downregulated genes from clusters 1 and 2, Green circle nodes are the upregulated genes from cluster 3, pink hexagons are the drugs with single hits, and purple diamonds are drugs that interact with more than one gene.
Figure 9.
Drug-Hub DEGs network. Blue circle nodes are downregulated genes from clusters 1 and 2, Green circle nodes are the upregulated genes from cluster 3, pink hexagons are the drugs with single hits, and purple diamonds are drugs that interact with more than one gene.
Figure 10.
Drug-Transcription factor network. Green box nodes are the transcription factors while the pink hexagon is the drugs with a single hit and purple diamond nodes are drugs with more than two hits.
Figure 10.
Drug-Transcription factor network. Green box nodes are the transcription factors while the pink hexagon is the drugs with a single hit and purple diamond nodes are drugs with more than two hits.
Figure 11.
MiRNA families enriched from the miRNAs considered in this study. The number inside the parenthesis is the miRNA count. -Log10 (P-value) describes the enrichment of the miRNA family.
Figure 11.
MiRNA families enriched from the miRNAs considered in this study. The number inside the parenthesis is the miRNA count. -Log10 (P-value) describes the enrichment of the miRNA family.
Figure 12.
Chord diagram of the top 5 genes in each of the nine CNS tumors corresponding KEGG and GO pathways. (a) upregulated genes and (b) downregulated genes.
Figure 12.
Chord diagram of the top 5 genes in each of the nine CNS tumors corresponding KEGG and GO pathways. (a) upregulated genes and (b) downregulated genes.
Table 1.
Summary of central nervous system tumor datasets used and analyzed in this study.
Table 1.
Summary of central nervous system tumor datasets used and analyzed in this study.
NCBI GEO Accession |
GSE66354 a |
GSE68848 b |
GSE74195 c |
GSE43290 d |
Published |
February 27, 2015 |
May 14, 2015 |
October 21, 2015 |
January 05, 2013 |
Type |
Expression profiling by array |
Conditions |
2 AA 17 ATRT 29 EPN-PFA 26 EPN-PFB 9 EPN-ST 19 GBM 4 MED-G3 7 MED-G4 8 MED-SHH 15 PA 13 normal brain tissue |
148 ACM 228 GBM 67 ODG 67 unknown 11 mixed 1 unclassified 30 tumor cell lines 28 normal brain tissue |
13 EPN 1 EPN-BM 5 PNET 27 MED 5 normal cerebellum tissue |
47 MEN 4 normal meninges
|
Platform |
GPL570 [HG-U133_Plus_2] Affymetrix Human Genome U133 Plus 2.0 Array |
GPL96 [HG-U133A] Affymetrix Human Genome U133A Array |
RNA Source |
Surgical CNS tumors and normal brain tissue |
No. of Samples |
149 |
580 |
51 |
51 |
Table 2.
List of common differentially expressed genes from four CNS tumors datasets.
Table 2.
List of common differentially expressed genes from four CNS tumors datasets.
Differentially expressed genes |
Total |
Genes |
Upregulated |
17 |
COL1A1, PTBP1, TYMS, KDELR2, SOX4, PDLIM7, FZD2, ZFP36L2, COL6A2, RUVBL1, PPIB, FN1, SLC16A1, FBN2, MDK, GRN, TCF3 |
Downregulated |
74 |
RTN1, PHYHIP, ALDOC, NEFH, PGBD5, CABP1, SYT13, DNM1, IDS, CAMK2B, MAP7, RUNX1T1, KCNAB1, SNCA, EPB41L3, MAGI1, KIF5C, GABRA1, ADARB1, ZNF365, TAGLN3, OPCML, MOBP, PDE4DIP, PEG3, TRPM3, RAPGEF5, NDRG4, STXBP1, NSG1, GABBR2, SH3GL2, EFR3A, PLP1, STMN2, CYFIP2, HSPA12A, MCTP1, SYNGR3, VSNL1, CACNA1A, RCAN2, OLFM1, MAPT, SEPTIN4, GUCY1B3, GABRB1, SNAP25, RIMS3, ITPR1, MYO5A, GABARAPL1, DUSP8, SLC17A7, DNAJC6, DNM3, SYT1, DOCK9, TUBB2A, HOMER1, FAIM2, EHD3, DYNC1I1, SLC12A5, NRIP3, PPP1R16B, GABARAPL1, PCP4, ADAM22, RUNDC3A, SNAP91, TUBB4A, GRIA2, SPOCK1 |
Table 3.
Genes in each module were identified by ClusterViz and the top 10 hub genes were calculated by cytoHubba. Numbers inside the parenthesis indicate node score.
Table 3.
Genes in each module were identified by ClusterViz and the top 10 hub genes were calculated by cytoHubba. Numbers inside the parenthesis indicate node score.
Cluster |
Gene names |
Cluster 1 (5) |
GRIA2 (7), STXBP1 (6), DNM1 (5), CACNA1A (5), GABRA1 (4), MAPT (3), SNAP91 (3), SLC12A5 (3) |
Cluster 2 (5) |
SNAP25 (10), SNCA (8), KIF5C (7), SH3GL2 (7), SYT1 (7), SLC17A7 (5), DYNC1I1 (5), NEFH (4), TUBB2A (4), STMN2 (3), TUBB4A (3), DNAJC6 (3), MOBP (2), CAMK2B (2), DNM3 (2) |
Cluster 3 (4) |
COL1A1 (3), COL6A2 (3), FBN2 (3), FN1 (3) |
Top 10 |
GRIA2 (9), SLC17A7 (9), SNAP25 (9), STXBP1 (9), SYT1 (9), DNM1 (8), CACNA1A (7), GABRA1 (6), MAPT (6), SNAP91 (6) |
Table 4.
Hub DEGs and their description.
Table 4.
Hub DEGs and their description.
Gene name |
Gene description |
CACNA1A |
Calcium voltage-gated channel subunit alpha1 A |
DNM1 |
Dynamin 1 |
GABRA1 |
Gamma-aminobutyric acid type A receptor subunit alpha1 |
GRIA2 |
Glutamate ionotropic receptor AMPA type subunit 2 |
MAPT |
Microtubule-associated protein tau |
SLC17A7 |
Solute carrier family 17-member 7 |
SNAP25 |
Synaptosome-associated protein 25
|
SNAP91 |
Synaptosome-associated protein 91
|
STXBP1 |
Syntaxin binding protein 1 |
SYT1 |
Synaptotagmin 1 |
COL1A1 |
Collagen type I alpha 1 chain |
COL6A2 |
Collagen type VI alpha 2 chain |
FBN2 |
Fibrillin 2 |
FN1 |
Fibronectin 1 |
Table 5.
Transcription factors co-express with hub genes.
Table 5.
Transcription factors co-express with hub genes.
Transcription factors |
Gene targets |
Cluster |
ARNT |
CAMK2B, SLC17A7, SYT1 |
Cluster 2 |
ATF1 |
NEFH, SNAP25, TUBB4A & FN1, COL1A1 |
Cluster 2 & Cluster 3 |
ATF2 |
NEFH, SH3GL2, SNAP25 |
Cluster 2 |
ATF4 |
CAMK2B, NEFH, SNAP25 |
Cluster 2 |
BCL11A |
SNAP25, SLC17A7, TUBB4A |
Cluster 2 |
CREB1 |
NEFH, SH3GL2, SLC17A7, SNAP25 |
Cluster 2 |
CTCF |
CAMK2B, SLC17A7, TUBB4A |
Cluster 2 |
E2F5 |
CAMK2B, SLC17A7, TUBB4A |
Cluster 2 |
EGR1 |
GRIA2, MAPT, STXBP1 & CAMK2B, NEFH, SLC17A7, TUBB2A & FN1, COL1A1 |
Cluster 1, Cluster 2 & Cluster 3 |
ELF1 |
DNAJC6, SLC17A7, TUBB4A |
Cluster 2 |
ELK1 |
DNAJC6, SLC17A7, SNCA |
Cluster 2 |
EZH2 |
GRIA2, SNAP91, SLC12A5 & KIF5C, SH3GL2, SLC17A7, STMN2, TUBB4A |
Cluster 1 & Cluster 2 |
KDM5B |
DNM3, SLC17A7, TUBB2A |
Cluster 2 |
KLF1 |
KIF5C, SLC17A7, TUBB2A |
Cluster 2 |
MAX |
GABRA1, MAPT, STXBP1 & DNAJC6, NEFH, SLC17A7 |
Cluster 1 & Cluster 2 |
MEF2A |
CAMK2B, SNAP25, SYT1 |
Cluster 2 |
PHF8 |
DNM3, SLC17A7, TUBB2A |
Cluster 2 |
PRKCA |
DNM1, GRIA2, STXBP1 |
Cluster 1 |
REST |
GRIA2, MAPT, SLC12A5 & DNAJC6, NEFH, SNAP25, STMN2, TUBB2A |
Cluster 1 & Cluster 2 |
RREB1 |
CAMK2B, NEFH, SYT1 |
Cluster 2 |
SAP30 |
DNM3, SLC17A7, TUBB2A |
Cluster 2 |
SP1 |
CAMK2B, DNAJC6, DYNC1I1, SLC17A7, TUBB2A& FN1, COL1A1 |
Cluster 2 & Cluster 3 |
SP3 |
SNAP25, STMN2, TUBB2A |
Cluster 2 |
SREBF1 |
DNM1, SNAP91, STXBP1 |
Cluster 1 |
TFAP2A |
MAPT, SNAP91, STXBP1 & SLC17A7, SNAP25, SYT1 |
Cluster 1 & Cluster 2 |
TFAP2C |
DNAJC6, SNAP25, SLC17A7 |
Cluster 2 |
TFAP4 |
DNAJC6, SH3GL2, SNCA |
Cluster 2 |
YY1 |
SLC17A7, SNAP25, SYT1 |
Cluster 2 |
ZBTB7A |
DNM1, MAPT, STXBP1 |
Cluster 1 |
ZEB1 |
SLC17A7, SNAP25, TUBB2A |
Cluster 2 |
ZFP2 |
COL1A1, FBN2, FN1 |
Cluster 3 |
ZNF501 |
SLC17A7, SNCA, STMN2 |
Cluster 2 |
ZNF71 |
DNM3, DYNC1I1, SLC17A7, TUBB4A |
Cluster 2 |
Table 6.
miRNAs co-regulating with hub genes.
Table 6.
miRNAs co-regulating with hub genes.
miRNAs |
Gene targets |
Cluster |
hsa-miR-137 |
GABRA1, SNAP91 ,SLC12A5 |
Cluster 1 |
hsa-miR-16 |
DYNC1I1, KIF5C, SH3GL2, SNAP25, SNCA |
Cluster 2 |
hsa-miR-181a |
GABRA1, GRIA2, SLC12A5 |
Cluster 1 |
hsa-miR-181b |
GABRA1, GRIA2, SLC12A5 |
Cluster 1 |
hsa-miR-181c |
GABRA1, GRIA2, SLC12A5 |
Cluster 1 |
hsa-miR-181d |
GABRA1, GRIA2, SLC12A5 |
Cluster 1 |
hsa-miR-195 |
DYNC1I1, KIF5C, SH3GL2, SNAP25, SNCA |
Cluster 2 |
hsa-let-7a-5p |
DNAJC6, SYT1, TUBB2A, TUBB4A |
Cluster 2 |
hsa-let-7b-5p |
DNM3, SYT1, TUBB2A, TUBB4A |
Cluster 2 |
hsa-let-7c-5p |
DNAJC6, SYT1, TUBB2A, TUBB4A |
Cluster 2 |
hsa-let-7d-5p |
SYT1, TUBB2A, TUBB4A |
Cluster 2 |
hsa-let-7e-5p |
SYT1, TUBB2A, TUBB4A |
Cluster 2 |
hsa-let-7f-5p |
SYT1, TUBB2A, TUBB4A |
Cluster 2 |
hsa-let-7g-5p |
SYT1, TUBB2A, TUBB4A |
Cluster 2 |
hsa-let-7i-5p |
SYT1, TUBB2A, TUBB4A |
Cluster 2 |
hsa-miR-1 |
DNM3, SNAP25, SYT1 |
Cluster 2 |
hsa-miR-103 |
DYNC1I1, KIF5C, SH3GL2 |
Cluster 2 |
hsa-miR-106a |
DNM3, KIF5C, SLC17A7 |
Cluster 2 |
hsa-miR-107 |
DYNC1I1, KIF5C, SH3GL2 |
Cluster 2 |
hsa-mir-124-3p |
DNM3, KIF5C, TUBB4A & COL1A1, COL6A2 |
Cluster 2 & Cluster 3 |
hsa-miR-130a |
DNM3, KIF5C, SNAP25, SYT1 |
Cluster 2 |
hsa-miR-130b |
DNM3, KIF5C, SNAP25, SYT1 |
Cluster 2 |
hsa-miR-135b |
DNM3, MOBP, SYT1 |
Cluster 2 |
hsa-miR-138 |
DNM3, SLC17A7, SNAP25 |
Cluster 2 |
hsa-miR-148b |
DNM3, MOBP, SYTI |
Cluster 2 |
hsa-miR-153 |
DNM3, SNAP25, SNCA, SYT1 |
Cluster 2 |
hsa-miR-15a |
DYNC1I1, NEFH, SH3GL2, SNCA |
Cluster 2 |
hsa-miR-206 |
DNM3, SNAP25, SYT1 |
Cluster 2 |
hsa-miR-221 |
DNM3, MOBP, NEFH |
Cluster 2 |
hsa-miR-222 |
DNM3, MOBP, NEFH |
Cluster 2 |
hsa-miR-23a |
DNAJC6, DNM3, NEFH, SNAP25 |
Cluster 2 |
hsa-miR-23b |
DNAJC6, DNM3, NEFH, SNAP25 |
Cluster 2 |
hsa-miR-27a |
DNM3, SNAP25, SYT1 |
Cluster 2 |
hsa-miR-27b |
DNM3, SNAP25, SYT1 |
Cluster 2 |
hsa-miR-301 |
DNM3, KIF5C, SNAP25 |
Cluster 2 |
hsa-miR-301b |
DNM3, KIF5C, SNAP25 |
Cluster 2 |
hsa-mir-335-5p |
DYNC1I1, NEFH, SLC17A7, SYT1 |
Cluster 2 |
hsa-miR-429 |
SNAP25, SYT1, TUBB2A |
Cluster 2 |
hsa-mir-4458 |
TUBB2A, TUBB4A, SYT1 |
Cluster 2 |
hsa-mir-4500 |
TUBB2A, TUBB4A, SYT1 |
Cluster 2 |
hsa-miR-497 |
DYNC1I1, KIF5C, SH3GL2 |
Cluster 2 |
hsa-miR-519b-3p |
NEFH, SLC17A7, SYT1 |
Cluster 2 |
hsa-miR-519c-3p |
KIF5C, NEFH, SLC17A7, SYT1 |
Cluster 2 |
hsa-miR-519d |
NEFH, KIF5C, SLC17A7 |
Cluster 2 |
hsa-mir-8485 |
MOBP, KIF5C, SYT1 |
Cluster 2 |
hsa-mir-98-5p |
TUBB2A, TUBB4A, SYT1 |
Cluster 2 |
Table 7.
Drugs with more than two gene interactions.
Table 7.
Drugs with more than two gene interactions.
Drug |
Degree |
Target HDEGs |
Curcumin |
3 |
MAPT, TUBB2A, TUBB4A |
Ocriplasmin |
3 |
COL1A1, COL6A2, FN1 |
Table 8.
Drugs with more than two transcription factor interactions.
Table 8.
Drugs with more than two transcription factor interactions.
Drug |
Degree |
Target TFs |
Target HDEGs |
Cisplatin |
5 |
GABPA, MYC, SMARCA4, STAT1, TP53 |
MAPT |
Daunorubicin |
5 |
CBFB, GATA1, IKZF1, THRB, WT1 |
MAPT |
Cytarabine |
4 |
GATA1, IKZF1, TP53, WT1 |
- |
Palbociclib |
4 |
CDK5, DYRK1A, SMARCA4, TP53 |
- |
Resveratrol |
4 |
NFKB1, PRKCA, RELA, TP53 |
MAPT |
Doxorubicin hydrochloride |
3 |
THRB, TP53, ZEB1 |
MAPT, SNCA |
Doxorubicin |
3 |
EZH2, SP3, ZEB1 |
- |
Gemcitabine |
3 |
CDC5L, POLR2A, TP53 |
- |
Indoprofen |
3 |
NFKB1, RELA, TP53 |
- |
Levothyroxine |
3 |
AHR, PPARG, THRB |
- |
Methotrexate |
3 |
IKZF1, PPARG, TP53 |
- |
Niclosamide |
3 |
AHR, STAT3, TP53 |
MAPT |
Quercetin |
3 |
NFKB1, PRKCA, RELA |
MAPT |
Vorinostat |
3 |
EZH2, MYC, TP53 |
TUBB2A, TUBB4A |
Table 9.
Drugs that interact with TFs and HDEGs with more than two hits.
Table 9.
Drugs that interact with TFs and HDEGs with more than two hits.
Drug |
Degree |
TFs |
HDEGs |
Paclitaxel |
4 |
FOS, TP53 |
TUBB2A, TUBB4A |
Colchicine |
3 |
JUN |
TUBB2A, TUBB4A |
Daunorubicin hydrochloride |
3 |
THRB, TP53 |
MAPT |
Docetaxel anhydrous |
3 |
TP53 |
TUBB2A, TUBB4A |
Epigallocatechin gallate |
3 |
DYRK1A, NFKB1 |
MAPT |
Fenretinide |
3 |
NFKB1, TP53 |
MAPT |
Hexachlorophene |
3 |
THRB, TP53 |
MAPT |
Masoprocol |
3 |
NFKB1, TP53 |
MAPT |
Nifedipine |
3 |
NFKB1 |
CAMK2B, MAPT |
Nitazoxanide |
3 |
AHR, TP53 |
MAPT |
Phenobarbital |
3 |
FOS |
GABRA1, GRIA2 |
Raloxifene hydrochloride |
3 |
PPARG, TP53 |
MAPT |
Trifluoperazine |
3 |
TP53 |
CAMK2B, SNAP91 |
Vinblastine sulfate |
3 |
JUN |
TUBB2A, TUBB4A |
Vinorelbine |
3 |
SMARCA4 |
TUBB2A, TUBB4A |
Vinorelbine tartrate |
3 |
JUN |
TUBB2A, TUBB4A |
Table 10.
Enriched miRNA families and their corresponding miRNAs.
Table 10.
Enriched miRNA families and their corresponding miRNAs.
miRNA Family |
miRNAs |
let-7 family |
hsa-let-7a-1, hsa-let-7a-2, hsa-let-7a-3, hsa-let-7b, hsa-let-7c, hsa-let-7d, hsa-let-7e, hsa-let-7f-1, hsa-let-7f-2, hsa-let-7g, hsa-let-7i, hsa-mir-98 |
mir-124 family |
hsa-mir-124-1, hsa-mir-124-2, hsa-mir-124-3 |
mir-1 family |
hsa-mir-1-1, hsa-mir-1-2, hsa-mir-206 |
mir-103 family |
hsa-mir-130a, hsa-mir-130b, hsa-mir-301b |
mir-27 family |
hsa-mir-27a, hsa-mir-27b |
Table 11.
Validated candidate repurposed drugs by CMAP analysis and their regulatory effects.
Table 11.
Validated candidate repurposed drugs by CMAP analysis and their regulatory effects.
Drugs |
Upregulated gene |
Downregulated gene |
Colchicine |
- |
DNM3, SLC12A5, TUBB2A |
Doxorubicin |
COL1A1, FN1, SNCA |
- |
Indoprofen |
COL1A1 |
- |
Levothyroxine |
COL1A1, SLC17A7 |
- |
Methotrexate |
CAMK2B, KIF5C, SNCA |
SLC12A5, STXBP1 |
Nifedipine |
COL1A1, DYNC1I1, FN1, KIF5C |
DYNC1I1, STXBP1 |
Paclitaxel |
CAMK2B, DNAJC6, SLC12A5, SLC17A7 |
DNM1 |
Quercetin |
SNCA |
- |
Raloxifene |
SLC12A5 |
DNM1, DNM3 |
Resveratrol |
COL1A1, MAPT, SNCA |
- |
Trifluoperazine |
CAMK2B, COL1A1, FN1, TUBB2A |
DNM3 |
Vinblastine |
COL1A1, KIF5C |
TUBB2A |
Vorinostat |
DNAJC6, DNM3, KIF5C, SLC17A7, STXBP1, SYT1, TUBBA2A |
- |
Table 12.
The top 5 upregulated and downregulated differentially expressed genes for each type of CNS or brain tumor.
Table 12.
The top 5 upregulated and downregulated differentially expressed genes for each type of CNS or brain tumor.
Regulation |
ATRT |
EPN |
PA |
MED |
PNET |
MEN |
ACM |
ODG |
GBM |
Upregulated |
TOP2A |
FAM81B |
POSTN |
SOX11 |
MEST |
YWHAE |
SOX4 |
SOX4 |
LTF |
TMSB15B |
CFAP126 |
CFI |
OTX2 |
VIM |
TNNC1 |
TOP2A |
TOP2A |
TOP2A |
MFAP2 |
CAPSL |
COL20A1 |
DACH1 |
COL3A1 |
EIF5A |
NKAIN4 |
HES6 |
ASPM |
MELK |
ARMC3 |
TRPM8 |
SOX4 |
MGP |
COL9A3 |
HES6 |
NDC80 |
IGFBP2 |
HMGA2 |
SPAG6 |
PLA2G2A |
COL1A1 |
COL1A1 |
CLIC3 |
LCAT |
TIMP4 |
NDC80 |
Downregulated |
VSNL1 |
RAB3C |
SLC12A5 |
MBP |
SNAP25 |
MBP |
MFSD4A |
MFSD4A |
MFSD4A |
GABRG2 |
SLC12A5 |
GJB6 |
CRTAM |
DIRAS2 |
CXCL2 |
RGS4 |
GJB6 |
GABRA1 |
GJB6 |
MYT1L |
PPP2R2C |
VSNL1 |
STMN2 |
MAFF |
CACNG3 |
RGS4 |
GJB6 |
PACSIN1 |
NEFL |
SYNPR |
GABRB2 |
ANK3 |
SNAP25 |
GABRA1 |
GABRA1 |
CACNG3 |
SV2B |
SV2B |
PACSIN1 |
PVALB |
MBP |
MYH11 |
DDN |
VSNL1 |
SLITRK4 |