Identification of crucial genes associated with Parkinson's disease using microarray data

  • Authors:
    • Yongqi Sun
    • Linlin Ye
    • Yonghui Zheng
    • Zichao Yang
  • View Affiliations

  • Published online on: December 18, 2017     https://doi.org/10.3892/mmr.2017.8305
  • Pages: 3775-3782
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study aimed to examine potential crucial genes associated with Parkinson's disease (PD) in addition to the interactions and regulators of these genes. The chip data (GSE7621) were obtained from the Gene Expression Omnibus and standardized using the robust multi‑array average in the Affy package of R software. The differentially expressed genes (DEGs) were then screened using the Samr package with a false discovery rate (FDR) <0.05 and |log2 fold change (FC)|>1. Crucial PD‑associated genes were predicted using the Genetic Association Database in the Database for Annotation, Visualization and Integrated Discovery and sequence alignment. Furthermore, transcription factors (TFs) of the crucial PD‑associated genes were predicted, and protein‑protein interactions (PPIs) between the crucial PD‑associated genes were analyzed using the Search Tool for the Retrieval of Interacting Genes/Proteins. Additionally, another dataset of PD was used to validate the expression of crucial PD‑associated genes. A total of 670 DEGs (398 upregulated and 272 downregulated genes) were identified in the PD samples. Of these, 10 DEGs enriched in pathways associated with the nervous system were predicted to be crucial in PD, including C‑X‑C chemokine receptor type 4 (CXCR4), deleted in colorectal cancer (DCC) and NCL adaptor protein 2 (NCK2). All 10 genes were associated with neuron development and differentiation. They were simultaneously modulated by multiple TFs, including GATA, E2F and E4 promoter‑binding protein 4. The PPI networks showed that DCC and CXCR4 were hub proteins. The DCC‑netrin 1‑roundabout guidance receptor 2‑slit guidance ligand 1 interaction pathway, and several genes, including TOX high mobility group box family member 4, kinase insert domain receptor and zymogen granule protein 16B, which interacted with CXCR4, were novel findings. Additionally, CXCR4 and NCK2 were upregulated in another dataset (GSE8397) of PD. These genes, interactions of proteins and TFs may be important in the progression of PD.

Introduction

Parkinson's disease (PD) is a common chronic neurodegenerative disorder, which affects >0.1% of individuals >40 years of age (1,2). There is a loss of dopaminergic nigrostriatal neurons in the substantia nigra (SN) pars compacta in PD (3,4), and clinical characteristics for the majority of patients with PD include motor disorder, slowness of movement, tremor at rest, rigidity and disturbances in balance. However, it is difficult to diagnose PD in its early stages due to the high rate of misdiagnosis with other movement disorders, which can share certain features with PD, including essential tremor, multiple system atrophy (MSA) and progressive supranuclear palsy (5). Therefore, it is necessary and urgent to screen diagnostic or therapeutical biomarkers, which can be utilized in the diagnosis or treatment of PD.

In previous years, numerous significant factors have been correlated with the development of PD, which has improved current understanding of PD. For example, the level of S100 calcium binding protein B (S100B) is higher in the post-mortem SN of patients with PD, compared with control tissue, and the ablation of S100B results in neuroprotection, reduces microgliosis and decreases the expression of tumor necrosis factor-α and the receptor for advanced glycation end products (6). The reduced expression level of lysosomes-associated membrane protein 2A in dopaminergic cell lines leads to reduced chaperone-mediated autophagy and decreases the metabolism of wild-type α-synuclein (SNCA), which is a cause of PD (7). In addition, a set of molecules, which offer potential in identifying individuals at risk for PD, and assist in the diagnosis and management of PD have been identified, including SNCA (8) and DJ-1 (9). Furthermore, based on microarray analysis, Papapetropoulos et al identified 11 candidate genes with deregulated expression in brain regions from patients with PD, and the significant role of mitochondria ribosomal protein S6 (MRPS6) in the oxidative phosphorylation system was reported (10). In addition, a previous study identified a set of differentially expressed genes (DEGs) in PD, and the candidate compound, alvespimycin, was selected as a lead compound for PD, based on the gene expression data from the study by Papapetropoulos et al (10,11). In organisms, each protein exerts its functions via interactions with other proteins and regulation of a set of regulators. However, interactions and regulators of DEGs in PD, which assist in obtaining a more comprehensive understanding of the pathogenesis of PD, were not fully investigated in the studies of Papapetropoulos et al and Gao et al (10,11); there may be additional potential key genes associated with PD, which remain undetected.

The present study aimed to examine additional potential key genes associated with PD, in addition to examining their interactions and regulation. The microarray data of Papapetropoulos et al (10) were used to identify DEGs between patients with PD and those without PD. The potential crucial genes associated with PD were then examined. Subsequently, transcription factors (TFs) and protein-protein interactions (PPIs) of the DEGs were analyzed. A second microarray dataset (12) of PD, which included additional post-mortem brain samples, was selected to validate the expression of key genes in PD. The results may provide novel information for investigation of the molecular mechanisms underlying the pathogenesis of PD.

Materials and methods

Gene expression data of PD

The GSE7621 gene expression data (10) were obtained from the Gene Expression Omnibus (GEO; http://www.ncbi.nlm.nih.gov/geo/) based on [HG-U133_Plus_2] Affymetrix Human Genome U133 Plus 2.0 Array platform (GPL570; Affymetrix, Inc., Santa Clara, CA, USA). A total of 25 post-mortem SN samples, including 16 samples from patients with idiopathic PD and nine samples from aged controls without PD, were included in the GSE7621 expression data. For the patients with PD, the ratio of men to women was 13:3, and the mean age was 74 years, ranging between 60 and 88. For the controls, the ratio of men to women was 4:5, and the mean age was 78.2 years, ranging between 46 and 90. The age, post-mortem interval (PMI), and brain pH of the two subject groups were matched as closely as possible. Analysis of the quality control parameters showed no significant differences in age, gender, brain pH, PMI or RNA QC values between the aged control and PD groups (10).

Another gene expression dataset, GSE8397 (12), was used for validation, which was also downloaded from the GEO, which was based on the platforms of Affymetrix Human Genome U133A Array (GPL96) and Affymetrix Human Genome U133B Array (GPL97; Affymetrix, Inc.). A total of 47 individual tissue samples analyzed using one A and one B GeneChip per sample [15 medial SN (MSN) samples and nine lateral SN (LSN) samples obtained from patients with idiopathic PD, eight MSN samples and seven LSN samples obtained from controls without PD, five superior frontal gyrus samples from patients with PD, and three superior frontal gyrus samples from controls] were included in this dataset. The LSN and MSN samples were obtained from the same cohort of patients with idiopathic PD or controls. For the patients with PD, the ratio of men to women was 9:6, and the mean age was 80 years, ranging between 68 and 89; for the controls, the ratio of men to women was 6:2, and the mean age was 70.6 years, ranging between 46 and 81. The subjects in the PD and control groups were well matched in age, PMI and brain pH (12). Among the 47 brain tissue samples, only the data of 16 LSN samples and 23 MSN samples from the patients with PD and controls produced by the GPL96 platform were used in the present study.

Data preprocessing and identification of DEGs

The CEL files and probe annotation files were downloaded from the database. The raw data were preprocessed via background correction, quantile normalization, and calculating expression using the robust multi-array average (RMA) algorithm (13). The probes were then mapped into corresponding gene names. The probe, which corresponded to a plurality of genes was removed. If one gene symbol was matched by multiple probe IDs, the mean expression value was selected as the expression level of this gene. The Samr package in R software (version 3.1; http://cran.r-project.org/web/packages/samr/index.html) (14) was used to select the DEGs between the PD and control samples. The P-value for each gene was calculated using an unpaired t-test, and adjusted into a false discovery rate (FDR) using the Benjamini-Hochberg (BH) method (15). Only the genes with FDR<0.05 and |log2fold change (FC)|>1, as commonly used criteria, were selected as DEGs.

Prediction of crucial PD-associated genes

The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database (http://www.kegg.jp/kegg/pathway.html) is a collection of manually drawn pathway maps representing current knowledge on the molecular interaction and reaction networks for metabolism, cellular processes and human diseases (16). Firstly, the online Database for Annotation, Visualization and Integrated Discovery (DAVID; https://david.ncifcrf.gov/), which integrates a set of functional annotation tools to understand the biological meaning of numerous genes (17), was used to perform KEGG pathway enrichment analysis for the identified DEGs. The P-value calculated by Fisher's exact test of P<0.05, and a gene count >2 were set as the cut-off criteria.

Secondly, in order to obtain the PD-associated genes, the DEGs were submitted to the Genetic Association Database (http://geneticassoiationdb.nih.gov/), which is a database of genetic association data from complex diseases and disorders. PD-associated genes with P<0.05 were considered significant.

Finally, to identify the crucial PD-associated genes, sequence alignment of the protein sequences of the genes, which were involved in the nervous system-associated KEGG pathways and PD-associated genes was performed using the BLASTP program (version 2.2.31; https://blast.ncbi.nlm.nih.gov/Blast.cgi?; E<10−10). The smaller the E value was, the more reliable the alignment result. If the protein sequence of the gene involved in the nervous system-associated KEGG pathways was matched to multiple protein sequences of PD-associated genes, and had the lowest E-value, this gene was considered to be a crucial PD-associated gene.

Gene ontology (GO) enrichment analysis of crucial PD-associated genes

The GO database collects biological data in the format of gene-to-annotation, and is a suitable and commonly used method for high-throughput bioinformatics scanning for enrichment analysis (18). GO enrichment analysis of the predicted crucial PD-associated genes was performed using DAVID. Only the GO terms with the common criterion of FDR<0.01 were considered significant.

Prediction of TFs for the crucial PD-associated genes

TF binding sites (TFBSs) are short DNA motifs in genes, which bind with TFs. TFBSs often occur in close proximity to each other forming cis-regulatory modules, which suggest the existence of a combinatorial code for transcriptional regulation (19). Based on the known TFBS information in the University of California Santa Cruz (UCSC) database (http://genome.ucsc.edu/) in DAVID (20), the TFs in the UCSC database targeting the crucial PD-associated genes were identified. FDR<0.01 was set as the cut-off criterion.

PPI network analysis

PPI analysis of the screened DEGs was performed using the online Search Tool for the Retrieval of Interacting Genes/Proteins (STRING; version 10.0; http://string-db.org/) software, which integrates known and predicted protein interaction data (21) (combined score>0.4). The PPI networks containing the predicted crucial PD-associated genes were then extracted and visualized using Cytoscape software (version 3.2.0; http://www.cytoscape.org/), which is an open source software project for integrating biomolecular interaction networks (22). In a network, a node represents a protein (gene), and lines represent interactions of the proteins. The ‘degree’ of each node is equal to the number of nodes interacting with this node. The higher the degree is, the closer the connections with other nodes are, indicating a higher importance of the node in the network.

Validation of the expression level of crucial PD-associated genes

The DEGs were identified in the PD and control samples from another dataset, GSE8397 (Moran et al. 2006) using the same method as that used for GSE7621. The identified DEGs were then compared with those in the GSE7621 dataset.

Results

Anlayses of DEGs and crucial PD-associated genes

Following normalization of the raw data in the GSE7621 dataset, a total of 670 DEGs were identified in the SN of PD, including 398 upregulated genes and 272 downregulated genes (data not shown). Based on the criteria of P<0.05 and gene count >2, these DEGs were associated with the pathways of axon guidance (P=0.003347; gene count, 12; http://www.genome.jp/dbget-bin/show_pathway? hsa04360+1630+4893+8440+3265 +219699+80031+7852+56920+6092+9423+4775+6585) and endocytosis (P=0.017984; gene count=13; http://www.genome.jp/dbget-bin/show_pathway? hsa04144+5979+3265+ 79643+80230+3303+3791+3305+7852+2870+57154+10938+ 8218+22905), which were associated with the nervous system. The DEGs, including deleted in colorectal cancer (DCC), NCL adaptor protein 2 (NCK2) and C-X-C chemokine receptor type 4 (CXCR4), were significantly involved in the axon guidance pathway, and a set of DEGs, including CXCR4 and clathrin heavy chain like 1 (CLTCL1), were associated with the endocytosis pathway (Table I).

Table I.

Kyoto Encyclopedia of Genes and Genomes pathways associated with the nervous system for differentially expressed genes.

Table I.

Kyoto Encyclopedia of Genes and Genomes pathways associated with the nervous system for differentially expressed genes.

TermP-valueGenes
hsa04360 Axon guidance0.003347DCC, NRAS, NCK2, HRAS, UNC5B, SEMA6D, CXCR4, SEMA3G, ROBO2, NTN1, NFATC3, SLIT1
hsa04144 Endocytosis0.017984RET, HRAS, CHMP6, RUFY1, HSPA1A, KDR, HSPA1L, CXCR4, GRK6, SMURF1, EHD1, CLTCL1, EPN2

A total of 10 DEGs, which may be correlated with PD were identified from the Genetic Association Database, including heat shock 70 kDa protein 1-like (HSPA1L), Cytochrome P450 family 27 subfamily A1 (CYP27A1), dopamine receptor D2 (DRD2), solute carrier family 6 member 3 (SLC6A3), PARK2 coregulated (PACRG), tyrosine hydroxylase (TH), nuclear receptor subfamily 4 A2 (NR4A2), solute carrier family 18 member A2 (SLC18A2), lutathione S-transferase ζ1 (GSTZ1) and heat shock protein family A member 1A (HSPA1A). Based on the DEGs enriched in the pathways of axon guidance and endocytosis, in addition to DEGs correlated with PD, which were identified using the Genetic Association Database, a total of 10 genes were predicted to be closely associated with PD via sequence alignment, including CXCR4, DCC, EH domain-containing 1 (EHD1), epsin-2 (EPN2), G protein-coupled receptor kinase 6 (GRK6), NCK2, roundabout guidance receptor 2 (ROBO2), RUN and FYVE domain containing 1 (RUFY1), semaphorin 6D (SEMA6D) and SLIT1. These were defined as crucial PD-associated genes.

GO enrichment analysis

To further reveal the biological functions of the predicted crucial PD-associated genes, GO enrichment analysis was performed. The majority of GO terms of the predicted crucial PD-associated genes, including NCK2, CXCR4, SL1T1, ROBO2 and DCC, were involved in neuron development and differentiation, and cell morphogenesis. EHD1, RUFY1, and EPN2 were associated with GO terms associated with endocytosis (Table II). No significant GO terms were enriched by the other two crucial PD-associated genes (SEMA6D and GRK6).

Table II.

Gene ontology enrichment analysis of the crucial Parkinson's disease-associated genes.

Table II.

Gene ontology enrichment analysis of the crucial Parkinson's disease-associated genes.

GeneGene Ontology terms
SLIT1Cell morphogenesis, neuron development, neuron projection development, cell part morphogenesis, cell projection morphogenesis, cell morphogenesis involved in differentiation, neuron projection morphogenesis, cell morphogenesis involved in neuron differentiation, axonogenesis, cell motion, cell projection organization, axon guidance, neuron differentiation, cellular component morphogenesis
RUFY1Regulation of endocytosis, membrane organization, membrane invagination, endocytosis
ROBO2Cell morphogenesis, neuron development, neuron projection development, cell part morphogenesis, cell projection morphogenesis, cell morphogenesis involved in differentiation, neuron projection morphogenesis, cell morphogenesis involved in neuron differentiation, axonogenesis, cell motion, cell projection organization, axon guidance, neuron projection, neuron differentiation, cellular component morphogenesis
NCK2Cell motion, cell projection organization, cell motility, localization of cell, cell migration
EPN2Endocytic vesicle, regulation of endocytosis, membrane organization, membrane invagination, endocytosis
EHD1Endocytic vesicle, membrane organization, membrane invagination, endocytosis
DCCCellular component morphogenesis, cell morphogenesis, neuron development, neuron projection development, cell part morphogenesis, cell projection morphogenesis, cell morphogenesis involved in differentiation, neuron projection morphogenesis, cell morphogenesis involved in neuron differentiation, axonogenesis, cell motion, cell projection organization, axon guidance, neuron migration, neuron projection, cell motility, localization of cell, cell migration, neuron differentiation
CXCR4Cellular component morphogenesis, cell morphogenesis, neuron development, neuron projection development, cell part morphogenesis, cell projection morphogenesis, cell morphogenesis involved in differentiation, neuron projection morphogenesis, cell morphogenesis involved in neuron differentiation, axonogenesis, cell motion, cell projection organization, axon guidance, neuron migration, neuron projection, cell motility, localization of cell, cell migration, neuron differentiation

[i] False discovery rate of all terms is <0.01.

Analyses of TFs and PPI interaction networks

To further investigate the regulatory factors and interactions of the crucial PD-associated genes, TFs targeting the genes were predicted. A total of 55 TFs were found to regulate the 10 crucial PD-associated genes. DCC, CXCR4, NCK2, SLIT1, ROBO2 and GRK6 were all regulated by a set of TFs, including GATA, E2F and E4 promoter-binding protein 4 (E4BP4) (Table III).

Table III.

List of the 55 transcription factors targeting the crucial Parkinson's disease-associated genes.

Table III.

List of the 55 transcription factors targeting the crucial Parkinson's disease-associated genes.

GeneTranscription factors
DCCIK2, SP1, HNF3B, MYOGNF1, TATA, TCF11, GFI1, NKX25, FAC1, PAX2, MEIS1B, HOXA9, OLF1, ARP1, HEN1, CEBPB, GATA1, HOXA3, HAND1E47, S8, CDP, RSRFC4, SOX5, NFY, CDP, CR3HD, SREBP1, MZF1, NRSF, MYC, MAX, AHRARNT, PAX5, MSX1, TAX, CREB, MYB, COUP, GATA2, IRF1, AP4, GATA, MRF2, PAX3, AP2REP, AREB6, SRY, MEF2, GRE, MIF1, USF, BRACH, PAX6, COMP1, AML1, LMO2COM, E2F, YY1, E4BP4
EPN2IK2, SP1, HNF3B, TCF11, GFI1, NKX25, PAX2, ARP1, HEN1, GATA1, HAND1E47, S8, RSRFC4, CDP, CR3HD, MZF1, NRSF, MYC, MAX, PAX5, MYB, AP4, AP2REP, SRY, MEF2, GRE, MIF1, USF, BRACH, PAX6, AML1, LMO2COM, E4BP4
CXCR4IK2, MYOGNF1, TATA, TCF11, GFI1, NKX25, FAC1, PAX2, MEIS1B, HOXA9, HAND1E47, S8, CDP, SOX5, NRSF, MYC, MAX, AHRARNT, MSX1, MYB, GATA2, IRF1, GATA, PAX3, AP2REP, AREB6, SRY, MEF2, USF, BRACH, PAX6, COMP1, AML1, LMO2COM, E2F, E4BP4
NCK2IK2, HNF3B, MYOGNF1, TATA, TCF11, NKX25, PAX2, MEIS1B, HOXA9, CEBPB, GATA1, HOXA3, S8, CDP, RSRFC4, SOX5, NFY, CDP, CR3HD, MZF1, NRSF, MYC, MAX, PAX5, MSX1, IRF1, AP4, GATA, MRF2, AP2REP, AREB6, MEF2, GRE, USF, BRACH, COMP1, AML1, E2F, E4BP4
SLIT1IK2, HNF3B, MYOGNF1, TATA, TCF11, GFI1, NKX25, FAC1, PAX2, MEIS1B, HOXA9, OLF1, ARP1, HEN1, CEBPB, GATA1, HAND1E47, S8, CDP, RSRFC4, SOX5, CDP, CR3HD, SREBP1, NRSF, MYC, MAX, AHRARNT, PAX5, MSX1, TAX, CREB, MYB, COUP, GATA2, AP4, GATA, MRF2, PAX3, AP2REP, AREB6, SRY, MEF2, GRE, MIF1, USF, BRACH, PAX6, COMP1, AML1, LMO2COM, E2F, YY1, E4BP4
SEMA6DIK2, HNF3B, TATA, TCF11, GFI1, NKX25, FAC1, PAX2, MEIS1B, HOXA9, ARP1, HEN1, CEBPB, GATA1, HOXA3, HAND1E47, S8, CDP, NFY, CDP, CR3HD, SREBP1, MZF1, MYC, MAX, PAX5, MSX1, TAX, CREB, MYB, COUP, GATA2, IRF1, AP4, GATA, MRF2, AREB6, SRY, MEF2, GRE, MIF1, USF, BRACH, PAX6, COMP1, AML1, LMO2COM, E2F, YY1, E4BP4
RUFY1SP1, HNF3B, TATA, NKX25, PAX2, MEIS1B, HOXA9, OLF1, ARP1, CEBPB, GATA1, HOXA3, HAND1E47, S8, CDP, RSRFC4, NFY, CDP, CR3HD, SREBP1, MZF1, NRSF, AHRARNT, PAX5, MSX1, TAX, CREB, IRF1, AP4, GATA, MRF2, AP2REP, AREB6, SRY, MEF2, GRE, USF, BRACH, PAX6, AML1, LMO2COM, YY1, E4BP4
EHD1SP1, MYOGNF1, TATA, TCF11, GFI1, PAX2, OLF1, ARP1, HEN1, GATA1, HOXA3, SOX5, NFY, SREBP1, MZF1, NRSF, MYC, MAX, AHRARNT, PAX5, TAX, CREB, MYB, COUP, IRF1, AP4, PAX3, AP2REP, AREB6, MEF2, MIF1, USF, BRACH, PAX6, COMP1, AML1, LMO2COM, E2F, YY1, E4BP4
ROBO2HNF3B, MYOGNF1, TATA, TCF11, GFI1, NKX25, FAC1, PAX2, MEIS1B, HOXA9, OLF1, HEN1, CEBPB, GATA1, HOXA3, HAND1E47, S8, CDP, RSRFC4, SOX5, NFY, CDP, CR3HD, SREBP1, MZF1, NRSF, MYC, MAX, AHRARNT, MSX1, TAX, CREB, MYB, COUP, GATA2, IRF1, AP4, GATA, MRF2, PAX3, AP2REP, AREB6, SRY, MEF2, GRE, MIF1, USF, BRACH, PAX6, COMP1, AML1, LMO2COM, E2F, YY1, E4BP4
GRK6MYOGNF1, TATA, TCF11, GFI1, FAC1, PAX2, MEIS1B, HOXA9, OLF1, ARP1, HEN1, CEBPB, GATA1, HOXA3, HAND1E47, S8, NFY, SREBP1, MZF1, NRSF, MYC, MAX, AHRARNT, PAX5, MSX1, TAX, CREB, COUP, AP4, PAX3, AREB6, MEF2, GRE, MIF1, USF, BRACH, PAX6, COMP1, AML1, LMO2COM, E2F, YY1, E4BP4

Furthermore, the PPIs of the crucial PD-associated genes were searched, and a PPI network was constructed. In the PPI networks, the downregulated DCC and upregulated CXCR4 were the hub nodes, interacting with multiple genes (Fig. 1). DCC, CXCR4 and NCK2 interacted with breast cancer anti-estrogen resistance 1 (BCAR1), and netrin 1 (NTN1) interacted with ROBO2, which interacted with SLIT1.

Validation of the expression level of crucial PD-associated genes

To confirm the expression level of the potential crucial PD-associated genes, DEGs in another microarray dataset, GSE8397 (12), were identified, and the mean probe expression levels of each common DEG in the two datasets were compared manually. In the other microarray dataset of PD (12), two crucial PD-associated genes (CXCR4 and NCK2) were also upregulated in the PD samples, compared with the controls, which was consistent with that in the first dataset (10). CXCR4 was significantly upregulated in the LSN and MSN samples (Fig. 2A and B), and NCK2 was upregulated in the MSN samples (Fig. 2B).

Discussion

In the present study, based on the analysis of the GSE7621 dataset, 398 upregulated genes and 272 downregulated genes were identified in the SN from the patients with PD, compared with the controls. A total of 10 DEGs (CXCR4, DCC, EHD1, EPN2, GRK6, NCK2, ROBO2, RUFY1, SEMA6D and SLIT1) were predicted to be closely associated with PD. Among these 10 genes, downregulated DCC and upregulated CXCR4 were the hub proteins, which interacted with multiple genes in the PPI network. These two proteins were distinctly enriched in the axon guidance pathway, and GO terms associated with neuron development and differentiation.

In the PPI network, DCC interacted with NTN1. DCC encodes an NTN1 receptor, a member of the immunoglobulin superfamily, which mediates the axon guidance of neuronal growth cones towards sources of NTN1 ligand (23). NTN1 binds to the DCC receptor and signals through downstream proteins, which regulate cytoskeletal reorganization, causing growth cone extension and neurite growth (24). Another study reported that NTN1 and DCC are able to attract axons to the nervous system midline, and mutations in DCC lead to midline guidance defects in humans (25). There is also evidence that DCC-mediated NTN1 signaling affects the formation/maintenance of mesocorticolimbic DA topography (26). Polymorphisms in NTN1 and DCC are associated with the loss of multiple functions and increased the risk of PD (27,28). These results suggested that the decreased expression of DCC and NTN1 may lead to a loss of dopaminergic nigrostriatal neurons in the SN, resulting in the onset of PD. In the PPI network, NTN1 interacted with ROBO2, which interacted with SLIT1. The ROBO2 protein belongs to the Robo family, which is important for axon guidance across the midline during central nervous system development (29). ROBO2 has also previously been found to be deregulated in PD (30). Loss of Slit1 expression results in an abnormal course of the nigrostriatal pathway through the diencephalon (31). Although the genes of the DCC-NTN1-ROBO2-SLIT1 interaction pathway have been previously reported to be associated with the nervous system, their interactions in the present study are a novel finding.

CXCR4 encodes a CXC chemokine receptor specific for stromal cell-derived factor-1 (32). In the SN of patients with PD, higher expression of CXCR4 and its natural ligand, CXCL12, have been detected, compared with control subjects (28,33), which is consistent with the results of the present study. The activation of CXCR4 has been shown to reduce dopamine levels in the striatum, and induce cell death and tissue loss in the SN and striatum (34,35). Furthermore, CXCL12 and CXCR4 are neurotoxic to dopamine neuronal populations through the release of neurotoxins from microglia (36). Therefore, CXCR4 may be pivotal in the etiology of PD by reducing dopamine levels in the striatum and/or releasing neurotoxins from microglia to induce the death of dopamine neurons. In the PPI network, CXCR4 interacted with other gene proteins, including TOX high mobility group box family member 4 (TOX4), YY1, GRK6, kinase insert domain receptor (KDR) and zymogen granule protein 16B (ZG16B). Among these genes, YY1 has been reported to regulate the expression of SNCA, which is an established susceptibility gene for PD (37). In the present study, YY1 was predicted as a TF targeting DCC, ROBO2, SLIT1 and GRK6. Therefore, YY1 may also be involved in PD via regulating the expression of these genes. In an animal model of PD, the expression of GRK6 was found to be altered in the caudate/putamen and ventral striatum (38), and the altered expression of GRK6 can result in the dysregulation of DA receptors, thus contributing to the core motor deficits observed in PD (39). Other genes, including TOX4, KDR and ZG16B, and their interactions with CXCR4, have not been correlated with PD in previous studies; thus, these are novel potential genes, which may be associated with PD.

In the present study, NCK2 was upregulated in the SN of PD based on the analysis of the two datasets. NCK2 encodes a member of the NCK family of adaptor proteins, which are involved in the regulation of receptor protein tyrosine kinases (40). It has been reported that NCK2 is expressed in neurons and glia (41). There is evidence that NCK2 interacts with particularly interesting cysteine histidine-rich protein and Dock180 protein, which are involved in neuropathological processes (42,43). In the PPI network, NCK2, CXCR4 and DCC all interacted with BCAR1. BCAR1 is involved in various cellular events, including migration, survival and transformation (44). A previous study has reported the differential expression of BCAR1 in PD (45). The cerebellum may have certain roles in the pathophysiology of PD (46). p130Cas/BCAR1 was found to be involved in cellular mechanisms regulating different stages of cerebellar development (47), suggesting that the abnormal expression of BCAR1 may be associated with PD. However, there is no evidence to confirm the association of NCK2 with the progression of PD. Collectively, these findings indicated that the upregulation of NCK2 may be responsible for the progression of PD, via interaction with BCAR1 or regulation by TFs.

In addition, according to the TF analysis for crucial PD-associated genes in the present study, a series of TFs were predicted to target the 10 crucial PD-associated genes. DCC, CXCR4, NCK2, SLIT1, ROBO2 and GRK6 were all regulated by a set of TFs, including GATA, E2F and E4BP4. A previous study reported that GATA-1 and GATA-2 regulate the expression of SNCA dopaminergic cells (48). The pRb/E2F cell-cycle pathway mediates cell death in PD, and E2F-1-deficient mice are significantly more resistant to dopaminergic cell death, compared with their wild-type littermates (49). E4BP4, also known as nuclear factor, interleukin 3 regulated is a transcriptional regulator of the human interleukin-3 promoter (50). A previous study reported that E4BP4 is reportedly associated with neuron disease (51), and is upregulated in the SN tissue from patients with PD (52). Collectively, the DCC, CXCR4 and NCK2 genes are involved in the pathogenesis of PD, not only via the axon guidance pathway, but also likely via the regulation of multiple TFs, including GATA, E2F and E4BP4.

Of note, although the present study reanalyzed the public data deposited by others, the analytical procedure of the present study was different from that of previous studies. The criteria of the FDR<0.05 and |log2 FC|>1 were more strict, compared with those (P<0.05; |FC|≥1.3) used in the previous study by Papapetropoulos et al (10), indicating that the DEGs identified in the present study show a higher level of differential expression. In the present study, the P-value of each gene was adjusted using the BH method to reduce false positives, however, the P-value was not adjusted in the previous study. Furthermore, in the present study, the protein interactions of DEGs and the TFs regulating DEGs were analyzed, which assisted in providing a more comprehensive understanding of the pathogenesis of PD, whereas these were not investigated in the previous study. Despite the significance, there were certain limitations in the present study. For example, only two datasets were analyzed, whereas performing a meta-analysis using multiple datasets, including multiple types of data, may produce additional significant results. This is an important aim of further investigations.

In conclusion, the present study identified a set of potential crucial PD-associated genes, including CXCR4, DCC and NCK2, which were associated with neuron development and differentiation. The DCC-NTN1-ROBO2-SLIT1 interaction pathway and several genes, including TOX4, KDR and ZG16B, which interacted with CXCR4, were novel findings. Genes, including DCC, CXCR4, NCK2, SLIT1 and ROBO2 were also regulated by multiple TFs, including GATA, E2F and E4BP4. Additionally, CXCR4 and NCK2 were detected to be upregulated in the SN of patients with PD following the analysis of two datasets. These genes, interactions of proteins and TFs may be key in the progression of PD. These findings contribute to an improved understanding of the pathogenesis of PD. However, further experiments are required to confirm the results of the present study and further elucidate the pathogenesis of PD.

References

1 

Dawson TM and Dawson VL: Molecular pathways of neurodegeneration in Parkinson's disease. Science. 302:819–822. 2003. View Article : Google Scholar : PubMed/NCBI

2 

Armentero MT, Pinna A, Ferré S, Lanciego JL, Müller CE and Franco R: Past, present and future of A(2A) adenosine receptor antagonists in the therapy of Parkinson's disease. Pharmacol Ther. 132:280–299. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Michell AW, Lewis SJ, Foltynie T and Barker RA: Biomarkers and Parkinson's disease. Brain. 127:1693–1705. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Berendse HW, Booij J, Francot CM, Bergmans PL, Hijman R, Stoof JC and Wolters EC: Subclinical dopaminergic dysfunction in asymptomatic Parkinson's disease patients' relatives with a decreased sense of smell. Ann Neurol. 50:34–41. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Galvin JE, Lee VM and Trojanowski JQ: Synucleinopathies: Clinical and pathological implications. Arch Neurol. 58:186–190. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Sathe K, Maetzler W, Lang JD, Mounsey RB, Fleckenstein C, Martin HL, Schulte C, Mustafa S, Synofzik M, Vukovic Z, et al: S100B is increased in Parkinson's disease and ablation protects against MPTP-induced toxicity through the RAGE and TNF-α pathway. Brain. 135:3336–3347. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Alvarez-Erviti L, Rodriguez-Oroz MC, Cooper JM, Caballero C, Ferrer I, Obeso JA and Schapira AH: Chaperone-mediated autophagy markers in Parkinson disease brains. Arch Neurol. 67:1464–1472. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Malek N, Swallow D, Grosset KA, Anichtchik O, Spillantini M and Grosset DG: Alpha-synuclein in peripheral tissues and body fluids as a biomarker for Parkinson's disease-a systematic review. Acta Neurol Scand. 130:59–72. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Devic I, Hwang H, Edgar JS, Izutsu K, Presland R, Pan C, Goodlett DR, Wang Y, Armaly J, Tumas V, et al: Salivary α-synuclein and DJ-1: Potential biomarkers for Parkinson's disease. Brain. 134:e1782011. View Article : Google Scholar : PubMed/NCBI

10 

Papapetropoulos S, Ffrench-Mullen J, McCorquodale D, Qin Y, Pablo J and Mash DC: Multiregional gene expression profiling identifies MRPS6 as a possible candidate gene for Parkinson's disease. Gene Expr. 13:205–215. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Gao L, Zhao G, Fang JS, Yuan TY, Liu AL and Du GH: Discovery of the neuroprotective effects of alvespimycin by computational prioritization of potential anti-Parkinson agents. FEBS J. 281:1110–1122. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Moran LB, Duke DC, Deprez M, Dexter DT, Pearce RK and Graeber MB: Whole genome expression profiling of the medial and lateral substantia nigra in Parkinson's disease. Neurogenetics. 7:1–11. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U and Speed TP: Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 4:249–264. 2003. View Article : Google Scholar : PubMed/NCBI

14 

Tibshirani R, Chu G, Narasimhan B and Li J: SAM: Significance Analysis of Microarrays. Version 2.0. The Comprehensive R Archive Network. 2011.

15 

Glueck DH, Mandel J, Karimpour-Fard A, Hunter L and Muller KE: Exact calculations of average power for the Benjamini-Hochberg procedure. Int J Biostat. 4:Article 112008.PubMed/NCBI

16 

Kanehisa M, Goto S, Sato Y, Furumichi M and Tanabe M: KEGG for integration and interpretation of large-scale molecular data sets. Nucleic Acids Res. 40:D109–D114. 2012. View Article : Google Scholar : PubMed/NCBI

17 

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

18 

Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al: Gene ontology: Tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 25:25–29. 2000. View Article : Google Scholar : PubMed/NCBI

19 

Levine M and Tjian R: Transcription regulation and animal diversity. Nature. 424:147–151. 2003. View Article : Google Scholar : PubMed/NCBI

20 

Martin TM, Plautz SA and Pannier AK: Network analysis of endogenous gene expression profiles after polyethyleneimine-mediated DNA delivery. J Gene Med. 15:142–154. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Serrato-Combe A: Lindebmayer Systems-experimenting with software string rewriting as an assist to the study and generation of architectural form. Proceedings of the 9th Iberoamerican Congress of Digital Graphics. SIGRADI; Lima. pp. 161–166. 2005;

22 

Smoot ME, Ono K, Ruscheinski J, Wang PL and Ideker T: Cytoscape 2.8: New features for data integration and network visualization. Bioinformatics. 27:431–432. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Keino-Masu K, Masu M, Hinck L, Leonardo ED, Chan SS, Culotti JG and Tessier-Lavigne M: Deleted in colorectal cancer (DCC) encodes a netrin receptor. Cell. 87:175–185. 1996. View Article : Google Scholar : PubMed/NCBI

24 

Xu B, Goldman JS, Rymar VV, Forget C, Lo PS, Bull SJ, Vereker E, Barker PA, Trudeau LE, Sadikot AF and Kennedy TE: Critical roles for the netrin receptor deleted in colorectal cancer in dopaminergic neuronal precursor migration, axon guidance, and axon arborization. Neuroscience. 169:932–949. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Engle EC: Human genetic disorders of axon guidance. Cold Spring Harb Perspect Biol. 2:a0017842010. View Article : Google Scholar : PubMed/NCBI

26 

Manitt C, Mimee A, Eng C, Pokinko M, Stroh T, Cooper HM, Kolb B and Flores C: The netrin receptor DCC is required in the pubertal organization of mesocortical dopamine circuitry. J Neurosci. 31:8381–8394. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Lin L, Lesnick TG, Maraganore DM and Isacson O: Axon guidance and synaptic maintenance: Preclinical markers for neurodegenerative disease and therapeutics. Trends Neurosci. 32:142–149. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Lesnick TG, Papapetropoulos S, Mash DC, Ffrench-Mullen J, Shehadeh L, de Andrade M, Henley JR, Rocca WA, Ahlskog JE and Maraganore DM: A genomic pathway approach to a complex disease: Axon guidance and Parkinson disease. PLoS Genet. 3:e982007. View Article : Google Scholar : PubMed/NCBI

29 

Sundaresan V, Mambetisaeva E, Andrews W, Annan A, Knöll B, Tear G and Bannister L: Dynamic expression patterns of Robo (Robo1 and Robo2) in the developing murine central nervous system. J Comp Neurol. 468:467–481. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Bossers K, Meerhoff G, Balesar R, van Dongen JW, Kruse CG, Swaab DF and Verhaagen J: Analysis of gene expression in Parkinson's disease: Possible involvement of neurotrophic support and axon guidance in dopaminergic cell death. Brain Pathol. 19:91–107. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Bagri A, Marı́n O, Plump AS, Mak J, Pleasure SJ, Rubenstein JL and Tessier-Lavigne M: Slit proteins prevent midline crossing and determine the dorsoventral position of major axonal pathways in the mammalian forebrain. Neuron. 33:233–248. 2002. View Article : Google Scholar : PubMed/NCBI

32 

Wegner SA, Ehrenberg PK, Chang G, Dayhoff DE, Sleeker AL and Michael NL: Genomic organization and functional characterization of the chemokine receptor CXCR4, a major entry co-receptor for human immunodeficiency virus type 1. J Biol Chem. 273:4754–4760. 1998. View Article : Google Scholar : PubMed/NCBI

33 

Shimoji M, Pagan F, Healton EB and Mocchetti I: CXCR4 and CXCL12 expression is increased in the nigro-striatal system of Parkinson's disease. Neurotox Res. 16:318–328. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Bachis A, Aden SA, Nosheny RL, Andrews PM and Mocchetti I: Axonal transport of human immunodeficiency virus type 1 envelope protein glycoprotein 120 is found in association with neuronal apoptosis. J Neurosci. 26:6771–6780. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Nosheny RL, Bachis A, Aden SA, De Bernardi MA and Mocchetti I: Intrastriatal administration of human immunodeficiency virus-1 glycoprotein 120 reduces glial cell-line derived neurotrophic factor levels and causes apoptosis in the substantia nigra. J Neurobiol. 66:1311–1321. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Bezzi P, Domercq M, Brambilla L, Galli R, Schols D, De Clercq E, Vescovi A, Bagetta G, Kollias G, Meldolesi J and Volterra A: CXCR4-activated astrocyte glutamate release via TNFalpha: Amplification by microglia triggers neurotoxicity. Nat Neurosci. 4:702–710. 2001. View Article : Google Scholar : PubMed/NCBI

37 

Mizuta I, Takafuji K, Ando Y, Satake W, Kanagawa M, Kobayashi K, Nagamori S, Shinohara T, Ito C, Yamamoto M, et al: YY1 binds to α-synuclein 3′-flanking region SNP and stimulates antisense noncoding RNA expression. J Hum Genet. 58:711–719. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Bezard E, Gross CE, Qin L, Gurevich VV, Benovic JL and Gurevich EV: L-DOPA reverses the MPTP-induced elevation of the arrestin2 and GRK6 expression and enhanced ERK activation in monkey brain. Neurobiol Dis. 18:323–335. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Managò F, Espinoza S, Salahpour A, Sotnikova TD, Caron MG, Premont RT and Gainetdinov RR: The role of GRK6 in animal models of Parkinson's disease and L-DOPA treatment. Sci Rep. 2:3012012. View Article : Google Scholar : PubMed/NCBI

40 

Tu Y, Li F and Wu C: Nck-2, a novel Src homology2/3-containing adaptor protein that interacts with the LIM-only protein PINCH and components of growth factor receptor kinase-signaling pathways. Mol Biol Cell. 9:3367–3382. 1998. View Article : Google Scholar : PubMed/NCBI

41 

Ritchie MD: Using prior knowledge and genome-wide association to identify pathways involved in multiple sclerosis. Genome Med. 1:652009. View Article : Google Scholar : PubMed/NCBI

42 

Rearden A, Hurford R, Luu N, Kieu E, Sandoval M, Perez-Liz G, Del Valle L, Powell H and Langford TD: Novel expression of PINCH in the central nervous system and its potential as a biomarker for human immunodeficiency virus-associated neurodegeneration. J Neurosci Res. 86:2535–2542. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Shi L: Dock protein family in brain development and neurological disease. Commun Integr Biol. 6:e268392013. View Article : Google Scholar : PubMed/NCBI

44 

Brinkman A, van der Flier S, Kok EM and Dorssers LC: BCAR1, a human homologue of the adapter protein p130Cas, and antiestrogen resistance in breast cancer cells. J Natl Cancer Inst. 92:112–120. 2000. View Article : Google Scholar : PubMed/NCBI

45 

Hourani M, Mendes A, Berretta R and Moscato P: Genetic biomarkers for brain hemisphere differentiation in Parkinson's disease. AIP Conference Proceedings. 952:pp. 207–216. 2007; View Article : Google Scholar

46 

Wu T and Hallett M: The cerebellum in Parkinson's disease. Brain. 136:696–709. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Furuichi T, Shiraishi-Yamaguchi Y, Sato A, Sadakata T, Huang J, Shinoda Y, Hayashi K, Mishima Y, Tomomura M, Nishibe H and Yoshikawa F: Systematizing and cloning of genes involved in the cerebellar cortex circuit development. Neurochem Res. 36:1241–1252. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Scherzer CR, Grass JA, Liao Z, Pepivani I, Zheng B, Eklund AC, Ney PA, Ng J, McGoldrick M, Mollenhauer B, et al: GATA transcription factors directly regulate the Parkinson's disease-linked gene alpha-synuclein. Proc Natl Acad Sci USA. 105:pp. 10907–10912. 2008; View Article : Google Scholar : PubMed/NCBI

49 

Höglinger GU, Breunig JJ, Depboylu C, Rouaux C, Michel PP, Alvarez-Fischer D, Boutillier AL, Degregori J, Oertel WH, Rakic P, et al: The pRb/E2F cell-cycle pathway mediates cell death in Parkinson's disease. Proc Natl Acad Sci USA. 104:pp. 3585–3590. 2007; View Article : Google Scholar : PubMed/NCBI

50 

Zhang W, Zhang J, Kornuc M, Kwan K, Frank R and Nimer SD: Molecular cloning and characterization of NF-IL3A, a transcriptional activator of the human interleukin-3 promoter. Mol Cell Biol. 15:6055–6063. 1995. View Article : Google Scholar : PubMed/NCBI

51 

Hulme DJ, Blair IP, Dawkins JL and Nicholson GA: Exclusion of NFIL3 as the gene causing hereditary sensory neuropathy type I by mutation analysis. Hum Genet. 106:594–596. 2000. View Article : Google Scholar : PubMed/NCBI

52 

Hu WC: Parkinson disease is a TH17 dominant autoimmune disorder against accumulated alpha-synuclein. Nature Preced. 61762011.

Related Articles

Journal Cover

March-2018
Volume 17 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sun Y, Ye L, Zheng Y and Yang Z: Identification of crucial genes associated with Parkinson's disease using microarray data. Mol Med Rep 17: 3775-3782, 2018
APA
Sun, Y., Ye, L., Zheng, Y., & Yang, Z. (2018). Identification of crucial genes associated with Parkinson's disease using microarray data. Molecular Medicine Reports, 17, 3775-3782. https://doi.org/10.3892/mmr.2017.8305
MLA
Sun, Y., Ye, L., Zheng, Y., Yang, Z."Identification of crucial genes associated with Parkinson's disease using microarray data". Molecular Medicine Reports 17.3 (2018): 3775-3782.
Chicago
Sun, Y., Ye, L., Zheng, Y., Yang, Z."Identification of crucial genes associated with Parkinson's disease using microarray data". Molecular Medicine Reports 17, no. 3 (2018): 3775-3782. https://doi.org/10.3892/mmr.2017.8305