Open Access

Identification of key genes and pathways for peri-implantitis through the analysis of gene expression data

  • Authors:
    • Huang Zhang
    • Xiong Zhang
    • Jie Huang
    • Xusheng Fan
  • View Affiliations

  • Published online on: March 2, 2017     https://doi.org/10.3892/etm.2017.4176
  • Pages: 1832-1840
  • Copyright: © Zhang 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

The present study attempted to identify potential key genes and pathways of peri-implantitis, and to investigate the possible mechanisms associated with it. An array data of GSE57631 was downloaded, including six samples of peri-implantitis tissue and two samples of normal tissue from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) in the peri‑implantitis samples compared with normal ones were analyzed with the limma package. Moreover, Gene Ontology annotation and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses for DEGs were performed by DAVID. A protein‑protein interaction (PPI) network was established using Cytoscape software, and significant modules were analyzed using Molecular Complex Detection. A total of 819 DEGs (759 upregulated and 60 downregulated) were identified in the peri‑implantitis samples compared with normal ones. Moreover, the PPI network was constructed with 413 nodes and 1,114 protein pairs. Heat shock protein HSP90AA1 (90 kDa α, member 1), a hub node with higher node degrees in module 4, was significantly enriched in antigen processing, in the presentation pathway and nucleotide‑binding oligomerization domain (NOD)‑like receptor‑signaling pathway. In addition, nuclear factor‑κ‑B1 (NFKB1) was enriched in the NOD‑like receptor‑signaling pathway in KEGG pathway enrichment analysis for upregulated genes. The proteasome is the most significant pathway in module 1 with the highest P‑value. Therefore, the results of the present study suggested that HSP90AA1 and NFKB1 may be potential key genes, and the NOD‑like receptor signaling pathway and proteasome may be potential pathways associated with peri‑implantitis development.

Introduction

Peri-implantitis is a destructive inflammatory disease that affects the tissues surrounding dental implants (1,2). It has been demonstrated that peri-implantitis is a crucial element in implant failure. In total, ~30% of patients that receive dental implants develop peri-implantitis (3). However, there are currently no effective therapeutic strategies against peri-implantitis. Moreover, the use of dental implants is constantly increasing, therefore an effective therapy to treat peri-implantitis is required. Thus, further investigations into the molecular pathophysiology of peri-implantitis are necessary in order to provide novel options for effective treatment (1).

Recently, a number of studies have investigated the pathological mechanisms underlying peri-implantitis progression. Becker et al (4) indicated that serglycin (SRGN) expression was significantly upregulated in peri-implantitis when compared with healthy individuals. It has been suggested that this gene may inhibit bone mineralization in vitro (5). Another study indicated that concentrations of the nuclear factor-κB (NF-B), soluble RANK ligand (sRANKL), osteoprotegerin (OPG) and sclerostin are significantly increased in patients with peri-implantitis (6). A number of typical bone matrix molecules, including collagen, type IX, α 1 (COL9A1), bone gamma-carboxyglutamate (Gla) protein (BGLAP) and secreted phosphoprotein 1 (SPP1) are decreased in the peri-implantitis tissues (1). Furthermore, it has been identified that fibroblasts are involved in the pathogenesis of peri-implantitis (7). The regulation of inflammatory mediators and matrix metalloproteinases (MMPs) in peri-implantitis fibroblasts function in the pathogenesis of the disease (8), and levels of the anti-inflammatory cytokine interleukin (IL)-10 are decreased in peri-implantitis (9). Furthermore, peroxisome proliferator-activated receptor γ (PPARγ) that can inhibit inflammation and promote osteoblast function is downregulated in the peri-implantitis tissues (10). However, other mechanisms associated with peri-implantitis have not been identified. Therefore, further research should focus on elucidating other potential mechanisms and investigate target genes for the treatment of peri-implantitis.

The microarray data of GSE57631 was used to confirm the similarities and differences of inflamed peri-implantitis tissues vs. normal peri-implantitis tissues at the mRNA level (1). In contrast to results from a previous study (1), the array data of GSE57631 was downloaded and the differentially expressed genes (DEGs) associated with peri-implantitis were analyzed using a biological informatics approach. In addition, functional enrichment analyses were performed for DEGs. In addition, a protein-protein interaction (PPI) network was established and four significant modules were analyzed. The present study aimed to identify the key genes and pathways of peri-implantitis, and identify possible significant mechanisms associated with it.

Materials and methods

Affymetrix microarray data

The array data of GSE57631 was downloaded from the Gene Expression Omnibus (GEO; http://www.ncbi.nlm.nih.gov/geo/) database, which was deposited by Schminke et al (1). Six samples of peri-implantitis tissues and two samples of normal tissues were included in the present study. The raw data and annotation files were downloaded for subsequent analysis, based on the GPL15034 platform (Affymetrix Human Gene 1.0 ST Array [HuGene10stv1_Hs_ENTREZ, Brainarray v14]; Affymetrix, Inc., Santa Clara, CA, USA).

Data preprocessing

The raw expression data was preprocessed using the robust multiarray average (11) algorithm by applying an oligo (12) in the R statistical software program in Bioconductor (http://www.bioconductor.org/). Background correction, normalization and a calculating expression were included in the process of preprocessing. A total of 18,977 gene expression values were obtained.

DEG analysis

The limma package (13) in Bioconductor was used to analyze DEGs in peri-implantitis samples compared with controls. In the process of the analysis, the P-values of DEGs were calculated using a t-test in the limma package. |log2FC|≥1 and P<0.05 were used as a cut-off criteria.

Gene Ontology (GO) and pathway enrichment analyses

GO is a tool that is used for gene annotation by collecting defined, structured, controlled vocabulary, which includes three main categories: Molecular function (MF), biological process (BP) and cellular component (CC) (14). Kyoto Encyclopedia of Genes and Genomes (KEGG) is a database used for associating related gene sets with their pathways (15). Moreover, DAVID, an integrated data-mining environment, is used to analyze gene lists (16).

GO annotation and KEGG pathway enrichment analyses were conducted for upregulated and downregulated genes by DAVID. Moreover, EASE ≤0.05 and gene counts ≥ were set as the threshold value.

PPI network analysis

The Search Tool for the Retrieval of Interacting Genes (STRING) (17) database provides information regarding the predicted and experimental interactions of proteins. The prediction method of this database came from neighborhood, gene fusion, co-occurrence, co-expression experiments, databases and text mining. Moreover, the interactions of protein pairs in the database are presented with a combined score. In the present study, DEGs were mapped into PPIs and a combined score of >0.9 was used as the cut-off value. In addition, PPI networks were constructed using Cytoscape software version 3.2.1 (18).

Topological properties of the PPI network, including degree (19), subgraph (20) and betweenness (21) centralities were determined using the R software package igraph (22), in order to analyze key genes in the network. A degree was used for describing the importance of protein nodes in the network. Subgraph centrality based on combining network topology and information of protein complexes was used to measure the importance of nodes in the network. The higher the degree and subgraph values were, the more important the nodes were in the network. Moreover, the betweenness centrality is an index describing the global topological properties of the network and could be used to describe how the nodes affect the connectivity between two nodes. Betweenness was defined as the ratio of the number of every path passed per node and the number of the shortest paths. The higher the betweenness values are, the greater the impact of the node in the network is. In addition, R software package igraph version 1.0.1 (22) was used for these three methods.

Module analysis

Network module was one of the characteristics of the protein network and may contain specific biological significance. The Cytoscape software package Molecular Complex Detection (MCODE) (23) was used to analyze the most notable clustering module. Next, the KEGG pathway enriched by DEGs in different modules was analyzed using DAVID online tool. EASE ≤0.05 and count ≥2 were set as the cutoff values.

Results

Data processing and DEG analysis

As shown in Fig. 1, a total of 819 DEGs including 759 upregulated and 60 downregulated genes, were identified in the peri-implantitis samples compared with the control ones. As a result, the number of upregulated genes was found to be significantly higher than that of the downregulated ones.

GO and pathway enrichment analyses

GO and KEGG pathway analyses were performed for upregulated and downregulated DEGs, respectively. The GO terms of upregulated DEGs were mainly associated with the proteasomal protein catabolic process, endoplasmic reticulum, melanosome and structural constituent of the cytoskeleton (Table IA). The downregulated DEGs were mainly enriched in the epidermis and ectoderm development, and the extracellular region part and cadmium ion binding (Table IB).

Table I.

GO for differentially expressed genes.

Table I.

GO for differentially expressed genes.

A, Upregulated

TermsDescriptionCountsP-value
GO-BP
  GO: 0010498Proteasomal protein catabolic process  24 5.32×10−11
  GO: 0043161Proteasomal ubiquitin-dependent protein catabolic process  24 5.32×10−11
  GO: 0032269Negative regulation of cellular protein metabolic process  30 9.32×10−10
GO-CC
  GO: 0005783Endoplasmic reticulum115 1.37×10−21
  GO: 0042470Melanosome  30 1.77×10−17
  GO: 0048770Pigment granule  30 1.77×10−17
GO-MF
  GO: 0005200Structural constituent of cytoskeleton  15 2.22×10−6
  GO: 0070003Threonine-type peptidase activity  8 1.13×10−5

B, Downregulated

TermsDescriptionCountsP-value

GO-BP
  GO:0008544Epidermis development20 2.57×10−10
  GO:0007398Ectoderm development20 9.84×10−10
  GO:0006954Inflammatory response19 9.40×10−6
GO-CC
  GO:0044421Extracellular region part35 1.05×10−5
  GO:0005576Extracellular region54 1.33×10−4
  GO:0005615Extracellular space25 2.79×10−4
GO-MF
  GO:0046870Cadmium ion binding  4 5.27×10−4
  GO:0005507Copper ion binding  6 6.22×10−3

[i] Terms represent the identification number of GO term; description represents the names of GO term; counts represent the number of genes enriched in GO terms. GO, Gene ontology; BP, biological process; CC: cellular component; MF, molecular function.

Two pathways that were significantly enriched by upregulated DEGs were Alzheimer's disease and the proteasome (Table II). Moreover, an important gene HSP90AA1 was significantly enriched in the antigen processing and presentation pathway and the nucleotide-binding oligomerization domain (NOD)-like receptor signaling pathway, and NFKB1 was enriched in NOD-like receptor signaling pathway in KEGG pathway enrichment analysis for upregulated genes. However, downregulated DEGs did not significantly enriched any pathways.

Table II.

KEGG pathway enrichment analysis for upregulated differentially expressed genes.

Table II.

KEGG pathway enrichment analysis for upregulated differentially expressed genes.

TermDescriptionCountsP-value
hsa05010Alzheimer's disease25 1.69×10−5
hsa03050Proteasome12 5.36×10−5
hsa00190Oxidative phosphorylation18 1.19×10−3
hsa05012Parkinson's disease17 2.65×10−3
hsa05016Huntington's disease21 3.41×10−3
hsa00510N-Glycan biosynthesis  9 4.31×10−3
hsa04142Lysosome15 7.11×10−3
hsa00480Glutathione metabolism  9 7.27×10−3
hsa04612Antigen processing and presentation12 7.74×10−3
hsa04621NOD-like receptor signaling pathway10 8.63×10−3
hsa00970Aminoacyl-tRNA biosynthesis  7 2.89×10−2
hsa00020Citrate cycle (TCA cycle)  6 3.07×10−2
hsa01040Biosynthesis of unsaturated fatty acids  5 3.51×10−2
hsa05130Pathogenic Escherichia coli infection  8 4.45×10−2

[i] Term represents the identification number of the KEGG pathway; Description represents the name of the KEGG pathway; Counts represent the number of genes enriched in the KEGG pathway. KEGG, Kyoto Encyclopedia of Genes and Genomes; NOD, nucleotide-binding oligomerization domain-like; TCA, tricarboxylic acid.

PPI network analysis

Based on the STRING database, a total of 413 nodes and 1,114 protein pairs were obtained with a combined score >0.9 (Fig. 2). As shown in Fig. 2, the majority of the nodes in the network were upregulated DEGs in peri-implantitis samples. The top 20 most up- and dowregulated nodes are presented in Table III. A number of protein enzyme families, such as the proteasome subunit, α type 1 (PSMA1), proteasome subunit, beta type 1 (PSMB1) and proteasome subunit, α type 4 (PSMA4) were hub nodes based on the subgraph and degree centralities. Heat shock protein HSP90AA1 (90 kDa α, member 1), Ras-related C3 botulinum toxin substrate 1, NFKB1, Jun proto-oncogene were hub nodes with higher betweenness values.

Table III.

Nodes with higher values in subgraph centrality, betweenness centrality and degree centrality.

Table III.

Nodes with higher values in subgraph centrality, betweenness centrality and degree centrality.

NodesSubgraghNodesBetweennessNodesDegree
PSMA1 2.35×107HSP90AA1 2.78×104HSP90AA134
PSMB1 2.32×107RAC1 1.77×104JUN27
PSMA4 2.31×107NFKB1 1.38×104RAC127
PSMB4 2.30×107JUN 1.21×104PSMA125
PSMA3 2.30×107HIF1A 1.07×104PSMA425
PSMB3 2.30×107CDH1 9.21×103PSMB125
PSMB6 2.30×107SOS1 8.93×103PSMC225
PSMC2 2.16×107CANX 8.55×103PSMB424
PSMD8 2.12×107HDAC1 8.35×103PSMA324
PSMD5 2.08×107ATP5B 8.24×103PSMB324
PSME4 2.08×107GTF2B 8.17×103PSMB624
PSMD10 2.04×107HSPA5 7.84×103PSMD1024
PSME3 1.96×107STAT3 7.46×103PSMD823
UBE2D1 1.54×107VCP 7.32×103SEC61A123
CDC16 1.36×107RPS5 7.14×103RPN223
ANAPC5 1.36×107NME1 6.31×103NFKB123
BUB3 1.35×107POLR2B 5.90×103PSMD522
SEC61A1 1.23×107P4HB 5.90×103PSME422
SKP1 1.16×107TXN 5.67×103UBE2D122
HSPB1 1.05×107RPN2 5.67×103RPN122

[i] PSM, proteasome subunit.

Module analysis

In total, four modules (modules 1, 2, 3 and 4) with score >6 were detected by MCODE (Fig. 3). As shown in Table IV, PSMA1, PSMA4 and PSMB1 were hub nodes with higher node degrees in module 1, and ribophorin 1 (RPN1), ribophorin 2 (RPN2) and dolichyl-diphosphooligosaccharide-protein glycosyltransferase subunit (DDOST) were hub nodes in module 2. The hub nodes with higher node degrees in module 3 were pre-mRNA processing factor 8 (PRPF8), small nuclear ribonucleoprotein D1 polypeptide (SNRPD1; 16 kDa) and small nuclear ribonucleoprotein polypeptides B and B1 (SNRPB), and the hub nodes in module 4 were HSP90AA1, ATP synthase, H+ transporting, mitochondrial Fo complex, Subunit F2 (ATP5J2) and ATP synthase, H+ transporting, mitochondrial F1 complex and beta polypeptide (ATP5B). Furthermore, the proteasome pathway was identified as the most significant pathway in module 1.

Table IV.

KEGG pathway enriched by differentially expressed genes in different modules (P<0.05).

Table IV.

KEGG pathway enriched by differentially expressed genes in different modules (P<0.05).

TermDescriptionP-valueGenes
Module1
  hsa03050Proteasome 6.77×10−17PSMB4, PSMA1, PSMB6, PSMB1, PSMC2, PSMA4, PSMB3, PSMA3, PSME3, PSME4, PSMD8
  hsa04110Cell cycle 9.03×10−3ANAPC5, SKP1, CDC16, BUB3
  hsa04120Ubiquitin mediated proteolysis 1.16×10−2ANAPC5, SKP1, CDC16, UBE2D1
Module2
  hsa00510N-Glycan biosynthesis 1.63×10−3RPN1, RPN2, DDOST
  hsa03060Protein export 1.10×10−2SRP68, SRP72
Module3
  hsa03040Spliceosome 5.19×10−6PRPF8, SNRPD1, SNRPB, TXNL4A, SF3A3
  hsa03020RNA polymerase 3.26×10−2POLR2H, POLR2B
Module4
  hsa00190Oxidative phosphorylation 1.68×10−5ATP5J2, ATP5B, ATP5C1, ATP5G1, ATP5H, PPA1
  hsa04612Antigen processing and presentation 1.34×10−3HSP90AB1, HSP90AA1, IFI30, HLA-DRB5
  hsa05012Parkinson's disease 4.63×10−3ATP5B, ATP5C1, ATP5G1, ATP5H
  hsa05010Alzheimer's disease 9.07×10−3ATP5B, ATP5C1, ATP5G1, ATP5H
  hsa05016Huntington's disease 1.19×10−2ATP5B, ATP5C1, ATP5G1, ATP5H
  hsa04514Cell adhesion molecules 5.00×10−2NCAM1, ICAM1, HLA-DRB5

[i] Term represents the identification number of the KEGG pathway; description represents the name of the KEGG pathway. KEGG, Kyoto Encyclopedia of Genes and Genomes.

Discussion

In the present study, the gene expression patterns obtained from the GEO database revealed a total of 819 genes, including 759 upregulated and 60 downregulated genes, that were differently expressed in peri-implantitis samples compared with controls. The results of the present study demonstrated that HSP90AA1, which had the highest degrees in the PPI network, was significantly identified in module 4. In addition, NFKB1 were also hub nodes with higher betweenness values in the PPI network. Moreover, the proteasome pathway was the most significant pathway in module 1, and may be key mechanisms associated with peri-implantitis progression.

HSP90, a member of the heat shock family of proteins, is essential in determining cell cycle control and survival, hormone and a number of signaling pathways (24). In addition, released HSP90, which functions as a danger signal, can elicit secretion of inflammatory cytokines (25). A cell-impermeable HSP90 inhibitor can prevent inflammatory responses, indicating that extracellular HSP90 is involved in mediating and initiating sterile inflammatory responses (26). Moreover, previous data indicate that HSP90-targeted agents may be helpful for the treatment of inflammatory disease, and that an HSP90 inhibitor can affect multiple signaling processes associated with inflammation (27). Furthermore, inhibition of HSP90 is able to reduce innate immunity responses and diminishes proinflammatory mediator production in immune-stimulated macrophages (28,29). In addition, analysis of inflammatory mediators in crevicular fluid can be used to distinguish peri-implantitis from normal tissue (30).

Systemic markers of inflammation are increased in patients with peri-implantitis (30) and high levels of inflammatory cytokines, such as IL-1β, are associated with signs of early peri-implantitis development (3133). Furthermore, it has been suggested that fibroblasts express HSP90 that also participates in the pathogenesis of peri-implantitis (7,34). Moreover, HSPs together with vascular and inflammatory biomarkers may be useful as biomarkers of peri-implantitis development (35,36). In the present study, HSP90AA1, a hub node with higher node degrees in module 4, was enriched in antigen processing and the presentation pathway, as well as the NOD-like receptor-signaling pathway. Therefore, the results of the present study are in line with results from former previous studies and indicate that HSP90AA1 may be directly or indirectly important in peri-implantitis development.

In the present study, NFKB1 was shown to also have hub nodes with higher betweenness values in the PPI network. The NFKB1 gene is known to encode the NF-KB p105/p50 isoforms (37). The central pathological pattern of peri-implantitis is inflammatory osteoclastogenesis, which is mediated by proinflammatory mediators and performed by the regulators of osteoclastogenesis, including NF-B, sRANKL and OPG (38,39). A prior study indicated that the NF-B concentration increased 1.5–4-fold in patients with peri-implantitis compared to those with mucositis (6). Moreover, recent studies demonstrated that high levels of NF-B were associated with peri-implantitis (40,41). In addition, NF-B is upregulated in inflammatory bowel disease and is a transcription regulator of the immune response (42). Zou et al (43) reported that NFKB1 could regulate the transcription of genes in the immune response, and was also a key part in coordinating the immune system. Numerous studies have indicated that the NFKB1-94ins/del ATTG promoter polymorphism is associated with inflammatory disease (37,42,44,45). Furthermore, as mentioned in the aforementioned paragraph, inflammation is associated with the pathogenesis of peri-implantitis. Therefore, the results of the present study are in accordance with those from previous studies and suggest that NFKB1 may be a key gene associated with peri-implantitis development. Notably in the current study, HSP90AA1 and NFKB1 were enriched in the NOD-like receptor-signaling pathway. Although the important roles of the NOD-like receptor-signaling pathway have not been fully discussed, it is speculated that HSP90AA1 and NFKB1 may be involved in peri-implantitis progression via the NOD-like receptor-signaling pathway.

In addition to the two aforementioned genes, the proteasome pathway containing the proteasome subunits (PSM) A1, PSMA3, PSMA4, PSMB1, PSMB3, PSMB4, PSMB6, PSMC2, PSMD8, PSME3 and PSME4, were found to be enriched in the most significant pathway in module 1 and to have the highest P-value. Elliott et al (46) indicated that key proteins modulated by the proteasome were involved in controlling inflammatory processes. A prior study demonstrated that the activation of the ubiquitin-proteasome by macrophages is involved in an NF-B-dependent increase in inflammation (47). Moreover, previous studies have indicated that the proteasomal pathway as a targeting goal may be a potential way to treat autoimmune and inflammatory diseases (4850). In addition, Wang and Maldonado (51) demonstrated that proteasome inhibitors could serve as novel drugs by regulating proinflammatory protein catabolism. Furthermore, as stated above, inflammation has been associated with the pathogenesis of peri-implantitis. Finally, the PSMA1, PSMA3, PSMA4, PSMB1, PSMB3, PSMB4, PSMB6, PSMC2, PSMD8, PSME3 and PSME4 proteins are essential subunits that contribute to the assembly of the proteasome complex (5255). Therefore, the proteasome and its subunit genes may be important in peri-implantitis progression.

In conclusion, HSP90AA1, NFKB1 and the NOD-like receptor signaling pathway, as well as the proteasome pathway and its subunits genes may be important in the development of peri-implantitis. However, there were certain limitations in the present study, such as no experimental verification and the relatively small sample size used. Therefore, further research investigating the potential mechanisms involved in peri-implantitis are required.

References

1 

Schminke B, Vom Orde F, Gruber R, Schliephake H, Bürgers R and Miosge N: The pathology of bone tissue during peri-implantitis. J Dent Res. 94:354–361. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Mombelli A: Microbiology and antimicrobial therapy of peri-implantitis. Periodontol 2000. 28:177–189. 2002. View Article : Google Scholar : PubMed/NCBI

3 

Berglundh T, Persson L and Klinge B: A systematic review of the incidence of biological and technical complications in implant dentistry reported in prospective longitudinal studies of at least 5 years. J Clin Periodontol. 29:(Suppl 3). 197–212, 232–233. 2002. View Article : Google Scholar : PubMed/NCBI

4 

Becker ST, Beck-Broichsitter BE, Graetz C, Dörfer CE, Wiltfang J and Häsler R: Peri-implantitis versus periodontitis: Functional differences indicated by transcriptome profiling. Clin Implant Dent Relat Res. 16:401–411. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Theocharis AD, Seidel C, Borset M, Dobra K, Baykov V, Labropoulou V, Kanakis I, Dalas E, Karamanos NK, Sundan A and Hjerpe A: Serglycin constitutively secreted by myeloma plasma cells is a potent inhibitor of bone mineralization in vitro. J Biol Chem. 281:35116–35128. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Rakic M, Struillou X, Petkovic-Curcin A, Matic S, Canullo L, Sanz M and Vojvodic D: Estimation of bone loss biomarkers as a diagnostic tool for peri-implantitis. J Periodontol. 85:1566–1574. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Bordin S, Flemmig TF and Verardi S: Role of fibroblast populations in peri-implantitis. Int J Oral Maxillofac Implants. 24:197–204. 2009.PubMed/NCBI

8 

Irshad M, Scheres N, Anssari Moin D, Crielaard W, Loos BG, Wismeijer D and Laine ML: Cytokine and matrix metalloproteinase expression in fibroblasts from peri-implantitis lesions in response to viable Porphyromonas gingivalis. J Periodontal Res. 48:647–656. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Casado PL, Canullo L, de Almeida Filardy A, Granjeiro JM, Barboza EP and Leite Duarte ME: Interleukins 1β and 10 expressions in the periimplant crevicular fluid from patients with untreated periimplant disease. Implant Dent. 22:143–150. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Park SY, Kim KH, Gwak EH, Rhee SH, Lee JC, Shin SY, Koo KT, Lee YM and Seol YJ: Ex vivo bone morphogenetic protein 2 gene delivery using periodontal ligament stem cells for enhanced re-osseointegration in the regenerative treatment of peri-implantitis. J Biomed Mater Res A. 103:38–47. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Gautier L, Cope L, Bolstad BM and Irizarry RA: affy-analysis of Affymetrix GeneChip data at the probe level. Bioinformatics. 20:307–315. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Carvalho BS and Irizarry RA: A framework for oligonucleotide microarray preprocessing. Bioinformatics. 26:2363–2367. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

14 

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

15 

Altermann E and Klaenhammer TR: PathwayVoyager: pathway mapping using the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. BMC Genomics. 6:602005. View Article : Google Scholar : PubMed/NCBI

16 

da Huang W, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2008. View Article : Google Scholar

17 

Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP, et al: STRING v10: Protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 43:D447–D452. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Jeong H, Mason SP, Barabási AL and Oltvai ZN: Lethality and centrality in protein networks. Nature. 411:41–42. 2001. View Article : Google Scholar : PubMed/NCBI

20 

Estrada E and Rodríguez-Velázquez JA: Subgraph centrality in complex networks. Phys Rev E Stat Nonlin Soft Matter Phys. 71:0561032005. View Article : Google Scholar : PubMed/NCBI

21 

Goh KI, Oh E, Kahng B and Kim D: Betweenness centrality correlation in social networks. Phys Rev E Stat Nonlin Soft Matter Phys. 67:0171012003. View Article : Google Scholar : PubMed/NCBI

22 

Csardi G and Nepusz T: The igraph software package for complex network research. Inter Journal Complex Systems. 1695:1–9. 2006.

23 

Bader GD and Hogue CW: An automated method for finding molecular complexes in large protein interaction networks. BMC Bioinformatics. 4:22003. View Article : Google Scholar : PubMed/NCBI

24 

Bucci M, Roviezzo F, Cicala C, Sessa WC and Cirino G: Geldanamycin, an inhibitor of heat shock protein 90 (Hsp90) mediated signal transduction has anti-inflammatory effects and interacts with glucocorticoid receptor in vivo. Br J Pharmacol. 131:13–16. 2000. View Article : Google Scholar : PubMed/NCBI

25 

Wallin RP, Lundqvist A, Moré SH, von Bonin A, Kiessling R and Ljunggren HG: Heat-shock proteins as activators of the innate immune system. Trends Immunol. 23:130–135. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Qin S, Ni M, Wang X, Maurier-Mahé F, Shurland DL and Rodrigues GA: Inhibition of RPE cell sterile inflammatory responses and endotoxin-induced uveitis by a cell-impermeable HSP90 inhibitor. Exp Eye Res. 93:889–897. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Rice JW, Veal JM, Fadden RP, Barabasz AF, Partridge JM, Barta TE, Dubois LG, Huang KH, Mabbett SR, Silinski MA, et al: Small molecule inhibitors of Hsp90 potently affect inflammatory disease pathways and exhibit activity in models of rheumatoid arthritis. Arthritis Rheum. 58:3765–3775. 2008. View Article : Google Scholar : PubMed/NCBI

28 

De Nardo D, Masendycz P, Ho S, Cross M, Fleetwood AJ, Reynolds EC, Hamilton JA and Scholz GM: A central role for the Hsp90· Cdc37 molecular chaperone module in interleukin-1 receptor-associated-kinase-dependent signaling by Toll-like receptors. J Biol Chem. 280:9813–9822. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Shimp SK III, Parson CD, Regna NL, Thomas AN, Chafin CB, Reilly CM and Rylander Nichole M: HSP90 inhibition by 17-DMAG reduces inflammation in J774 macrophages through suppression of Akt and nuclear factor-κB pathways. Inflamm Res. 61:521–533. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Hultin M, Gustafsson A, Hallström H, Johansson LÅ, Ekfeldt A and Klinge B: Microbiological findings and host response in patients with peri-implantitis. Clin Oral Implants Res. 13:349–358. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Tokoro Y, Yamamoto T and Hara K: IL-1 beta mRNA as the predominant inflammatory cytokine transcript: Correlation with inflammatory cell infiltration into human gingiva. J Oral Pathol Med. 25:225–231. 1996. View Article : Google Scholar : PubMed/NCBI

32 

Panagakos FS, Aboyoussef H, Dondero R and Jandinski JJ: Detection and measurement of inflammatory cytokines in implant crevicular fluid: A pilot study. Int J Oral Maxillofac Implants. 11:794–799. 1996.PubMed/NCBI

33 

Aboyoussef H, Carter C, Jandinski JJ and Panagakos FS: Detection of prostaglandin E2 and matrix metalloproteinases in implant crevicular fluid. Int J Oral Maxillofac Implants. 13:689–696. 1998.PubMed/NCBI

34 

Lee SS, Jang JH, Kim KS, Yoo YJ, Kim YS and Lee SK: Failure of bone regeneration after demineralized bone matrix allograft in human maxillary sinus floor elevation. Basic Appl Pathol. 2:125–130. 2009. View Article : Google Scholar

35 

Bullon P, Fioroni M, Goteri G, Rubini C and Battino M: Immunohistochemical analysis of soft tissues in implants with healthy and peri-implantitis condition, and aggressive periodontitis. Clin Oral Implants Res. 15:553–559. 2004. View Article : Google Scholar : PubMed/NCBI

36 

Borsani E, Salgarello S, Stacchiotti A, Mensi M, Boninsegna R, Ricci F, Zanotti L, Rezzani R, Sapelli P, Bianchi R and Rodella LF: Altered immunolocalization of heat-shock proteins in human peri-implant gingiva. Acta Histochem. 109:221–227. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Borm ME, van Bodegraven AA, Mulder CJ, Kraal G and Bouma G: A NFKB1 promoter polymorphism is involved in susceptibility to ulcerative colitis. Int J Immunogenet. 32:401–405. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Cochran DL: Inflammation and bone loss in periodontal disease. J Periodontol. 79:(8 Suppl). 1569–1576. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Theoleyre S, Wittrant Y, Tat SK, Fortun Y, Redini F and Heymann D: The molecular triad OPG/RANK/RANKL: Involvement in the orchestration of pathophysiological bone remodeling. Cytokine Growth Factor Rev. 15:457–475. 2004. View Article : Google Scholar : PubMed/NCBI

40 

Rakić M, Nikolić-Jakoba N, Struillout X, Petković-Curcin A, Stamatović N, Matić S, Janković S, Aleksić Z, Vasilić D, Leković V and Vojvodić D: Receptor activator of nuclear factor kappa B (RANK) as a determinant of peri-implantitis. Vojnosanit Pregl. 70:346–351. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Rakic M, Lekovic V, Nikolic-Jakoba N, Vojvodic D, Petkovic-Curcin A and Sanz M: Bone loss biomarkers associated with peri-implantitis. A cross-sectional study. Clin Oral Implants Res. 24:1110–1116. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Karban AS, Okazaki T, Panhuysen CI, Gallegos T, Potter JJ, Bailey-Wilson JE, Silverberg MS, Duerr RH, Cho JH, Gregersen PK, et al: Functional annotation of a novel NFKB1 promoter polymorphism that increases risk for ulcerative colitis. Hum Mol Genet. 13:35–45. 2004. View Article : Google Scholar : PubMed/NCBI

43 

Zou YF, Wang F, Feng XL, Tao JH, Zhu JM, Pan FM and Su H: Association of NFKB1-94ins/delATTG promoter polymorphism with susceptibility to autoimmune and inflammatory diseases: A meta-analysis. Tissue antigens. 77:9–17. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Li H, Gao L, Shen Z, Li CY, Li K, Li M, Lv YJ, Li CX, Gao TW and Liu YF: Association study of NFKB1 and SUMO4 polymorphisms in Chinese patients with psoriasis vulgaris. Arch Dermatol Res. 300:425–433. 2008. View Article : Google Scholar : PubMed/NCBI

45 

Kosoy R and Concannon P: Functional variants in SUMO4, TAB2, and NFkappaB and the risk of type 1 diabetes. Genes Immun. 6:231–235. 2005. View Article : Google Scholar : PubMed/NCBI

46 

Elliott PJ, Zollner TM and Boehncke WH: Proteasome inhibition: A new anti-inflammatory strategy. J Mol Med (Berl). 81:235–245. 2003. View Article : Google Scholar : PubMed/NCBI

47 

Marfella R, D'Amico M, Di Filippo C, Baldi A, Siniscalchi M, Sasso FC, Portoghese M, Carbonara O, Crescenzi B, Sangiuolo P, et al: Increased activity of the ubiquitin-proteasome system in patients with symptomatic carotid disease is associated with enhanced inflammation and may destabilize the atherosclerotic plaque: Effects of rosiglitazone treatment. J Am Coll Cardiol. 47:2444–2455. 2006. View Article : Google Scholar : PubMed/NCBI

48 

Goldberg AL and Rock K: Not just research tools-proteasome inhibitors offer therapeutic promise. Nat Med. 8:338–340. 2002. View Article : Google Scholar : PubMed/NCBI

49 

Lecker SH, Goldberg AL and Mitch WE: Protein degradation by the ubiquitin-proteasome pathway in normal and disease states. J Am Soc Nephrol. 17:1807–1819. 2006. View Article : Google Scholar : PubMed/NCBI

50 

Ciechanover A: The ubiquitin proteolytic system and pathogenesis of human diseases: A novel platform for mechanism-based drug targeting. Biochem Soc Trans. 31:474–481. 2003. View Article : Google Scholar : PubMed/NCBI

51 

Wang J and Maldonado MA: The ubiquitin-proteasome system and its role in inflammatory and autoimmune diseases. Cell Mol Immunol. 3:255–261. 2006.PubMed/NCBI

52 

Nothwang HG, Tamura T, Tanaka K and Ichihara A: Sequence analyses and inter-species comparisons of three novel human proteasomal subunits, HsN3, HsC7-I and HsC10-II, confine potential proteolytic active-site residues. Biochim Biophys Acta. 1219:361–368. 1994. View Article : Google Scholar : PubMed/NCBI

53 

Gallastegui N and Groll M: The 26S proteasome: Assembly and function of a destructive machine. Trends Biochem Sci. 35:634–642. 2010. View Article : Google Scholar : PubMed/NCBI

54 

Le Tallec B, Barrault MB, Courbeyrette R, Guérois R, Marsolier-Kergoat MC and Peyroche A: 20S proteasome assembly is orchestrated by two distinct pairs of chaperones in yeast and in mammals. Mol Cell. 27:660–674. 2007. View Article : Google Scholar : PubMed/NCBI

55 

Murata S, Yashiroda H and Tanaka K: Molecular mechanisms of proteasome assembly. Nat Rev Mol Cell Biol. 10:104–115. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2017
Volume 13 Issue 5

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
Zhang H, Zhang X, Huang J and Fan X: Identification of key genes and pathways for peri-implantitis through the analysis of gene expression data. Exp Ther Med 13: 1832-1840, 2017
APA
Zhang, H., Zhang, X., Huang, J., & Fan, X. (2017). Identification of key genes and pathways for peri-implantitis through the analysis of gene expression data. Experimental and Therapeutic Medicine, 13, 1832-1840. https://doi.org/10.3892/etm.2017.4176
MLA
Zhang, H., Zhang, X., Huang, J., Fan, X."Identification of key genes and pathways for peri-implantitis through the analysis of gene expression data". Experimental and Therapeutic Medicine 13.5 (2017): 1832-1840.
Chicago
Zhang, H., Zhang, X., Huang, J., Fan, X."Identification of key genes and pathways for peri-implantitis through the analysis of gene expression data". Experimental and Therapeutic Medicine 13, no. 5 (2017): 1832-1840. https://doi.org/10.3892/etm.2017.4176