Identification The Low Oxidative Stress Subtype of Periodontitis

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

international dental journal 7 4 ( 2 0 2 4 ) 1 1 9 − 1 2 8

Scientific Research Report

Identification the Low Oxidative Stress Subtype of


Periodontitis

Yuchen Wu a, Xianfang Zhang a, Yunong Chen a, Weiting Chen b,


Wenhao Qian c*
a
Department of Prosthodontics, Shanghai Xuhui District Dental Center, Shanghai, People’s Republic of China
b
Department of Periodontology, Shanghai Xuhui District Dental Center, Shanghai, People’s Republic of China
c
Department of Oral Implantology, Shanghai Xuhui District Dental Center, Shanghai, People’s Republic of China

A R T I C L E I N F O A B S T R A C T

Article history: Objective: The aim of this research was to identify the low oxidative stress−related genes
Received 25 April 2023 expression (L-OS) subtype in patients with periodontitis.
Received in revised form Methods: Microarray data (MA) were retrieved from the Gene Expression Omnibus database.
7 July 2023 The different oxidative stress (OS) subtypes of periodontitis were identified by K-means
Accepted 20 July 2023 clustering analysis and gene set variation analysis (GSVA). Differentially expressed genes
Available online 10 October 2023 (DEGs) (|Log fold change (FC)| >1, q < 0.05) amongst the OS subtypes and healthy controls
(HCs) were identified by Limma R package. The genomic feature of L-OS subtype and corre-
Key words: sponding medicines were evaluated and visualised with Drug-Gene Interaction Database
Periodontitis and cytoscape-v3.7.2 software (Pearson correlation coefficient > 0.4). Finally, the LASSO-
Low oxidative stress Logistic regression model was adopted to evaluate and predict patients’ OS phenotype in
Microarray data routine clinical practice.
TFs-Genes-Medicines network Results: The 241 periodontitis samples and 69 HCs were included. Thirty-three DEGs
Machine learning between the L-OS and high oxidative stress−related genes expression (H-OS) subtypes and
96 DEGs, including 8 transcription factors, between L-OS subtype and HCs were identified,
respectively. Then, the network of TFs-Genes-Drugs was constructed to determine geno-
mic feature of L-OS subtype. Finally, a 4-gene signature formula and the cutoff value were
identified by ML with LASSO model to predict patients’ classification.
Conclusions: For the first time, we identified L-OS subtype of periodontitis and evaluated its
genomic feature with MA.
Ó 2023 The Authors. Published by Elsevier Inc. on behalf of FDI World Dental Federation.
This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/)

Introduction periodontitis is not fully understood, and no effective drugs


have been approved for clinical treatment.7
Periodontitis involves various chronic inflammations of tooth- Previous studies showed that the oxidative stress (OS) caused
supporting structures including ligaments, gums, and bones.1,2 by the imbalance of intracellular oxidant−antioxidant system
Periodontitis can cause tooth loss and exacerbate many medi- was entwined with periodontitis.8,9 When invaded by pathogenic
cal conditions, such as atherosclerosis, type 2 diabetes mellitus, microorganism, the periodontal tissue releases pathogenic fac-
and even cancer by increasing the inflammatory burden in the tors and recruited inflammatory cells, such as neutrophils,
periodontal microenvironment or throughout the body.3-5 Stud- monocytes, and macrophages. Phagocytes bring about bacteri-
ies have shown that the incidence of periodontitis in the popu- cidal activity through “respiratory bursts” and released reactive
lation is about 30%, and more than one-third of them had oxygen species (ROS).10,11 As the result of persistent chronic
severe periodontitis, which confers major health and financial inflammatory response, ROS produced excessively in the peri-
burden to individuals and societies.6 To date, the aetiology of odontal microenvironment leading to OS, and eventual oxidative
damage to DNA, proteins, lipids. These may in turn lead to the
degeneration of periodontal ligament, fibrous connective tissue,
* Corresponding author. No. 500 Fenglin Road, Xuhui District
and alveolar bone.12 Hence reducing tissue OS damage should be
Shanghai 200032, People’s Republic of China.
an effective method for managing periodontitis, but the out-
E-mail address: ORCID
Yuchen Wu: http://orcid.org/0000-0002-7596-9524 comes are variable and unsatisfactory, and the molecular
https://doi.org/10.1016/j.identj.2023.07.011
0020-6539/Ó 2023 The Authors. Published by Elsevier Inc. on behalf of FDI World Dental Federation. This is an open access article under
the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/)
120 wu et al.

mechanisms underlying this phenomenon are not clear.13 To which was completed by R package ConsensusClusterPlus.21
date, whether the expression levels of OS-related genes are dis- Pam cluster algorithm, which is more robust than the K-m
crepant in periodontitis or critical for routine clinical practice is method and effective for a small dataset due to its insensitivity
still unknown, and it is challenging to obtain reliable and quanti- to outliers, was adopted here. GSVA, a method for the unsuper-
tative OS measurement values from a considerable number of vised classification of samples, was adopted to evaluate the
human samples. Further efforts are therefore needed to explore expression of OS gene sets between subgroups.
the pathogenesis of periodontal disease and develop novel thera-
peutic drugs. Identification of the genomic features for the L-OS subtype of
Recent advances in high-throughput sequencing and data periodontitis
analysis have made it possible to monitor, quantify, and corre-
late biological signals with cellular phenotypes.14,15 As one of To identify the genomic feature of the L-OS subtype of peri-
the transcriptomics, microarray gene expression analysis can odontitis, differential analysis was applied between Cluster 1,
detect differential genes expression for a multitude of RNAs lowly enriched in OS pathways, and normal periodontal tis-
simultaneously, which makes it feasible to assess the activation sues. Limma R package (version 4.0.4.), a commonly used differ-
of OS-related pathways comprehensively.16 Data-driven entially expressed genes (DEGs) identification method, was
machine learning (ML) has gained increased attention from adopted here, and genes with Log fold change (FC) >1 or < 1,
researchers due to its superior performance in transforming and adjusted P value< .05 were deemed to be DEGs. Subse-
biological detection and treatment of human diseases.17 Vari- quently, those DEGs were divided into transcription factors
ous clustering methods have been used in combination with (TFs) and their targeting genes by the Pearson correlation anal-
ML, such as K-means and Pam cluster analysis. The application ysis (PCA). Significant TFs−genes interaction pairs were
of combination high-throughput phenotypic screening and ML selected if their Pearson correlation coefficient (PCC) was >0.4.
could contribute to deep understanding the pathogenesis of Next, by integrating data from the Drug-Gene Interaction Data-
periodontitis. base (DGIdb), a public database providing information of inter-
Thus, the aim of the current study is to assess the expres- action between drugs and genes (https://dgidb.genome.wustl.
sion levels of OS-related genes in patients with periodontitis edu/), the potential medicines for the key TFs and genes in the
and identify the low oxidative stress−related genes expres- L-OS subtype were also located. At last, the TFs-Genes-Medi-
sion (L-OS) subtype as well as the related genomic features by cines network was visualised by cytoscape-v3.7.2 software22 to
using microarray data and ML. guide drug application in the clinical practice.

ML-based 4 genes signature to recognise the L-OS subtype of


Methods patients with periodontitis in clinical practice

Data acquisition and processing To guide clinical classification, we further constructed an ML-
based 4-gene signature to commodiously distinguish patients
In all, 241 samples with periodontitis and 69 control samples into L-OS and high oxidative stress−related genes expression
were included in GSE16134, which was downloaded from the (H-OS) group, and subsets of marker genes and OS score were
Gene Expression Omnibus (GEO) database of National Center chosen by using the Lasso regression algorithm. First, differen-
for Biotechnology Information (NCBI)18,19 (http://www.ncbi. tial analysis was carried out between Cluster 1 and Cluster 3,
nlm.nih.gov/geo/). The inclusion criteria for all participants which are lowly and highly enriched in OS pathways, respec-
were as follows: at least 13 years old, not pregnant, not using tively. Then, the DEGs between the 2 clusters were subjected to
tobacco products or nicotine substitutes, have more than 24 logistic regression model with LASSO penalisation. Accordingly,
teeth, and have not been on systemic antibiotics or anti- the OS score was computed for each patient by the following
inflammatory medications for more than 6 months. The formula: Sni Coefi*Xi (Coefi: logistic regression coefficient, Xi:
patients with periodontitis also need to meet the following expression value of corresponding gene). Then, 70% of periodon-
criteria: bleeding on probing, probing pocket depth ≥4 mm, titis samples were assigned to the training set to generate the 4-
and clinical attachment loss ≥3 mm.18,19 The healthy and gene signature formula by using the ML, and the remaining 30%
periodontal pathology gingival tissue biopsies were obtained of periodontitis samples were assigned as the test set to test the
from healthy controls and patients with periodontitis.18,19 performance of this model. Finally, receiver operating character-
Microarray data were centralised by log2 (x+1) transformation istic (ROC) curve and area under the curve (AUC) of ROC were
and normalised by using the “normalize between array” func- taken into consideration to evaluate the model’s performance in
tion of R package “limma.”20 Meanwhile, 16 OS pathways and both training and validation sets.
the 580 OS-related genes were obtained from the Gene Ontol-
ogy (GO) resource (http://geneontology.org/). Statistics

Identification of the L-OS subtype of periodontitis Data processing and all analyses were accomplished by R 4.0.4.
(Package: limma, ggplot2, GSVA, glmnet, ConsensusClusterPlus,
As an unsupervised learning technique, K-means clustering is a Goplot, and so on). Chi-square test was used for counting data.
commonly used clustering method. The value of 580 OS-related Wilcox or Kruskal−Wallis test were applied for comparisons
genes were applied as the input feature of K-means clustering between groups, whilst Pearson and Spearman rank correlation
to identify the L-OS of the 241 samples with periodontitis, were adopted to estimate the statistical correlation of
i d e n t i fi c a t i o n t h e l o w o x i d a t i v e s t r e s s s u b t y p e o f p e r i o d o n t i t i s 121

parametric or nonparametric variables. Two-sided P < .05 was cluster was excluded due to its small sample size (only 3 sam-
considered as significant threshold for all statistical tests. ples for each analysis) (Figure 2D). These 3 remaining clusters
This study was based on secondary database which is pub- demonstrated huge differences in GSVA pathway enrichment
licly available in GEO (https://www.ncbi.nlm.nih.gov/geo/ analysis (Supplementary Table 2). Cluster 1 (67 of 241 samples)
query/acc.cgi?acc=GSE16134). was seen to be lowly enriched in oxidative-related pathways,
whilst Cluster 3 (98 of 241 samples) displayed higher enrich-
ment scores in the same genesets (Figure 2E). Accordingly,
Results about one-quarter of samples in Cluster 1 seem to be the L-OS
subtype of periodontal disease, which is less likely to respond
Identification of the L-OS subtype of periodontitis to antioxidant than Cluster 3 (Figure 2E).

The workflow is depicted in Figure 1. The baseline features of The genomic features of the L-OS subtype of periodontial
samples collected from the GEO database are described in Sup- disease
plementary Table 1. First, microarray data of GSE1613418,19
were retrieved and included. In all, 580 genes were extracted We identified L-OS subtype genomic signatures by comparing
from 16 OS Go items, subjected to cluster analysis. Four optimal periodontitis tissues with healthy controls (HCs). As the
clusters were obtained from Pam cluster analysis (Figure 2A) results show in Figure 3A, 96 DEGs were identified between
according to the results of consensus of Cumulative Distribu- Cluster 1 and HCs, and the list of the DEGs is described in Sup-
tion Function (CDF) and area under CDF (Figure 2 B and C). One plementary Table 3. Amongst them, 53 genes were up-

Fig. 1 – The flow chart of this study.


122 wu et al.

Fig. 2 – Identification of the low oxidative stress−related genes expression subtype of periodontitis. A, Pam-clustering analysis of
all samples according to their enrichment degree of oxidative stress (OS) genes. B and C, The consensus Cumulative Distribution
Function (CDF) and delta area of the CDF curve. D, The tracking plot of CDF. E, GSVA pathway enrichment analysis.

regulated and 43 genes were down-regulated (Figure 3B) in Amongst this 96 DEGs, 8 TFs (ZBTB43, TET2, BNC1, CEBPG,
Cluster 1. The functional enrichment analysis showed the GRHL3, ISL1, FLI1, and ZNF57) came into our view for their
most significant different Kyoto Encyclopedia of Genes and close connection with other genes in PCA (Figure 4A). Espe-
Genomes (KEGG) item were seen to be mainly enriched in cially, TET2, BNC1, and FLI1 seemed to be in the dominant
“cell adhesion,” “leukocyte migration,” “macrophage migra- position, for they had dozens of connections with other
tion pathways,” and so on (Figure 3C), and the related genes genes. Then, a network of TFs-Genes-Drugs was constructed
are illustrated in Figure 3D. As leukocytes have been given by integrating drug−gene interaction information from DGIdb
a key role in the process of oxidation in periodontal dis- (Figure 4B).
ease, these results also provided an indirect evidence for
Cluster 1 to be the L-OS subtype. The GO analysis revealed ML-based 4-gene signature to recognise the L-OS subtype of
that the prominent biological processes “extracellular periodontal disease in clinical practice
matrix organisation” and “extracellular structure organ-
isation” were significantly different between these 2 We applied an ML algorithm to develop a predictive diagnos-
groups (Figure 3E). tic model using periodontitis samples. First, 33 DEGs between
i d e n t i fi c a t i o n t h e l o w o x i d a t i v e s t r e s s s u b t y p e o f p e r i o d o n t i t i s 123

Fig. 2 Continued.

the L-OS and H-OS subtypes (Cluster 1 and Cluster 3) were score was calculated to help guide clinical classification, and
identified by limma and admitted as the input features 1.41 was the best cutoff value to stratify patients into L-OS
(Figure 5A). The least absolute shrinkage and selection opera- and H-OS subtypes (Figure 5C and D).
tor (LASSO)-Logistic regression model was adopted to predict
patients’ OS phenotype (Cluster 1 or Cluster 3), with 70% of
samples being the training set and the remaining 30% as the Discussion
test set. Then, the 4-gene (AADACL2, FCRL5, SLAMF7, and
COMP) signature formula was constructed and applied to pre- Periodontitis is the sixth most prevalent disease in humans,
dict patients’ classification by LASSO model, and the coeffi- and as a chronic common disease amongst the elderly popula-
cient for each gene is described in Supplementary Table 4. tion, the incidence of periodontitis can reach to 70% in people
ROC was applied to evaluate the performance of the LASSO older than 65 years, which brings an escalating burden to the
logistic regression model in both datasets. The obtained diag- health care economy, especially with the ageing of the global
nostic prediction model discriminated patients with L-OS population.23 Periodontitis has irreversible adverse effects on
from periodontitis samples with high accuracy, and the AUC tooth-supporting structures and can cause tooth loss, poor
was 0.902 for the training set and 0.863 for the validation set nutritional status, and reduced quality of life.1,2 Studies have
(Figure 5B). In addition, the optimal threshold value of OS proved that severe periodontitis is a potential risk factor for
124 wu et al.

Fig. 3 – The differentially expressed genes (DEGs) and functional enrichment analysis between low oxidative stress−related
genes expression (L-OS) and control group. A, The heatmap and volcano plot (B) of DEGs between L-OS and control group. The
functional enrichment analysis showed the most significant different Kyoto Encyclopedia of Genes and Genomes (KEGG)
item and related genes (C, D) and most significant Gene Ontology (GO) items (E).

many systematic diseases, such as hypertension, atherosclero- tooth scaling, periodontal pocket flush, and guided tissue
sis, type 2 diabetes mellitus, and cancer.3,4 As a complex group regeneration) are variable and unsatisfactory.13
of inflammatory diseases, the aetiology of periodontitis is not In this study, we identified L-OS subtype of periodontitis and
fully understood, and the outcomes of clinical procedures (eg, evaluated its genomic feature by using bioinformatics
i d e n t i fi c a t i o n t h e l o w o x i d a t i v e s t r e s s s u b t y p e o f p e r i o d o n t i t i s 125

Fig. 4 – The network of TFs-Genes-Drugs. The heatmap of 8 transcription factors (TFs) (A), and the network was constructed
by integrating drug−gene interaction information from DGIdb, and the genes with red background were key differentially
expressed genes (DEGs), the yellow background were TFs, and the compounds marked with blue background were drugs for
low oxidative stress−related genes expression (L-OS) (B).

algorithm with microarray data in a pioneering way. We then pathogens appear to be insufficient, and providing additional
identified a 4-gene signature formulae and the cutoff values to antioxidants, such as resveratrol—which react with ROS stoi-
identify this subtype in clinical practice using ML. Our data may chiometrically—has emerged as a promising treatment for
provide a theoretical justification for use of antioxidants for periodontitis.9,26 But recently, studies have shown that the vari-
periodontitis. Due to the short survival time, ROS are difficult to able therapeutic effect of antioxidants in patients with peri-
detect. In previous studies, the level of OS in periodontitis was odontitis.27-29 Evaluating the expression levels of OS-related
analysed using indirect OS markers, such as the tissue expres- genes for clinicians before using antioxidants in periodontal
sion levels of superoxide dismutase and catalase.24 In our study, treatment might therefore be clinically relevant.
we took an innovative approach to examine the expression lev- KEGG pathway enrichment analysis of 96 DEGs in L-OS vs
els of all OS-related genes with high-throughput microarray controls suggested that the signaling pathways were essen-
data and successfully categorised the expression levels of OS- tially enriched in cell adhesion, leukocyte migration and mac-
related genes into 2 distinct subgroups: the L-OS and H-OS sub- rophage migration pathways. It has been confirmed that cell
group. To the best of our knowledge, this is the first study to adhesion molecules (such as vascular cell adhesion molecule
report the L-OS subtype of periodontitis. 1) can promote arterial inflammation by trapping inflamma-
Many studies have proved that periodontitis is initiated by tory cells such as macrophages and T lymphocytes at sites of
uncontrolled inflammatory response, and elevated OS plays a inflammation.30 Since the inflammatory environment
vital role in periodontitis by increasing periodontal microenvi- strongly enhances phagocyte migration and activities, leuko-
ronmental inflammation.12,25 That means that conventional cyte migration and macrophage migration have been given a
treatment for periodontitis that focus on combating bacterial key role in the process of oxidation in periodontal disease,
126 wu et al.

Fig. 5 – The identification of low oxidative stress−related genes expression (L-OS) subtype of periodontal disease in clinical
practice. A, The volcano plot of differentially expressed genes (DEGs) between L-OS and high oxidative stress−related genes
expression (H-OS). B, The AUC of 4-genes-signature to predict patients’ classification. C,The performance identification of
LASSO model in training group and validation group. AUC, area under the curve.

and these results provided indirect evidence for Cluster 1 to Recently, several automated ML strategies have been
be L-OS subtypes.31,32 GO analysis revealed that the promi- developed to excavate potential prognostic biomarkers.38
nent biological processes “extracellular matrix organisation” Pam cluster analysis is a multivariate statistical analysis
and “extracellular structural organisation,” which were method that classifies samples or indicators without prior
related to tissue fibrosis atrophy, were also upregulated in knowledge. The LASSO model regression algorithm can real-
periodontitis.33,34 Amongst the 8 TFs predicted in the present ise the selection of independent variables and estimates
study, TET2, BNC1, and FLI1 have been verified as key modu- model parameters at the same time by using L1-regularised
lators and potential therapeutic targets in a wide variety of linear regression.39 In this study, we integrated microarray
inflammatory diseases.35-37 data and ML algorithm to identify the L-OS subtype of
i d e n t i fi c a t i o n t h e l o w o x i d a t i v e s t r e s s s u b t y p e o f p e r i o d o n t i t i s 127

periodontitis and its genomic feature, as well as the diagnos- Wenhao Qian designed this study and revised articles. All
tic criteria. First, we used OS pathway genes as a reference to authors have read and approved the final manuscript.
detect the validity of its subsets in the range of 2 to 10 and
combined with CDF and delta area of CDF curve to determine
the optimal number of clusters through Pam cluster analysis. Funding
When the optimal number of subsets is 4, the area under CDF
no longer changes significantly (Figure 2B and C). For each This study was funded by the National Natural Science Foun-
event, the leftmost clustering, containing a very small num- dation of China (no. 52272283) and Xuhui District Medical
ber of samples, was stable amongst analyses (Figure 2D), Research Project of Shanghai (no. SHXH202141).
which is estimated to be outliers. After being excluded from
the leftmost clustering, the distribution of the remaining 3
types of samples was found to be more uniform. Further- Supplementary materials
more, 16 OS gene sets enrichment scored were evaluated by
GSVA in 3 remaining clusters, and L-OS and H-OS units were Supplementary material associated with this article can be
determined. Then, the clinical diagnosis model was estab- found in the online version at doi:10.1016/j.identj.2023.07.011.
lished by LASSO. We identified a 4-gene diagnostic prediction
models that can specifically distinguish the L-OS whilst the
value of OS score is <1.4. This diagnostic model provides a R E F E R E N C E S
reference for clinical treatment of antioxidant stress in
patients with periodontitis.
There are some limitations to this study. Given the inher- 1. Kinane DF, Stathopoulou PG, Papapanou PN. Periodontal dis-
ent issues related to bioinformatics analysis— the conclu- eases. Nat Rev Dis Primers 2017;3:17038.
2. Slots J. Periodontitis: facts, fallacies and the future. Periodon-
sions of this study may be constrained. The levels of
tol 2000 2017;75.
traditional OS indicators, superoxide dismutase, total antioxi-
3. Hajishengallis G. Periodontitis: from microbial immune sub-
dant capacity, and malondialdehyde, for example, also need version to systemic inflammation. Nat Rev Immunol 2015;15:
evauation to test the OS state of periodontal tissue in the 30–44.
L-OS group. Further multicentric clinical studies are war- 4. Romandini M, Baima G, Antonoglou G, Bueno J, Figuero E,
ranted to validate our results under varying circumstances Sanz M. Periodontitis, edentulism, and risk of mortality: a sys-
and confirm whether these subtypes have different clinic tematic review with meta-analyses. J Dent Res 2021;100:37–49.
5. Michaud DS, Kelsey KT, Papathanasiou E, Genco CA, Giovan-
prognosis as well as their response to antioxidant therapy.
nucci E. Periodontal disease and risk of all cancers among
male never smokers: an updated analysis of the Health Pro-
fessionals Follow-up Study. Ann Oncol 2016;27:941–7.
Conclusions 6. Ganesan SM, Joshi V, Fellows M, et al. A tale of two risks:
smoking, diabetes and the subgingival microbiome. ISME J
For the first time, we identified L-OS subtype of periodontitis 2017;11:2075–89.
7. Chapple ILC. Time to take periodontitis seriously. BMJ
and evaluated its genomic features with microarray data. We
2014;348:g2645.
first identified a 4-gene signature formula and its cutoff value
8. Darveau RP. Periodontitis: a polymicrobial disruption of host
to identify this subtype in clinical practice by ML algoruthms. homeostasis. Nat Rev Microbiol 2010;8:481–90.
Our findings may provide a theoretical justification for the 9. Sczepanik FSC, Grossi ML, Casati M, et al. Periodontitis is an
use of antioxidants to manage peridontal disease. inflammatory disease of oxidative stress: we should treat it
that way. Periodontol 2000 2020;84:45–68.
10. Pan W, Wang Q, Chen Q. The cytokine network involved in
the host immune response to periodontitis. Int J Oral Sci
Conflict of interest
2019;11:30.
11. Sui L, Wang J, Xiao Z, Yang Y, Yang Z, Ai K. ROS-scavenging
None disclosed. nanomaterials to treat periodontitis. Front Chem 2020;8:
595530.
12. Wang Y, Andrukhov O, Rausch-Fan X. Oxidative stress and
Acknowledgements antioxidant system in periodontitis. Front Physiol 2017;8:910.
13. Suvan J, Leira Y, Moreno Sancho FM, Graziani F, Derks J, Tom-
asi C. Subgingival instrumentation for treatment of periodon-
We acknowledge the GEO database for providing their plat-
titis. A systematic review. J Clin Periodontol 2020;47(Suppl
forms and Papapanou et al. for uploading their meaningful 22)):155–75.
datasets. 14. Yang JH, Wright SN, Hamblin M, et al. A white-box machine
learning approach for revealing antibiotic mechanisms of
action. Cell 2019:177.
Author contributions 15. Shokhirev MN, Johnson AA. An integrative machine-learning
meta-analysis of high-throughput omics data identifies age-
specific hallmarks of Alzheimer’s disease. Ageing Res Rev
All authors contributed to various parts of this manuscript.
2022;81:101721.
Yuchen Wu was the major contributor in writing the manu- 16. Mao K, Wu X. Microarray analysis of small extracellular vesi-
script. Xianfang Zhang, Yunong Chen, and Weiting Chen helped cle-derived miRNAs involved in oxidative stress of RPE cells.
perform the bioinformatics analysis and machine learning. Oxid Med Cell Longev 2020;2020:7658921.
128 wu et al.

17. Sengupta PP, Shrestha S. Machine learning for data-driven oral care regimen- a randomized placebo-controlled clinical
discovery: the rise and relevance. JACC Cardiovasc Imaging trial. BMC Complement Altern Med 2019;19:327.
2019;12:690–2. 29. El-Sharkawy H, Elmeadawy S, Elshinnawi U, Anees M. Is die-
18. Kebschull M, Demmer RT, Gru € n B, Guarnieri P, Pavlidis P, tary melatonin supplementation a viable adjunctive therapy
Papapanou PN. Gingival tissue transcriptomes identify dis- for chronic periodontitis?-A randomized controlled clinical
tinct periodontitis phenotypes. J Dent Res 2014;93:459–68. trial. J Periodontal Res 2019;54:190–7.
19. Papapanou PN, Behle JH, Kebschull M, et al. Subgingival bacte- 30. Shen J, Yang M, Ju D, et al. Disruption of SM22 promotes
rial colonization profiles correlate with gingival tissue gene inflammation after artery injury via nuclear factor kappaB
expression. BMC Microbiol 2009;9:221. activation. Circ Res 2010;106:1351–62.
20. Ritchie ME, Phipson B, Wu D, et al. limma powers differential 31. Liu P, Zhu W, Chen C, et al. The mechanisms of lysophospha-
expression analyses for RNA-sequencing and microarray tidylcholine in the development of diseases. Life Sci
studies. Nucleic Acids Res 2015;43 e47-e. 2020;247:117443.
21. Wilkerson MD, Hayes DN. ConsensusClusterPlus: a class dis- 32. de Oliveira S, Rosowski EE, Huttenlocher A. Neutrophil migra-
covery tool with confidence assessments and item tracking. tion in infection and wound repair: going forward in reverse.
Bioinformatics (Oxford, England) 2010;26:1572–3. Nat Rev Immunol 2016;16:378–91.
22. Shannon P, Markiel A, Ozier O, et al. Cytoscape: a software 33. Dempsey LA. Defective fibrolysis in periodontitis. Nat Immu-
environment for integrated models of biomolecular interac- nol 2022;23:150.
tion networks. 2003;13:2498−504. 34. Takeuchi H, Kubota S, Murakashi E, et al. Nicotine-induced
23. Eke PI, Wei L, Borgnakke WS, et al. Periodontitis prevalence in CCN2: from smoking to periodontal fibrosis. J Dent Res
adults ≥ 65 years of age, in the USA. Periodontol 2000 2010;89:34–9.
2016;72:76–95. 35. Cai Z, Kotzin JJ, Ramdas B, et al. Inhibition of inflammatory
24. Chen S, Gao X, Song J. Oxidative stress-related biomarkers in signaling in tet2 mutant preleukemic cells mitigates stress-
chronic periodontitis patients with or without type 2 diabetes: induced abnormalities and clonal hematopoiesis. Cell Stem
a systematic review and meta-analysis. J Periodontal Res Cell 2018;23.
2023;58:780–90. 36. Gao Y, Li J, Jiao J, et al. Upregulation of basonuclin1 is associated
25. Zhang Z, Zheng Y, Bian X, et al. Identification of key genes and with p63-involved epithelial barrier impairment and type-2
pathways associated with oxidative stress in periodontitis. helper T-cell inflammation in chronic rhinosinusitis with nasal
Oxid Med Cell Longev 2022;2022:9728172. polyps. Int Arch Allergy Immunol 2021;182:1046–57.
26. Vo TTT, Chu P-M, Tuan VP, Te JS-L, Lee IT. The promising role of 37. Chen M, Wang Y, Deng S, Lian Z, Yu K. Skeletal muscle oxidative
antioxidant phytochemicals in the prevention and treatment of stress and inflammation in aging: focus on antioxidant and
periodontal disease via the inhibition of oxidative stress path- anti-inflammatory therapy. Front Cell Dev Biol 2022;10:964130.
ways: updated insights. Antioxidants (Basel) 2020;9. 38. Jiang C, Liu Y, Wen S, Xu C, Gu L. In silico development and
27. Seydanur Dengizek E, Serkan D, Abubekir E, Aysun Bay K, clinical validation of novel 8 gene signature based on lipid
Onder O, Arife C. Evaluating clinical and laboratory effects of metabolism related genes in colon adenocarcinoma. Pharma-
ozone in non-surgical periodontal treatment: a randomized col Res 2021;169:105644.
controlled trial. J Appl Oral Sci 2019;27:e20180108. 39. Vitzthum LK, Riviere P, Sheridan P, et al. Predicting persistent
28. Nayak N, Varghese J, Shetty S, et al. Evaluation of a mouth- opioid use, abuse, and toxicity among cancer survivors. J Natl
rinse containing guava leaf extract as part of comprehensive Cancer Inst 2020;112:720–7.

You might also like