Skip to main content
PLOS One logoLink to PLOS One
. 2024 Jan 19;19(1):e0296925. doi: 10.1371/journal.pone.0296925

Distinctive genes and signaling pathways associated with type 2 diabetes-related periodontitis: Preliminary study

Poliana Mendes Duarte 1,*,#, Bruno César de Vasconcelos Gurgel 2, Tamires Szeremeske Miranda 3, Juliana Sardenberg 1, Tongjun Gu 4, Ikramuddin Aukhil 5,*,#
Editor: Víctor Sánchez-Margalet6
PMCID: PMC10798476  PMID: 38241313

Abstract

The biological mechanisms underlying the pathogenesis of type 2 diabetes (T2DM)-related periodontitis remain unclear. This cross-sectional study evaluated the distinctive transcriptomic changes between tissues with periodontal health and with periodontitis in patients with T2DM. In this cross-sectional study, whole transcriptome sequencing was performed on gingival biopsies from non-periodontitis and periodontitis tissues from non-diabetic and diabetic patients. A differentially expressed gene (DEG) analysis and Ingenuity Pathway Analysis (IPA) assessed the genes and signaling pathways associated with T2DM-related periodontitis. Immunohistochemistry was performed to validate selected DEGs possibly involved in T2DM-related periodontitis. Four hundred and twenty and one thousand five hundred and sixty-three DEGs (fold change ≥ 2) were uniquely identified in the diseased tissues of non-diabetic and diabetic patients, respectively. The IPA predicted the activation of Phagosome Formation, Cardiac β-adrenergic, tRNA Splicing, and PI3K/AKT pathways. The IPA also predicted the inhibition of Cholesterol Biosynthesis, Adrenomedullin, and Inositol Phosphate Compounds pathways in T2DM-related periodontitis. Validation of DEGs confirmed changes in protein expression of PTPN2, PTPN13, DHCR24, PIK3R2, CALCRL, IL1RN, IL-6R and ITGA4 in diseased tissues in diabetic subjects. Thus, these preliminary findings indicate that there are specific genes and functional pathways that may be involved in the pathogenesis of T2DM-related periodontitis.

Introduction

Diabetes mellitus (DM) has long been documented as a risk factor for periodontitis, a highly prevalent infectious-inflammatory disease that can cause periodontal tissue breakdown and, ultimately, tooth loss [1, 2]. DM is considered a disease grade modifier by the classification system of periodontal diseases, used as a critical indicator of periodontitis progression [3].

Previous studies have attempted to ascertain the changes in specific transcripts and proteins associated with the pathogenesis of DM-related periodontitis [4]. Overall evidence indicates that chronic hyperglycemia may stimulate the pathophysiological determinants of periodontitis onset and progression, including microvascular dysfunction, oxidative stress, hyperinflammation, and dysregulation of cell dynamics [5]. Nevertheless, the specific genes and biological pathways underlying the pathogenesis of type 2 DM (T2DM)-related periodontitis are still undetermined. To date, most of the studies focusing on the pathogenesis of the T2DM-related periodontitis had restricted gene or protein coverage due to the use of low- to mid-plex or microarray-based techniques, thus, failing to assess the biological processes involved in T2DM-related periodontitis comprehensively.

The emergence of high-throughput technologies and bioinformatic approaches enhanced the possibility of understanding the complexity of the eukaryotic transcriptome related to several diseases and identifying broader gene expression profiles, specific signaling pathways, and novel transcript variants [6]. To the best of our knowledge, there are no previous studies on whole transcriptome datasets from human gingival biopsies of patients with periodontitis and T2DM. Thus, the aim of this study is to assess the changes in the whole transcriptome that occur from periodontal health to periodontitis in patients with T2DM. We hypothesize that diabetic and non-diabetic patients exhibit significant differences in gene expression profiles that may translate into different immunoinflammatory responses and, ultimately, distinct susceptibility to periodontal breakdown. The transcriptional characterization of the T2DM-related periodontitis is important to identify key genes and signaling pathways that can be used as targets for diagnosis, preventive, and therapeutic strategies in the future.

Material and methods

Population

In this cross-sectional study, 36 patients (45–85 years old) were selected from a pool of patients seeking treatment at the University of Florida’s Dental Clinics from January 2020 to January 2022. The study was approved by the Institutional Review Board (IRB201901176), and written informed consent was obtained from all eligible individuals. All experiments were performed in accordance with the Declaration of Helsinki (Ethical Principles for Medical Research Involving Human Subjects) and relevant guidelines and regulations. The Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) guidelines were adopted for study conducting and reporting findings.

Based on periodontal and DM status, patients were assigned into one of the following four groups: non-diabetic patients without periodontitis (HH; n = 9), non-diabetic patients with periodontitis (HP; n = 9), T2DM patients without periodontitis (DH; n = 9), and T2DM patients with periodontitis (DP: n = 9). The periodontally-healthy patients had no history of periodontitis, <10% of sites with marginal bleeding (MB) or bleeding on probing (BoP), and probing depths (PD) ≤3mm (assuming no pseudopockets) [7]. Furthermore, the periodontally-healthy patients should have needed at least one crown lengthening surgery or tooth extraction due to non-periodontitis reasons. The periodontitis patients had localized or generalized stage III or IV grade B or C periodontitis [8] and a referral for at least one tooth extraction due to severe untreated periodontitis. The diabetic patients were required to have a diagnosis of T2DM confirmed by a physician, dating from at least the last three years, and confirmed by HbA1c levels ≥7% and a fasting plasma glucose (FPG) >99 mg/dl in last two months. The non-diabetic patients were required to have no history of pre-DM or DM, confirmed by an HbA1c <5.5% and an FPG <99 mg/dl.

The exclusion criteria were history of smoking (current or past); pregnancy; lactation; use of antibiotics, anti-inflammatory, immunosuppressant, or other immune regulators within six months; use of bone metabolism-related medications (e.g., bisphosphonates, or denosumab); subgengival periodontal treatment in the past 12 months; regular use of mouth rinses containing antimicrobials; and other systemic conditions that could affect periodontitis progression [9].

The demographic and clinical data of the participants were obtained from their records, including age, sex, HbA1C levels, duration of DM, and full-mouth periodontal parameters. In order to have accuracy in the periodontal diagnosis of the sampled teeth, periodontal parameters were recorded around the sampled teeth by the same trained and calibrated examiner (P.M.D). The study examiner participated in a previous calibration exercise, and the standard error of measurement was estimated. Intra-examiner variability was recorded as 0.20 mm for PD and 0.23 mm for CAL.

Tissue collection

Gingival biopsies (surgical waste specimes), including oral, junctional, and sulcular epitheliums, and connective tissue were obtained during surgical procedures. In patients with periodontitis, the biopsies were obtained from sites with a PD and clinical attachment level (CAL) ≥5 mm and with BoP, mobility, and/or bone loss of more than half of the root during tooth extraction due to severe periodontitis. In the periodontally-healthy patients, the biopsies were obtained from teeth with a PD ≤3 mm, no bone loss, and no BoP or marginal bleeding during crown lengthening surgery or tooth extraction due to non-periodontitis reasons. Briefly, internal bevel and intrasulcular incisions were performed around selected teeth using a scalpel blade. Subsequently, the collar of gingival tissue was carefully removed. After removing the blood from the biopsies by washing them in a saline solution, samples were immediately stored in an RNA later solution (Ambion Inc., Austin, TX, USA) at −80oC for future RNA extraction.

RNA extraction

Total RNA was isolated from the biopsies using the Trizol method (TRIzol™ Reagent, Thermo Fisher Scientific, Waltham, MA, USA), according to the manufacturer’s instructions. RNA samples were resuspended in diethylpyrocarbonate-treated (DEPC) water and stored at −80oC. The RNA concentration was determined by the optical density using a micro-volume spectrophotometer (Nanodrop 1000, Nanodrop Technologies LLC, Wilmington, NC, USA). The total RNA was DNase treated (Turbo DNA-free, Ambion Inc., Austin, TX, USA) to rid the RNA samples of contaminating DNA.

Library construction, sequencing, and bioinformatics

Illumina sequencing library construction

The RNA-seq library construction was performed at the University of Florida’s ICBR Gene Expression and Genotyping Core Facility (RRID: SCR_019145). The RNA samples were measured by the QUBIT fluorescent method (Invitrogen, Waltham, MA, USA) and Agilent Bioanalyzer (Agilent, Santa Clara, CA, USA). An amount of 500 ng of protein-free, total RNA was used for the library construction using the reagents provided in the NEBNext rRNA depletion kit (catalog # E6350) and the Ultra Directional RNA Library Prep Kit for Illumina (New England Biolabs, Ipswich, MA, USA), following the manufacturer’s recommendations. Two microliters of probe hybridization buffer were added to the total RNA samples, followed by RNase H and DNAase I digestion. Subsequently, the RNA was purified using NEBNext RNA purification beads (New England Biolabs, catalog # E6350). This step was followed by the RNA library construction using the NEBNext Ultra II Directional Lib Prep (New England Biolabs, catalog # E7760), according to the manufacturer’s user guide. Briefly, RNA was fragmented in a solution containing divalent cations and incubated at 94oC, followed by first-strand cDNA synthesis using reverse transcriptase and oligo dT primers. The synthesis of the ds-cDNA was done using the 2nd strand master mix provided in the kit, followed by end-repair and dA-tailing. Next, Illumina adaptors were ligated to the samples. Finally, the library was amplified, followed by purification with AMPure beads (Beckman Coulter, Pasadena, CA, USA, catalog # A63881). The library size and mass were assessed by analysis in an Agilent TapeStation using a DNA5000 Screen Tape (Agilent, Santa Clara, CA, USA). A 200–1000 broad library peak was observed, with the highest peak at around 500 bp. Quantitative PCR was used to validate the library’s functionality, using the KAPA library quantification kit (Kapa Biosystems, Wilmington, MA, USA catalog # KK4824) and monitored on a BioRad CFX 96 real-time PCR system (Bio-Rad Laboratories, Hercules, CA, USA). Eighteen barcoded samples were pooled equimolarly for simultaneous sequencing, as described below.

Illumina NovaSeq6000 sequencing

The sequencing run was performed at the University of Florida’s ICBR NextGen DNA Sequencing Core Facility (RRID: SCR_019152). Briefly, normalized libraries were submitted to the Illumina Free Adapter Blocking (FAB) Reagent protocol (Illumina, San Diego, CA, USA, catalog # 20024145) to minimize the presence of adaptor-dimers and index hopping rates. The library pool was diluted to 0.8 nM and sequenced on the one S4 flow cell lane (2×150 cycles) of an Illumina NovaSeq6000 (Illumina, San Diego, CA, USA). The instrument’s computer utilized NovaSeq Control Software v1.6. Cluster and SBS consumables were v1.5. The final loading concentration of the library was 120 pM with a 1% PhiX spike-in control. One lane generated approximately 2.5 billion paired-end reads (∼750 Gb) with an average Q30% greater or equal to 92.5% and a Cluster PF of 85.4%. FastQ files were generated using the BCL2fastQ function in the Illumina BaseSpace portal. An average of 75 million demultiplexed, paired-end reads were used for the data analyses.

Sequence quality control

The quality of the RNA-Seq sequence data was first evaluated using FastQC (FASQC) [10] prior to further downstream analysis. Low-quality sequences were trimmed, and poor-quality reads were removed using the Trimmomatic tool [11].

Differential gene expression analysis

Data analyses were performed at the University of Florida’s ICBR Bioinformatics core. A Star Aligner [12] was used to map the high-quality single-end reads to the genome, GRCh38 (https://useast.ensembl.org/Homo_sapiens/Info/Index/). The gene expression was obtained using RSEM [13]. The expected read counts and fragments per kilobase of transcript per million mapped (FPKM) were extracted for further analysis. The estimated read counts were used as the input for edgeR [14] to perform the differential gene expression analysis. The exact test was developed to identify DEGs, and the thresholds were set at an FDR of 0.05 and a fold change of greater than or equal to 2. Pairwise comparisons were made between the healthy controls and periodontitis in the non-diabetic and diabetic patients (HP versus HH and DP versus DH). Prior to the DE analysis, a PCA was performed to identify outlier samples. No obvious outlier samples were found.

Pathway analysis

An Ingenuity Pathway Analysis (IPA) (Qiagen, Redwood City, CA) was performed on the DEGs expressed exclusively in the diseased tissues in diabetic and non-diabetic patients to identify the canonical pathways dysregulated in periodontitis in the presence and absence of T2DM. The transcripts that were identified to be differently expressed in the tissues with periodontitis compared with the tissues with periodontal heath in non-diabetic (HP versus HH) and diabetic patients (DP versus DH), along with their respective fold changes, were input into the IPA, for subsequent bioinformatics analysis. The Canonical Pathways were used for interpreting the functions of the DEGs. For the Ingenuity Canonical Pathway analysis, a −log (P-value) greater than 2 was taken as the significant threshold for the diabetic and non-diabetic groups. A Z-score over 2 was defined as the threshold of significant activation, while a Z-score under −2 was defined as the threshold of significant inhibition for patients with T2DM. No significant signaling pathways in the non-diabetic patients presented a Z-score over 2 or under −2. Therefore, a Z-score under −1.3 was defined as the threshold of significant inhibition for the non-diabetic patients.

Clinical and demographic data analyses

The mean percentages of sites with plaque and BoP, the mean full-mouth PD and CAL, the mean PD and CAL of the sampled teeth, the HbA1c levels, and the duration of DM were computed for each subject and, subsequently, across the groups. Data were examined for normality by the Shapiro-Wilk test, and parametric methods were used as data achieved a normal distribution. The significance of differences for age, HbA1c, and clinical parameters was compared by a one-way ANOVA, followed by a Tukey test. A t-test was used to compare the duration of DM between both diabetic groups. The significance of differences for sex was compared by a Chi-square test. The level of significance for all these analyses was set at 5%.

IHC

IHC experiments on four tissues per group were performed in an independent set of patients to confirm the transcriptome findings at the protein level. The selection of the genes for validation was performed based on their biological relevance within the representative signaling pathways observed in the diseased tissues of patients with T2DM and the magnitude of difference in mRNA levels between healthy and disease tissues in diabetic and non-diabetic patients. Gingival samples were collected as described above and fixed in 10% neutral-buffered formalin. Four micrometers-thick formalin-fixed paraffin-embedded (FFPE) sections were set on positively charged glass slides. All the staining steps were performed on a Leica RX Autostainer (Leica Biosystems, Wetzlar, Germany), following the vendor’s instructions. The slides were de-paraffinized, re-hydrated, and heat-induced antigen retrieval treated with either epitope retrieval (ER)1 or ER2 buffers for 20 minutes. The slides were treated with hydrogen peroxide to eliminate endogenous peroxidases for five minutes. Afterward, the sections were incubated with primary antibodies against PTPN2 (1:500, polyclonal, rabbit, PA582516, ThermoFisher Scientific, Waltham, MA, USA), PTPN13 (1:500, polyclonal, rabbit, 25944-1-AP, Proteintech, Rosemont, IL, USA), DHCR24 (1:750, polyclonal, rabbit, 10471-1-AP), PIK3R2 (1:100, monoclonal, mouse, 67644-1-Ig, Proteintech, Rosemont, IL, USA), CALCRL (1:400, polyclonal, rabbit, PA550644, ThermoFisher Scientific, Waltham, MA, USA), IL1RN (1:2000, monoclonal, mouse, CF803396, ThermoFisher Scientific, Waltham, MA, USA), IL-6R alpha (1:350, polyclonal, rabbit, 23457-1-AP, Proteintech, Rosemont, IL, USA), and ITGA4 (1:500, polyclonal, rabbit, 10471-1-AP, ThermoFisher Scientific, Waltham, MA, USA) for 30 minutes. The sections were then treated with an HRP Polymer (Leica Biosystems, Wetzlar, Germany) for eight minutes. The specific reaction for each antibody was observed through the application of 3,3’diaminobenzidine (DAB) for ten minutes. The sections were counter-stained with hematoxylin (Leica Biosystems, Wetzlar, Germany), dehydrated through graded ethanol, cleared in xylene, and mounted on slides with the aid of Cytoseal mounting media (Richard-Allan Scientific, San Diego, CA, USA). The negative controls were obtained by the omission of the primary antibodies. Results are presented in a descriptive form, including the description of the histological picture regarding which cells or tissue components were immunopositive, patterns of staining (membrane, cytoplasm, or nuclear), and intensity of IHC expression (weak, moderate, or strong).

Results

The mean age and frequency of sex did not differ among the groups (P > 0.05; S1 Table). The periodontal parameters were significantly higher in both groups with periodontitis compared with the groups without periodontitis (P < 0.05; S1 Table). There were no differences between non-diabetic patients without periodontitis (HH) and T2DM patients without periodontitis (DH) and between non-diabetic patients with periodontitis (HP) and T2DM patients with periodontitis (DP) in relation to the periodontal parameters (P > 0.05; S1 Table)

Differentially expressed genes (DEGs) analysis

S1 Fig presents the Venn diagram of the differentially expressed genes (DEGs) in the non-diabetic (HH vs. HP) and diabetic (DH vs. DP) patients exclusively and the overlapping DEGs between the two groups. Four hundred and twenty DEGs (S2 Table; 211 upregulated and 209 downregulated) were identified in tissues with periodontitis compared with those with periodontal health in non-diabetic patients from the 16,164 background genes. One thousand five hundred and sixty-three DEGs (S3 Table; 654 upregulated and 909 downregulated) were identified in tissues with periodontitis compared with those with periodontal health in diabetic patients from the 16,299 background genes. One thousand two hundred and forty-two DEGs were concomitantly expressed in tissues with periodontitis in the non-diabetic and diabetic patients (S4 Table; 708 upregulated and 534 downregulated).

The top 15 up and downregulated DEGs in the non-diabetic (HP vs. HH) and diabetic (DP vs. DH) groups are listed in Table 1. Overall, immunoglobulin (Ig)-related genes were highly expressed in the non-diabetic and diabetic patients. Excluding the Ig-related DEGs, inhibin βE (INHBE) and C-X-C motif chemokine ligand 5 (CXCL5) were the most upregulated DEGs in non-diabetic and diabetic patients, respectively. Adhesion G protein-coupled receptor G7 (ADGRG7) and AL356488.2 were the DEGs with the strongest downregulation in non-diabetic and diabetic patients, respectively. Five genes from the late cornified envelope (LCE) family were amongst the top 15 downregulated DEGs in tissues with periodontitis in diabetic patients.

Table 1. Top 15 downregulated and upregulated DEGs in periodontitis in relation to periodontal health in non-diabetic and in patients with T2DM exclusively.

Downregulated Upregulated

DEGs exclusively in non-diabetics
(HP vs HH)
Gene Q value P value Log2 FC Gene Q value P value Log2 FC
ADGRG7 6.08E-05 1.47E-06 -9.65 IGKV1D-17 0.0071315 0.000790428 4.80
AADAC 0.00024201 9.12E-06 -7.02 IGLV10-54 0.002906844 0.000228862 4.67
TYRP1 8.23E-06 1.17E-07 -5.22 IGKV5-2 9.14E-06 1.34E-07 4.39
CCER2 0.0001552 4.92E-06 -4.77 MTND2P28 (lnc)
ENSG00000225630
0.001346243 8.27E-05 3.75
AC036176.3 (lnc)
ENSG00000269989
3.60E-05 7.64E-07 -4.10 IGKV6D-21 1.07E-07 4.19E-10 3.54
DCT 0.00022835 8.50E-06 -3.76 IGHV4-31 0.000355956 1.51E-05 3.24
PAX1 0.00143649 8.98E-05 -3.32 INHBE 0.000189732 6.60E-06 3.20
AC073896.1 0.00016101 5.27E-06 -3.20 AC026202.2 (lnc)
ENSG00000233912
1.74E-06 1.65E-08 2.99
TM4SF19 0.00018363 6.28E-06 -3.20 CBSL 0.009032748 0.001093897 2.97
FOSB 1.74E-06 1.64E-08 -2.93 IGLV3-9 0.003098993 0.000247676 2.89
ARC 0.00528802 0.00051847 -2.92 CU633967.1 (lnc)
ENSG00000274333
2.61E-07 1.48E-09 2.79
NPIPA8 0.03595036 0.00706707 -2.86 AMPD1 1.23E-05 1.94E-07 2.77
SMARCA5-AS1 (lnc)
ENSG00000245112
0.00016881 5.61E-06 -2.82 IGKV2D-30 0.00849601 0.00100104 2.73
AC126773.2 (lnc)
ENSG00000260577
0.0007186 3.67E-05 -2.77 MAP3K19 3.52E-05 7.38E-07 2.70
AC106886.2 (lnc)
ENSG00000260899
0.0001565 4.99E-06 -2.71 JCHAIN 8.20E-05 2.19E-06 2.60

DEGs exclusivaly in diabetics
(DP vs DH)
SCARNA2 (lnc)
ENSG00000270066
5.91E-26 2.18E-29 -10.8 IGHV3-20 4.21E-09 8.12E-11 7.12
LCE2C 7.91E-08 2.34E-09 -9.89 IGHV1-69 0.00040398 4.51E-05 5.46
AC105001.2 6.80E-13 4.26E-15 -9.88 IGHV3-72 1.36E-08 3.08E-10 5.44
LCE2A 9.99E-06 5.82E-07 -9.05 IGHV3-33 5.08E-10 7.47E-12 4.80
LCE2B 2.43E-06 1.15E-07 -8.93 IGKV1D-16 0.0001974 1.94E-05 4.66
AC138811.2 1.47E-07 4.67E-09 -8.73 IGHV3-48 3.36E-13 1.94E-15 4.22
LCE2D 3.71E-05 2.71E-06 -8.688 IGHV3-74 4.01E-16 8.12E-19 4.04
SMAD1-AS1(lnc)
ENSG00000250902
0.00034329 3.72E-05 -8.63 IGKV2D-29 1.29E-05 7.76E-07 3.48
SENP3-EIF4A1(lnc)
ENSG00000277957
1.47E-05 9.02E-07 -8.30 AC244226.1 0.00201771 0.00029938 3.46
ATP12A 5.27E-06 2.79E-07 -8.25 CXCL5 0.0001365 1.25E-05 3.45
KLRG2 1.98E-05 1.29E-06 -7.96 IGHV3-66 0.028498749 0.007819436 3.21
AP000350.6 (lnc)
ENSG00000273295
6.86E-06 3.79E-07 -6.57 IGHV1-46 2.04E-05 1.33E-06 3.18
LCE3C 2.03E-06 9.40E-08 -5.79 IGLV3-21 0.003652181 0.000613812 3.18
KRT76 0.00013426 1.23E-05 -5.19 P2RX5 7.01E-16 1.55E-18 3.15
PLA2G2F 3.51E-05 2.54E-06 -4.97 GDF15 5.80E-07 2.25E-08 3.02

Ingenuity Pathway Analysis (IPA)

The list of ingenuity canonical pathways related to the DEGs that were uniquely expressed in non-diabetic patients is presented in S5 Table. Twenty-one ingenuity canonical pathways were identified in non-diabetics by applying a −log (P-value) threshold of greater than two. The top ten representative pathways ranked according to the −log (P‑value) are presented in Fig 1A. B Cell Receptor Signaling was the highest-ranking signaling pathway in the non-diabetic patients. None of the significant signaling pathways in non-diabetic patients presented a Z-score over 2 or under −2. Considering a Z-score of less than −1.3 as significant, the Tumor Microenvironment and PI3K Signaling in B Lymphocytes pathways were significantly inhibited (Fig 1B and 1C). Within these enriched pathways, FOS, JUN, activating transcription factor 3 (ATF3), and matrix metalloproteinase (MMP)-13 were the DEGs with a log2 fold change (FC) less than or equal to −1.5.

Fig 1.

Fig 1

IPA analysis of the canonical pathways related to the DEGs altered in tissues with periodontitis compared with tissues with periodontal health in non-diabetic patients. (A) The top ten dysregulated canonical signaling pathways are presented in descending order of statistical significance of −log(P-values). (B) The canonical pathways that were predicted to be modulated in the non-diabetic patients, as defined by a −log(P-value) >2 and Z-score <−1.3 (inhibited). (C) The number of upregulated and downregulated DEGs involved in each of the representative canonical pathways.

The list of ingenuity canonical pathways related to the DEGs exclusively expressed in diabetic patients is presented in S6 Table. Forty-three ingenuity canonical pathways were identified by applying a −log (P-value) threshold of greater than two. The top ten signaling pathways are presented in Fig 2A. Granulocyte Adhesion and Diapedesis was the highest-ranking pathway (−log [P-value] = 5.55), followed by Phagosome Formation (−log [P-value] = 4.75) and Atherosclerosis Signaling (−log [P-value] = 4.55). Taking Z-scores greater than 2 and less than −2, Phagosome Formation, Cardiac β-adrenergic, tRNA, and PI3K/AKT signaling pathways were significantly activated. In contrast, the Superpathway of Cholesterol Biosynthesis, Adrenomedullin (ADM), and Superpathway of Inositol Phosphate Compounds (SIPC) were inhibited in tissues with periodontitis in the diabetic patients (Fig 2B and 2C).

Fig 2. IPA analysis of the canonical pathways related to the DEGs altered in tissues with periodontitis compared with tissues with periodontal health in patients with T2DM.

Fig 2

(A) The top ten dysregulated canonical signaling pathways are presented in descending order of statistical significance of −log(P-values). (B) The canonical pathways that were predicted to be modulated in the non-diabetic patients, as defined by a −log(P-value) >2 and Z-score >2 (activated) or <−2 (inhibited). (C) The number of upregulated and downregulated DEGs involved in each of the representative canonical pathways.

Table 2 presents the DEGs that function within the abovementioned canonical pathways in diabetic patients. The upregulated DEGs that function in the Phagosome Formation pathway included mostly adhesion G protein-coupled receptors (GPCRs), complement-related factors, chemokine, pathogen pattern-recognition and Ig receptors, and integrin (ITG) subunits. The downregulated DEGs involved in this pathway were comprised mainly of GPCRs, mitogen-activated protein kinase (MAPK), and phospholipases (PL). ITG subunits were the predominant upregulated gene group, while interleukin (IL) receptors and phosphatase-related components were the leading downregulated DEGs functioning in PI3K/AKT signaling. Most of the upregulated genes in the Cardiac β-Adrenergic and tRNA Splicing pathways were phosphodiesterases. Of the 12 genes downregulated in Cardiac β-Adrenergic Signaling, six were phosphatase-related, and two were calcium voltage-gated channel auxiliary-related genes. The primary downregulated genes observed in the ADM pathway were comprised of MAPKs, Phospholipase C (PLC), and transcription factors. Notably, in the ADM pathway, IL1A was upregulated while IL1 receptor antagonist (IL1RN) was downregulated in the diabetic patients. The mRNA for different enzyme functional classes were downregulated in Superpathway of Cholesterol Biosynthesis signaling. Phosphatase and PL genes were generally downregulated in SIPC signaling.

Table 2. The DEGs that are a part of the most critical canonical pathways in patients with T2DM and periodontitis compared with the patients with T2DM and without periodontitis.
Upregulated Downregulated
Gene symbol Gene name Q value Log2 FC Gene symbol Gene name Q value Log2 FC
Phagosome Formation: -log (p-value) = 4.75; z-score = 3.25
ABHD3 Abhydrolase Domain Containing 3, Phospholipase 8.31E-05 1.23 ADGRF4 Adhesion G Protein-Coupled Receptor F4 0.001502735 -1.34
ADGRD1 Adhesion G Protein-Coupled Receptor D1 0.000221199 1.49 ADGRG1 Adhesion G Protein-Coupled Receptor G1 7.43E-05 -1.14
ADGRE5 Adhesion G Protein-Coupled Receptor E5 0.000103337 1.01 AP1M2 Adaptor Related Protein Complex 1 Subunit Mu 2 0.01001205 -1.10
ADGRF5 Adhesion G Protein-Coupled Receptor F5 2.95E-10 1.47 CELSR1 Cadherin EGF LAG Seven-Pass G-Type Receptor 1 2.25E-05 -1.23
ADGRL4 Adhesion G Protein-Coupled Receptor L4 7.13E-08 1.69 CCR7 C-C Motif Chemokine Receptor 7 0.020276507 -1.20
AP1S2 Adaptor Related Protein Complex 1 Subunit Sigma 2 1.46E-06 1.35 F2RL1 F2R Like Trypsin Receptor 1 0.000861457 -2.03
APBB1IP Amyloid Beta Precursor Protein Binding Family B Member 1 Interacting Protein 2.58E-05 1.41 LGR4: Leucine Rich Repeat Containing G Protein-Coupled Receptor 4 3.95E-05 -1.08
APLNR Apelin Receptor 1.22E-05 1.43 MAPK15 Mitogen-Activated Protein Kinase 15 0.010420729 -1.52
AVPR1A Arginine Vasopressin Receptor 1A 0.000286265 1.55 MAPK7 Mitogen-Activated Protein Kinase 7 6.82E-05 -1.17
C3 Complement C3 0.000197404 1.44 MCHR1 Melanin Concentrating Hormone Receptor 1 0.036117633 -1.69
C5AR1 Complement C5a Receptor 1 0.000391725 1.57 P2RY11 Purinergic Receptor P2Y11 0.015989797 -1.06
CALCRL Calcitonin receptor-like receptor 9.96E-06 1.48 PIK3R2 Phosphoinositide-3-Kinase Regulatory Subunit 2 3.02E-07 -1.24
CCR1 C-C Motif Chemokine Receptor 1 0.006780284 1.15 P2RY2 Purinergic Receptor P2Y2 4.27E-07 -1.71
CCR2 C-C Motif Chemokine Receptor 2 0.000328093 1.40 PLA2G2F Phospholipase A2 Group IIF 3.51E-05 -4.97
CLEC4E C-Type Lectin Domain Family 4 Member E 0.005497415 1.83 PLA2G4B Phospholipase A2 Group IVB 0.001044935 -1.68
CR1 Complement receptor 1 8.19E-05 1.73 PLD2 Phospholipase D2 0.000178173 -1.11
CR2 Complement C3d Receptor 2 0.035193615 1.61 SMO Smoothened, Frizzled Class Receptor 2.48E-05 -1.38
EDNRB Endothelin receptor type B 0.002427503 1.06 SRC SRC Proto-Oncogene, Non-Receptor Tyrosine Kinase 3.84E-05 -1.08
F2RL2 Coagulation Factor II Thrombin Receptor Like 2 3.37E-05 1.47 VAV3 Vav Guanine Nucleotide Exchange Factor 3 0.000301819 -1.16
FCER1G Fc Fragment of IgE Receptor Ig 0.02219775 1.01 VIPR1 Vasoactive Intestinal Peptide Receptor 1 0.01962491 -1.15
FCGR1A Fc Fragment of IgG Receptor Ia 0.004828328 1.27
FCGR1B Fc Fragment of IgG Receptor Ib 0.008226445 1.73
FCGR2C Fc Fragment of IgG Receptor IIc (Gene/Pseudogene) 0.010786066 1.33
FZD3 Frizzled Class Receptor 3 1.49E-07 1.38
GPR162 G Protein-Coupled Receptor 162 0.023897665 1.15
GPR182 G Protein-Coupled Receptor 182 0.00076005 1.70
GPR180 G Protein-Coupled Receptor 180 0.00169883 1.06
GPR22 G Protein-Coupled Receptor 22 0.008043412 1.10
GPR34 G Protein-Coupled Receptor 34 0.001864425 1.29
GPR4 G Protein-Coupled Receptor 4 0.02343798 1.00
GPR52 G Protein-Coupled Receptor 52 0.008604626 1.24
GPR65 G Protein-Coupled Receptor 65 0.001368858 1.60
HRH2 Histamine Receptor H2 0.006393526 1.35
ITGA1 Integrin Subunit Alpha 1 3.91E-05 1.21
ITGA4 Integrin Subunit Alpha 4 0.003952353 1.05
ITGAM Integrin Subunit Alpha M 0.002551803 1.29
ITGAX Integrin Subunit Alpha X 0.007791008 1.07
LBP Lipopolysaccharide Binding Protein 0.011434944 2.43
LYN LYN Proto-Oncogene, Src Family Tyrosine Kinase 0.000863943 1.01
MSR1 Macrophage Scavenger Receptor 1 0.001405248 1.24
PLA2G4C Phospholipase A2 Group IVC 0.004463955 1.23
PTGFR Prostaglandin F Receptor 0.00014384 1.41
S1PR3 Sphingosine-1-Phosphate Receptor 3 0.002548839 1.33
S1PR4 Sphingosine-1-Phosphate Receptor 4 0.000299743 1.45
TLR2 Toll Like Receptor 2 0.00426382 1.01
TLR4 Toll Like Receptor 4 5.63E-07 1.66
TLR8 Toll Like Receptor 8 0.013887157 1.39
WIPF1 WAS/WASL Interacting Protein Family Member 1 0.000146684 1.13
PI3K/AKT Signaling: -log (p-value) = 3.69; z-score = 2.11
BCL2A1 BCL2 Related Protein A1 0.001296469 1.66 CCND1 Cyclin D1 0.000369428 -1.00
CDKN1B Cyclin Dependent Kinase Inhibitor 1B 3.28E-07 1.22 IKBKE Inhibitor Of Nuclear Factor Kappa B Kinase Subunit Epsilon 4.78E-06 -1.19
FOXO1 Forkhead Box O1 1.69E-05 1.01 IL12RB2 Interleukin 12 Receptor Subunit Beta 2 0.000404413 -1.79
GDF15 Growth Differentiation Factor 15 5.80E-07 3.02 IL17RC Interleukin 17 Receptor C 1.80E-05 -1.36
IL18RAP Interleukin 18 Receptor Accessory Protein 0.00028751 2.10 IL17RD Interleukin 17 Receptor D 0.000389797 -1.15
IL6R Interleukin 6 Receptor 2.05E-08 1.30 IL20RA Interleukin 20 Receptor Subunit Alpha 0.015230566 -1.06
ITGA1 Integrin Subunit Alpha 1 3.91E-05 1.21 IL20RB Interleukin 20 Receptor Subunit Beta 0.000300872 -1.12
ITGA4 Integrin Subunit Alpha 4 0.003952353 1.05 IL22RA1 Interleukin 22 Receptor Subunit Alpha 1 5.43E-07 -2.26
ITGAM Integrin Subunit Alpha M 0.002551803 1.29 IL4R Interleukin 4 Receptor 6.36E-06 -1.14
ITGAX Integrin Subunit Alpha X 0.007791008 1.07 IL9R Interleukin 9 Receptor 0.00360178 -2.34
PTGS2 Prostaglandin-Endoperoxide Synthase 2 0.005655989 1.44 INPP5J Inositol Polyphosphate-5-Phosphatase J 0.00134853 -1.78
PIK3R2 Phosphoinositide-3-Kinase Regulatory Subunit 2 3.02E-07 -1.29
PPM1J Protein Phosphatase, Mg2+/Mn2+ Dependent 1J 7.81E-05 -2.40
PPP2R2B Protein Phosphatase 2 Regulatory Subunit Bbeta 0.006000897 -1.74
PPP2R3A Protein Phosphatase 2 Regulatory Subunit B’’Alpha 1.06E-05 -1.02
Cardiac β-adrenergic Signaling: -log (p-value) = 2.90; z-score = 3.0
AKAP12 A-Kinase Anchoring Protein 12 4.23E-06 1.37 CACNA2D3 Calcium Voltage-Gated Channel Auxiliary Subunit Alpha2delta 3 3.29E-06 -1.85
PDE10A Phosphodiesterase 10A 0.000450127 1.18 CACNB4 Calcium Voltage-Gated Channel Auxiliary Subunit Beta 4 0.001979528 -1.39
PDE1A Phosphodiesterase 1A 2.77E-06 2.26 GDPD3 Glycerophosphodiester Phosphodiesterase Domain Containing 3 0.005953332 -1.33
PDE4A Phosphodiesterase 4A 0.000197404 1.05 GNA11 G Protein Subunit Alpha 11 9.91E-07 -1.15
PDE4B Phosphodiesterase 4B 3.64E-05 1.30 PDE6A Phosphodiesterase 6A 0.032414648 -1.44
PDE4D Phosphodiesterase 4D 0.000401816 1.02 PPM1J Protein Phosphatase, Mg2+/Mn2+ Dependent 1J 7.81E-05 -2.40
PDE5A Phosphodiesterase 5A 2.69E-05 1.05 PPP1R14B Protein Phosphatase 1 Regulatory Inhibitor Subunit 14B 1.36E-05 -1.12
PDE7B Phosphodiesterase 7B 0.002560175 1.27 PPP1R14C Protein Phosphatase 1 Regulatory Inhibitor Subunit 14C 0.000537905 -1.43
PRKACB Protein Kinase CAMP-Activated Catalytic Subunit Beta 1.02E-06 1.50 PPP1R3C Protein Phosphatase 1 Regulatory Subunit 3C 2.72E-05 -1.48
PPP2R2B Protein Phosphatase 2 Regulatory Subunit B beta: 0.006000897 -1.74
PPP2R3A Protein Phosphatase 2 Regulatory Subunit B’’Alpha: 1.06E-05 -1.02
PRKAR1B Protein Kinase CAMP-Dependent Type I Regulatory Subunit Beta 0.005500148 -1.03
tRNA Splicing: -log (p-value) = 2.83; z-score = 2.12
PDE10A Phosphodiesterase 10A 0.000450127 1.18 GDPD3 Glycerophosphodiester Phosphodiesterase Domain Containing 3 0.005953332 -1.33
PDE1A Phosphodiesterase 1A 2.77E-06 2.26 PDE6A Phosphodiesterase 6A 0.032414648 -1.44
PDE4A Phosphodiesterase 4A 0.000197404 1.05
PDE4B Phosphodiesterase 4B 3.64E-05 1.30
PDE4D Phosphodiesterase 4D 0.000401816 1.02
PDE5A Phosphodiesterase 5A 2.69E-05 1.05
PDE7B Phosphodiesterase 7B 0.002560175 1.27
Adrenomedullin signaling pathway: -log (p-value) = 3.80; z-score = -2.2
BCL2 BCL2 Apoptosis Regulator 3.64E-05 1.16 GNA11 G Protein Subunit Alpha 11 9.91E-07 -1.15
C3 Complement C3 0.000197404 1.44 GUCY2C Guanylate Cyclase 2C 0.006407274 -1.22
CALCRL Calcitonin receptor-like receptor 9.96E-06 1.48 IL1RN Interleukin 1 Receptor Antagonist 0.029831597 -1.01
CASP3 Caspase 3 4.78E-07 1.22 IL36A Interleukin 36 Alpha 0.002517721 -1.52
GPR182 G Protein-Coupled Receptor 182 0.000760052 1.70 MAPK13 Mitogen-Activated Protein Kinase 13 5.38E-07 -1.38
IL1A Interleukin 1 Alpha 9.31E-05 1.22 MAPK15 Mitogen-Activated Protein Kinase 15 0.010420729 -1.52
PLCL2 Phospholipase C Like 2 2.00E-07 1.42 MAPK7 Mitogen-Activated Protein Kinase 7 6.82E-05 -1.17
PRKACB Protein Kinase CAMP-Activated Catalytic Subunit Beta 1.02E-06 1.50 PIK3R2 Phosphoinositide-3-Kinase Regulatory Subunit 2 3.02E-07 -1.24
RAMP2 Receptor Activity Modifying Protein 2 4.27E-05 1.35 PLCD1 Phospholipase C Delta 1 3.66E-05 -1.38
PLCD3 Phospholipase C Delta 3 0.000111424 -1.22
PLCH2 Phospholipase C Eta 2 2.34E-08 -1.75
PPARG Peroxisome Proliferator Activated Receptor Gamma 0.033030069 -1.31
PRKAR1B Protein Kinase CAMP-Dependent Type I Regulatory Subunit Beta 0.005500148 -1.03
RXRA Retinoid X receptor alpha 9.18E-05 -1.08
TFAP2A Transcription Factor AP-2 Alpha 7.30E-08 -1.73
TFAP2C Transcription factor AP-2 gamma 8.19E-08 -1.37
Superpathway of Cholesterol Biosynthesis: -log (p-value) = 3.68; z-score = -2.82
ACAT2 Acetyl-CoA Acetyltransferase 2 2.67E-06 -1.61
DHCR24 24-Dehydrocholesterol Reductase 0.00019843 -1.44
DHCR7 7-Dehydrocholesterol Reductase 0.00073273 -1.04
MVD Mevalonate Diphosphate Decarboxylase 8.06E-06 -1.53
MVK Mevalonate Kinase 2.19E-05 -1.47
PMVK Phosphomevalonate kinase 3.19E-06 -1.10
SQLE Squalene Epoxidase 2.76E-05 -1.28
TM7SF2 Transmembrane 7 Superfamily Member 2 5.64E-06 -1.48
Superpathway of Inositol Phosphate Compounds: -log (p-value) = 2.73; z-score = -2.13
ALPL Alkaline Phosphatase, Biomineralization Associated 2.25E-08 1.49 DUSP14 Dual Specificity Phosphatase 14 0.004823952 -1.20
EYA4 EYA Transcriptional Coactivator And Phosphatase 4 0.003920438 1.25 EPHX2 Epoxide Hydrolase 2 0.001017046 -1.06
NUDT4 Nudix Hydrolase 4 7.46E-07 1.12 INPP5J Inositol Polyphosphate-5-Phosphatase J 0.00134853 -1.78
PPP1R16B Protein Phosphatase 1 Regulatory Subunit 16B 0.001087609 1.02 IPPK Inositol-Pentakisphosphate 2-Kinase 2.59E-06 -1.37
PTPN2 Protein Tyrosine Phosphatase Non-Receptor Type 2 8.70E-06 1.03 ITPKC Inositol-Trisphosphate 3-Kinase C 0.001218582 -1.25
NUDT1 Nudix Hydrolase 1 0.003150706 -1.09
PIK3R2 Phosphoinositide-3-Kinase Regulatory Subunit 2 3.02E-07 -1.24
PLCD1 Phospholipase C Delta 1 3.66E-05 -1.38
PLCD3 Phospholipase C Delta 3 0.000111424 -1.22
PLCH2 Phospholipase C Eta 2 2.34E-08 -1.75
PPFIA3 PTPRF interacting protein alpha 3 0.000150788 -1.36
PPP1R14B Protein Phosphatase 1 Regulatory Inhibitor Subunit 14B 1.36E-05 -1.12
PPP1R14C Protein Phosphatase 1 Regulatory Inhibitor Subunit 14C 0.000537905 -1.43
PPP1R3C Protein Phosphatase 1 Regulatory Subunit 3C 2.72E-05 -1.47
PPP2R2B Protein Phosphatase 2 Regulatory Subunit B beta 0.006000897 -1.74
PPP2R3A Protein Phosphatase 2 Regulatory Subunit B’’Alpha 1.06E-05 -1.02
PTPN13 Protein Tyrosine Phosphatase Non-Receptor Type 13 0.00191405 -1.24
SGPP2 Sphingosine-1-Phosphate Phosphatase 2 0.000186653 -1.26
SSH3 Slingshot Protein Phosphatase 3 1.47E-07 -1.45
PPM1J Protein Phosphatase, Mg2+/Mn2+ Dependent 1J 7.81E-05 -2.40

Validation of transcriptome data by immunohistochemistry (IHC)

The immunohistochemistry (IHC) are shown in Figs 3 and 4.

Fig 3. The immunohistochemical staining using antibodies against PTPN2, PTPN13, DHCR24 (Seladin), and PIK3R2 in representative gingival biopsies from the HH, HP, DH, and DP groups.

Fig 3

Bar length = 50 μm, original magnification × 20. HH: non-diabetic patients without periodontitis; HP: non-diabetic patients with periodontitis; DH: T2DM patients without periodontitis; DP: T2DM patients with periodontitis.

Fig 4. The immunohistochemical staining using antibodies against CALCRL, IL1RN, IL6R, and ITGA4 in representative gingival biopsies from the HH, HP, DH, and DP groups.

Fig 4

Bar length = 50 μm, original magnification × 20. HH: non-diabetic patients without periodontitis; HP: non-diabetic patients with periodontitis; DH: T2DM patients without periodontitis; DP: T2DM patients with periodontitis.

Protein tyrosine phosphatase non-receptor (PTPN) 2 showed slightly decreased expression in the DH group compared with the HH group. PTPN2 expression was increased in both periodontitis groups but was more pronounced in the DP group, particularly in the connective tissue (Fig 3). PTPN13 had a noticeable nuclear and cytoplasmatic protein expression reduction in the DP group (Fig 3). DHCR24 presented with considerable expression in the cytoplasm and nucleus in the basal and supra-basal layers of epithelial and endothelial cells in the HH group. However, its expression was significantly reduced in the DH group, especially in the basal cell layer. The DP group showed the lowest expression levels of the DHCR24 protein in intensity and distribution (Fig 3). The DH group presented noticeably lower Phosphoinositide-3-Kinase Regulatory Subunit 2 (PIK3R2) protein expression than the HH group. While the PIK3R2 levels decreased in the HP group (mainly in the basal cell layer), the DP group showed increased cytoplasmatic and nuclear PIK3R2 expression in the epithelium and connective tissues (Fig 3).

The calcitonin receptor-like receptor (CALCRL) protein expression was overall stronger in diseased tissues than in healthy tissues (particularly in epithelial and endothelial cells). However, it was even stronger in the DP group, with nuclear and cytoplasmatic distribution. Notably, while CALCRL expression was clearly noticed in the basal cell layers in the HH group, it was barely expressed in the basal layers in the DH group. IL1RN showed intense staining in the cytoplasm and nucleus of all the epithelial cell layers in both periodontally-healthy groups; however, it had an even stronger staining in the DH group (Fig 4). The IL1RN protein expression was considerably reduced in the cytoplasm and was more prominent in the nucleus of epithelial cells in the diseased tissues, especially in the DP group (Fig 4). IL6R expression was overall stronger in the periodontitis groups than the non-periodontitis groups; however, it was even more intense in the DP group, particularly in the basal layer of the epithelium and connective tissue (Fig 4). There was a robust ITGA4 expression in the cytoplasm and the membrane of supra-basal cell layers of the epithelium in the HH, HP, and DH groups. Remarkably, the basal cell layer in the DH group exhibited no or mild ITGA4 protein expression. Moreover, the DP group presented a more noticeable ITGA4 expression even in the basal layers of the epithelium and underlying connective tissue cells (Fig 4).

Discussion

We show here that periodontitis in non-diabetic and diabetic patients is a distinct process at the whole transcript level. T2DM significantly alters the expression of specific genes and signaling pathways involved in immune and inflammatory responses that may affect the course of periodontitis.

Some of the most upregulated and downregulated genes are worthy of attention. CXCL5 was among the top 15 upregulated DEGs, while LCE2 and LCE3 were among the top 15 downregulated DEGs observed in T2DM-related periodontitis. CXCL5 is a chemokine ligand of the IL-8 receptor type 2 that has proinflammatory activities, including chemotaxis and angiogenesis [15, 16]. In support of our findings, previous studies observed an association between increased CXCL5 levels and DM complications [16, 17]. Noteworthy, lipopolysaccharide (LPS) from Porphyromonas gingivalis, in combination with advanced glycation end products (AGE), induced greater CXCL5 expressions from monocyte and oral keratinocyte cells [18]. Although the biological functions of LCE are not entirely understood, they seem to have antibacterial activities and a role in the epithelial physical barrier [19, 20]. The lack of LCE3B/C causes dysbiotic changes in the oral and cutaneous microbiota in patients with psoriasis [20]. Other possibly relevant downregulated genes to the pathogenesis of T2DM-related periodontitis are keratin 76, critical for epidermal integrity [21], and the PLA2G2F, as PLA2s are involved in host defense and barrier function [22].

The IPA of the DEGs that were exclusively expressed in periodontitis in diabetic patients revealed a significant upregulation of the Phagosome Formation pathway through the alterations in the expression of genes required for actin cytoskeleton rearrangements, membrane ruffle formation, and phagosome closure (e.g., complement, complement receptor (CR), ITGs, GPCRs, scavenger receptor, toll-like receptor, LPS binding protein, Fc receptor, C-type lectin domain family 4 member E, chemokine receptor, and PL) [23]. Noteworthily, some of the phagosome formation-associated genes that were upregulated in T2DM-related periodontitis (e.g., C3a, C5ar1, and LYN) were also found to be upregulated in other DM complications [24, 25], functioning against microbial invasion and as proinflammatory factors and regulators of immunometabolism [25].

Despite the significant activation of the Phagosome Formation pathway in T2DM-related periodontitis, some critical pathways in response to infections may be impaired in diabetic hosts, including the completion of phagocytosis [26, 27]. Hyperglycemia/DM can affect the expression of complement-related factors required for bacterial opsonization and internalization [26], and reduce the phagosome maturation which succeeds phagosome formation [27]. Finally, the dysregulation of PI3Ks, herein represented by PIK3R2, may also affect phagocytosis [28]. Thus, despite the effort to recruit immune cells and form phagosomes in disease tissues in patients with T2DM, under hyperglycemia, microorganisms may evade phagosome maturation and degradation in the phagolysosomes, leading to persistent infection and inflammation. The impact of DM on different stages of phagocytosis represents an interesting area for further research in the context of periodontitis.

DEGs uniquely expressed in T2DM-related periodontitis were related to the activation of the PI3K/AKT signaling pathway that is activated through various stimuli (e.g., cytokines, growth factors, cellular stresses, and insulin) and regulates several biological processes (e.g., cellular survival, apoptosis, autophagy, proliferation, growth, motility, transcription, and translation) involved in periodontitis and other diseases [29, 30]. Additionally, the PI3K/AKT pathway is reported to regulate insulin and hyperglycemia, and the insulin resistance [29] and the AGE-RAGE axis can activate PI3K/AKT signaling [31]. Finally, the activation of the PI3K/AKT signaling pathway has been associated with DM-related complications [32] and with osteoclastogenesis and bone resorption, an important hallmark of periodontitis [33].

The relevant genes that were involved in the PI3K/AKT pathway activation observed in the T2DM-related periodontitis included those that affect apoptosis (e.g., FOXO1, BCL2, and growth differentiation factor 15 [GDF15]) and cell cycle passage (e.g., CDKN1B and CCND1). Also abundant were the dysregulated expression of pro- and anti-inflammatory cytokine receptors (e.g., IL18RAP, IL6R, IL12RB2, IL17RC, IL17RD, IL20RA, IL20RB, IL22RA1, IL4R, and IL9R), supporting the notion that cytokines play pivotal roles in the pathogenesis of T2DM-related periodontitis, a typical infectious-inflammatory disorder. The concurrent increased expression of IL-6R at the protein level observed in diseased tissues in diabetic patients indicates the role of the pro-inflammatory IL-6/IL-6R axis in disease pathogenesis. ITGs, which mediate cell-matrix interactions and leukocyte migration and adhesion [34], were consistently overexpressed in T2DM-related periodontitis, and the ITGA4 upregulation was confirmed at the protein level. T2DM-related periodontitis also exhibited marked upregulation of GDF15 (also known as macrophage inhibitory cytokine-1) (Table 1), which was reported to play a role in regulating inflammatory and apoptotic pathways and whose expression was found to be significantly higher in multiple tissues of diabetic patients [35]. Finally, the downregulation of kinases and phosphatase-related genes in diseased tissues in diabetic patients is consistent with the role of the PI3K/AKT signaling as a classical phosphorylation cascade, where the protein kinases regulate phosphorylation and the phosphatases (e.g., PPP family) control dephosphorylation reactions. Remarkably, PIK3R2, a PI3K subunit involved in multiple canonical pathways, was downregulated at the mRNA level but was upregulated at the protein level in T2DM-related periodontitis. This downregulation could be due to the low stability of mRNA and/or the reduced proteasomal degradation of PIK3R2 since PTPN13, which dephosphorylates PIK3R2 at Tyr-655 [36], was decreased at the mRNA and protein levels. However, further studies are needed to confirm this assumption.

The IPA also predicted the activation of the Cardiac β-adrenergic and tRNA Splicing pathways in the T2DM-related periodontitis, which were predominantly driven by the upregulation of phosphodiesterases (PDE 1, 4, 5, 7, and 10), a superfamily of cAMP/cGMP hydrolyzing enzymes. The upregulation of PDEs has been linked to the cellular decrease of cAMP and cGMP and to the increase of oxidative stress and pro-inflammatory factors. Hence, the dysregulation of PDEs has been associated with inflammation, DM, DM complications, and imbalanced bone homeostasis [37] whereas the inhibition of PDEs has been suggested as a strategy for treating infectious and inflammatory conditions [3739]. Thus, the use of PDE inhibitors as an add-on treatment for T2DM-related periodontitis seems to be an attractive research topic.

The inactivation of the ADM signaling pathway was predicted in T2DM-related periodontitis. The downregulated genes associated with the ADM pathway included MAPKs, PLCs, the PIK3R2, the guanylate cyclase C, implicated in infection and inflammation control and the IL1RN, which encodes the anti-inflammatory protein IL-1Ra. Worthy of note, IL1RN mRNA downregulation was validated at the protein level by IHC. The ADM signaling pathway was also related to the upregulation of BCL2 and caspase-3, regulators of apoptosis, C3, an activator of the complement system, and IL1A, a pro-inflammatory mediator. Importantly, ADM has antimicrobial properties in the oral mucosal epithelia [40] and acts as an angiogenic, anti-apoptotic, and anti-inflammatory factor through the G protein-coupled receptor 182 (GPR182) and the receptor activity modifying protein 2 (RAMP2)/CALCRL complex [41]. While ADM2 was upregulated in periodontitis in the diabetic and non-diabetic patients (S4 Table), GPR182, CALCRL (validated at the protein level), and RAMP2 were significantly upregulated in the diabetic patients only, suggesting that ADM may perform its biological functions mainly in T2DM-related periodontitis. This hypothesis needs to be tested in further studies.

The superpathway of Cholesterol Biosynthesis and some of its related DEGs were downregulated in the T2DM-related periodontitis, suggesting the existence of a transcriptional regulation connecting this metabolic pathway with the immune system. DM and insulin deficiency have been reported to suppress the production of cholesterol synthesis enzymes and the transcriptional regulator sterol regulatory element-binding protein-2 (SREBF2; here, downregulated in the diabetic patients only; S3 Table) [42]. Overall, the reduction of cholesterol at a cellular level and the dysregulation of its precursors might affect cell proliferation, cycle progression, apoptosis, inflammation, and phagocytosis [43]. Notably, cholesterol intermediates may play biological functions besides cholesterol biosynthesis, which may affect inflammation and immunity [44]. Dehydrocholesterol reductase (DHCR) 7, for example, is involved in vitamin D synthesis, macrophage polarization, and regulation of IL-10 expression. Transmembrane 7 superfamily member 2 (TM7SF2) acts on the cell cycle, differentiation, and apoptosis and directs an anti-inflammatory loop [43, 44]. DHCR24, here, downregulated at the mRNA and protein levels in the T2DM-related periodontitis, has antiapoptotic functions and inhibits the generation of intracellular reactive oxygen species [43, 44] and LPS-induced osteoclastogenesis [45]. Future in-depth studies are required to identify the actual role of cholesterol biosynthesis-related genes on the pathogenesis of T2DM-related periodontitis.

The inhibition of SIPC was predicted in the T2DM-related periodontitis. Inositol phosphates are membrane components that have functions in cell growth, differentiation, migration, apoptosis, and endocytosis. Inositols are considered possible mediators of insulin signaling, and their insufficiency may contribute to insulin resistance, T2DM, and DM complications [46]. Among the proteins encoded by the downregulated DEGs associated with SIPC observed in the T2DM-related periodontitis were predominantly kinases and phosphatases, which not only control phosphorylation but also have further biological functions that can affect periodontitis progression. Dual-specificity phosphatase 14 (DUSP14) and inositol 1,4,5-trisphosphate 3-kinase C (ITPKC), for example, are both negative regulators of T-cell activation [47]. DUSP14 also has inhibitory effects on inflammation and osteoclastogenesis [45]. PTPN13, downregulated in diseased tissues in the diabetic patients at the mRNA and protein levels, is involved in apoptosis, cell migration, epithelial cell junction stabilization, and epithelial barrier regulation [48]. PTPN2, a vitamin-D-responsive tyrosine-specific phosphatase, acts as a protection factor controlling glucose homeostasis and pro-inflammatory conditions [48]. Therefore, the transcript-level and protein-level overexpression of PTPN2 observed in this study may represent an attempt to control the periodontal breakdown in the diabetic patients. Lastly, the observed downregulation of PLCs may also have implications for the pathogenesis of T2DM-related periodontitis. For instance, the lack or reduction of PLCD1 and PLCD3 was associated with increased cell apoptosis and the upregulation of LPS-induced proinflammation [49].

The IPA on the DEGs in the non-diabetic patients revealed less of an impact on the canonical pathways (there were no pathways with a Z‐score >2 or <−2). Among the dysregulated genes associated with the Tumor Microenvironment pathway observed in the non-diabetic patients were those that encoded MMPs, which play well-known roles in inflammation and immunity [50]. Importantly, ATF3, FOS, and JUN, that function in Tumor Microenvironment and PI3K Signaling in B Lymphocytes pathways, were exclusively downregulated in periodontitis in the non-diabetic patients. JUN, FOS, ATF, and FOSB (among the top ten downregulated genes in the non-diabetic patients; Table 1) are members of the activator protein-1 (AP-1) transcription factor complex, which is involved in cell proliferation, migration, apoptosis, inflammation, and bone remodeling [51]. The analysis of the transcriptomic datasets of human periodontitis (GSE16134 and GSE10334) and the immunosuppression genes by deep learning-based autoencoder techniques suggested that FOS and JUN were downregulated transcription factors with possible roles in periodontitis [52]. Using the same transcriptomic database (GESE16134), FOSB was identified as an upregulated DEG with potential participation in periodontitis [53]. Future studies are needed to explore the actual involvement of the AP-1 complex-related genes in periodontitis.

Long non-coding RNAs (lncRNAs) may have coding potential and affect the transcription, post-transcription, and translation of other genes and the activation or suppression of various encoding molecules and signaling pathways [5457]. In this study, tissues with periodontitis in diabetic and non-diabetic patients exhibited dysregulation in the expression of several lncRNAs, supporting the findings of previous investigations [57, 58]. lncRNAs were also suggested to play critical roles in DM complications [59, 60]. Intriguingly, two lncRNAs ranked in the top 15 downregulated DEGs (SCARNA2 and SMAD1-AS1) were only expressed in the healthy tissues in diabetic patients (Table 1). SCARNA2 has been reported to participate in DNA repair [61], whereas the biological functions of SMAD1-AS1 are still unknown. To understand the roles of lncRNAs in the pathogenesis of T2DM-related periodontitis is a future challenge.

A strength of this study is to be the first RNA-seq experiment evaluating the exclusively DEGs in T2DM-related periodontitis. Moreover, data analyses were expanded by using IPA to distinguish the associated signaling pathways. Furthermore, a valuable database for the future functional validation of DEGs and signaling pathways was generated.. Although a previous study indorsed a minimum of five samples per group for RNA-Seq datasets [62], one of the limitations of the present study is the small sample size. Hence our findings should be interpreted with caution and may need to be confirmed in a large-scale study. Another limitation is the analysis of the entire gingival transcriptomic profiles from a mix of cells that tends to reflect the gene expression patterns of predominant cell types (e.g., epithelial cells and fibroblasts). Thus, additional studies are needed to elucidate individual cell transcription profile involved in T2DM-related periodontitis. Furthermore, our study focused on uncontrolled T2DM. Future studies should also evaluate the effects of well-controlled T2DM on the transcriptomic profile of periodontium. Also, the cross-sectional nature of this study does not allow for causal and temporal inferences on the effects of a given gene/pathway on disease progression. Finally, to obtain a comprehensive understanding of the biological system underling the pathogenesis of the T2DM-related periodontitis, additional omics-based methodologies (e.g., proteomics) should be considered in future studies.

Conclusion

CXCL5 was one of the most upregulated genes, while LCE2 and LCE3 were considerably downregulated T2DM-related periodontitis. Furthermore, T2DM-related periodontitis was associated with the dysregulation of functional pathways, including the activation of Phagosome Formation, Cardiac β-adrenergic, tRNA Splicing, and PI3K/AKT pathways and inhibition of Cholesterol Biosynthesis, Adrenomedullin, and Inositol Phosphate Compounds pathways. The idenfication of unique transcriptomic signatures can be used as potential targets for the development of future diagnostic, preventive and therapeutic approaches.

Supporting information

S1 Fig. The DEGs expressed in the non-diabetic patients (yellow), in patients with T2DM (blue) and the commonly expressed in both the non-diabetic and diabetic patients.

HH: non-diabetic patients without periodontitis; HP: non-diabetic patients with periodontitis; DH: T2DM patients without periodontitis; DP: T2DM patients with periodontitis.

(JPEG)

S1 Table. Demographic characteristics, glycemic and periodontal parameters (mean ± SD) of the study population.

(DOCX)

S2 Table. DEGs in tissues with periodontitis in non-diabetics relative to healthy tissues in non-diabetics exclusively.

(XLSX)

S3 Table. DEGs in tissues with periodontitis relative to healthy tissues observed in patients with T2DM exclusively.

(XLSX)

S4 Table. DEGs in tissues with periodontitis in relation to healthy tissues observed in non-diabetic patients and in patients with T2DM concurrently.

(XLSX)

S5 Table. Ingenuity canonical pathways related to DEGs expressed exclusively in non-diabetic patients and the respective genes that function within each of these signaling pathways.

(DOCX)

S6 Table. Ingenuity canonical pathways related to the DEGs expressed exclusively in patients with T2DM and the respective genes that function within these signaling pathways.

(DOCX)

Acknowledgments

We thank Dr. Ann Fu Dongtao for her technical assistance with immunohistochemistry.

Data Availability

All relevant data are within the manuscript and its Supporting Information files.

Funding Statement

PMD received a University of Florida College of Dentistry Faculty grant. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Emrich LJ, Shlossman M, Genco RJ. Periodontal disease in non-insulin-dependent diabetes mellitus. J Periodontol. 1991;62: 123–131. doi: 10.1902/jop.1991.62.2.123 [DOI] [PubMed] [Google Scholar]
  • 2.Graziani F, Gennai S, Solini A, Petrini M. A systematic review and meta-analysis of epidemiologic observational evidence on the effect of periodontitis on diabetes An update of the EFP-AAP review. J Clin Periodontol. 2018;45: 167–187. doi: 10.1111/jcpe.12837 [DOI] [PubMed] [Google Scholar]
  • 3.Tonetti MS, Greenwell H, Kornman KS. Staging and grading of periodontitis: Framework and proposal of a new classification and case definition. J Clin Periodontol. 2018; 45 Suppl 20: S149–S161. [DOI] [PubMed] [Google Scholar]
  • 4.Sonnenschein SK, Meyle J. Local inflammatory reactions in patients with diabetes and periodontitis. Periodontol 2000. 2015;69: 221–254. doi: 10.1111/prd.12089 [DOI] [PubMed] [Google Scholar]
  • 5.Graves DT, Ding Z, Yang Y. The impact of diabetes on periodontal diseases. Periodontol 2000. 2020;82: 214–224. doi: 10.1111/prd.12318 [DOI] [PubMed] [Google Scholar]
  • 6.Park SJ, Saito-Adachi M, Komiyama Y, Nakai K. Advances, practice, and clinical perspectives in high-throughput sequencing. Oral Dis. 2016;22: 353–364. doi: 10.1111/odi.12403 [DOI] [PubMed] [Google Scholar]
  • 7.Chapple ILC, Mealey BL, Van Dyke TE, Bartold PM, Dommisch H, Eickholz P, et al. Periodontal health and gingival diseases and conditions on an intact and a reduced periodontium: Consensus report of workgroup 1 of the 2017 World Workshop on the Classification of Periodontal and Peri-Implant Diseases and Conditions. J Clin Periodontol. 2018;45 Suppl 20: S68–S77. doi: 10.1111/jcpe.12940 [DOI] [PubMed] [Google Scholar]
  • 8.Papapanou PN, Sanz M, Buduneli N, Dietrich T, Feres M, Fine DH, et al. Periodontitis: Consensus report of workgroup 2 of the 2017 World Workshop on the Classification of Periodontal and Peri-Implant Diseases and Conditions. J Clin Periodontol. 2018;45 Suppl 20: S162–S170. doi: 10.1111/jcpe.12946 [DOI] [PubMed] [Google Scholar]
  • 9.Jepsen S, Caton JG, Albandar JM, Bissada NF, Bouchard P, Cortellini P, et al. Periodontal manifestations of systemic diseases and developmental and acquired conditions: Consensus report of workgroup 3 of the 2017 World Workshop on the Classification of Periodontal and Peri-Implant Diseases and Conditions. J Periodontol. 2018; 89 Suppl 1: S237–S248. doi: 10.1002/JPER.17-0733 [DOI] [PubMed] [Google Scholar]
  • 10.FASQC, http://www.bioinformatics.babraham.ac.uk/projects/fastqc/.
  • 11.Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014; 30: 2114–2120. doi: 10.1093/bioinformatics/btu170 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013; 29:15–21. doi: 10.1093/bioinformatics/bts635 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. 2011;12: 323. doi: 10.1186/1471-2105-12-323 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–40. doi: 10.1093/bioinformatics/btp616 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Koch AE. Chemokines and their receptors in rheumatoid arthritis: future targets? Arthritis Rheum. 2005;52:710–721. doi: 10.1002/art.20932 [DOI] [PubMed] [Google Scholar]
  • 16.Chavey C, Fajas L. CXCL5 drives obesity to diabetes, and further. Aging (Albany NY). 2009;1: 674–677. doi: 10.18632/aging.100064 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zychowska M, Rojewska E, Pilat D, Mika J. The role of some chemokines from the CXC subfamily in a mouse model of diabetic neuropathy. J Diabetes Res. 2015;2015: 750182. doi: 10.1155/2015/750182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lappin DF, Robertson D, Hodge P, Treagus D, Awang RA, Ramage G, et al. The Influence of Glycated Hemoglobin on the Cross Susceptibility Between Type 1 Diabetes Mellitus and Periodontal Disease. J Periodontol. 2015;86:1249–1259. doi: 10.1902/jop.2015.150149 [DOI] [PubMed] [Google Scholar]
  • 19.de Koning HD, van den Bogaard EH, Bergboer JG, Kamsteeg M, van Vlijmen-Willems IM, Hitomi K, et al. Expression profile of cornified envelope structural proteins and keratinocyte differentiation-regulating proteins during skin barrier repair. Br J Dermatol. 2012; 166: 1245–1254. doi: 10.1111/j.1365-2133.2012.10885.x [DOI] [PubMed] [Google Scholar]
  • 20.Niehues H, van der Krieken DA, Ederveen THA, Jansen PAM, van Niftrik L, Mesman R, et al. Antimicrobial Late Cornified Envelope Proteins: The Psoriasis Risk Factor Deletion of LCE3B/C Genes Affects Microbiota Composition. J Invest Dermatol. 2022;142: 1947–1955.e6. doi: 10.1016/j.jid.2021.11.036 [DOI] [PubMed] [Google Scholar]
  • 21.DiTommaso T, Cottle DL, Pearson HB, Schlüter H, Kaur P, Humbert PO, et al. Keratin 76 is required for tight junction function and maintenance of the skin barrier. PLoS Genet. 2014;10: e1004706. doi: 10.1371/journal.pgen.1004706 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Shao S, Chen J, Swindell WR, Tsoi LC, Xing X, Ma F, et al. Phospholipase A2 enzymes represent a shared pathogenic pathway in psoriasis and pityriasis rubra pilaris. JCI Insight. 2021;6: e151911. doi: 10.1172/jci.insight.151911 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lee HJ, Woo Y, Hahn TW, Jung YM, Jung YJ. Formation and Maturation of the Phagosome: A Key Mechanism in Innate Immunity against Intracellular Bacterial Infection. Microorganisms. 2020;8: 1298. doi: 10.3390/microorganisms8091298 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Tang S, Wang X, Deng T, Ge H, Xiao X. Identification of C3 as a therapeutic target for diabetic nephropathy by bioinformatics analysis. Sci Rep. 2020. 10(1): 13468. doi: 10.1038/s41598-020-70540-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Tan SM, Snelson M, Østergaard JA, Coughlan MT. The Complement Pathway: New Insights into Immunometabolic Signaling in Diabetic Kidney Disease. Antioxid Redox Signal. 2022;37:781–801. doi: 10.1089/ars.2021.0125 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hodgson K, Morris J, Bridson T, Govan B, Rush C, Ketheesan N. Immunological mechanisms contributing to the double burden of diabetes and intracellular bacterial infections. Immunology. 2015;144: 171–185. doi: 10.1111/imm.12394 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Dowey R, Iqbal A, Heller SR, Sabroe I, Prince LR. A Bittersweet Response to Infection in Diabetes; Targeting Neutrophils to Modify Inflammation and Improve Host Immunity. Front Immunol. 2021;12: 678771. doi: 10.3389/fimmu.2021.678771 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Gillooly DJ, Simonsen A, Stenmark H. Phosphoinositides and phagocytosis. J Cell Biol. 2001;155: 15–17. doi: 10.1083/jcb.200109001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Huang X, Liu G, Guo J, Su Z. The PI3K/AKT pathway in obesity and type 2 diabetes. Int J Biol Sci. 2018;14:1483–1496. doi: 10.7150/ijbs.27173 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kumar M, Bansal N. Implications of Phosphoinositide 3-Kinase-Akt (PI3K-Akt) Pathway in the Pathogenesis of Alzheimer’s Disease. Mol Neurobiol. 2022;59: 354–385. doi: 10.1007/s12035-021-02611-7 [DOI] [PubMed] [Google Scholar]
  • 31.Dariya B, Nagaraju GP. Advanced glycation end products in diabetes, cancer and phytochemical therapy. Drug Discov Today. 2020;25: 1614–1623. doi: 10.1016/j.drudis.2020.07.003 [DOI] [PubMed] [Google Scholar]
  • 32.Xing X, Jiang Y, Wang H, Zhang Y, Niu T, Qu Y, et al. Identification of novel differentially expressed genes in retinas of STZ-induced long-term diabetic rats through RNA sequencing. Mol Genet Genomic Med. 2020;8: e1115. doi: 10.1002/mgg3.1115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Leibbrandt A, Penninger JM. RANK/RANKL: regulators of immune responses and bone physiology. Ann N Y Acad Sci. 2008;1143: 123–150. doi: 10.1196/annals.1443.016 [DOI] [PubMed] [Google Scholar]
  • 34.Mezu-Ndubuisi OJ, Maheshwari A. The role of integrins in inflammation and angiogenesis. Pediatr Res. 2021;89: 1619–1626. doi: 10.1038/s41390-020-01177-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Lu J, Zhang Y, Dong X, Lu J, Zhang C, Liu J, et al. Association between MIC-1 and Type 2 Diabetes: A Combined Analysis. Dis Markers. 2019;2019: 7284691. doi: 10.1155/2019/7284691 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kuchay S, Duan S, Schenkein E, Peschiaroli A, Saraf A, Florens L, et al. FBXL2- and PTPL1-mediated degradation of p110-free p85β regulatory subunit controls the PI(3)K signalling cascade. Nat Cell Biol. 2013;15:472–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kilanowska A, Ziółkowska A. Role of Phosphodiesterase in the Biology and Pathology of Diabetes. Int J Mol Sci. 2020;21: 8244. doi: 10.3390/ijms21218244 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Su H, Ma C, Li H. Anti-infection mechanism of phosphodiesterase-5 inhibitors and their roles in coronavirus disease 2019 (Review). Exp Ther Med. 2021. Apr;21(4):320. doi: 10.3892/etm.2021.9751 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Ookawara M, Nio Y. Phosphodiesterase 4 inhibitors in diabetic nephropathy. Cell Signal. 2022;90: 110185. doi: 10.1016/j.cellsig.2021.110185 [DOI] [PubMed] [Google Scholar]
  • 40.Hinson JP, Kapas S, Smith DM. Adrenomedullin, a multifunctional regulatory peptide. Endocr Rev. 2000;21: 138–167 doi: 10.1210/edrv.21.2.0396 [DOI] [PubMed] [Google Scholar]
  • 41.Xu M, Qi F, Zhang S, Ma X, Wang S, Wang C, et al. Adrenomedullin promotes the growth of pancreatic ductal adenocarcinoma through recruitment of myelomonocytic cells. Oncotarget. 2016;7: 55043–55056. doi: 10.18632/oncotarget.10393 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Suzuki R, Lee K, Jing E, Biddinger SB, McDonald JG, Montine TJ, et al. Diabetes and insulin in regulation of brain cholesterol metabolism. Cell Metab. 2010;12: 567–579. doi: 10.1016/j.cmet.2010.11.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Bah SY, Dickinson P, Forster T, Kampmann B, Ghazal P. Immune oxysterols: Role in mycobacterial infection and inflammation. J Steroid Biochem Mol Biol. 2017;169: 152–163. doi: 10.1016/j.jsbmb.2016.04.015 [DOI] [PubMed] [Google Scholar]
  • 44.Brown AJ, Ikonen E, Olkkonen VM. Cholesterol precursors: more than mere markers of biosynthesis. Curr Opin Lipidol. 2014; 25:133–139. doi: 10.1097/MOL.0000000000000038 [DOI] [PubMed] [Google Scholar]
  • 45.Khuda II, Koide N, Noman AS, Dagvadorj J, Tumurkhuu G, Naiki Y, et al. Seladin-1 is a novel lipopolysaccharide (LPS)-responsive gene and inhibits the tumour necrosis factor-alpha production and osteoclast formation in response to LPS. Immunology. 2010;131:59–66. doi: 10.1111/j.1365-2567.2010.03274.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Caputo M, Bona E, Leone I, Samà MT, Nuzzo A, Ferrero A, et al. Inositols and metabolic disorders: From farm to bedside. J Tradit Complement Med. 2020;10: 252–259. doi: 10.1016/j.jtcme.2020.03.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zhao Z, Yang J, Zhang L, Zhou Y. Enhancement of DUSP14 (dual specificity phosphatase 14) limits osteoarthritis progression by alleviating chondrocyte injury, inflammation and metabolic homeostasis. Bioengineered. 2021;12: 7495–7507. doi: 10.1080/21655979.2021.1979355 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Tang X, Qi C, Zhou H, Liu Y. Critical roles of PTPN family members regulated by non-coding RNAs in tumorigenesis and immunotherapy. Front Oncol. 2022;12: 972906. doi: 10.3389/fonc.2022.972906 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Kudo K, Uchida T, Sawada M, Nakamura Y, Yoneda A, Fukami K. Phospholipase C δ1 in macrophages negatively regulates TLR4-induced proinflammatory cytokine production and Fcγ receptor-mediated phagocytosis. Adv Biol Regul. 2016;61: 68–79. [DOI] [PubMed] [Google Scholar]
  • 50.Cui N, Hu M, Khalil RA. Biochemical and Biological Attributes of Matrix Metalloproteinases. Prog Mol Biol Transl Sci. 2017;147: 1–73 doi: 10.1016/bs.pmbts.2017.02.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Cai M, Chen N. The Roles of IRF-8 in Regulating IL-9-Mediated Immunologic Mechanisms in the Development of DLBCL: A State-of-the-Art Literature Review. Front Oncol. 2022;12: 817069. doi: 10.3389/fonc.2022.817069 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Ning W, Acharya A, Sun Z, Ogbuehi AC, Li C, Hua S, et al. Deep Learning Reveals Key Immunosuppression Genes and Distinct Immunotypes in Periodontitis. Front Genet. 2021;12: 648329. doi: 10.3389/fgene.2021.648329 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Song L, Yao J, He Z, Xu B. Genes related to inflammation and bone loss process in periodontitis suggested by bioinformatics methods. BMC Oral Health. 2015. Sep 4;15: 105. doi: 10.1186/s12903-015-0086-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Statello L, Guo CJ, Chen LL, Huarte M. Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol. 2021; 22: 96–118. doi: 10.1038/s41580-020-00315-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Della Bella E, Koch J, Baerenfaller K. Translation and emerging functions of non-coding RNAs in inflammation and immunity. Allergy. 2022;77: 2025–2037. doi: 10.1111/all.15234 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Sayad A, Mirzajani S, Gholami L, Razzaghi P, Ghafouri-Fard S, Taheri M. Emerging role of long non-coding RNAs in the pathogenesis of periodontitis. Biomed Pharmacother. 2020;129: 110362. doi: 10.1016/j.biopha.2020.110362 [DOI] [PubMed] [Google Scholar]
  • 57.Xu J, Yin Y, Lin Y, Tian M, Liu T, Li X, et al. Long non-coding RNAs: Emerging roles in periodontitis. J Periodontal Res. 2021; 56: 848–862. doi: 10.1111/jre.12910 [DOI] [PubMed] [Google Scholar]
  • 58.Chang W, Wang M, Zhang Y, Yu F, Hu B, Goljanek-Whysall K, et al. Roles of long noncoding RNAs and small extracellular vesicle-long noncoding RNAs in type 2 diabetes. Traffic. 2022; 23: 526–537. doi: 10.1111/tra.12868 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Chen Y, He Y, Zhou H. The potential role of lncRNAs in diabetes and diabetic microvascular complications. Endocr J. 2020. Jul 28;67(7):659–668. doi: 10.1507/endocrj.EJ19-0574 [DOI] [PubMed] [Google Scholar]
  • 60.Tanwar VS, Reddy MA, Natarajan R. Emerging Role of Long Non-Coding RNAs in Diabetic Vascular Complications. Front Endocrinol (Lausanne). 2021;12: 665811. doi: 10.3389/fendo.2021.665811 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Bergstrand S, O’Brien EM, Coucoravas C, Hrossova D, Peirasmaki D, Schmidli S, et al. Small Cajal body-associated RNA 2 (scaRNA2) regulates DNA repair pathway choice by inhibiting DNA-PK. Nat Commun. 2022;13: 1015. doi: 10.1038/s41467-022-28646-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Ching T, Huang S, Garmire LX. Power analysis and sample size estimation for RNA-Seq differential expression. RNA. 2014;20:1684–1696. doi: 10.1261/rna.046011.114 [DOI] [PMC free article] [PubMed] [Google Scholar]

Decision Letter 0

Víctor Sánchez-Margalet

7 Nov 2023

PONE-D-23-19661Distinctive genes and signaling pathways associated with type 2 diabetes-related periodontitis: a preliminary studyPLOS ONE

Dear Dr. Duarte,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

==============================

As you may find in the reviewers comments the manuscript should be changed throughly before it may be considered for publication. The conclusions should be better explained and the limitations of the study should be stressed. Besides the methodological suggestions of the reviewr 2should be followed.

==============================

Please submit your revised manuscript by Dec 22 2023 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols. Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols.

We look forward to receiving your revised manuscript.

Kind regards,

Víctor Sánchez-Margalet

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at 

https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and 

https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2. Thank you for stating the following in the Acknowledgments Section of your manuscript: 

"All funding was provided by University of Florida College of Dentistry Faculty Seed Grant. We thank Dr. Ann Fu Dongtao for her technical assistance with immunohistochemistry."

We note that you have provided funding information that is not currently declared in your Funding Statement. However, funding information should not appear in the Acknowledgments section or other areas of your manuscript. We will only publish funding information present in the Funding Statement section of the online submission form. 

Please remove any funding-related text from the manuscript and let us know how you would like to update your Funding Statement. Currently, your Funding Statement reads as follows: 

"PMD received a University of Florida College of Dentistry Faculty grant. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript."

Please include your amended statements within your cover letter; we will change the online submission form on your behalf.

3. Your ethics statement should only appear in the Methods section of your manuscript. If your ethics statement is written in any section besides the Methods, please delete it from any other section. 

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: No

Reviewer #2: Yes

Reviewer #3: Yes

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: No

Reviewer #2: Yes

Reviewer #3: Yes

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: No

Reviewer #2: Yes

Reviewer #3: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: The biological mechanisms underlying the pathogenesis of type 2 diabetes (T2DM) related periodontitis remain unclear. This cross-sectional study evaluated the distinctive transcriptomic changes between tissues with periodontal health and with periodontitis in patients with T2DM. In this cross-sectional study, whole transcriptome sequencing was performed on gingival biopsies from non-periodontitis and periodontitis tissues from non-diabetic and diabetic patients. A differentially expressed gene (DEG) analysis and Ingenuity Pathway Analysis (IPA) assessed the genes and signaling pathways associated with T2DM-related periodontitis.

It is preliminary study. It may be submitted again after completion of the project.

Reviewer #2: This study evaluated the distinctive transcriptomic changes between tissues with periodontal health and with periodontitis in patients with T2DM, through the analysis of the transcriptome performed on gingival biopsies from non-periodontitis and periodontitis tissues from non-diabetic and diabetic patients.

The manuscript is well written, the study well designed and the discussion of the findings was carried out in a detailed and precise manner.

I made some comments that can be found in the attached file.

Reviewer #3: Dear Authors,

I appreciate your time and effort in conducting this study.

In this study, the authors evaluated the whole transcriptome profile and related histopathological analysis of the periodontitis patients with and without T2DM and non-periodontitis patients with and without T2DM from gingival tissue samples.

The determination of the numbers of study groups, collection of samples during which surgical procedures, how the samples for IHC were stored, the reason why 4 samples were selected from each group for IHC, and why proteomics was not conducted have not been explained.

I believe that the results of this study necessitate researchers to write a more specific conclusion. What is expected to be served by the results of this study?

I hope I could provide some support.

Kind Regards

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

Reviewer #3: Yes: Esra Guzeldemir Akcakanat

**********

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step.

Attachment

Submitted filename: PONE-D-23-19661_revised.pdf

Attachment

Submitted filename: Review.docx

PLoS One. 2024 Jan 19;19(1):e0296925. doi: 10.1371/journal.pone.0296925.r002

Author response to Decision Letter 0


27 Nov 2023

AUTHOR’S RESPONSE

We thank the Associate Editor and the Referees for their time and for the constructive comments regarding this manuscript. Please, find below the point-by-point answers to the Reviewers' comments.

Reviewer’s comments: 1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at

https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

Author’s response: PLOS ONE's style requirements were revised accordingly.

Reviewer’s comments: 2. Thank you for stating the following in the Acknowledgments Section of your manuscript: "All funding was provided by University of Florida College of Dentistry Faculty Seed Grant. We thank Dr. Ann Fu Dongtao for her technical assistance with immunohistochemistry." We note that you have provided funding information that is not currently declared in your Funding Statement. However, funding information should not appear in the Acknowledgments section or other areas of your manuscript. We will only publish funding information present in the Funding Statement section of the online submission form.

Please remove any funding-related text from the manuscript and let us know how you would like to update your Funding Statement. Currently, your Funding Statement reads as follows:

"PMD received a University of Florida College of Dentistry Faculty grant. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript." Please include your amended statements within your cover letter; we will change the online submission form on your behalf.

Author’s response: Acknowledgments Section was revised accordingly. Please, keep the same funding statement.

Reviewer’s comments: 3. Your ethics statement should only appear in the Methods section of your manuscript. If your ethics statement is written in any section besides the Methods, please delete it from any other section.

Author’s response: Ethics statement was revised accordingly. It is now only in the Methods section.

Review Comments to the Author

Reviewer #1:

Reviewer’s comments: The biological mechanisms underlying the pathogenesis of type 2 diabetes (T2DM) related periodontitis remain unclear. This cross-sectional study evaluated the distinctive transcriptomic changes between tissues with periodontal health and with periodontitis in patients with T2DM. In this cross-sectional study, whole transcriptome sequencing was performed on gingival biopsies from non-periodontitis and periodontitis tissues from non-diabetic and diabetic patients. A differentially expressed gene (DEG) analysis and Ingenuity Pathway Analysis (IPA) assessed the genes and signaling pathways associated with T2DM-related periodontitis. It is preliminary study. It may be submitted again after completion of the project.

Author’s response: Thank you so much for taking the time to review our manuscript. We classified our study as preliminary because of its exploratory nature and sample size (please, note that the study was performed using a University of Florida Seed Grant). However, considering the magnitude and richness of the data generated, we believe that the study is certainly powered to provide relevant information to the scientific community. Based on these initial validated data, we are going to definitely continue to investigate the potential pathways and mechanisms involved in the pathogenesis of type 2 DM-related periodontitis by applying additional methodologies and a larger population. However, we believe that data generated in this initial study is important and valuable and can contribute to other research groups that investigate the pathogenesis of periodontitis in patient with diabetes. Thus, we humbly would like to ask this reviewer to consider the publication of these findings as they are.

Reviewer #2

Reviewer’s comments: The authors presented the periodontal parameters with mean and SD. The authors could stratify the sample regarding the periodontal parameters to better characterize the sample, especially for CAL and PD. For example: 2-3 mm, 4-5 mm and greater than 6 mm.

Author’s response: Information about the full-mouth percentage of sites with different PD (PD ≤ 3mm, PD=4-6mm and PD ≥ 7 mm), and clinical attachment loss (no clinical attachment loss and with CA loss of 1-2 mm, 3-4mm and ≥ 5 mm) categories was added in S1 table, as requested by this reviewer. Please, note that some periodontally healthy patients could have sites with PD > 3mm, assuming pseudopockets. Furthermore, they could also present some sites with CAL, but those were non-interdental and attributed to non-periodontitis reasons. This information was taken from patients records.

Reviewer’s comments: Writing gene names in humans should be written in italics.

Author’s response: Gene names were revised accordingly.

Reviewer’s comments: Have you ever think about GSEA (Gene Set Enrichment Analysis)? I believe that the statistical approach of GSEA strengthened the results regarding the gene sets involved in the biological process of the diseases studied here.

Author’s response: Considering the exploratory nature of this study, we chose the Ingenuity pathway analysis (IPA, QIAGEN) to initially explore our transcriptomic data and to provide information about pathways, genes and other signatures that may be significantly altered across our groups. The authors are aware that GSEA employs gene sets and features that have been assumed to be associated with a given disease or pathway in order to suggest biological applications. Thus, the GSEA analysis suggested by this Reviewer is a valuable complementary/confirmatory analysis that our group intends to apply in future studies. However, at this stage, we would need to start the study from the very beginning, doing new bioinformatics, results presentation (tables and figures), data interpretation and discussion. So, given the impracticality of carrying out the suggested analysis at this moment, we would be grateful if this reviewer agrees that the IPA analyses presented in our preliminary study are sufficient and provide relevant information that may guide future studies in the field.

Reviewer’s comments: The authors could provide more information about their findings in the conclusion section, drawing attention to the main pathways found and some differently expressed genes.

Author’s response: Conclusion section was revised as suggest by this reviewer and reviewer #3.

Reviewer’s comments: I suggest reporting the study methodology according to STROBE guidelines.

Author’s response: Thank you very much for this suggestion. STROBE guidelines were adopted for study conducting and reporting findings

Reviewer’s comments: How did the authors set the sample size?

Author’s response: This reviewer raised an important question about our study sample size. Studies using bulk RNA sequencing in human gingival tissues with periodontitis varies considerably in terms of sample size (from 6 to more than 10 sample per group) (e.g. Chen et al. doi: 10.1007/s00784-023-05017-y, Oh et al. doi: 10.1002/JPER.23-0289, Kim et al. doi: 10.1186/s40246-016-0084-0). However, so far, no previous study performed RNA sequencing of human gingival samples with periodontitis and diabetes to be used as a reference for our sample size calculation. Thus, as we did not know the actual difference in the means and variance of genes that are significantly changed between our case and control samples from prior studies, a precise sample size could not be calculated. Considering the exploratory characteristic of our study (preliminary study), sample size was established based on previous recommendation of a minimum of five samples per group for RNA-Seq datasets (Ching et al. 2014). It is important to highlight that we are aware that sample size is a limitation of our study and that our findings should be confirmed in a large-scale study. This is stated in the discussion section (last paragraph).

Reviewer’s comments: Who performed the diagnosis of the periodontal condition? Was it one or more examiners? Were the examiners who performed the periodontal examination calibrated to each other?

Author’s response: Our transcriptome analysis focused on waste gingival tissues from specific teeth not on patient’s full-mouth periodontal condition. Full-mouth data are presented in S1 Table in order to characterize the severity of periodontitis of the study population. Thus, full-mouth clinical examination was not performed by a single examiner but obtained from patient’s records. However, in order to have accuracy in the periodontal diagnosis of the sampled teeth, periodontal parameters (i.e., probing depth, clinical attachment level, bleeding on probing, furcation involvement and mobility) were recorded around the specific sampled teeth by the same trained and calibrated examiner (P.M.D). The study examiner participated in a previous calibration exercise, and the standard error of measurement was estimated. Intra-examiner variability was recorded as 0.20 mm for PD and 0.23 mm for CAL. This information was included in the Material and Methods section – Population.

Reviewer’s comments: I believe that a group of patients with well-controlled DM could bring important information about the influence of DM on the pathogenesis of Periodontitis. The absence of this group does not make the results obtained unfeasible, but a discussion about this would be interesting.

Author’s response: Thank you very much for this suggestion. At this moment, we only focused on uncontrolled T2DM because we assumed that it would affect more the pathogenesis of periodontitis. But we agree with this reviewer that a group of well-controlled patients would be very interesting. Thus, it will be considered in future studies. This consideration was included in the discussion section, as suggested by this reviewer (Please, see Discussion Section – last paragraph).

Reviewer’s comments: Do the authors intend to deposit the transcriptome data in a public domain database, such as Gene Expression Omnibus (GEO)?

Author’s response: According to our data availability statement, all data are available in the main text or in the supplementary materials. We do not intend to deposit it in a public domain database at this time.

Reviewer #3

Reviewer’s comments: This study evaluated the distinctive transcriptomic changes between tissues with periodontal health and with periodontitis in patients with T2DM, through the analysis of the transcriptome performed on gingival biopsies from non-periodontitis and periodontitis tissues from non-diabetic and diabetic patients. The manuscript is well written, the study well designed, and the discussion of the findings was carried out in a detailed and precise manner. I made some comments that can be found in the attached file.

Author’s response: Thank you so much for the encouraging comments about our manuscript.

Reviewer’s comments: The determination of the numbers of study groups, collection of samples during which surgical procedures, how the samples for IHC were stored, the reason why 4 samples were selected from each group for IHC, and why proteomics was not conducted have not been explained.

Author’s response: All explanations requested by this reviewer was included in the Material and Methods section (please, see tack changes).

Determination of the number of study groups: Considering that we would like to observe the differently expressed genes and pathways in patients with periodontitis and in patients with DM, we had to use periodontally-healthy patients and patients without DM as references for the transcriptome analysis and IHC. Thus, “based on the periodontal and DM conditions, patients were assigned into one of the following four groups: non-diabetic patients without periodontitis (HH; n=9), non-diabetic patients with periodontitis (HP; n=9), T2DM patients without periodontitis (DH; n=9), and T2DM patients with periodontitis (DP: n=9).” (Please, see Material and Methods - Population).

Collection of samples during which surgical procedures: Collection of samples was detailed, as requested by this reviewer (Please, see Material and Methods – Tissue collection).

How the samples for IHC were stored: Thank you for this observation. The method used for sample fixation and storage was described accordingly (Please, see Material and Methods – IHC).

The reason why 4 samples were selected from each group for IHC: In this study, IHC was performed for a total of eight antibodies in four different groups, totalizing 128 sections. At this exploratory phase, we sought to present only descriptive analyses of the IHC sections, which does not require a large number of repetitions. Thus, sample size calculation was not performed. Considering the investigative characteristic of our study, we thought that the descriptive method would give us valuable details (staining location in terms of tissue [epithelium, connective tissue, or vessels], cell type and cell site [membrane, cytoplasm or nuclear], which may be hidden by quantitative methods, using scoring system categorization.

Why proteomics was not conducted: This reviewer raised an important question about the performance of additional omics analyses. Unfortunately, due to a budget constraint, at this moment, our analysis limited to transcriptome analysis, followed by validation of the main results at the protein level by IHC. Additional omics analyses will be certainly considered in future studies. This was stated in the discussion section. (Please, see Discussion Section – last paragraph).

Reviewer’s comments: I believe that the results of this study necessitate researchers to write a more specific conclusion. What is expected to be served by the results of this study?

Author’s response: Conclusion section was revised, as suggested by this reviewer and reviewer #2.

Attachment

Submitted filename: Response to Reviewers.docx

Decision Letter 1

Víctor Sánchez-Margalet

26 Dec 2023

Distinctive genes and signaling pathways associated with type 2 diabetes-related periodontitis: a preliminary study

PONE-D-23-19661R1

Dear Dr. Mendes Duarte

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice for payment will follow shortly after the formal acceptance. To ensure an efficient process, please log into Editorial Manager at http://www.editorialmanager.com/pone/, click the 'Update My Information' link at the top of the page, and double check that your user information is up-to-date. If you have any billing related questions, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Víctor Sánchez-Margalet

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #2: All comments have been addressed

Reviewer #3: All comments have been addressed

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #2: Yes

Reviewer #3: Yes

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #2: Yes

Reviewer #3: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #2: Yes

Reviewer #3: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #2: Yes

Reviewer #3: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #2: I would like to congratulate the authors for their effort in properly reviewing the manuscript. I believe that with the reviewers' suggestions the study became more robust and accurate.

Reviewer #3: (No Response)

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #2: No

Reviewer #3: No

**********

Attachment

Submitted filename: Review1_Editor.docx

Acceptance letter

Víctor Sánchez-Margalet

10 Jan 2024

PONE-D-23-19661R1

PLOS ONE

Dear Dr. Duarte,

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now being handed over to our production team.

At this stage, our production department will prepare your paper for publication. This includes ensuring the following:

* All references, tables, and figures are properly cited

* All relevant supporting information is included in the manuscript submission,

* There are no issues that prevent the paper from being properly typeset

If revisions are needed, the production department will contact you directly to resolve them. If no revisions are needed, you will receive an email when the publication date has been set. At this time, we do not offer pre-publication proofs to authors during production of the accepted work. Please keep in mind that we are working through a large volume of accepted articles, so please give us a few weeks to review your paper and let you know the next and final steps.

Lastly, if your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

If we can help with anything else, please email us at customercare@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Víctor Sánchez-Margalet

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Fig. The DEGs expressed in the non-diabetic patients (yellow), in patients with T2DM (blue) and the commonly expressed in both the non-diabetic and diabetic patients.

    HH: non-diabetic patients without periodontitis; HP: non-diabetic patients with periodontitis; DH: T2DM patients without periodontitis; DP: T2DM patients with periodontitis.

    (JPEG)

    S1 Table. Demographic characteristics, glycemic and periodontal parameters (mean ± SD) of the study population.

    (DOCX)

    S2 Table. DEGs in tissues with periodontitis in non-diabetics relative to healthy tissues in non-diabetics exclusively.

    (XLSX)

    S3 Table. DEGs in tissues with periodontitis relative to healthy tissues observed in patients with T2DM exclusively.

    (XLSX)

    S4 Table. DEGs in tissues with periodontitis in relation to healthy tissues observed in non-diabetic patients and in patients with T2DM concurrently.

    (XLSX)

    S5 Table. Ingenuity canonical pathways related to DEGs expressed exclusively in non-diabetic patients and the respective genes that function within each of these signaling pathways.

    (DOCX)

    S6 Table. Ingenuity canonical pathways related to the DEGs expressed exclusively in patients with T2DM and the respective genes that function within these signaling pathways.

    (DOCX)

    Attachment

    Submitted filename: PONE-D-23-19661_revised.pdf

    Attachment

    Submitted filename: Review.docx

    Attachment

    Submitted filename: Response to Reviewers.docx

    Attachment

    Submitted filename: Review1_Editor.docx

    Data Availability Statement

    All relevant data are within the manuscript and its Supporting Information files.


    Articles from PLOS ONE are provided here courtesy of PLOS

    RESOURCES