Next Article in Journal
Spontaneous Bone Marrow Edema: Perfusion Abnormalities and Treatment with Surgical Decompression
Next Article in Special Issue
Er:YAG Laser Alleviates Inflammaging in Diabetes-Associated Periodontitis via Activation CTBP1-AS2/miR-155/SIRT1 Axis
Previous Article in Journal
Sirtuin 6 Regulates the Activation of the ATP/Purinergic Axis in Endothelial Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Evidence Linking PPARG Genetic Variants with Periodontitis and Type 2 Diabetes Mellitus in a Brazilian Population

by
Thamiris Cirelli
1,2,
Ingra G. Nicchio
2,3,
Diego G. Bussaneli
3,
Bárbara R. Silva
2,3,
Rafael Nepomuceno
2,
Silvana R. P. Orrico
2,4,
Joni A. Cirelli
2,
Letícia H. Theodoro
5,
Silvana P. Barros
6 and
Raquel M. Scarel-Caminaga
3,*
1
Department of Dentistry, School of Dentistry, University Center—UNIFAE, São João da Boa Vista 13870-377, SP, Brazil
2
Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University—UNESP, Araraquara 14801-903, SP, Brazil
3
Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, School of Dentistry at Araraquara, São Paulo State University—UNESP, Araraquara 14801-903, SP, Brazil
4
Advanced Research Center in Medicine, Union of the Colleges of the Great Lakes—UNILAGO, São José do Rio Preto 15030-070, SP, Brazil
5
Department of Diagnosis and Surgery, School of Dentistry at Araçatuba, São Paulo State University—UNESP, Araçatuba 16015-050, SP, Brazil
6
Department of Periodontology, School of Dentistry, University of North Carolina at Chapel Hill—UNC, Chapel Hill, NC 27599, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(7), 6760; https://doi.org/10.3390/ijms24076760
Submission received: 15 February 2023 / Revised: 3 March 2023 / Accepted: 5 March 2023 / Published: 5 April 2023

Abstract

:
The peroxisome proliferator-activated receptor gamma (PPARG) gene encodes a transcription factor involved in the regulation of complex metabolic and inflammatory diseases. We investigated whether single nucleotide polymorphisms (SNPs) and haplotypes of the PPARG gene could contribute with susceptibility to develop periodontitis alone or together with type 2 diabetes mellitus (T2DM). Moreover, we evaluated the gene–phenotype association by assessing the subjects’ biochemical and periodontal parameters, and the expression of PPARG and other immune response–related genes. We examined 345 subjects with a healthy periodontium and without T2DM, 349 subjects with moderate or severe periodontitis but without T2DM, and 202 subjects with moderate or severe periodontitis and T2DM. PPARG SNPs rs12495364, rs1801282, rs1373640, and rs1151999 were investigated. Multiple logistic regressions adjusted for age, sex, and smoking status showed that individuals carrying rs1151999-GG had a 64% lower chance of developing periodontitis together with T2DM. The CCGT haplotype increased the risk of developing periodontitis together with T2DM. The rs1151999-GG and rs12495364-TC were associated with reduced risk of obesity, periodontitis, elevated triglycerides, and elevated glycated hemoglobin, but there was no association with gene expression. Polymorphisms of the PPARG gene were associated with developing periodontitis together with T2DM, and with obesity, lipid, glycemic, and periodontal characteristics.

1. Introduction

Periodontitis is a complex chronic immunoinflammatory disease of the oral cavity caused by dysbiosis of the oral microbiota [1]. It is characterized by a broad spectrum of systemic implications, including increased cardiovascular risk [2], and often coexists with several chronic metabolic diseases, such as T2DM, obesity, and metabolic syndrome [3]. T2DM is a complex endocrine and metabolic disease caused by defects in insulin secretion or insulin resistance due to genetic and/or environmental factors [3]. An important role in the evolution of many inflammatory diseases is attributed to reactive oxygen species (ROS), and changes in the activity of antioxidants in periodontal disease are influenced by systemic conditions. Furthermore, oxidative status markers and proinflammatory biomarkers are modified in the saliva of diabetic patients [4].
The peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors involved in a wide variety of regulatory functions. Dysregulation of PPAR-gamma (PPAR-γ) is linked to the development of complex diseases, such as obesity, type 2 diabetes mellitus (T2DM), atherosclerosis, osteoporosis [5], and Alzheimer’s disease [6]. PPAR-γ activation was reported to downregulate periodontal bone resorption [7], as mentioned by Schaefer et al., 2013 [8]; PPAR-γ was also implicated in the pathology of numerous co-morbidities of periodontitis, including obesity, diabetes and atherosclerosis [8]. Obese mice infected by Porphyromonas gingivalis showed the expression of PPAR-γ upregulated in the liver [9]; biopsies of gingiva from healthy individuals’ samples also had higher PPAR-γ expressions [10]. PPAR-γ agonist rosiglitazone controls blood glucose and has the ability to downregulate proinflammatory cytokine secretion through activation of PPAR-γ receptors, while also maintaining intracellular homeostasis by increasing insulin sensitivity, reducing oxidative stress, and suppressing inflammation [11]. T2DM, obesity, and periodontitis are strongly linked, and the mechanism that can explain this link is inflammation [12,13]. The inflammation underlying the relationship between periodontal disease and T2DM is mainly due to the individual’s response to chronic oral infection, which is influenced by her/his genetic makeup [14].
Single nucleotide polymorphisms (SNPs) represent the most common type of genetic variations among individuals. There is a lot of information about the genetic background of T2DM, including the high degree of polygenicity and the very small effect sizes of most genetic risk variants [15], but there is scarce evidence on the genetic association of some SNPs with susceptibility to develop periodontitis together with T2DM [14,16,17]. Given the identification of novel loci associated with common diseases such as T2DM, it is important to investigate their translation, despite several obstacles that complicate these kind of studies [15].
PPAR-γ plays a prominent role in regulating inflammatory reactions. In particular, it influences the differentiation of monocytes and attenuates the expression of pro-inflammatory mediators (e.g., tumor necrosis factor alpha [TNF-a], interleukin [IL]-1β, and IL-6 [18]). In this context, and because of the biological links between T2DM and periodontitis, as well as the many pieces of evidence pointing to rs1801282 SNP as a risk marker of T2DM [19,20], we hypothesize that this SNP and others in the PPARG gene might be markers of susceptibility to periodontitis together with T2DM. TagSNPs on the haplotype blocks of the PPARG gene evidenced by the data of validated SNPs from CEU + TSI were searched on the minor allele frequencies (MAF) higher than 0.05 in these CEU and TSI populations (CEU = individuals residing in Utah with Western and Northern European ancestry, and TSI = individuals from Tuscany in Italy). These data were obtained from the International HapMap Project database (www.hapmap.org). The selected tagSNPs were rs12495364, rs1373640, and rs1151999, in the respective first, third and fourth main haplotype blocks of the PPARG gene. For the selection of SNPs in the PPARG gene, we were also interested in those reported as potentially clinically relevant, as observed for the rs1151999 SNP that was associated with protection against Alzheimer’s disease [6]. In line with this prerogative, in the second haplotype block, we chose to investigate the rs1801282 SNP, which is a well-established risk marker of T2DM [19,20], and a relatively common SNP.
Despite the known association between periodontitis, T2DM and PPARG, and its biological importance in glucose metabolism and in the inflammatory response, nothing has been reported regarding the PPARG genetic variants (SNPs) in the risk and developing of periodontitis together with T2DM. We investigated whether SNPs and haplotypes of the PPARG gene could contribute, or not, with susceptibility to develop periodontitis, alone or together with T2DM. Moreover, we assessed gene–phenotype associations by using the biochemical, obesity, and periodontal data of subjects. We also evaluated whether circulating expression of PPARG and immune response–related genes are associated with the PPARG gene variants.

2. Results

We observed that the P + T2DM group presented higher visible plaque index than the Periodontitis group, which might contribute with a worse periodontal condition (higher percentage of sites with PPDi ≥ 5 mm and CALi between 4 and 5 mm and ≥6 mm). The P + T2DM group had the worst glycemic and lipid profiles and the physical examination parameters (Table 1).
Table 2 presents multiple logistic regressions adjusted for age, sex, and smoking status. The G allele of rs1151999 (G > T) in the PPARG gene was associated with a lower risk of developing periodontitis together with T2DM. Considering all subjects, those carrying rs1151999-GG had a 64% lower risk of developing periodontitis together with T2DM than individuals carrying the more common TT genotype (OR = 0.36, 95% CI = 0.18–0.73, p = 0.004). When separated by sex, male carriers of rs1151999-GG had an 89% lower risk of developing periodontitis together with T2DM (OR = 0.11, 95% CI = 0.03–0.39, p = 0.001). In the Heathy versus Periodontitis + P + T2DM group comparison, male carriers of rs1151999-GG were confirmed to be at significant lower risk (79%) of developing periodontitis with or without T2DM (OR = 021, 95% CI = 0.07–0.56, p = 0.002). We also found that ever-smokers carrying rs1151999-GG had a markedly lower risk of developing periodontitis together with T2DM (OR = 0.08, 95% CI = 0.02–0.37, p = 0.001) than those ever-smokers carrying rs1151999-TT. In the Periodontitis versus P + T2DM group comparison, besides rs1151999-GG, carriers of rs12495364-TC had about twice the risk to develop periodontitis together with T2DM, whether they are men (OR = 2.54; 95% CI = 1.25–5.19, p = 0.010) that is still significant after Bonferroni correction (p < 0.0125), or never-smokers (OR = 1.87; 95% CI = 1.08–3.21, p = 0.02).
We assessed the gene–phenotype associations with the periodontal, biochemical, and obesity parameters of the subjects. Based on linear regression, both rs1151999-GG and rs12495364-TC were significantly associated with reduced risk of hyperglycemia (elevated HbA1c), hyperlipidemia (elevated triglycerides), obesity (BMI and the waist-to-hip ratio) (Table 3), and periodontal parameters (bleeding on probing and the number of remaining teeth) (Table 4), even after adjustments for age, sex, and smoking status.
We evaluated whether the PPARG SNPs are transmitted together (as haplotypes), and it was confirmed by the strong LD among them (Figure S1B). The most frequent haplotype in the entire population was TCGG (about 33%), followed by TCAT (about 24.8%) (Figure S2). We performed multiple logistic regressions adjusted for age, sex, and smoking status similar to the analysis we performed for each SNP individually. For the Healthy versus P + T2DM group comparison, individuals carrying the CCGT haplotype were twice as likely to develop periodontitis together with T2DM (OR = 2.02; 95% CI = 1.28–3.16, p = 0.0027, Figure S2B). For the Healthy versus Periodontitis and P + T2DM group comparison, the CCGT haplotype was significantly associated with the disease phenotype (OR = 1.73; 95% CI = 1.21–2.48, p = 0.0029, Figure S2C).
Considering the more significant findings related to rs1151999 (Table 2), and that it is in LD with rs1801282 (D’ = 0.758, meaning approximately high LD, Figure S1B), we evaluated whether it could be associated with the circulating leukocyte expression of PPARG, IL6, TNF, IL10 and IL12 genes (Figure 1). Only individuals carrying rs1151999-GT had significant increased IL6 gene expression (Figure 1F, p-value ≤ 0.05) compared with the other genotypes. No significant results were found for the other analyzed genes. Alternatively, we assessed the circulating leukocyte gene expression in each studied group, independently of the rs1151999 genotype, but again, no significant finding was observed.

3. Discussion

Although several studies investigating the role of SNPs in the PPARG gene in T2DM have been published, to our knowledge, this is the first study to investigate the potential contribution of the PPARG SNPs with the occurrence of periodontitis together with T2DM. The most investigated SNP in the PPARG gene regarding the association with T2DM is rs1801282 (Pro12Ala). Many case-control studies enrolling different ancestries have reported that the Pro12Ala (Ala12) variant is associated with protection against T2DM in East Asian (Japanese) [21,22], Greater Middle Eastern [23], and European ancestries such as Finnish [24], Czech [25] and White Scottish [26]. A meta-analysis from 73 studies, involving 62,250 cases and 69,613 controls, provided substantial evidence that the minor allele (G) of rs1801282 is associated with an approximately 18% decreased risk of developing T2DM under different genetic models and in different ethnicities [27]. In agreement, our study found the opposite allele (C) of rs1801282, composing the haplotype CCGT (the underlined C is the rs1801282-C), associated with two-fold odds to develop periodontitis together with T2DM (Figure S2B,C).
There have been few studies of how SNPs in the PPARG gene are related to periodontitis; most have investigated rs1801282. Researchers have shown a significant association between periodontitis and rs1801282 in obese elderly women, indicating a gene–gene or gene–environment interaction between periodontitis and rs1801282 [28]. A study enrolling a group of postmenopausal women with mild periodontitis showed no association between rs1801282 and periodontal clinical parameters [29]. In the present study, we did not find a significant association between rs1801282 and the development of periodontitis or periodontitis together with T2DM, but the rs1151999-GG was associated with a significantly reduced risk of developing periodontitis together with T2DM when considering all subjects or only men (Table 2). Besides the rs1151999-GG, other interesting results were found after stratifying the population by sex: the rs1373640 SNP was related to male carriers of the rs1373640-GA in the Periodontitis group, who showed a 47% lower risk of developing periodontitis (OR = 0.53, 95% CI = 0.30–0.92, p = 0.03), and male carriers of the rs12495364-TC had about twice the risk to develop periodontitis together with T2DM (Table 2). For a while, we were not able to directly explain these gene-phenotype results, but the literature demonstrates that PPAR-γ can influence androgen biosynthesis [30]. The PPAR-γ is a ligand-activated transcription factor expressed in adipose tissue, macrophages and ovaries that influences adipocyte differentiation, insulin sensitivity, and lipid metabolism [5,18]. There are differences in the distribution of adipose tissue between males and females, and the expression of PPAR-γ in perigonadal adipose tissues was significantly higher in females than in males. Further studies are necessary to investigate the functional role of these SNPs in the periodontal disease context, and to verify the possible influence of sex at the transcriptional or translational levels of the encoded proteins.
In the present study, the main significant findings are related to rs11519999-GG: considering the entire population, carriers of that minor allele in homozygosis were 64% less likely to develop periodontitis together with T2DM than carriers of rs11519999-TT. Consistently, carriers of rs1151999-G, together with three other PPARG SNPs, were strongly protected against Alzheimer’s disease when they did not carry the ε4 allele of apolipoprotein E (APOE) [6], indicating a gene–gene interaction. In the additive model, an increase in the minor allele of rs1151999-G marginally reduced the risk of high LDL cholesterol in women [31]. We also employed the additive model and found that rs1151999-GG reduced triglycerides, BMI, the waist-to-hip ratio, and HbA1c (Table 3). In agreement, PPAR-γ also regulates adipogenesis, and the rs1801282 SNP has been suggested to be a risk for obesity, since it was associated with BMI [32]. Moreover, for the first time the rs12495364-TC in the PPARG gene was associated with glycemic, lipid, obesity, and periodontal parameters (Table 3). On the other hand, further studies focusing on the investigation of genetic susceptibility to periodontitis might access whether, for example, the obesity phenotype could partially influence the results.
In addition to the PPARG gene–phenotype associations for T2DM and cardiovascular diseases, researchers have reported a PPARG SNP interaction with diet (fat intake), with a significant influence on bone mineral density (BMD) [33]. While rs1151999-G tends to reduce the risk of high LDL cholesterol [31], in the Framingham Offspring cohort the researchers found that the common allele rs1151999-T interacts with energy derived from dietary fat (p = 0.0004) as well as with the BMD of femoral neck in men [33]. There was a higher risk of vertebral fracture in Danish individuals heterozygous for the rs1151999 SNP (OR = 1.76; 95% CI = 1.18–2.63, p < 0.01) [5]. A recent meta-analysis enrolling 2,157,037 participants demonstrated that osteoporosis patients were more exposed to periodontitis (OR = 1.96; 95% CI, 1.50–2.54) [34]. Osteoporosis was significantly associated with severe alveolar crestal bone loss and the prevalence of periodontitis cases in postmenopausal Jordanian women [35]. In addition, osteoporotic women presented severe periodontitis with greater gingival inflammation, greater clinical attachment level and greater gingival recession than the women with normal BMD [36].
It is not uncommon to produce different results when analyzing SNPs as haplotypes rather than each one separately. Despite the importance of spanning the genetic region under study as much as possible to get better insight into the gene–disease association, only a few studies have focused on haplotypes of the PPARG gene. No previous report has evaluated the same combination of SNPs in the PPARG gene as we have done here. The only study focusing on periodontitis that evaluated haplotypes of the PPARG gene (A-C-C = rs2067819-A, rs1801282-C, and rs3856806-C) demonstrated a slight association with chronic periodontitis (OR = 1.53; 95% CI, 1.01–2.32, p = 0.04) [37]. The TCGG haplotype was most frequent in our population, followed by TCAT. Individuals carrying the CCGT haplotype were twice as susceptible to develop periodontitis together with T2DM, which we confirmed based on the occurrence of periodontitis when comparing the Periodontitis and P + T2DM groups with the Healthy group (OR = 1.73). Further discussion regarding this can be found in the Supplementary material [20,38,39,40,41,42,43,44,45,46,47,48,49,50].
Considering that rs1151999 is associated with the periodontitis together with T2DM phenotype, we examined its potential influence on the host’s inflammatory response. We noted only a tendency of higher TNF, IL10 and IL12 expression in PBMC of carriers of rs1151999-TT regardless of their phenotype, but no influence in the PPARG expression. Further discussion regarding this can be found in the Supplementary material. Again, our study is novel because no other researchers have evaluated the potential influence of this SNP on gene expression. Future studies enrolling more subjects who have been genotyped for this SNP, as well as SNPs in other exonic areas, could more accurately confirm these results.
The present study has a limitation of not investigating individuals solely affected by T2DM. We made this decision because of the high prevalence of periodontitis in individuals with T2DM, which motivated us to evaluate these diseases as comorbidities. Moreover, we were not able to investigate gene expression in a larger number of subjects, nor in gingival biopsies. However, we were interested in PBMC because information for these cells might reveal the potential influence of PPARG variants on the gene expression of circulating leukocytes. This is important because it is easier to assess venous blood at routine medical checkups compared with gingival biopsies. We acknowledge that additional studies are necessary to investigate other SNPs in the PPARG gene and the haplotypes, in larger and ethnically diverse cohorts of individuals affected by only T2DM or periodontitis, as well as by these two diseases as comorbidities, than we evaluated here (even though our experiment was sufficiently powered). It will also be important to verify the potential influence of sex and smoking in those populations, as well as parameters of obesity, such as BMI, and to evaluate the functionality of SNPs, mainly haplotypes, at the transcriptional and translational levels after inflammatory stimuli.

4. Materials and Methods

4.1. Subject Selection

Figure 2 shows the flowchart of the study. From the pool of individuals at the School of Dentistry at Araraquara, São Paulo State University (UNESP), from the southeast region of Brazil, the total number of screened participants was 1158, of which 269 were excluded because of the following exclusion criteria: younger than 30 years; with less than 10 teeth (excluding third molars); with important systemic diseases, such as type 1 diabetes mellitus, hepatitis, HIV (human immunodeficiency virus) infection, and anemia; participants with reports of systemic antibiotics in the previous three months, immunosuppressive chemotherapy, chronic use of anti-inflammatory drugs, pregnant or lactating subjects; or who received periodontal treatment in the previous six months. A total of 896 unrelated participants were selected, and they were required to have some periodontal clinical and biochemical evaluations, as described below.
Each participant completed a questionnaire about their smoking status. We used the answers to classify them as never-smokers or ever-smokers (current smokers + former smokers). Regarding smoking status, participants were classified as a smoker when they reported smoking on a daily basis for at least one year with a mean of 20 cigarettes per day, and a former smoker when they had stopped smoking for at least one year [16,51]. Each participant also self-reported their skin color, as white (67.4%), black (10.83%), brown (19.31%) or mulatto (2.46%). We assessed the clinical periodontal parameters—interproximal probing pocket depth (PPDi), interproximal clinical attachment level (CALi), bleeding on probing (BOP), the visible plaque index, and marginal bleeding—by utilizing a University of North Carolina-15 periodontal probe at four sites (interproximal sites) around each tooth [16,17,51]. We followed the Centers for Disease Control and Prevention/American Academy of Periodontology (CDC/AAP) criteria to define periodontitis [52].
Peripheral blood was collected from all subjects in the morning after they had fasted for at least 8 h. After that, each subject was submitted to the periodontal examination. We used these samples to assess biochemical parameters. We assessed the glycemic profile, including fasting glycemia (using the modified Bondar and Mead method, Labtest Kit), glycated hemoglobin (HbA1c, using the turbidimetric inhibition immunoassay method, Roche), and insulin (using a chemiluminescence method); and the lipid profile, including triglycerides (using the enzymatic Trinder method, Labtest Kit), total cholesterol (using the enzymatic Trinder method, Labtest Kit), high-density lipoprotein cholesterol (HDL, using the enzymatic method, Labtest Kit), very-low-density lipoprotein cholesterol (VLDL), and low-density lipoprotein cholesterol (LDL). We used the Friedewald equation to determine the lipid parameters [53].
The T2DM diagnosis was obtained from their physicians and confirmed by the blood biochemical parameters tested on the day of the periodontal examination, according to the American Diabetes Association (ADA) parameters, fasting plasma glucose concentration ≥ 126 mg/dL (7.0 mmol/L) and/or an HbA1c ≥ 6.5% [54]. Based on the periodontal status and glycemic profile of each individual, we divided the subjects into three groups: 345 individuals with healthy periodontium or mild periodontitis and without T2DM (the Healthy group); 349 individuals with moderate or severe periodontitis and without T2DM (the Periodontitis group); and 202 individuals with moderate or severe periodontitis as well as T2DM (P + T2DM group). The inclusion and exclusion criteria have been described in previous studies [16,17,51].
This study was performed in accordance with the Declaration of Helsinki of 1964, as revised in 1983, and was approved by the Ethics in Human Research Committee of the Araraquara School of Dentistry (UNESP; CAAE 26839019.6.0000.5416), All participants gave oral and written consent to participate in the study. To ensure our experiment was sufficiently powered to evaluate the association of PPARG SNPs with periodontitis and T2DM, we calculated the required sample size by using the G*Power Calculator (version 3.182, Heinrich Heine University, Düsseldorf, Germany). We considered the following parameters: logistic regression; two tails; an odds ratio (OR) of 1.5; Pr (Y = 1/X = 1), H0 = 0.2; α of 0.0125 (0.05 divided by four SNPs); and 80% power, R2 = 0. We found that each group should contain at least 159 subjects.

4.2. DNA Extraction, Polymerase Chain Reaction (PCR) Amplification, and SNP Genotyping

Genomic DNA of each subject, obtained at least 1 h after they had brushed their teeth, was obtained from buccal epithelial cells after a mouthwash with glucose (3%) for 2 min. We extracted the DNA of each subject using standard protocols involving proteinase K digestion, 8 M ammonium acetate, and subsequent ethanol precipitation [55]. We quantified the genomic DNA with a NanoDrop® 2000 spectrophotometer (Thermo Fisher Scientific, Waltham, MA, USA) and a Qubit® 2.0 fluorometer (Thermo Fisher Scientific, Waltham, MA, USA). We used samples showing an A260/280 ratio between 1.7 and 2.0. We genotyped the rs12495364, rs1801282, rs1373640, and rs1151999 SNPs of the PPARG gene with 0.63 μL of the genotyping-specific TaqMan assay (Applied Biosystems, Thermo Fisher Scientific, Waltham, MA, USA) (including forward and reverse primers and VIC- and FAM-labeled probes, detailed in Table S1) as described previously [56].

4.3. Gene-Phenotype Evaluation

To obtain insight into the influence of the rs1151999 SNP on the expression of PPARG and pro- and anti-inflammatory genes, we recalled some individuals carrying each rs1151999 genotype (GG, GT or TT), and repeated the physical, biochemical and periodontal evaluations. Because we were interested in assessing the potential influence of rs1151999 on gene expression, we calculated the sample size with the DDS Research sample size calculator (SPH Analytics, Alpharetta, GA, USA), utilizing gene expression values from a previous study [57]. Based on this calculation, we required at least six subjects carrying each genotype (GG, GT, and TT) to have the ability to detect significant (at the 80% confidence level) differences of 0.3 units in the average gene expression values, with an estimated variation of 0.6 units. Fortunately, we were able to increase from the minimum sample size of 54 subjects to a total of 73 individuals, (Healthy, n = 26; P, n = 29; and P + T2DM, n = 18). At the same time blood was collected from each volunteer for the biochemical analyses, peripheral blood mononuclear cells (PBMC) were obtained by a Histopaque density gradient (Histopaque 1077 over Histopaque 1119, Sigma-Aldrich Co., St. Louis, MO, USA). After centrifugation (700× g for 30 min at room temperature), we washed PBMC twice with saline. Then, we resuspended PBMC in TRIzol and followed the manufacturer’s instructions to isolate total RNA. We determined the concentration and purity of total RNA with a UV microvolume spectrophotometer (Nanovue Plus, GE Health Sciences, Chicago, IL, USA).
We synthesized complementary DNA (cDNA) using 500 ng of total RNA and the High-Capacity Reverse Transcriptase Kit (Applied Biosystems, Foster City, CA, USA) according to the manufacturer’s instructions. We performed real-time quantitative PCR (qPCR) with TaqMan gene expression assays (Applied Biosystems, Thermo Fisher Scientific, Waltham, MA, USA) in duplicate to detect the target genes PPARG (Hs01115513_m1), IL6 (Hs00174131_m1), TNFA (Hs01113624_g1), IL10 (Hs00961622_m1), and IL12 (Hs01073447_m1), and the endogenous control gene GAPDH (Hs02758991_g1). The thermocycling conditions in the StepOne Plus machine (Thermo Fisher Scientific) were: initial denaturation for 10 min at 95 °C, followed by 40 cycles of 15 s at 95 °C and 1 min at 60 °C.

4.4. Statistical Analyses

We have presented demographics, and periodontal, biochemical, and physical parameters, as the median and interquartile range (IQR) for numerical data, and as distribution frequencies for categorical data. We evaluated differences in the frequencies using the Kruskal-Wallis test, followed by Dunn’s test (age, periodontal, biochemical, and physical data), or the chi-square test (sex and smoking data), using SPSS Statistics (version 20, IBM Corp., Armonk, NY, USA) and GraphPad Prism (version 5.01, GraphPad Software, San Diego, CA, USA). To detect differences in the allele and genotype distributions of each SNP between the groups, and to test the Hardy-Weinberg equilibrium, we utilized the PLINK software (version 1.9, Harvard University, Cambridge, MA, USA) [58], as can be viewed in Table S2.
We determined the association between genotypes and disease phenotypes using multivariate logistic regression analyses assuming the additive model and adjusting for age, sex, and smoking status (as confounding factors), when we considered the entire population. We have expressed the effect size as an OR with a 95% confidence interval (CI). Furthermore, we stratified the same analysis (i) by sex, to identify the sex-specific effects of each SNPs on the pathological phenotype, but adjusting multiple logistic regressions by age and smoking; and (ii) by smoking status, adjusting the multiple logistic regressions by age and sex. To assess the gene–clinical–phenotype interrelationship, we performed multiple linear regressions to verify the independent association of each SNP with periodontal parameters (the number of teeth remaining, marginal bleeding, PPDi, and CALi) and with glycemic, lipid, and obesity parameters (fasting glucose, insulin level, HbA1c, triglycerides, total cholesterol, HDL, LDL, body mass index (BMI), and the waist-to-hip ratio). We used STATA (version 12.0, Stata Corporation, College Station, TX, USA) for logistic and linear regressions. We determined experiment-wise significance (p < 0.0125) by using Bonferroni’s correction because we genotyped four SNPs (0.05/4 = 0.0125).
We used the web-based tool SNPStats (www.SNPstats.com) [59] to calculate the linkage disequilibrium (LD) between the investigated SNPs to determine any co-segregation and to estimate the haplotype frequency. We used logistic regressions (adjusted for age, sex, and smoking status) to test associations between haplotypes and diseased phenotypes. We used the Expression Suite software (version 1.1, Applied Biosystems, Foster City, CA, USA) to normalize the cycle threshold (Ct) value of each target gene to GAPDH gene expression, by the 2−ΔCt method. We used the Kruskal-Wallis test followed by the Dunn’s test for statistical comparison (using GraphPad Prism 5.01). We considered p < 0.05 to be significant.

5. Conclusions

In conclusion, our multiple logistic regressions adjusted for age, sex, and smoking status indicate that rs1151999-GG may contribute to reducing the chance to develop periodontitis together with T2DM, while the CCGT haplotype increases this disease phenotype. The rs1151999-GG and rs12495364-TC together are associated with reduced risk of obesity, periodontitis, high triglycerides, and high HbA1c, but there is no association with gene expression in circulating leukocytes.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms24076760/s1. Additional discussion regarding the haplotypes of the PPARG gene and the expression of PPARG and other immune response-related genes in PBMC of subjects can be found in the Supplementary material, with the References [20,38,39,40,41,42,43,44,45,46,47,48,49,50].

Author Contributions

T.C., R.N., I.G.N., D.G.B., B.R.S. and R.M.S.-C. were involved in data collection and data analysis. T.C., R.N., S.P.B. and R.M.S.-C. were involved in data interpretation. T.C., R.N., I.G.N., D.G.B., B.R.S., S.R.P.O., J.A.C., L.H.T., S.P.B. and R.M.S.-C. drafted the manuscript, revised it critically, and have given final approval of the version to be published. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Foundation for Research Support of State of São Paulo—FAPESP, Grants 2014/13295-1, 2016/08070-6, 2016/03753-8, 2016/18313-3 and 2018/26367-1, and National Council for Scientific and Technological Development (CNPq) Grants 445336/2014-5, 304570/2017-6 and 306073/2020-0.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Ethics in Human Research Committee of the Araraquara School of Dentistry (UNESP; CAAE 26839019.6.0000.5416).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Conflicts of Interest

The authors have no relevant conflict of interest to disclose.

References

  1. Kinane, D.F.; Stathopoulou, P.G.; Papapanou, P.N. Periodontal diseases. Nat. Rev. Dis. Primers 2017, 3, 17038. [Google Scholar] [CrossRef] [PubMed]
  2. Carrizales-Sepúlveda, E.F.; Ordaz-Farías, A.; Vera-Pineda, R.; Flores-Ramírez, R. Periodontal Disease, Systemic Inflammation and the Risk of Cardiovascular Disease. Heart Lung Circ. 2018, 27, 1327–1334. [Google Scholar] [CrossRef] [PubMed]
  3. Zhang, S.; Philips, K.H.; Moss, K.; Wu, D.; Adam, H.S.; Selvin, E.; Demmer, R.T.; Norby, F.L.; Mustapha, I.Z.; Beck, J.D. Periodontitis and Risk of Diabetes in the Atherosclerosis Risk In Communities (ARIC) Study: A BMI-Modified Association. J. Clin. Endocrinol. Metab. 2021, 106, e3546–e3558. [Google Scholar] [CrossRef]
  4. Maftei, G.-A.; Martu, M.-A.; Martu, M.-C.; Popescu, D.; Surlin, P.; Tatarciuc, D.; Popa, C.; Foia, L.-G. Correlations between Salivary Immuno-Biochemical Markers and HbA1c in Type 2 Diabetes Subjects before and after Dental Extraction. Antioxidants 2021, 10, 1741. [Google Scholar] [CrossRef] [PubMed]
  5. Harslof, T.; Tofteng, C.L.; Husted, L.B.; Nyegaard, M.; Borglum, A.; Carstens, M.; Stenkjaer, L.; Brixen, K.; Eiken, P.; Jensen, J.E.; et al. Polymorphisms of the peroxisome proliferator-activated receptor gamma (PPARgamma) gene are associated with osteoporosis. Osteoporos Int. 2011, 22, 2655–2666. [Google Scholar] [CrossRef]
  6. Combarros, O.; Rodriguez-Rodriguez, E.; Mateo, I.; Vazquez-Higuera, J.L.; Infante, J.; Berciano, J.; Sanchez-Juan, P. APOE dependent-association of PPAR-gamma genetic variants with Alzheimer’s disease risk. Neurobiol. Aging 2011, 32, 547e1-6. [Google Scholar] [CrossRef]
  7. Hassumi, M.Y.; Silva-Filho, V.J.; Campos-Junior, J.C.; Vieira, S.M.; Cunha, F.Q.; Alves, P.M.; Alves, J.B.; Kawai, T.; Goncalves, R.B.; Napimoga, M.H. PPAR-gamma agonist rosiglitazone prevents inflammatory periodontal bone loss by inhibiting osteoclastogenesis. Int. Immunopharmacol. 2009, 9, 1150–1158. [Google Scholar] [CrossRef]
  8. Schaefer, A.S.; Bochenek, G.; Manke, T.; Nothnagel, M.; Graetz, C.; Thien, A.; Jockel-Schneider, Y.; Harks, I.; Staufenbiel, I.; Wijmenga, C.; et al. Validation of reported genetic risk factors for periodontitis in a large-scale replication study. J. Clin. Periodontol. 2013, 40, 563–572. [Google Scholar] [CrossRef]
  9. Ahn, J.S.; Yang, J.W.; Oh, S.J.; Shin, Y.Y.; Kang, M.J.; Park, H.R.; Seo, Y.; Kim, H.S. Porphyromonas gingivalis exacerbates the progression of fatty liver disease via CD36-PPARgamma pathway. BMB Rep. 2021, 54, 323–328. [Google Scholar] [CrossRef]
  10. Karatas, O.; Yuce, H.B.; Taskan, M.M.; Gevrek, F.; Yarkac, F.U.; Cacan, E. Detection of nuclear receptors in gingival samples of diabetic and nondiabetic periodontitis patients. Niger J. Clin. Pract. 2021, 24, 269–276. [Google Scholar]
  11. Qian, L.; Yin, X.; Lan, T.; Lu, Y. Peroxisome proliferator-activated receptor gamma preserves intracellular homeostasis of insulin-resistant periodontal ligament stem cells. Ann. Transl. Med. 2022, 10, 580. [Google Scholar] [CrossRef] [PubMed]
  12. Hajishengallis, G.; Chavakis, T. Local and systemic mechanisms linking periodontal disease and inflammatory comorbidities. Nat. Rev. Immunol. 2021, 21, 426–440. [Google Scholar] [CrossRef] [PubMed]
  13. Pamuk, F.; Kantarci, A. Inflammation as a link between periodontal disease and obesity. Periodontology 2000 2022, 90, 186–196. [Google Scholar] [CrossRef] [PubMed]
  14. Shi, L.; Zhang, L.; Zhang, D.; Zhou, J.; Jiang, X.; Jin, Y.; Chang, W. Association between TNF-α G-308A (rs1800629) polymorphism and susceptibility to chronic periodontitis and type 2 diabetes mellitus: A meta-analysis. J. Periodontal. Res. 2020, 56, 226–235. [Google Scholar] [CrossRef]
  15. Ingelsson, E.; McCarthy, M.I. Human Genetics of Obesity and Type 2 Diabetes Mellitus: Past, Present, and Future. Circ. Genom. Precis. Med. 2018, 11, e002090. [Google Scholar] [CrossRef] [Green Version]
  16. Cirelli, T.; Nepomuceno, R.; Rios, A.C.S.; Orrico, S.R.P.; Cirelli, J.A.; Theodoro, L.H.; Barros, S.P.; Scarel-Caminaga, R.M. Genetic polymorphisms in the Interleukins IL1B, IL4, and IL6 are associated with concomitant periodontitis and type 2 diabetes mellitus in Brazilian patients. J. Periodontal Res. 2020, 55, 918–930. [Google Scholar] [CrossRef]
  17. Cirelli, T.; Nepomuceno, R.; Goveia, J.M.; Orrico, S.R.P.; Cirelli, J.A.; Theodoro, L.H.; Barros, S.P.; Scarel-Caminaga, R.M. Association of type 2 diabetes mellitus and periodontal disease susceptibility with genome-wide association–identified risk variants in a Southeastern Brazilian population. Clin. Oral Investig. 2021, 25, 3873–3892. [Google Scholar] [CrossRef]
  18. Román-Malo, L.; Bullon, P. Influence of the Periodontal Disease, the Most Prevalent Inflammatory Event, in Peroxisome Proliferator-Activated Receptors Linking Nutrition and Energy Metabolism. Int. J. Mol. Sci. 2017, 18, 1438. [Google Scholar] [CrossRef] [Green Version]
  19. Meirhaeghe, A.; Amouyel, P. Impact of genetic variation of PPARgamma in humans. Mol. Genet. Metab. 2004, 83, 93–102. [Google Scholar] [CrossRef]
  20. Sugawara, K.; Olson, T.S.; Moskaluk, C.A.; Stevens, B.K.; Hoang, S.; Kozaiwa, K.; Cominelli, F.; Ley, K.F.; McDuffie, M. Linkage to peroxisome proliferator-activated receptor-gamma in SAMP1/YitFc mice and in human Crohn’s disease. Gastroenterology 2005, 128, 351–360. [Google Scholar] [CrossRef] [Green Version]
  21. Mori, H.; Ikegami, H.; Kawaguchi, Y.; Seino, S.; Yokoi, N.; Takeda, J.; Inoue, I.; Seino, Y.; Yasuda, K.; Hanafusa, T.; et al. The Pro12 -->Ala substitution in PPAR-gamma is associated with resistance to development of diabetes in the general population: Possible involvement in impairment of insulin secretion in individuals with type 2 diabetes. Diabetes 2001, 50, 891-4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Hara, K.; Okada, T.; Tobe, K.; Yasuda, K.; Mori, Y.; Kadowaki, H.; Hagura, R.; Akanuma, Y.; Kimura, S.; Ito, C.; et al. The Pro12Ala polymorphism in PPAR gamma2 may confer resistance to type 2 diabetes. Biochem. Biophys. Res. Commun. 2000, 271, 212–216. [Google Scholar] [CrossRef] [PubMed]
  23. Motavallian, A.; Andalib, S.; Vaseghi, G.; Mirmohammad-Sadeghi, H.; Amini, M. Association between PRO12ALA polymorphism of the PPAR-gamma2 gene and type 2 diabetes mellitus in Iranian patients. Indian J. Hum. Genet. 2013, 19, 239–244. [Google Scholar] [PubMed] [Green Version]
  24. Douglas, J.A.; Erdos, M.R.; Watanabe, R.M.; Braun, A.; Johnston, C.L.; Oeth, P.; Mohlke, K.L.; Valle, T.T.; Ehnholm, C.; Buchanan, T.A.; et al. The peroxisome proliferator-activated receptor-gamma2 Pro12A1a variant: Association with type 2 diabetes and trait differences. Diabetes 2001, 50, 886–890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Pintérová, D.; Cerná, M.; Kolostová, K.; Novota, P.; Cimburová, M.; Romzová, M.; Kubena, A.; Andel, M. The frequency of alleles of the Pro12Ala polymorphism in PPARgamma2 is different between healthy controls and patients with type 2 diabetes. Folia Biol. 2004, 50, 153–156. [Google Scholar]
  26. Doney, A.S.F.; Fischer, B.; Cecil, J.E.; Boylan, K.; McGuigan, F.E.; Ralston, S.H.; Morris, A.D.; Palmer, C.N.A. Association of the Pro12Ala and C1431T variants of PPARG and their haplotypes with susceptibility to Type 2 diabetes. Diabetologia 2004, 47, 555–558. [Google Scholar] [CrossRef] [Green Version]
  27. Sarhangi, N.; Sharifi, F.; Hashemian, L.; Doabsari, M.H.; Heshmatzad, K.; Rahbaran, M.; Jamaldini, S.H.; Meybodi, H.R.A.; Hasanzad, M. PPARG (Pro12Ala) genetic variant and risk of T2DM: A systematic review and meta-analysis. Sci. Rep. 2020, 10, 12764. [Google Scholar] [CrossRef]
  28. Yoshihara, A.; Sugita, N.; Iwasaki, M.; Wang, Y.; Miyazaki, H.; Yoshie, H.; Nakamura, K. The Interaction Between beta-3 Adrenergic Receptor and Peroxisome Proliferator-Activated Receptor Gamma Gene Polymorphism to Periodontal Disease in Community-Dwelling Elderly Japanese. J. Periodontol. 2015, 86, 955–963. [Google Scholar] [CrossRef]
  29. Wang, Y.; Sugita, N.; Yoshihara, A.; Iwasaki, M.; Miyazaki, H.; Nakamura, K.; Yoshie, H. PPARγ gene polymorphism, C-reactive protein level, BMI and periodontitis in post-menopausal Japanese women. Gerodontology 2014, 33, 44–51. [Google Scholar] [CrossRef]
  30. Watanabe, M.; Inukai, K.; Katagiri, H.; Awata, T.; Oka, Y.; Katayama, S. Regulation of PPAR gamma transcriptional activity in 3T3-L1 adipocytes. Biochem. Biophys. Res. Commun. 2003, 300, 429–436. [Google Scholar] [CrossRef]
  31. Matsunaga, T.; Naito, M.; Yin, G.; Hishida, A.; Okada, R.; Kawai, S.; Sasakabe, T.; Kadomatsu, Y.; Tsukamoto, M.; Kubo, Y.; et al. Associations between peroxisome proliferator-activated receptor gamma (PPAR-gamma) polymorphisms and serum lipids: Two cross-sectional studies of community-dwelling adults. Gene 2020, 762, 145019. [Google Scholar] [CrossRef] [PubMed]
  32. Maeda, A.; Gohda, T.; Funabiki, K.; Horikoshi, S.; Tomino, Y. Peroxisome proliferator-activated receptor gamma gene polymorphism is associated with serum triglyceride levels and body mass index in Japanese type 2 diabetic patients. J. Clin. Lab. Anal. 2004, 18, 317–321. [Google Scholar] [CrossRef]
  33. Ackert-Bicknell, C.L.; Demissie, S.; De Evsikova, C.M.; Hsu, Y.-H.; DeMambro, V.; Karasik, D.; Cupples, L.A.; Ordovas, J.M.; Tucker, K.L.; Cho, K.; et al. PPARG by Dietary Fat Interaction Influences Bone Mass in Mice and Humans. J. Bone Miner. Res. 2008, 23, 1398–1408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Xu, S.; Zhang, G.; Guo, J.F.; Tan, Y.H. Associations between osteoporosis and risk of periodontitis: A pooled analysis of observational studies. Oral Dis. 2021, 27, 357–369. [Google Scholar] [CrossRef] [PubMed]
  35. Al Habashneh, R.; Alchalabi, H.; Khader, Y.; Hazza’A, A.; Odat, Z.; Johnson, G.K. Association Between Periodontal Disease and Osteoporosis in Postmenopausal Women in Jordan. J. Periodontol. 2010, 81, 1613–1621. [Google Scholar] [CrossRef]
  36. Pepelassi, E.; Nicopoulou-Karayianni, K.; Archontopoulou, A.; Mitsea, A.; Kavadella, A.; Tsiklakis, K.; Vrotsos, I.; Devlin, H.; Horner, K. The relationship between osteoporosis and periodontitis in women aged 45-70 years. Oral Dis. 2011, 18, 353–359. [Google Scholar] [CrossRef]
  37. Folwaczny, M.; Manolis, V.; Markus, C.; Glas, J. Variants of the human PPARG locus and the susceptibility to chronic periodontitis. J. Endotoxin Res. 2011, 17, 541–547. [Google Scholar] [CrossRef] [Green Version]
  38. Taskan, M.M.; Gevrek, F. PPAR-γ, RXR, VDR, and COX-2 Expressions in gingival tissue samples of healthy individuals, periodontitis and peri-implantitis patients. Niger. J. Clin. Pract. 2020, 23, 46–53. [Google Scholar]
  39. Di Paola, R.; Briguglio, F.; Paterniti, I.; Mazzon, E.; Oteri, G.; Militi, D.; Cordasco, G.; Cuzzocrea, S. Emerging role of PPAR-beta/delta in inflammatory process associated to experimental periodontitis. Mediat. Inflamm. 2011, 2011, 787159. [Google Scholar] [CrossRef] [Green Version]
  40. Lu, Y.; Zhou, Q.; Shi, Y.; Liu, J.; Zhong, F.; Hao, X.; Li, C.; Chen, N.; Wang, W. SUMOylation of PPARgamma by rosiglitazone prevents LPS-induced NCoR degradation mediating down regulation of chemokines expression in renal proximal tubular cells. PLoS ONE 2013, 8, e79815. [Google Scholar] [CrossRef] [Green Version]
  41. Qiao, W.; Wang, C.; Huang, W.; Hu, Y.; Song, Y. Peroxisome proliferator-activated receptor gamma plays dual roles on experimental periodontitis in rats. J. Clin. Periodontol. 2018, 45, 514–523. [Google Scholar] [CrossRef] [PubMed]
  42. Kukolj, T.; Trivanović, D.; Djordjević, I.O.; Mojsilović, S.; Krstić, J.; Obradović, H.; Janković, S.; Santibanez, J.F.; Jauković, A.; Bugarski, D. Lipopolysaccharide can modify differentiation and immunomodulatory potential of periodontal ligament stem cells via ERK1,2 signaling. J. Cell. Physiol. 2017, 233, 447–462. [Google Scholar] [CrossRef] [PubMed]
  43. Muniandy, S.; Qvist, R.; Ismail, I.; Kaviarasan, S. Gene expression profile in leucocytes of type 2 diabetic subjects. Biomed. Res. India 2009, 20, 158. [Google Scholar] [CrossRef]
  44. Safi, S.Z.; Qvist, R.; Chinna, K.; Ashraf, M.A.; Paramasivam, D.; Ismail, I.S. Gene expression profiling of the peripheral blood mononuclear cells of offspring of one type 2 diabetic parent. Int. J. Diabetes Dev. Ctries. 2015, 36, 407–419. [Google Scholar] [CrossRef]
  45. Sadeghabadi, Z.A.; Nourbakhsh, M.; Alaee, M.; Larijani, B.; Razzaghy-Azar, M. Peroxisome proliferator-activated receptor gamma expression in peripheral blood mononuclear cells and angiopoietin-like protein 4 levels in obese children and adolescents. J. Endocrinol. Investig. 2017, 41, 241–247. [Google Scholar] [CrossRef]
  46. Kim, Y.S.; Lee, H.-M.; Kim, J.K.; Yang, C.-S.; Kim, T.S.; Jung, M.; Jin, H.S.; Kim, S.; Jang, J.; Oh, G.T.; et al. PPAR-α Activation Mediates Innate Host Defense through Induction of TFEB and Lipid Catabolism. J. Immunol. 2017, 198, 3283–3295. [Google Scholar] [CrossRef] [Green Version]
  47. Sharaf, S.A.; Kantoush, N.A.; Ayoub, D.F.; Ibrahim, A.A.; Abdelaal, A.A.; Aziz, R.A.; ElHefnawi, M.M.; Ahmed, A.N. Altered expression of WFS1 and NOTCH2 genes associated with diabetic nephropathy in T2DM patients. Diabetes Res. Clin. Pr. 2018, 140, 304–313. [Google Scholar] [CrossRef]
  48. Liu, B.; Tan, P. PPAR gamma/TLR4/TGF-beta1 axis mediates the protection effect of erythropoietin on cyclosporin A-induced chronic nephropathy in rat. Ren. Fail. 2020, 42, 216–224. [Google Scholar] [CrossRef]
  49. Oliver, P.; Reynés, B.; Caimari, A.; Palou, A. Peripheral blood mononuclear cells: A potential source of homeostatic imbalance markers associated with obesity development. Pflugers Arch. 2013, 465, 459–468. [Google Scholar] [CrossRef]
  50. Pniewska, E.; Sokolowska, M.; Kupryś-Lipińska, I.; Kacprzak, D.; Kuna, P.; Pawliczak, R. Exacerbating Factors Induce Different Gene Expression Profiles in Peripheral Blood Mononuclear Cells from Asthmatics, Patients with Chronic Obstructive Pulmonary Disease and Healthy Subjects. Int. Arch. Allergy Immunol. 2014, 165, 229–243. [Google Scholar] [CrossRef]
  51. Cirelli, T.; Nepomuceno, R.; Orrico, S.R.P.; Rossa, C.; Cirelli, J.A.; North, K.E.; Graff, M.; Barros, S.P.; Scarel-Caminaga, R.M. Validation in a Brazilian population of gene markers of periodontitis previously investigated by GWAS and bioinformatic studies. J. Periodontol. 2020, 92, 689–703. [Google Scholar] [CrossRef] [PubMed]
  52. Page, R.C.; Eke, P.I. Case Definitions for Use in Population-Based Surveillance of Periodontitis. J. Periodontol. 2007, 78, 1387–1399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Nordestgaard, B.G.; Langsted, A.; Mora, S.; Kolovou, G.; Baum, H.; Bruckert, E.; Watts, G.F.; Sypniewska, G.; Wiklund, O.; Borén, J.; et al. Fasting is not routinely required for determination of a lipid profile: Clinical and laboratory implications including flagging at desirable concentration cut-points—A joint consensus statement from the European Atherosclerosis Society and European Federation of Clinical Chemistry and Laboratory Medicine. Eur. Heart J. 2016, 37, 1944–1958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. American Diabetes, A. 2. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes-2019. Diabetes Care 2019, 42 (Suppl. 1), S13–S28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Aidar, M.; Line, S.R.P. A simple and cost-effective protocol for DNA isolation from buccal epithelial cells. Braz. Dent. J. 2007, 18, 148–152. [Google Scholar] [CrossRef] [Green Version]
  56. da Silva, B.R.; Cirelli, T.; Nepomuceno, R.; Theodoro, L.H.; Orrico, S.R.; Cirelli, J.A.; Barros, S.P.; Scarel-Caminaga, R.M. Functional haplotype in the Interleukin8 (CXCL8) gene is associated with type 2 Diabetes Mellitus and Periodontitis in Brazilian population. Diabetes Metab. Syndr. Clin. Res. Rev. 2020, 14, 1665–1672. [Google Scholar] [CrossRef] [PubMed]
  57. Anovazzi, G.; de Medeiros, M.C.; Pigossi, S.C.; Finoti, L.S.; Mayer, M.P.A.; Rossa, C.; Scarel-Caminaga, R.M. Functional Haplotypes in Interleukin 4 Gene Associated with Periodontitis. PLoS ONE 2017, 12, e0169870. [Google Scholar] [CrossRef] [Green Version]
  58. Purcell, S.; Neale, B.; Todd-Brown, K.; Thomas, L.; Ferreira, M.A.R.; Bender, D.; Maller, J.; Sklar, P.; de Bakker, P.I.W.; Daly, M.J.; et al. PLINK: A Tool Set for Whole-Genome Association and Population-Based Linkage Analyses. Am. J. Hum. Genet. 2007, 81, 559–575. [Google Scholar] [CrossRef] [Green Version]
  59. Solé, X.; Guinó, E.; Valls, J.; Iniesta, R.; Moreno, V. SNPStats: A web tool for the analysis of association studies. Bioinformatics 2006, 22, 1928–1929. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Expression of the PPARG, IL6, TNF, IL10, and IL12 genes. The mean and standard error of 2−ΔCt expression values of each gene after normalization by the GAPDH. In the first column (A,D,G,J,M), the subjects are divided according to the rs1151999 genotype, namely GG, GT, or TT, and the phenotype (Healthy, periodontitis [P], or periodontitis together with type 2 diabetes mellitus [P + T2DM], named Groups. In the second column (B,E,H,K,N), the subjects are divided based only on their genotypes, irrespective of the phenotypic classification. In the third column (C,F,I,L,O), the expression of each target gene is presented only according to the subjects’ phenotype, independently of the rs1151999 genotype. Comparisons among groups were made with the Kruskal-Wallis test, followed by Dunn’s test. * p-value ≤ 0.05.
Figure 1. Expression of the PPARG, IL6, TNF, IL10, and IL12 genes. The mean and standard error of 2−ΔCt expression values of each gene after normalization by the GAPDH. In the first column (A,D,G,J,M), the subjects are divided according to the rs1151999 genotype, namely GG, GT, or TT, and the phenotype (Healthy, periodontitis [P], or periodontitis together with type 2 diabetes mellitus [P + T2DM], named Groups. In the second column (B,E,H,K,N), the subjects are divided based only on their genotypes, irrespective of the phenotypic classification. In the third column (C,F,I,L,O), the expression of each target gene is presented only according to the subjects’ phenotype, independently of the rs1151999 genotype. Comparisons among groups were made with the Kruskal-Wallis test, followed by Dunn’s test. * p-value ≤ 0.05.
Ijms 24 06760 g001
Figure 2. Flowchart of the study.
Figure 2. Flowchart of the study.
Ijms 24 06760 g002
Table 1. Comparison of demographic characteristics, and periodontal, biochemical, and physical parameters among the groups.
Table 1. Comparison of demographic characteristics, and periodontal, biochemical, and physical parameters among the groups.
Demographic CharacteristicsHealthyPeriodontitisPeriodontitis + T2DM
n = 345n = 349n = 202
Age—Median (IQR)41.0 (35.0–50.0) a49.0 (43.0–55.0) b56.0 (50.0–62.0) c
Sex n (%) *
Males114 (32.5)143 (40.9)85 (42.1)
Females233 (67.5)206 (59.1)117 (57.9)
Smoking habits (%) *
Never268 (77.7)224 (64.2)114 (56.4)
Ever77 (22.3)125 (35.8)88 (43.6)
Periodontal Parameters—Median (IQR)
Number of teeth27.0 (24.0–28.0) a24.0 (20.0–26.0) b21.0 (16.0–25.0) c
Visible plaque (% of sites)20.4 (8.2–36.6) a43.4 (22.0–67.9) b56.7 (36.5–77.5) c
Marginal bleeding (% of sites)4.2 (0.0–11.8) a11.0 (3.1–28.3) b36.5 (21.8–52.8) c
BOP (% of sites)1.0 (0.0–4.7) a37.0 (20.6–55.6) b52.3 (28.1–72.4) c
PPDi ≤ 4 mm (% of sites)100.0 (100.0–100.0) a91.3 (80.9–97.0) b87.5 (68.4–97.8) b
PPDi ≥ 5 mm (% of sites)0.0 (0.0–0.0) a8.6 (2.9–19.1) b12.5 (2.2–31.6) b
CALi ≤ 3 mm (% of sites)110.0 (99.1–100.0) a72.4 (55.5–85.4) b58.3 (33.3–80.4) c
CALi = 4–5 mm (% of sites)0.0 (0.0–0.93) a17.9 (10.1–28.3) b24.1 (11.2–34.8) b
CALi ≥ 6 mm (% of sites)0.0 (0.0–2.1) a6.3 (2.1–15.4) b11.1 (2.5–28.8) b
Biochemical and Physical data—Median (IQR)
Fasting blood glucose (mg/dL)92.0 (86.0–96.0) a94.5 (89.0–99.0) a153.1 (118.0–217.0) b
HbA1c (percentage)5.4 (5.2–5.6) a5.6 (5.3–5.6) a7.9 (6.6–9.7) b
Insulin (UI/mL)7.4 (3.6–37.8) a9.9 (4.1–32.4) a13.8 (3.2–161.0) b
Total cholesterol (mg/dL)174.0 (152.0–194.0) a189.0 (160.5–208.5) a,b191.0 (160.0–227.0) b
HDL cholesterol (mg/dL)54.0 (40.0–61.0) a51.5 (42.0–60.2) a44.0 (37.0–55.5) b
LDL cholesterol (mg/dL)97.0 (78.0–121.2) a98.4 (79.4–116.0) a108.2 (70.9–142.5) a
Triglycerides (mg/dL)105.0 (81.0–156.0) a138.5 (94.5–203.0) b155.0 (105.8–240.3) b
BMI (kg/m2)27.1 (22.4–29.7) a26.2 (24.2–29.5) a29.5 (26.3–33.4) b
Waist/hip ratio (cm)0.87 (0.78–0.95) a0.89 (0.83–0.97) a0.95 (0.91–1.0) b
Abbreviations: BOP, bleeding on probing; CALi, interproximal clinical attachment level; IQR, interquartile range (25th percentile to 75th percentile); P, periodontitis; PPDi, interproximal probing pocket depth. a, b, c = different letters indicate significant differences among groups (p < 0.05). * Significant difference in sex and smoking status distribution between groups (p < 0.0001). Categorical sex and smoking status intergroup comparisons were made using the chi-square test. Age, and periodontal, biochemical, and physical parameters, were analyzed by the Kruskal–Wallis test, followed by Dunn’s test.
Table 2. Multiple logistic regressions adjusted for age, sex, and smoking status and stratified by sex and smoking status.
Table 2. Multiple logistic regressions adjusted for age, sex, and smoking status and stratified by sex and smoking status.
PPARG SNPHealthy vs. PeriodontitisHealthy vs. P + T2DMPeriodontitis vs. P + T2DMHealthy vs. Periodontitis + P + T2DM
rs12495364 (T > C)
All subjects Adjusted OR Adjusted OR Adjusted OR Adjusted OR
TTRef Ref
TC0.75 (0.56–1.05)0.101.53 (0.94–2.47)0.081.50 (0.96–2.33)0.071.57 (0.04–2.36)0.05
CC0.62 (0.36–1.08)0.091.73 (0.79–3.80)0.171.51 (0.76–3.02)0.241.67 (0.87–3.19)0.12
Male Adjusted OR Adjusted OR Adjusted OR Adjusted OR
CCRef Ref Ref Ref
TC0.62 (0.35–1.12)0.112.13 (0.89–5.05)0.092.54 (1.25–5.19)0.010 *2.29 (1.18–4.44)0.014
TT0.84 (0.33–2.14)0.721.24 (0.32–4.84)0.751.86 (0.62–5.58)0.271.73 (0.61–4.90)0.29
Female
CCRef Ref Ref Ref
TC1.02 (0.67–1.55)0.901.32 (0.73–2.39)0.351.07 (0.61–1.88)0.811.24 (0.74–2.09)0.40
TT1.42 (0.65–3.12)0.382.14 (0.79–5.75)0.131.35 (0.56–3.31)0.511.67 (0.73–3.84)0.22
Never Smoking §Adjusted OR § Adjusted OR § Adjusted OR § Adjusted OR§
CCRef Ref Ref Ref
TC0.78 (0.53–1.17)0.241.70 (0.96–3.01)0.071.87 (1.08–3.21)0.021.87 (1.13–3.09)0.02
TT1.11 (0.54–2.27)0.772.04 (0.82–5.07)0.131.94 (0.83–4.47)0.122.17 (0.99–3.09)0.20
Ever Smoking §
CCRef Ref Ref Ref
TC1.01 (0.57–2.12)0.761.32 (0.52–3.41)0.561.12 (0.51–2.48)0.771.23 (0.59–2.55)0.57
TT1.25 (0.42–3.76)0.691.35 (0.25–7.16)0.731.05 (0.28–3.86)0.941.03 (0.30–3.59)0.95
rs1801282 (C > G)
All subjects Adjusted OR Adjusted OR Adjusted OR Adjusted OR
CCRef Ref Ref Ref
CG0.75 (0.48–1.15)0.190.77 (0.43–1.39)0.390.97 (0.56–1.68)0.920.87 (0.53–1.45)0.60
GG0.27 (0.04–1.62)0.151.01 (0.21–4.84)0.993.46 (0.61–19.74)0.161.68 (0.44–6.51)0.45
Male Adjusted OR Adjusted OR Adjusted OR Adjusted OR
CCRef Ref Ref Ref
CG0.52 (0.26–1.02)0.060.64 (0.25–1.61)0.341.51 (0.63–3.59)0.351.08 (0.49–2.36)0.84
GGempty 0.48 (0.04–6.47)0.58empty 2.36 (0.19–29.28)0.50
Female
CCRef Ref Ref Ref
CG0.99 (0.57–1.74)0.990.92 (0.43–1.99)0.840.72 (0.34–1.50)0.380.79 (0.39–1.53)0.47
GG0.53 (0.08–3.28)0.491.28 (0.19–8.34)0.791.92 (0.26–14.22)0.521.36 (0.25–7.37)0.71
Never Smoking §Adjusted OR § Adjusted OR § Adjusted OR § Adjusted OR §
CCRef Ref Ref Ref
CG0.70 (0.42–1.17)0.180.83 (0.42–1.67)0.611.07 (0.55–2.08)0.841.01 (0.55–2.83)0.98
GG0.25 (0.02–2.42)0.231.66 (0.30–9.06)0.565.89 (0.59–58.4)0.132.69 (0.57–12.64)0.21
Ever Smoking §
CCRef Ref Ref Ref
CG0.87 (0.39–1.93)0.740.71 (0.23–2.15)0.540.95 (0.34–2.71)0.940.72 (0.28–1.84)0.49
GG0.35 (0.02–6.43)0.510.16 (0.007–3.04)0.221.17 (0.06–22.51)0.910.43 (0.03–6.04)0.53
rs1373640 (A > G)
All subjects Adjusted OR Adjusted OR Adjusted OR Adjusted OR
GGRef Ref
GA0.72 (0.87–0.99)0.050.74 (0.47–1.16)0.191.01 (0.67–1.53)0.950.85 (0.58–1.25)0.42
AA1.11 (0.55–2.26)0.760.87 (0.33–2.29)0.770.98 (0.41–2.33)0.960.95 (0.42–2.11)0.89
Male Adjusted OR Adjusted OR Adjusted OR Adjusted OR
GGRef Ref Ref Ref
GA0.53 (0.30–0.92)0.030.87 (0.38–1.97)0.751.30 (0.66–2.56)0.441.16 (0.62–2.19)0.47
AA1.72 (0.56–5.27)0.343.65 (0.81–16.41)0.092.25 (0.70–7.24)0.172.77 (0.92–8.39)0.71
Female
GGRef Ref Ref Ref
GA0.85 (0.57–1.28)0.460.70 (0.40–1.23)0.220.84 (0.49–1.43)0.530.72 (0.44–1.15)0.17
AA0.82 (0.32–2.11)0.680.24 (0.04–1.25)0.090.35 (0.0–1.73)0.200.28 (0.06–1.28)0.20
Never Smoking §Adjusted OR § Adjusted OR § Adjusted OR § Adjusted OR§
GGRef Ref Ref Ref
GA0.76 (0.51–1.12)0.160.63 (0.37– 1.09)0.100.75 (0.44–1.26)0.280.65 (0.40–1.06)0.09
AA0.94 (0.41–2.11)0.860.40 (0.11–1.44) 0.160.68 (0.21–2.23)0.530.59 (0.21–1.77)0.35
Ever Smoking §
GGRef Ref Ref Ref
GA0.62 (0.34–1.16)0.140.99 (0.43–2.27)0.981.67 (0.81–3.42)0.161.31 (0.68–2.53)0.41
AA1.99 (0.39–10.01)0.404.62 (0.72–29.66)0.112.19 (0.57–8.43)0.252.24 (0.64–7.87)0.21
rs1151999 (G > T)
All subjects Adjusted OR Adjusted OR Adjusted OR Adjusted OR
TTRef Ref
TG0.69 (0.49–0.99)0.050.72 (0.44–1.15) 0.170.93 (0.61–1.42)0.760.77 (0.52–1.14)0.20
GG0.68 (0.43–1.1.07) 0.100.36 (0.18–0.73)0.004 *0.67 (0.36–1.24)0.210.49 (0.27–1.19)0.20
Male Adjusted OR Adjusted OR Adjusted OR Adjusted OR
TTRef Ref Ref Ref
TG0.56 (0.29–1.04)0.070.35 (0.14–0.87)0.020.51 (0.25–1.03)0.060.42 (0.21–0.82)0.02
GG0.74 (0.35–1.60)0.350.11 (0.03–0.39)0.001 *0.28 (0.11–0.77)0.0140.21 (0.07–0.56)0.002 *
Female
TTRef Ref Ref Ref
TG0.77 (0.50–1.19)0.250.99 (0.56–1.76)0.981.29 (0.79–2.23)0.351.08 (0.65–1.78)0.75
GG0.59 (0.33–1.07)0.080.63 (0.28–1.43)0.271.21 (0.54–2.73)0.640.83 (0.40–1.73)0.63
Never Smoking §Adjusted OR § Adjusted OR § Adjusted OR § Adjusted OR §
TTRef Ref Ref Ref
TG0.77 (0.51–1.15)0.200.94 (0.53–1.65)0.820.97 (0.56–1.64)0.890.85 (0.52–1.40)0.54
GG0.61 (0.35–1.07)0.090.59 (0.27–1.30)0.191.27 (0.58–2.78)0.590.87 (0.43–1.75)0.71
Ever Smoking §
TTRef Ref Ref Ref
TG0.54 (0.27–1.10)0.090.32 (0.12–0.82)0.020.77 (0.36–1.64)0.490.59 (0.29–1.18)0.14
GG0.79 (0.34–1.82)0.590.08 (0.02–0.37)0.001 *0.20 (0.06–0.62)0.005 *0.16 (0.05–0.47)0.001 *
Abbreviations: P + T2DM = periodontitis together with type 2 diabetes mellitus; Adj OR (95% CI) = Adjusted odds ratio (95% confidence interval); Ref = reference or more frequent single nucleotide polymorphism. Never smokers = subjects who never smoked; ever smokers = current smokers + former smokers. * Indicates statistical significance after Bonferroni correction (p < 0.0125). ORs with 95% CIs were estimated by multiple logistic regression models after controlling for age, sex, and smoking status. ORs with 95% CIs were estimated by multiple logistic regression models after controlling for age and smoking status. § ORs with 95% CIs were estimated by multiple logistic regression models after controlling for age and sex. Bold font indicates p < 0.05.
Table 3. Multiple linear regression analysis of all subjects for effects of single nucleotide polymorphisms on glycemic, lipid, and obesity parameters.
Table 3. Multiple linear regression analysis of all subjects for effects of single nucleotide polymorphisms on glycemic, lipid, and obesity parameters.
PPARG SNPFasting GlucoseInsulinHbA1cTriglyceridesTotal CholesterolHDLLDLBMI (kg/m2)Waist-To-Hip Ratio (cm)
rs12495364Adjusted β (95% CI)p-ValueAdjusted β (95% CI)p-ValueAdjusted β (95% CI)p-ValueAdjusted β (95% CI)p-ValueAdjusted β (95% CI)p-ValueAdjusted β (95% CI)p-ValueAdjusted β (95% CI)p-ValueAdjusted β (95% CI)p-ValueAdjusted β (95% CI)p-Value
TTRef Ref Ref Ref Ref Ref Ref Ref Ref
TC−0.14
(−0.26–−0.03)
0.008−0.14
(−0.28–−0.01)
0.03−0.14
(−0.25–−0.03)
0.01−0.13
(−0.25–−0.02)
0.01−0.12
(−0.23–−0.01)
0.03−0.11
(−0.23–−0.001)
0.04−0.12
(−0.24–−0.001)
0.04−0.14
(−0.25–−0.03)
0.01−0.25
(−0.38–−0.11)
0.000 *
CC−0.01
(−0.25–0.21)
0.880.19
(−0.26–0.29)
0.89−0.06
(−0.29–0.16)
0.58−0.04
(−0.28–0.20)
0.740.02
(−0.22–0.27)
0.840.006
(−0.24–0.26)
0.950.03
(−0.22–0.29)
0.80−0.02
(−0.27–0.21)
0.82−0.01
(−0.28–0.25)
0.88
rs1801282
CCRef Ref Ref Ref Ref Ref Ref Ref Ref
CG0.02
(−0.07–0.13)
0.58−0.002
(−0.13–0.13)
0.970.004
(−0.09–0.10)
0.920.01
(−0.08–0.12)
0.710.02
(−0.08–0.12)
0.690.01
(−0.09–0.12)
0.750.008
(−0.10–0.12)
0.880.02
(−0.07–0.12)
0.68−0.01
(−0.14–0.11)
0.80
GG0.21
(0.01–0.41)
0.030.14
(−0.09–0.39)
0.230.17
(−0.01–0.37)
0.070.13
(−0.07–0.34)
0.200.17
(−0.03–0.38)
0.100.17
(−0.03–0.39)
0.100.17
(−0.05–0.40)
0.130.20
(−0.01–0.43)
0.070.24
(−0.04–0.53)
0.09
rs1373640
GGRef Ref Ref Ref Ref Ref Ref Ref Ref
GA0.01
(−0.09–0.12)
0.74−0.04
(−0.17–0.09)
0.56−0.001
(−0.10–0.10)
0.98−0.03
(−0.13–0.07)
0.57−0.03
(−0.14–0.07)
0.55−0.02
(−0.13–0.08)
0.66−0.01
(−0.12–0.10)
0.83−0.03
(−0.14–0.06)
0.48−0.04
(−0.18–0.09)
0.52
AA0.01
(−0.33–0.36)
0.92−0.03
(−0.53–0.46)
0.890.04
(−0.29–0.37)
0.81−0.01
(−0.37–0.34)
0.930.02
(−0.34–0.38)
0.900.01
(−0.36–0.38)
0.95−0.01
(−0.43–0.40)
0.940.005
(−0.33–0.35)
0.97−0.10
(−0.59–0.38)
0.68
rs1151999
TTRef Ref Ref Ref Ref Ref Ref Ref Ref
TG−0.005
(−0.11–0.10)
0.92−0.07
(−0.22–0.07)
0.30−0.04
(−0.14–0.06)
0.38−0.07
(−0.19–0.06)
0.31−0.04
(−0.16–0.09)
0.63−0.03
(−0.16–0.10)
0.65−0.04
(−0.18–0.09)
0.53−0.09
(−0.21–0.04)
0.17−0.14
(−0.29–0.01)
0.07
GG−0.07
(−0.22–0.08)
0.34−0.11
(−0.31–0.08)
0.28−0.15
(−0.28–−0.003)
0.04−0.19
(−0.37–−0.02)
0.03−0.16
(−0.34–0.02)
0.08−0.17
(−0.35–0.02)
0.07−0.16
(−0.34–0.03)
0.10−0.21
(−0.39–0.04)
0.02−0.23
(−0.44–0.02)
0.03
Abbreviations: β, regression coefficient; BMI, body mass index; CI, confidence interval; HbA1C, glycated hemoglobin; HDL, high-density lipoprotein; LDL, low-density lipoprotein. * Indicates statistical significance after Bonferroni correction (p < 0.0125). β the regression coefficient with 95% CI was estimated by multiple linear regression models after controlling for age, sex and smoking. Bold font indicates p < 0.05.
Table 4. Multiple linear regression analysis of all subjects for effects of single nucleotide polymorphisms on periodontal parameters.
Table 4. Multiple linear regression analysis of all subjects for effects of single nucleotide polymorphisms on periodontal parameters.
PPARG SNPNumber of Remaining TeethBleeding on Marginal Probing (% of Sites)Bleeding on Pocket Probing (% of Sites)PPDi ≥ 5 mm
(% of Sites)
CALi ≥ 6 mm
(% of Sites)
rs12495364Adjusted β * (95% CI)p-valueAdjusted β * (95% CI)p-ValueAdjusted β * (95% CI)p-ValueAdjusted β * (95% CI)p-ValueAdjusted β * (95% CI)p-Value
TTRef Ref Ref Ref Ref
TC−0.14
(−0.26–−0.03)
0.008−0.14
(−0.28–−0.01)
0.03−0.14
(−0.25–−0.03)
0.01−0.13
(−0.25–−0.02)
0.01−0.12
(−0.23–−0.01)
0.03
CC−0.01
(−0.25–0.21)
0.880.19
(−0.26–0.29)
0.89−0.06
(−0.29–0.16)
0.58−0.04
(−0.28–0.20)
0.740.02
(−0.22–0.27)
0.84
rs1801282
CCRef Ref Ref Ref Ref
CG0.02
(−0.07–0.13)
0.58−0.002
(−0.13–0.13)
0.970.004
(−0.09–0.10)
0.920.01
(−0.08–0.12)
0.710.02
(−0.08–0.12)
0.69
GG0.21
(0.01–0.41)
0.030.14
(−0.09–0.39)
0.230.17
(−0.01–0.37)
0.070.13
(−0.07–0.34)
0.200.17
(−0.03–0.38)
0.10
rs1373640
GGRef Ref Ref Ref Ref
GA0.01
(−0.09–0.12)
0.74−0.04
(−0.17–0.09)
0.56−0.001
(−0.10–0.10)
0.98−0.03
(−0.13–0.07)
0.57−0.03
(−0.14–0.07)
0.55
AA0.01
(−0.33–0.36)
0.92−0.03
(−0.53–0.46)
0.890.04
(−0.29–0.37)
0.81−0.01
(−0.37–0.34)
0.930.02
(−0.34–0.38)
0.90
rs1151999
TTRef Ref Ref Ref Ref
TG−0.03
(−0.09–0.02)
0.25−0.006
(−0.07–0.05)
0.83−0.01
(−0.06–0.04)
0.63−0.02
(−0.07–0.04)

0.54
−0.01
(−0.07–0.04)
0.59
GG−0.07
(−0.15–−0.002)
0.04−0.08
(−0.16–−0.12)
0.02−0.06
(−0.13–−0.01)
0.09−0.07
(−0.14–−0.008)
0.08−0.06
(−0.14–0.009)
0.09
Abbreviations: β, regression coefficient; CALi, interproximal clinical attachment level; CI, confidence interval; PPDi, interproximal probing pocket depth. * β the regression coefficient with 95% CI was estimated by multiple linear regression models after controlling for age, sex, and smoking status. Bold font indicates p < 0.05.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cirelli, T.; Nicchio, I.G.; Bussaneli, D.G.; Silva, B.R.; Nepomuceno, R.; Orrico, S.R.P.; Cirelli, J.A.; Theodoro, L.H.; Barros, S.P.; Scarel-Caminaga, R.M. Evidence Linking PPARG Genetic Variants with Periodontitis and Type 2 Diabetes Mellitus in a Brazilian Population. Int. J. Mol. Sci. 2023, 24, 6760. https://doi.org/10.3390/ijms24076760

AMA Style

Cirelli T, Nicchio IG, Bussaneli DG, Silva BR, Nepomuceno R, Orrico SRP, Cirelli JA, Theodoro LH, Barros SP, Scarel-Caminaga RM. Evidence Linking PPARG Genetic Variants with Periodontitis and Type 2 Diabetes Mellitus in a Brazilian Population. International Journal of Molecular Sciences. 2023; 24(7):6760. https://doi.org/10.3390/ijms24076760

Chicago/Turabian Style

Cirelli, Thamiris, Ingra G. Nicchio, Diego G. Bussaneli, Bárbara R. Silva, Rafael Nepomuceno, Silvana R. P. Orrico, Joni A. Cirelli, Letícia H. Theodoro, Silvana P. Barros, and Raquel M. Scarel-Caminaga. 2023. "Evidence Linking PPARG Genetic Variants with Periodontitis and Type 2 Diabetes Mellitus in a Brazilian Population" International Journal of Molecular Sciences 24, no. 7: 6760. https://doi.org/10.3390/ijms24076760

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop