REVIEW ARTICLE |
https://doi.org/10.5005/jp-journals-10015-2353 |
Exploring the Epigenetic Landscape—Insights from Epigenomics in Periodontitis and Stress-related Health Implications: A Review
1,2Department of Periodontology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (Deemed to be University), Chennai, Tamil Nadu, India
3Department of Orthodontics, Sri Sankara Dental College, Vettoor, Kerala, India
Corresponding Author: Smrithi Vishakha Varma, Department of Periodontology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (Deemed to be University), Chennai, Tamil Nadu, India, Phone: +91 9995982747, e-mail: smrithivishakhavarma@gmail.com
Received: 06 December 2023; Accepted: 08 January 2024 Published on: 20 February 2024
ABSTRACT
Background: The emerging field of epigenetics probes into the intricate modifications and information beyond the primary genetic code, investigating how gene function can be altered and inherited across cell generations without changes in deoxyribonucleic acid (DNA) sequences. This review specifically focuses on the epigenetic dimensions within two captivating areas—periodontitis and stress-related health implications.
Aim: The aim of this literature review is to comprehensively delve into current research, shedding light on the interplay between epigenetics, oral health, and overall well-being. It particularly explores the epigenetic intricacies associated with periodontitis and stress-related conditions.
Clinical significance: Despite the genetic complexity of periodontitis, the emergence of epigenetic biomarkers provides hope for early disease diagnosis and personalized management. Epigenetics, with established links to various diseases, including cancer, unfolds new therapeutic possibilities. While challenges persist in unraveling the intricacies of epigenetic regulation, it holds promise for advancing disease eradication, even for historically incurable conditions. Epigenetics stands as a hopeful avenue for treating stress-related disorders, although its full potential is still in the early stages of exploration.
How to cite this article: Varma SV, Varghese SS, Nair SV. Exploring the Epigenetic Landscape—Insights from Epigenomics in Periodontitis and Stress-related Health Implications: A Review. World J Dent 2024;15(1):72–78.
Source of support: Nil
Conflict of interest: None
Keywords: Chronic stress, Deoxyribonucleic acid methylation, Epigenetics, Epigenome, Histone modification, Immune response, Periodontitis.
INTRODUCTION
In the intricate tapestry of human health, the dynamic epigenetic landscape emerges as a pivotal terrain governing the interplay between genes and the environment. The “epigenome” extends beyond the primary genetic code, prompting exploration into how gene functions dynamically pass from one cell generation to the next without altering the deoxyribonucleic acid (DNA) sequence during cell division.1 This exploration, rooted in the scientific discipline of epigenetics, delves into the interplay between genetics and the environment, particularly investigating how epigenetic changes shape variations in localized gene expression associated with inflammation and disease susceptibility among individuals.2
Within this context, periodontitis, characterized by persistent gingival inflammation due to bacterial colonization, stands as a pertinent subject. Understanding the risk factors for periodontitis is crucial for discerning individual susceptibility, encompassing genetic variations, environmental influences, lifestyle choices, and the intricate realm of epigenetics.3 Stress, characterized as a cognitive perception of a lack of control and predictability, becomes a notable player, impacting physiological and behavioral reactions. Prolonged stress episodes can potentially influence inflammatory mechanisms, contributing to various conditions, including periodontal diseases, diabetes, and cardiovascular diseases.4,5
Recognizing the pivotal role of epigenetics in diseases, ranging from cancer to inflammatory conditions,6 this review aims to meticulously assess the current body of research concerning the epigenetic dimensions of periodontitis and stress-related health implications. By critically evaluating existing research, we aspire to identify gaps, highlight emerging trends, and provide a comprehensive overview, thereby guiding future investigations. The synthesis of epigenetic insights into these health conditions holds transformative potential, paving the way for more targeted interventions and personalized health strategies. In addressing the need for this study, we aim to contribute substantively to our understanding of the molecular intricacies of periodontitis and stress-related health conditions, fostering advancements in clinical approaches and personalized health strategies.
Immune System Dynamics: From Innate to Adaptive Responses to Genetic Regulations and Disease Pathways
The immune system orchestrates a complex interplay between innate and adaptive responses to combat invading microorganisms. Foundational components of this intricate defense mechanism encompass both inherent (innate) and acquired (adaptive) factors.7 In response to stressful situations, the anterior hypothalamus activates the hypothalamus–pituitary–adrenal (HPA) axis, initiating a cascade of hormonal events. Corticotropin-releasing hormone and arginine vasopressin are secreted, targeting the pituitary gland, ultimately leading to the release of adrenocorticotrophic hormone and cortisol. Notably, cortisol, a glucocorticoid hormone, plays a crucial role in modulating immune responses.8
Within the realm of immune responses, external triggers, such as bacterial substances, activate signaling pathways in cells. This activation prompts the binding of transcription factors to DNA regions known as promoters and enhancers, initiating the transcription process and activating specific genes. The genetic constitution of an individual contributes significantly to immune response diversity, involving polygenic interactions and concurrent variations at different genetic locations.9 Genetic changes, whether mutations, polymorphisms, or modifications in the DNA sequence, can alter transcription factor binding sites within the DNA’s promoter region, influencing the speed and extent of gene expression.10 The intricate control of gene expression spans chromatin structures, transcription factors, and posttranscriptional alterations, including ribonucleic acid (RNA) splicing, messenger RNA (mRNA) polyadenylation, and the involvement of regulatory molecules such as microRNA and long noncoding RNAs.11 Chronic inflammatory conditions like periodontitis exhibit particular target tissues where inflammation leads to tissue damage.12 This integration of genetic regulations into immune system dynamics underscores the complexity of disease pathways and emphasizes the need for a comprehensive understanding to advance therapeutic interventions. Figure 1 illustrates the complex interplay among external stressors, epigenetic modifications, genetics, immunity, inflammation, and periodontitis, showcasing the influence of stress on the immune response and epigenetic changes.
Fig. 1: The schematic diagram illustrates the intricate interplay between external stressors, epigenetic modifications, genetics, immunity, inflammation, and periodontitis. Stress influences both epigenetic changes and the immune response, while epigenetic modifications contribute to variations in periodontitis susceptibility. The genetic landscape shapes the immune response, and the resulting inflammation becomes a central factor in the development and aggravation of periodontitis
Molecular Choreography: Decoding Epigenetic Mechanisms
Coined by Waddington, “epigenetics” elucidates gene-phenotype cause-and-effect relationships.13,14 Heritable changes in gene expression involve DNA methylation, chromatin modifications, imprinting changes, and noncoding RNA, constituting key mechanisms.15,17 Epigenomes, influenced by stress, diet, and maternal care, dictate molecular changes and cell responsiveness.18 The epigenetic process begins with an “epigenetic initiator,” shaping chromatin interactions and guiding modifications independently of environmental cues.19 Additionally, the “epigenetic maintainer,” represented by histone acetyltransferases and DNA methyltransferases, forms binding site complexes upon the initiator’s activation. These maintainers induce modifications in histone amino acids and DNA cytosines, culminating in distinctive epigenomes that define cellular states and responsiveness to environmental factors.20 The “epigenetic code” concept emerges from specific patterns, their significance, and guiding pathways.21,28 DNA methylation involves gene silencing through modifications on amino acid tails, forming heritable epigenetic marks.29,30 H3-H4 tetramer stability allows diverse modifications on histone tails.31,32 Noncoding RNA, proposed to regulate gene expression, lacks an open reading frame, inhibiting expression through interactions with RNA molecules.33Figure 2 illustrates the epigenetic exposome, depicting external influences on gene functions, including repression of transposable elements and gene silencing pathways. The schematic outlines a three-stage mechanism—epigenator, epigenetic initiator, and epigenetic maintainer, providing a comprehensive view of epigenetic regulation.
Fig. 2: The schematic diagram illustrates the exposomes of epigenetics, encompassing external influences on gene regulation. It delineates three primary functions—the repression pathway of transposable element movement, gene silencing involving translational and post-translational processes, and imprinting for heritable gene marks. The mechanism of the epigenetic process is depicted through three stages—epigenator (external cues), epigenetic initiator (triggering chromatin modifications), and epigenetic maintainer (sustaining modifications via DNA methylation, histone modification, noncoding RNA, and ATP-dependent chromatin remodeling)
Periodontal Disease as an Epidisease with Significant Epigenetic Component: A Stride in the Direction of Better Understanding Periodontitis
Periodontitis is the inflammation of the gingival tissues caused by bacteria, leading to tissue and bone loss.12 According to the 2016 Global Burden of Disease Study, severe periodontal disease was ranked as the 11th most prevalent health condition worldwide.34 Periodontal disease was found to have a global prevalence ranging from 20 to 50%.35 From 1990 to 2010, there was a significant increase of 57.3% in the global burden of periodontal disease.36
Epithelial cells in the oral cavity encounter diverse bacteria, initiating signaling pathways that trigger complex inflammatory responses influenced by both genetic and environmental factors.2 Similar to other chronic diseases, periodontal disease is now seen as a multifactorial condition, as evidenced by twin and family studies and recent genome-wide association studies37 researchers have estimated the heritability of periodontal disease to be in the range of 30–50%. Researchers observed that individuals with periodontitis tend to exhibit a similar pattern of early tooth loss as one of their parents.38,39
Genetic and Epigenetic Insights into Periodontal Disease: Moving Beyond Pathogen-centric Approaches
The study conducted by Chatzopoulos et al. on a Caucasian population with chronic periodontal disease concluded that genetic variations in interleukin-6 (IL-6)—572 G/C and IL-10-592 C/A polymorphisms, both individually and in combination, did not exhibit a significant influence on the outcomes of nonsurgical periodontal therapy. Despite the presence of distinct genotypes, there were no statistically significant associations observed with clinical parameters after therapy, suggesting that these genetic polymorphisms may not be substantial predictors of treatment outcomes in this population.40,41 Hernández et al. study42 revealed 81 differentially hypermethylated genes and 21 differentially hypomethylated genes in patients with periodontitis compared to gingivally healthy controls. Notably, the intersection analysis highlighted three specific genes—zinc finger protein 718 (ZNF718) and Homeobox A4 (HOXA4) were differentially hypermethylated, while ZFP57 was differentially hypomethylated in individuals with periodontitis. The functional enrichment analysis of differentially methylated genes revealed immune-related ontologies such as “detection of a bacterium” and “antigen processing and presentation,” suggesting a potential link between altered DNA methylation patterns in peripheral leukocytes and immune processes in periodontitis. Kurushima et al. study43 focused on self-reported indicators, associating specific DNA methylation changes with periodontal traits, validated in buccal and adipose tissues. Collectively, these studies contribute valuable insights into periodontal disease at the epigenetic level, suggesting potential biomarkers, and personalized treatment avenues. This emphasizes the need for a multifaceted approach beyond pathogen targeting for effective disease management.
Evidence of Epigenetic Alterations and their Role in Development of Periodontal Disease: A Shifting Trend from Genetics to Epigenetics
The landscape of periodontal disease research underwent a significant transformation in the 1990s when the first evidence emerged, linking genetics to the development of periodontitis. This shift marked a departure from a purely genetic focus to an exploration of epigenetic mechanisms, introducing concepts of susceptibility and predisposition.44,45 Notable studies by Loos and Velden et al.,46 and Michalowicz et al.,47 provided crucial insights into the hereditary aspects of periodontitis. Loos and Velden, investigated the impact of sibling relationships on periodontal health, revealing a substantial siblingship effect on various clinical parameters. Michalowicz et al., in their assessment of 110 adult twins, highlighted those genetics appeared to be the root cause of familial aggregation.
Despite these genetic underpinnings, the complex interplay of genetic and environmental factors prompted a transition toward investigating epigenetic mechanisms.48,49 The field of epigenetics, considered a new frontier in dentistry, explores molecular mechanisms such as chromatin remodeling and selective gene activation or inactivation, offering a nuanced understanding of the pathogenesis of infectious and inflammatory diseases.50
Recognizing the intricacies of periodontitis, studies emphasized the role of both genetic and epigenetic factors, revealing varying susceptibility among individuals sharing the same microbial infection. The association between periodontal conditions and environmental factors, such as dietary habits and systemic diseases, remains an area of exploration, emphasizing the dynamic nature of epigenetic information responsive to environmental conditions.
The transition from genetics to epigenetics was underscored by advances in sequencing technology, enabling high-throughput DNA analysis. Studies like those by De Souza et al.51 provided insights into DNA methylation variations in immune-related genes, shedding light on their potential influence on periodontitis prognosis.
Further exploration into the gender-related distinctions in DNA methylation was presented by Li et al.,52 their use of pyrosequencing revealed a positive correlation between matrix metalloproteinase-9 (MMP-9) cytosine-phosphate-guanine islands methylation levels and chronic periodontitis severity. Additionally, they observed lower MMP-9 methylation in female patients but higher tissue inhibitor of metalloproteinase-1 (TIMP-1) methylation, with TIMP-1 methylation decreasing with age.
Martins et al.53 delved into the impact of lipopolysaccharides on histone modifications in oral epithelial cells. They identified potential epigenetic mechanisms involving DNA methylation and histone acetylation during dysbiosis, suggesting a delicate molecular balance between these processes. Although further characterization is needed, targeting these epigenetic mechanisms may offer therapeutic insights for periodontitis.
Zhu et al.54 provided critical insights into the role of the long noncoding RNA associated with periodontitis (lncR-APDC) in periodontitis. Their work, involving lncR-APDC knockout mice with induced experimental periodontitis, demonstrated exacerbated bone loss, disrupted cytokine regulation, altered immune cell proportions, and increased trefoil factor family 2 expression, highlighting the RNA’s critical role and therapeutic potential in periodontitis.
This evolving trend from genetics to epigenetics signifies a paradigm shift in understanding the intricate molecular mechanisms influencing the development of periodontal disease. Epigenetic alterations, as evidenced by various studies, present a dynamic and responsive framework that extends beyond static genetic information, offering new avenues for therapeutic interventions in periodontitis.
What are the Contributing Factors to Immune Fitness Dysregulation in Periodontitis?
The disruption of immune balance in periodontitis results from a combination of various risk factors. The causal factors for periodontitis can be grouped into five categories—subgingival bacterial biofilm, genetics and epigenetic changes, lifestyle factors, systemic illnesses, and miscellaneous factors. In older individuals, pathobionts are a significant influence on periodontitis, while younger patients, like those with early-onset periodontitis, are more closely linked to genetic factors.55 Epigenetic alterations and genetic mutations that occur over a lifetime can additionally influence an individual’s vulnerability to periodontitis.
Chronic Stress and its Detrimental Impact on Physiological Process
Stress is the body’s response to challenging life situations, influenced by factors like the stressor’s duration, intensity, and controllability, as well as the individual’s age and gender.56,57 From a Darwinian perspective, a person’s parental stress history significantly influences their descendants’ resilience.58 Stress-related memories are retained at the cellular level in brain regions like the hippocampus, highlighting the importance of past experiences. Neurons may preserve memories of stressful experiences through epigenetic mechanisms, including DNA methylation and histone modification.
The Impact of Chronic Stress on Periodontal Health: Mechanisms and Indirect Effects
Stress, a recognized trigger for mental and neurological disorders, exerts a profound impact on the epigenome and is implicated in an increased risk of periodontitis. The multifaceted mechanisms through which stress influences the periodontium encompass both indirect and direct pathways. Indirectly, stress instigates behavioral and lifestyle changes, such as heightened smoking, increased alcohol consumption, unhealthy dietary patterns, suboptimal oral hygiene practices, and diminished adherence to dental care recommendations. Simultaneously, stress directly impacts periodontal health by reshaping saliva composition, influencing gingival blood circulation, and modulating the host immune response.59
The intricate connection between stress and periodontal health stems from its influence on the immune response and health-related behaviors. Stress elevates the production of neuroendocrine hormones, including glucocorticoids and catecholamines, which detrimentally affect immune functions. This includes reductions in lymphocyte populations and natural killer cell activity, rendering individuals more susceptible to infections and contributing to the degradation of periodontal tissues.60
Insights from studies conducted on experimental animal models suggest that persistent stress may induce vascular inflammation, evidenced by an increase in circulating proinflammatory cytokines.61
Hilgert et al. observed a positive association between higher salivary cortisol levels and increased probing pocket depths and attachment loss in a previous study.62 Cakmak et al., found elevated salivary and gingival crevicular fluid (GCF) cortisol levels in groups with aggressive and chronic periodontitis compared to a healthy cohort.63 Jaiswal et al., explored the relationship between self-assessed stress levels and serum cortisol values, revealing a positive correlation with chronic periodontitis.64 Mousavijazi et al., investigated the connection between stress disorders and heightened levels of inflammatory mediators in periodontal disease, highlighting the significant role of stress in triggering inflammatory processes, as reflected in increased crevicular IL-1B levels in both aggressive and chronic periodontitis groups.65 Goyal et al., delved into the impact of psychosocial stress on periodontal health, establishing significant correlations between cortisol levels and clinical parameters in chronic periodontitis and stressed subjects. The study underscores stress as a potential contributor to periodontal disease.66
Genomic Stress Response and Its Epigenetic Influence
Epigenetics explains why individuals with similar genetic backgrounds can have varying disease susceptibility and coping abilities. Epigenetics encompasses both temporary and enduring gene expression changes not directly encoded in DNA, shaping how cells react at a molecular level.19,67 Epigenetic mechanisms, including DNA methylation and histone modifications, play a key role in regulating gene expression during stress, whether it’s a prolonged or rapid response.68 Epigenetics is particularly important in synaptic plasticity, memory formation, cognitive processes, and the development of stress-related traits and behavioral adjustments in response to chronic stress.69
Stress Markers and Receptors in Oral Cavity
Periodontal tissues contain glucocorticoid receptors that react to glucocorticoids released from the HPA axis. It has been shown that keratinocytes have the ability to directly react to adrenocorticotropic hormone and generate the glucocorticoid cortisol. This production can lead to localized immunosuppressive and anti-inflammatory effects,70 which encompass the inhibition of T lymphocyte formation71 and the inhibition of macrophage72 and natural killer cell functions.73
Stress and periodontal diseases have been studied through blood, saliva, and GCF. Catecholamines affect immune functions, influencing proinflammatory cytokines and inhibiting lymphocyte proliferation and natural killer cell activity.
Chromogranin A (CgA), an acidic secretory glycoprotein, is stored and released with catecholamines from the adrenal medulla and by sympathetic nerve endings and submandibular gland cells. It’s the precursor of bioactive peptides like catestatin, which may contribute to inflammation.
Salivary α-amylase (sAA) reflects sympathetic nervous system activity in response to stress and has antimicrobial properties. Neuropeptides like substance P-mediate neurogenic inflammation, initiating and sustaining inflammation by stimulating proinflammatory cytokine production.
Gallacher and Petersen reported that heightened concentrations of cortisol and β-endorphin might have significantly upregulated the expression of MMP-1, -2, -7, and -11, along with TIMP-1 in human gingival fibroblasts. This suggested mechanism could contribute to the increased periodontal breakdown associated with psychosocial stress status.74
Shared Pathways: Unveiling Common Biomarkers between Stress and Periodontitis
The intricate interplay between psychosocial stress and periodontitis reveals shared pathways mediated by a range of biomarkers. Cohen-Cole et al., have implicated proinflammatory cytokines such as IL-1 and IL-6, identified in the GCF, as potential bridges linking stress to periodontal disease.75 Additionally, cortisol, a well-established stress hormone, demonstrates associations with clinical periodontal parameters, including pocket depth and bleeding on probing, highlighting its role in the stress-periodontitis nexus. Salivary markers, such as CgA, cortisol, sAA, and β-endorphin, offer insights into the physiological responses to stress in the context of periodontal health. Moreover, the dysregulation of the HPA axis, autonomic nervous system, and central nervous system may influence the immune and inflammatory responses, contributing to the vulnerability of periodontal tissues to pathogenic microorganisms.76 These shared pathways underscore the complexity of the relationship between stress and periodontitis, shedding light on potential common biomarkers that warrant further exploration.
Challenges of Epigenetic Study
Studying genetic polymorphisms for periodontitis is challenging because variations associated with the disease in one population, like Caucasians, may not hold the same associations in different racial or ethnic groups, such as Asians, Brazilians, and Africans.45
Numerous investigations have indicated that the DNA methylation markers identified in peripheral tissues often lack precision in predicting the DNA methylation patterns found in the brain.77
Currently, most literature doesn’t distinguish between DNA methylation and hydroxymethylation, and common assays measure both modifications together. Future research should explore these modifications simultaneously for a more comprehensive understanding.
Using animal models and in vitro studies can be challenging when applying findings to patients due to the impact of environmental factors and significant individual disparities in epigenomic patterns. To establish a dependable benchmark, it’s essential to conduct in-depth analyses with well-organized and controlled conditions. A comprehensive understanding of epigenomic, gene expression, and phenotype profiles related to target diseases is crucial for effective mapping efforts. Additionally, developing user-friendly software for managing large datasets is essential for practical research implementation across various fields.78
Personalized Periodontics
Personalized periodontics is an innovative approach that considers variations in genetics, environment, lifestyle, and behavior among individuals. Diagnostic testing is often used to determine the most effective treatments based on factors like genetics or various analyzes encompassing epidemiology, sociology, molecular biology, physiology, or cellular studies.
Stress and allostatic load are gaining importance in personalized periodontics research. Repeated exposure to stressors leads to systemic effects, known as “allostatic load.”
Cutting-edge sequencing technologies and bioinformatics tools have accelerated the evolution of “omics” profiling, advancing the concept of personalized medicine.78 Recent advances in genomics allow for the study of complex interaction effects in individuals with multiple health conditions.
To achieve personalized medicine, it’s crucial to analyze multi-omics datasets, including genome, epigenome, transcriptome, metabolome, microbiome, proteome, and more. Effective collaboration between bioinformaticians, scientists, and clinicians is essential for this effort.78
CONCLUSION
In conclusion, this review highlights the intricate interplay between epigenetics, periodontitis, and stress-related health. It recognizes the potential of epigenetic biomarkers and pharmacogenomics for early disease detection and the tools available for personalized management. The practical application of epigenetics in treatments, particularly in modifying DNA methylation and histone patterns in cancer therapies, is acknowledged. The evolving focus from genetics to epigenetics in research is deemed promising, offering insights into disease understanding. However, it emphasizes that our comprehension of this intricate epigenetic orchestration is still at an early stage. The review underscores the importance of personalized approaches in treating and caring for individuals, particularly in the context of stress-related disorders.
ORCID
Smrithi Vishakha Varma https://orcid.org/0000-0002-5026-5555
Sheeja Saji Varghese https://orcid.org/0000-0003-4237-0002
Sajan Velayudhan Nair https://orcid.org/0009-0009-0490-2208
REFERENCES
1. Widschwendter M, Jones PA. DNA methylation and breast carcinogenesis. Oncogene 2002;21(35):5462–5482. DOI: 10.1038/sj.onc.1205606
2. Larsson L, Castilho RM, Giannobile WV. Epigenetics and its role in periodontal diseases: a state-of-the-art review. J Periodontol 2015;86(4):556–568. DOI: 10.1902/jop.2014.140559
3. Meyle J, Chapple I. Molecular aspects of the pathogenesis of periodontitis. Periodontol 2000 2015;69(1):7–17. DOI: 10.1111/prd.12104.PMID:26252398
4. Koolhaas JM, Bartolomucci A, Buwalda B, et al. Stress revisited: a critical evaluation of the stress concept. Neurosci Biobehav Rev 2011;35(5):1291–1301. DOI: 10.1016/j.neubiorev.2011.02.003
5. Stabholz A, Soskolne WA, Shapira L. Genetic and environmental risk factors for chronic periodontitis and aggressive periodontitis. Periodontol 2000 2010;53:138–153. DOI: 10.1111/j.1600-0757.2010.00340.x
6. Issa JP. Age-related epigenetic changes and the immune system. Clin Immunol 2003;109(1):103–108. DOI: 10.1016/s1521-6616(03)00203-1
7. Vivier E, Raulet DH, Moretta A, et al. Innate or adaptive immunity? The example of natural killer cells. Science 2011;331(6013):44–49. DOI: 10.1126/science.1198687
8. Tsigos C, Chrousos GP. Hypothalamic-pituitary-adrenal axis, neuroendocrine factors and stress. J Psychosom Res 2002;53(4):865–871. DOI: 10.1016/s0022-3999(02)00429-4
9. Loos BG, Van Dyke TE. The role of inflammation and genetics in periodontal disease. Periodontol 2000 2020;83(1):26–39. DOI: 10.1111/prd.12297
10. Smale ST. Transcriptional regulation in the innate immune system. Curr Opin Immunol 2012;24(1):51–57. DOI: 10.1016/j.coi.2011.12.008
11. Carpenter S, Ricci EP, Mercier BC, et al. Post-transcriptional regulation of gene expression in innate immunity. Nat Rev Immunol 2014;14(6):361–376. DOI: 10.1038/nri3682
12. Kornman KS. Mapping the pathogenesis of periodontitis: a new look. J Periodontol 2008;79(8 Suppl):1560–1568. DOI: 10.1902/jop.2008.080213
13. Waddington CH. The epigenotype. 1942. Int J Epidemiol 2012;41(1):10–13. DOI: 10.1093/ije/dyr184
14. Waddington CH. Towards a theoretical biology. Nature 1968;218(5141):525–527. DOI: 10.1038/218525a0
15. Egger G, Liang G, Aparicio A, et al. Epigenetics in human disease and prospects for epigenetic therapy. Nature 2004;429(6990):457–463. DOI: 10.1038/nature02625
16. Hamilton JP. Epigenetics: principles and practice. Dig Dis 2011;29(2):130–135. DOI: 10.1159/000323874
17. Feil R, Fraga MF. Epigenetics and the environment: emerging patterns and implications. Nat Rev Genet 2012;13(2):97–109. DOI: 10.1038/nrg3142
18. McGowan PO, Sasaki A, D’Alessio AC, et al. Epigenetic regulation of the glucocorticoid receptor in human brain associates with childhood abuse. Nat Neurosci 2009;12(3):342–348. DOI: 10.1038/nn.2270
19. Berger SL, Kouzarides T, Shiekhattar R, et al. An operational definition of epigenetics. Genes Dev 2009;23(7):781–783. DOI: 10.1101/gad.1787609
20. Tsankova NM, Berton O, Renthal W, et al. Sustained hippocampal chromatin regulation in a mouse model of depression and antidepressant action. Nat Neurosci 2006;9(4):519–525. DOI: 10.1038/nn1659
21. Turner BM. Defining an epigenetic code. Nat Cell Biol 2007;9(1):2–6. DOI: 10.1038/ncb0107-2
22. Holliday R, Pugh JE. DNA modification mechanisms and gene activity during development. Science 1975;187(4173):226–232. DOI: 10.1126/science.187.4173.226
23. Riggs AD. X inactivation, differentiation, and DNA methylation. Cytogenet Cell Genet 1975;14(1):9–25. DOI: 10.1159/000130315
24. Szyf M. The early life social environment and DNA methylation: DNA methylation mediating the long-term impact of social environments early in life. Epigenetics 2011;6(8):971–978. DOI: 10.4161/epi.6.8.16793
25. Héberlé É, Bardet AF. Sensitivity of transcription factors to DNA methylation. Essays Biochem 2019;63(6):727–741. DOI: 10.1042/EBC20190033
26. Kohli RM, Zhang Y. TET enzymes, TDG and the dynamics of DNA demethylation. Nature 2013;502(7472):472–479. DOI: 10.1038/nature12750
27. Kriaucionis S, Heintz N. The nuclear DNA base 5-hydroxymethylcytosine is present in Purkinje neurons and the brain. Science 2009;324(5929):929–930. DOI: 10.1126/science.1169786
28. Fischle W, Tseng BS, Dormann HL, et al. Regulation of HP1-chromatin binding by histone H3 methylation and phosphorylation. Nature 2005;438(7071):1116–1122. DOI: 10.1038/nature04219
29. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 2012;489(7414):57–74. DOI: 10.1038/nature11247
30. Jenuwein T, Allis CD. Translating the histone code. Science 2001;293(5532):1074–1080. DOI: 10.1126/science.1063127
31. Kimura H. Histone dynamics in living cells revealed by photobleaching. DNA Repair (Amst) 2005;4(8):939–950. DOI: 10.1016/j.dnarep.2005.04.012
32. Peterson CL, Laniel MA. Histones and histone modifications. Curr Biol 2004;14(14):R546–R551. DOI: 10.1016/j.cub.2004.07.007
33. Zaratiegui M, Irvine DV, Martienssen RA. Noncoding RNAs and gene silencing. Cell 2007;128(4):763–776. DOI: 10.1016/j.cell.2007.02.016
34. GBD 2016 Disease and Injury Incidence and Prevalence Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990-2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet 2017;390(10100):1211–1259. DOI: 10.1016/S0140-6736(17)32154-2
35. Sanz M, D’Aiuto F, Deanfield J, et al. European workshop in periodontal health and cardiovascular disease. Eur Heart J Suppl 2010;12(Suppl B):B3–B12 DOI: 10.1093/eurheartj/suq003
36. Jin LJ, Lamster IB, Greenspan JS, et al. Global burden of oral diseases: emerging concepts, management and interplay with systemic health. Oral Dis 2016;22(7):609–619. DOI: 10.1111/odi.12428
37. Nibali L, Bayliss-Chapman J, Almofareh SA, et al. What Is the heritability of periodontitis? A systematic review. J Dent Res 2019;98(6):632–641. DOI: 10.1177/0022034519842510
38. Michalowicz BS, Aeppli DP, Kuba RK, et al. A twin study of genetic variation in proportional radiographic alveolar bone height. J Dent Res 1991;70(11):1431–1435. DOI: 10.1177/00220345910700110701
39. Mucci LA, Björkman L, Douglass CW, et al. Environmental and heritable factors in the etiology of oral diseases–a population-based study of Swedish twins. J Dent Res 2005;84(9):800–805. DOI: 10.1177/154405910508400904
40. Chatzopoulos GS, Doufexi AE, Kouvatsi A. Clinical response to non-surgical periodontal treatment in patients with interleukin-6 and interleukin-10 polymorphisms. Med Oral Patol Oral Cir Bucal 2017;22(4):e446–e457. DOI: 10.4317/medoral.21795
41. Chatzopoulos GS, Doufexi AE, Zarenti S, et al. Periodontal disease progression and gene polymorphisms: results after 3 years of active periodontal treatment. Minerva Dent Oral Sci 2022;71(6):329–338. DOI: 10.23736/S2724-6329.22.04709-X
42. Hernández HG, Hernández-Castañeda AA, Pieschacón MP, et al. ZNF718, HOXA4, and ZFP57 are differentially methylated in periodontitis in comparison with periodontal health: epigenome-wide DNA methylation pilot study. J Periodontal Res 2021;56(4):710–725. DOI: 10.1111/jre.12868
43. Kurushima Y, Tsai PC, Castillo-Fernandez J, et al. Epigenetic findings in periodontitis in UK twins: a cross-sectional study. Clin Epigenetics 2019;11(1):27. DOI: 10.1186/s13148-019-0614-4
44. Hart TC. Genetic considerations of risk in human periodontal disease. Curr Opin Periodontol 1994:3–11. PMID: 8032464.
45. Schafer AS, Jepsen S, Loos BG. Periodontal genetics: a decade of genetic association studies mandates better study designs. J Clin Periodontol 2011;38(2):103–107. DOI: 10.1111/j.1600-051X.2010.01653.x
46. Loos BG, Velden UV. Clinical Periodontology and Implant Dentistry, 4th edition. Jaypee Brothers; 2003. pp. 387–399.
47. Michalowicz BS, Aeppli D, Virag JG, et al. Periodontal findings in adult twins. J Periodontal 1991;62(5):293–299. DOI: 10.1902/jop.1991.62.5.293
48. Larsson L. Current concepts of epigenetics and its role in periodontitis. Curr Oral Health Rep 2017;4(4):286–293. DOI: 10.1007/s40496-017-0156-9
49. Khouly I, Braun RS, Ordway M, et al. The role of DNA methylation and histone modification in periodontal disease: a systematic review. Int J Mol Sci 2020;21(17):6217. DOI: 10.3390/ijms21176217
50. Williams SD, Hughes TE, Adler CJ, et al. Epigenetics: a new frontier in dentistry. Aust Dent J 2014;59(Suppl 1):23–33. DOI: 10.1111/adj.12155
51. De Souza AP, Planello AC, Marques MR, et al. High-throughput DNA analysis shows the importance of methylation in the control of immune inflammatory gene transcription in chronic periodontitis. Clin Epigenet 2014;6(1):15. DOI: 10.1186/1868-7083-6-15
52. Li X, Lu J, Teng W, et al. Quantitative evaluation of MMP-9 and TIMP-1 promoter methylation in chronic periodontitis. DNA Cell Biol 2018;37(3):168–173. DOI: 10.1089/dna.2017.3948
53. Martins MD, Jiao Y, Larsson L, et al. Epigenetic modifications of histones in periodontal disease. J Dent Res 2016;95(2):215–222. DOI: 10.1177/0022034515611876
54. Zhu ZX, Liu Y, Wang J, et al. A novel lncRNA-mediated epigenetic regulatory mechanism in periodontitis. Int J Biol Sci 2023;19(16):5187–5203. DOI: 10.7150/ijbs.87977
55. Loos BG, Papantonopoulos G, Jepsen S, et al. What is the contribution of genetics to periodontal risk? Dent Clin North Am 2015;59(4):761–780. DOI: 10.1016/j.cden.2015.06.005
56. Radley JJ, Kabbaj M, Jacobson L, et al. Stress risk factors and stress-related pathology: neuroplasticity, epigenetics and endophenotypes. Stress 2011;14(5):481–497. DOI: 10.3109/10253890.2011.604751
57. Karatsoreos IN, McEwen BS. Annual research review: the neurobiology and physiology of resilience and adaptation across the life course. J Child Psychol Psychiatry 2013;54(4):337–347. DOI: 10.1111/jcpp.12054
58. Yehuda R, Bierer LM. Transgenerational transmission of cortisol and PTSD risk. Prog Brain Res 2008;167:121–135. DOI: 10.1016/S0079-6123(07)67009-5
59. Goyal S, Gupta G, Thomas B, et al. Stress and periodontal disease: the link and logic! Ind Psychiatry J 2013;22(1):4–11. DOI: 10.4103/0972-6748.123585
60. Warren KR, Postolache TT, Groer ME, et al. Role of chronic stress and depression in periodontal diseases. Periodontol 2000 2014;64(1):127–138. DOI: 10.1111/prd.12036
61. Lu XT, Zhao YX, Zhang Y, et al. Psychological stress, vascular inflammation, and atherogenesis: potential roles of circulating cytokines. J Cardiovasc Pharmacol 2013;62(1):6–12. DOI: 10.1097/FJC.0b013e3182858fac
62. Hilgert JB, Hugo FN, Bandeira DR, et al. Stress, cortisol, and periodontitis in a population aged 50 years and over. J Dent Res 2006;85(4):324–328. DOI: 10.1177/154405910608500408
63. Cakmak O, Tasdemir Z, Aral CA, et al. Gingival crevicular fluid and saliva stress hormone levels in patients with chronic and aggressive periodontitis. J Clin Periodontol 2016;43(12):1024–1031. DOI: 10.1111/jcpe.12614
64. Jaiswal R, Shenoy N, Thomas B. Evaluation of association between psychological stress and serum cortisol levels in patients with chronic periodontitis - estimation of relationship between psychological stress and periodontal status. J Indian Soc Periodontol 2016;20(4):381–385. DOI: 10.4103/0972-124X.193165
65. Mousavijazi M, Naderan A, Ebrahimpoor M, et al. Association between psychological stress and stimulation of inflammatory responses in periodontal disease. J Dent (Tehran) 2013;10(1):103–111. PMID: 23724208.
66. Goyal S, Jajoo S, Nagappa G, et al. Estimation of relationship between psychosocial stress and periodontal status using serum cortisol level: a clinico-biochemical study. Indian J Dent Res 2011;22(1):6–9. DOI: 10.4103/0970-9290.79966
67. Slatkin M. Epigenetic inheritance and the missing heritability problem. Genetics 2009;182(3):845–850. DOI: 10.1534/genetics.109.102798
68. Tsankova N, Renthal W, Kumar A, et al. Epigenetic regulation in psychiatric disorders. Nat Rev Neurosci 2007;8(5):355–367. DOI: 10.1038/nrn2132
69. Siegmund KD, Connor CM, Campan M, et al. DNA methylation in the human cerebral cortex is dynamically regulated throughout the life span and involves differentiated neurons. PLoS One 2007;2(9):e895. DOI: 10.1371/journal.pone.0000895
70. Cirillo N, Prime SS. Keratinocytes synthesize and activate cortisol. J Cell Biochem 2011;112(6):1499–1505. DOI: 10.1002/jcb.23081
71. Dhabhar FS, McEwen BS. Acute stress enhances while chronic stress suppresses cell-mediated immunity in vivo: a potential role for leukocyte trafficking. Brain Behav Immun 1997;11(4):286–306. DOI: 10.1006/brbi.1997.0508
72. Baybutt HN, Holsboer F. Inhibition of macrophage differentiation and function by cortisol. Endocrinology 1990;127(1):476–480. DOI: 10.1210/endo-127-1-476
73. Gatti G, Cavallo R, Sartori ML, et al. Inhibition by cortisol of human natural killer (NK) cell activity. J Steroid Biochem 1987;26(1):49–58. DOI: 10.1016/0022-4731(87)90030-6
74. Gallacher DV, Petersen OH. Stimulus-secretion coupling in mammalian salivary glands. Int Rev Physiol 1983;28:1–52. PMID: 6307913.
75. Cohen-Cole SA, Cogen RB, Stevens AW, et al. Psychiatric, psychosocial, and endocrine correlates of acute necrotizing ulcerative gingivitis (trench mouth): a preliminary report. Psychiatr Med 1983;1(2):215–225. PMID: 6599850.
76. Neupane SP, Virtej A, Myhren LE, et al. Biomarkers common for inflammatory periodontal disease and depression: a systematic review. Brain Behav Immun Health 2022;21:100450. DOI: 10.1016/j.bbih.2022.100450
77. Walton E, Hass J, Liu J, et al. Correspondence of DNA methylation between blood and brain tissue and its application to schizophrenia research. Schizophr Bull 2016;42(2):406–414. DOI: 10.1093/schbul/sbv074
78. Cho YD, Kim WJ, Ryoo HM, et al. Current advances of epigenetics in periodontology from ENCODE project: a review and future perspectives. Clin Epigenetics 2021;13(1):92. DOI: 10.1186/s13148-021-01074-w
________________________
© The Author(s). 2024 Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by-nc/4.0/), which permits unrestricted use, distribution, and non-commercial reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.