Open access peer-reviewed chapter

Role of Cellular Responses in Periodontal Tissue Destruction

Written By

Nam Cong-Nhat Huynh

Submitted: 28 June 2022 Reviewed: 19 July 2022 Published: 14 August 2022

DOI: 10.5772/intechopen.106645

From the Edited Volume

Periodontology - New Insights

Edited by Gokul Sridharan

Chapter metrics overview

204 Chapter Downloads

View Full Metrics

Abstract

Periodontal tissue destruction is the deterioration of tooth-supporting components, particularly the periodontal ligament (PDL) and alveolar bone, resulting in gingival recession, root exposure, tooth mobility and drifting, and, finally, tooth loss. The breakdown of the epithelial barriers by infection or mechanical damage allows bacteria and their toxins to enter and stimulates the immune response. The bacteria cause periodontal damage via the cascade of the host reaction which is crucial in the destruction of the connective tissue around the tooth. The OPG/RANKL/RANK system is the key player in bone regulation of periodontal tissue and was controlled by both immune and non-immune cells. This knowledge has predicated the successfulness of implant and orthodontics treatments with the predictable healing and regeneration of the bone and supporting tissues surrounding the teeth.

Keywords

  • RANKL
  • OPG
  • RANK
  • osteoimmunology
  • periodontal destruction
  • periodontitis
  • bone resorption
  • inflammation
  • immune response

1. Introduction

The periodontium is made up of four components: gingiva, alveolar bone, cementum, and periodontal ligament (PDL) (Figure 1) [1]. They differ in cellular composition, protein kinds and quantities, mineralization, metabolic activity, and disease susceptibility. Embryologically, the gingival component is produced from the pharyngeal arches’ ectoderm and contains stratified squamous epithelium while PDL, cementum, and alveolar bone are produced from neuro-mesenchymal stem cells and contain connective tissues [2]. Periodontal destruction is initiated by bacterial agents (periodontitis) or mechanism stimuli (such as orthodontic force or dental trauma). However, host response plays a very important role in the demolition of the connective tissue around the tooth. In this chapter, we concisely discussed the cellular responses, including immune and non-immune cells to the destruction of periodontal tissue, and how we applied the knowledge in dental practice.

Figure 1.

Periodontium is made up of four components: gingiva, alveolar bone, cementum, and periodontal ligament (PDL). Oral epithelium (OE): is the epithelium that exposes to the surface of the mouth. Sulcular epithelium (SE): is the epithelium that contacts the surface of the tooth. Paroxizmalnaya form epithelium (JE): the epithelium which connects the gingiva to the tooth. OE, SE, and JE are keratinized tissue while alveolar mucosa is non-keratinized. Created with BioRender.com.

Advertisement

2. Periodontal tissues destruction

The components of periodontal tissue can be divided into hard tissue (cementum and alveolar bone) and soft tissue (gingiva and PDL). The anchors to which the fibrous PDL binds the tooth into the skeleton are the two mineralized tissues, cementum and alveolar bone [3]. The gingiva is the periodontium’s covering tissue, providing immediate protection for the underlying tissues and extra tooth attachment. The cementum is the calcified avascular mesenchymal tissue that forms the anatomic root’s outer layer. The periodontium’s primary roles include protecting teeth, nerves, and blood vessels from mechanical stresses, attaching teeth to the bone, and transmitting occlusal forces and sensibility to stimuli such as temperature and pain [4].

Periodontitis (PD) is a common illness that can lead to tooth loss and causes a slew of problems for physicians. Periodontitis is caused by inflammation of the tooth-supporting tissues, which results in the gradual loss of PDL and alveolar bones [5]. Periodontal disease is a collection of chronic inflammatory diseases that affect the gingiva, bone, and ligament (the connective tissue collagen fibers that bind a tooth to the alveolar bone) that support the teeth [6]. Chronic periodontitis occurs when untreated gingivitis progresses to the loss of gingiva, bone, and ligament, resulting in the deep periodontal ‘pockets’ that are a characteristic of the condition and can eventually lead to tooth loss. Periodontal disease can aggravate inflammatory illnesses, such as diabetes and atherosclerosis by increasing the body’s total inflammatory load.

On the other hand, apical periodontitis (AP) is an infection that causes inflammation and destruction of the periradicular tissues. It happens as a result of a series of assaults on the tooth pulp, such as infection, physical and iatrogenic trauma, endodontic therapy, and the destructive effects of root canal filling materials [7]. Then, the host mounts a slew of defenses, including various cell types, intercellular messengers, antibodies, and effector chemicals. Microbial factors and host defense forces interact with, battle with, and destroy much of the periapical tissue, resulting in the production of several types of AP lesions, most often reactive granulomas and cysts, with simultaneous bone resorption around the roots of impacted teeth.

Periodontitis or dental trauma can result in periodontal tissue destruction. It is the destruction of the tooth-supporting structures especially the PDL, and alveolar bone leading to the recession of the marginal gingiva, root exposure, tooth mobility and drifting, and, eventually, tooth loss. They are the results of chronic periodontitis with host defense response, tissue inflammation, and bone destruction in response to bacterial aggression [8]. Especially, the receptor activator of nuclear factor – (KB) ligand (RANKL) is a member of the TNF cytokine family encoded by the gene TNFSF11. It expresses in a membrane-bound protein or secreted forms and plays an important role in bone resorption including periodontal bone destruction via cellular and immune responses under the term the cross-talk between bone biology and immunology - osteoimmunology [9, 10].

Advertisement

3. Cellular responses in periodontal tissue destruction

3.1 Immune cell responses

The first physical barrier that is against infection from bacterial invasion is the periodontal epithelium. The epithelial barriers prevent pathogenic invasion and protect the periodontal tissues. The cells are involved in an active role in the innate host defense as well as further acquired immune responses. When the pathogens overcome the fence, they cause inflammation and destruction of the connective tissue, with subsequent bone loss and tooth loss. In periodontal tissues, cytokines are produced by fibroblasts, endothelial cells, macrophages, osteoclasts, epithelial cells, and leukocytes. By those mediators, osteoclasts will be differentiated and accumulated to the injury (Figure 2) [12].

Figure 2.

The mechanisms involved in periodontal tissue destruction. The breakdown of the epithelial barriers by infection or mechanical damage allows bacteria and their toxins to enter and stimulates the immune response. The OPG/RANKL/RANK system plays a central role in regulating bone destruction in periodontal tissue. RANKL binds to the RANK receptor on osteoclast precursor to activate osteoclast while OPG neutralizes this interaction to inhibit osteoclastogenesis. RANKL was main produced by periodontal stromal cells, such as gingival fibroblasts, PDL cells, cementoblasts, osteoblasts, and especially osteocytes as well as immune cells, such as T cells and B cells. T cells communicate with other cells, such as stromal cells, macrophages, and dendritic cells via IL-17, IL-6, and IL-23 to regulate RANKL/OPG signaling resulting in bone destruction and tooth loss and resolution of infection and inflammation [9, 11]. Created with BioRender.com.

The cells of the adaptive immune system are special types of leukocytes, called lymphocytes. They produce a number of mediators of signaling to induce osteoclastogenesis, such as IL-1, -6, and -17; RANKL; and TNF-α. The differentiation of osteoclast is mainly regulated by RANKL and macrophage-colony stimulating factor (M-CSF). Both of these factors are secreted by immune cells and osteoblasts, odontoblasts, osteoclasts, and other cells in the PDL during alveolar bone resorption [13]. On the other hand, osteoprotegerin (OPG) is a member of the TNF receptor superfamily that prevents osteoclast differentiation, as well as its resorptive function, and stimulates osteoclast apoptosis. It is found in PDL cells and osteoblasts while Receptor activator of nuclear factor κ B (RANK) is detected in osteoclasts and osteoclast precursors. In normal bone remodeling and a variety of pathologic circumstances, RANKL/RANK signaling directs the generation of multinucleated osteoclasts from their progenitors, as well as their activation and survival. By attaching to RANKL and inhibiting it from connecting to its receptor, RANK, OPG protects the skeleton from excessive bone resorption. As a result, the RANKL/OPG ratio is a key predictor of bone mass and skeletal integrity [14].

Early, Baker et al. [15] found that following Porphyromonas gingivalis infection in periodontal disease, CD4+ T cells and their proinflammatory cytokines are important effectors of bone destruction as an adaptive immune response [15]. Directly, CD4+ T cells are the primary cells responsible for the increased levels of RANKL in chronic periodontitis patients contributing to the molecular local imbalance that results in periodontitis-induced alveolar bone resorption [16]. Besides the ability to support osteoclastogenesis directly, T lymphocytes secrete IL-1, IL-6, and IL-17 which can stimulate RANKL expression. The cells also inhibit osteoclast formation by a number of inhibitory molecules such as IL-4, -10, and -13 … and IFN-γ [17]. Horwood 2001 [18] inhibited osteoclast formation in cocultures of rat and human cells treated with M-CSF and RANKL by IL-12 and IL-18. When they were administered at synergistically low doses both GM-CSF and IFN-γ were secreted. T helper cells also play role in determining the effect of the T lymphocytes’ immune responses against periodontal diseases. Bainbridge et al. 2010 [19] tested the capacity of some P. gingivalis strains to induce gingival inflammation, immune responses, and alveolar bone resorption of periodontal disease. They found that these certain strains can induce significant systemic levels of IgG and isotypes IgG1, IgG2a, and IgG2b, indicating the involvement of both two types of T helper cell responses to infection.

In the bone resorption lesion of periodontal disease, regulatory T cell (Treg, FoxP3 + CD25+) was diminished and associated with the increased RANKL+ T cells [20]. Using animal models, Treg recruiting in periodontal tissue can inhibit periodontitis, however, Treg’s function might be suppressed [21]. Intriguingly, under inflammatory conditions, several Tregs can convert into Th17 cells called exFoxp3Th17 cells, which induce strongly osteoclastogenesis and bone destruction via IL-17 production [22]. These cells are also found in periodontal lesions in response to the oral commensal bacteria and play a central role in periodontal inflammation [23].

It was known that B cells and plasma cells are also able to produce RANKL to induce osteoclastogenesis [24, 25]. However, there was little evidence of the role of B cells in osteoclastogenesis. In the absence of T lymphocytes, Actinobacillus actinomycetemcomitans-responsive B lymphocytes can lead to accelerate periodontal bone resorption via the up-regulation of RANKL [26, 27]. In brief, bacterial antigen-specific T and B lymphocytes play a key role in RANKL- mediated bone loss in periodontal tissue destruction [24].

3.2 Non-immune cell responses

Regarding these oral pathogens, in PDL cells, P. gingivalis and/or nicotine activation increased reactive oxygen species (ROS) and superoxide production indicating oxidative stress [28]. It was confirmed clinically in the saliva of patients with chronic periodontitis [29, 30]. In gingival fibroblasts, high concentrations of saline (NaCl) inhibited cell migration but not proliferation [31]. These findings highlighted the susceptibility of these periodontal fibroblasts under oral stimulating factors. Injury to the periapical tissue can damage periodontal cells (gingival fibroblasts, PDL cells, cementoblasts), resulting in the production of cementoclasts/osteoclasts locally. P. gingivalis upregulated the RANKL expression in gingival fibroblasts and PDL cells, denoting enhanced osteoclastogenesis [32].

We previously separated cementoblasts from human third molars and co-cultivating them with mononuclear blood cells. Both osteoclast development and resorptive activity were studied in the absence and presence of IL-1. Cementoblasts could initiate osteoclastogenesis via RANKL production, which is heavily influenced by IL-1β explaining why osteoclasts can arise around the root of teeth [33]. RANKL was also produced primarily by osteocytes during the alveolar bone remodeling of the orthodontic tooth movement process. Using a newly developed approach for isolating periodontal tissue component cells from the alveolar bone, Shoji-Matsunaga et al. discovered that osteocytes expressed far more RANKL than other periodontal tissue cells [34]. The decrease of orthodontic tooth movement in mice particularly missing RANKL in osteocytes demonstrated the crucial function of osteocyte-derived RANKL. The study highlighted the critical involvement of osteocyte-derived RANKL in alveolar bone remodeling, laying the groundwork for orthodontic force-mediated bone resorption.

3.3 Pro-inflammatory molecule responses

Pro-inflammatory molecules including chemokines, interleukins, and TNF (Tumor Necrosis Factors) were secreted by immune and stromal cells during the inflammation and bone resorption of periodontal tissue destruction. Chemokines are chemotactic cytokines that stimulate the recruitment of inflammatory cells. Chemokines have a role in osteoclastogenesis by inducing the differentiation of osteoclasts, they also affect osteoclast functions/properties [35]. In this way, chemokines can affect periodontal bone loss by recruiting neutrophils to fight against bacteria. Yu et al. [36] use P. gingivalis in CXCR2-deficient mice, a type of chemokine receptor. The result showed that knockout mice were highly susceptible to alveolar bone loss. These mice also suggested a role for chemokines in maintaining normal bone homeostasis.

Interleukins are a group of cytokines that are expressed by leukocytes and function in the immune system. Many studies in human and animal patterns have indicated the mediating bone loss stimulated by periodontal pathogens. Delima [17] investigated the role of IL-1 in periodontal disease in monkeys by using an inhibitor. The results indicate that inhibition of IL-1 significantly reduces inflammation, connective tissue attachment loss, and bone resorption. Especially, the transgenic mice that overexpress a form of IL-1α in the oral mucosal epithelium develop a syndrome that possesses all of the major features of periodontal disease, including epithelial proliferation and apical migration, loss of attachment, and destruction of cementum and alveolar bone [37]. Using antibiotics did not reduce the disease, giving the role of IL-1 in mediating in promoting the destruction of the periodontium. However, other types of IL seem to reduce bone loss and symptoms of disease in the periodontium. For example, according to Shaker 2012 [38], the IL-11 concentration was significantly higher in control and chronic periodontitis groups in comparison to aggressive periodontitis groups.

Curiously, studies reported the opposing roles of IL-17 in periodontal bone resorption, especially with rheumatoid arthritis (RA) condition. Th17 cells produce mainly IL-17 leading to bone destruction in RA by mediating the osteoclastogenesis process [38, 39]. However, inflammatory mediators, including chemokines, cytokines, prostaglandin E2, and nitric oxide that IL-17 recruited in RA conditions have antibacterial properties that lead to bone protection in the context of periodontal infection. Yu et al. [36] examined IL-17’s role in inflammatory bone loss induced by P. gingivalis in IL-17–deficient mice in an autoimmune arthritis model. These animals had increased periodontal bone degradation, indicating that IL-17 has a bone-protective effect.

TNF refers to a group of cytokines that can cause cell death. In this family, TNF-α has various functions, such as cytolysis of some specific cells, cachexia, pyrogen, cell proliferation, and differentiation in some cases. It seems to be that reducing the level of TNF-α in the body leads to reduce bone loss, however, it will increase the number of bacteria in the lesion since the lessen immune response. Garlet et al. [40] examined how TNF-α modulates the periodontal disease by A. actinomycetemcomitans in TNF-α receptor-deficient mice. These mice had less severe periodontitis with less alveolar bone loss and inflammatory reaction. However, the higher level of bacteria compares with the wild-type group.

Advertisement

4. Relation in dental practice

Implants, orthodontics, and other areas of maxillofacial dentistry are more or less based on the predictable repair and regeneration of the bone and supporting tissues around the teeth. In the field of implantology, the process of bone repair and regeneration (osteogenesis) plays an important role in determining the success of treatment. Bone integration includes the initial stages of tissue response, peri-implant osteogenesis, and peri-implant bone remodeling, resulting in new bone formation on the implant surface. Composition, implant design, and surface treatment affect bone integration. Other factors include systemic condition, surgical technique, adequate healing time, and load-bearing properties. In dental implants, bone and implant integrate directly (Figure 3). There are no PDL and Sharpey’s fibers that help to absorb force and micro trauma. Instead, osteoblasts are attached to the mineralized collagen framework, forming a dense, mineralized area directly on the implant surface. The distance from the bone to the implant surface takes place in a continuous process of bone regeneration in response to stress and mechanical force over time. The cavity containing osteoclasts, osteoblasts, mesenchymal cells, and blood vessels is always present adjacent to the implant surface [41, 42]. Excessive micro-motion in healing leads to tension and torsion forces, stimulating the formation of a fibrin membrane around the implant, and displacing the bone-implant interface. This phenomenon loosens the implant, inhibiting bone integration.

Figure 3.

The anchor of a dental implant in the alveolar bone directly by the osteointegration without the PDL and Sharpey’s fibers in comparison to the real teeth (Figure 1). Note that there was also no connective tissue connection between the implant and the gingiva. Created with BioRender.com.

Using the bone repair and regeneration processes to accelerate tooth movement in orthodontics can significantly reduce treatment time and harmful effects. The ability to move teeth is mainly determined by the regeneration of periodontal tissue, under the regulation of molecular mechanisms by the response of cells in the alveolar bone and PDL. Fluid shear stress that PDL cells are constantly subjected to, is a form of mechanical loading on the cell level. Under compression, the fluids are forced through the voids and are relocated either to adjacent zones of the PDL or driven into the neighboring alveolar bone. This fluid movement creates the type of mechanical loading known as fluid shear stress [43]. This force appears during the mastication, speech, and orthodontic procedure. By their arrangement and structure, fibers of PDL can absorb forces that are harmful to teeth and surrounding apparatus. However, excessing the endured limitation of PDL (by duration and/or magnitude), PDL can respond to the compression by stimulating many compartments. By using mechanical stress (light orthodontic application) to activate the fluid shear stress response in PDL cells, Yarmolyuk 2012 [44] showed that it could alleviate the compromised periodontal status via down-regulation of TLR4.

Similar to the inflammation response triggered by bacteria, mechanical forces also cause increased cytokine secretion by PDL cells. By using an orthodontic model which can mimic real situations, many studies have proven that IL-1β, IL-6, and TNF-α have been revealed in the compression side of teeth [45]. In the results of this elevation, the OPG/RANKL/RANK system will function leading to bone remodeling. Depending on the kind of force and its magnitude, the bone will be resorbed or formed prominently (Figure 4).

Figure 4.

Orthodontics force induces periodontal and bone tissue remodeling. Mechanical stress applied to a tooth can destruct periodontal and bone tissue through the RANKL/RANK/OPG system. At the site of compression, periodontal tissue cells such as PDL cells, cementoblasts, osteoblasts, and osteocytes of alveolar bone can produce RANKL to active osteoclast via RANK leading to bone resorption. On the opposite side of the tooth where OPG will be produced increasingly by stromal cells to inhibit osteoclastogenesis leading to more bone formation. The application of too strong orthodontics force will enhance uncontrolled osteoclastogenesis/cementoclastogenesis leading to tooth’s root resorption (external resorption) and periodontal tissue destruction permanently. Created with BioRender.com.

Interestingly, Diercke et al. [46] indicated that static compressive forces significantly induced the expression of ephrin-A2, while the expression of ephrin -B2 was significantly down-regulated in PDL. Although these coupling factors (Ephrin and eph receptor) have been defined as osteoclast-derived molecules that induce osteoblastic bone formation. The more ephrin-A2, the more osteoclast, while reducing ephrin-B2 will reduce osteoblast formation [47].

Low-level lasers can also accelerate tooth migration, according to human and animal studies [48, 49, 50, 51, 52]. However, some studies suggest that low-level lasers do not accelerate tooth migration but also slow it down [53]. This difference may be explained by different treatment regimens including laser wavelength, radiation dose, location, and frequency. Several studies have reported that low-level lasers stimulate bone regeneration by increasing the number and function of osteoblasts and osteoclasts as well as markers, such as matrix metalloproteinase-9, cathepsin K, integrin [50], and the RANK/RANKL/OPG system [51, 54] at periodontal tissue. More research is needed to find the most effective regimen to extend its effectiveness.

Advertisement

5. Conclusions

The imbalance between bone resorption and formation leads to the loss of bone and PDL. This process is regulated by inflammatory infiltration followed by a dozen of factors, such as bacterial products, cytokines, chemokines, and complements from surrounding cells and blood vessels. Using our knowledge of periodontal tissue destruction, bone repair, and regeneration mechanisms, and the factors that influence them help us to better understand the molecular mechanisms, explain phenomena and applications, and develop approaches to new treatments, thereby helping to achieve optimal treatment results, and minimizing complications for patients.

Advertisement

Acknowledgments

The author thanks the Faculty of Odonto-Stomatology, University of Medicine and Pharmacy at Ho Chi Minh City, Vietnam for supporting this chapter.

Advertisement

Conflict of interest

The author declares that there is no conflict of interest regarding the publication of this chapter.

References

  1. 1. Melcher AH. On the repair potential of periodontal tissues. Journal of Periodontology. 1976;47(5):256-260
  2. 2. Rathee M, Jain P. Embryology, Teeth. Treasure Island (FL): StatPearls; 2022
  3. 3. Seo BM, Song IS, Um S, Lee J-H. Chapter 22 - Periodontal ligament stem cells. In: Vishwakarma A, Sharpe P, Shi S, Ramalingam M, editors. Stem Cell Biology and Tissue Engineering in Dental Sciences. Boston: Academic Press; 2015. pp. 291-296
  4. 4. Cho MI, Garant PR. Development and general structure of the periodontium. Periodontology 2000. 2000;2000(24):9-27
  5. 5. Nguyen TT, Ho TH, Huynh CN, Dien HAV, Vo LT. Hyaluronic acid 0.2% application enhanced periodontitis treatment in non-surgical phase. Journal of Stomatology. 2021;74(2):76-83
  6. 6. Kinane DF, Stathopoulou PG, Papapanou PN. Periodontal diseases. Nature Reviews Disease Primers. 2017;3:17038
  7. 7. Graunaite I, Lodiene G, Maciulskiene V. Pathogenesis of apical periodontitis: A literature review. Journal of Oral & Maxillofacial Research. 2012;2(4):e1
  8. 8. Bascones A, Noronha S, Gomez M, Mota P, Gonzalez Moles MA, Villarroel DM. Tissue destruction in periodontitis: Bacteria or cytokines fault? Quintessence International. 2005;36(4):299-306
  9. 9. Tsukasaki M. RANKL and osteoimmunology in periodontitis. Journal of Bone and Mineral Metabolism. 2021;39(1):82-90
  10. 10. Chen B, Wu W, Sun W, Zhang Q, Yan F, Xiao Y. RANKL expression in periodontal disease: Where does RANKL come from? BioMed Research International. 2014;2014:731039
  11. 11. Tsukasaki M, Takayanagi H. Osteoimmunology: Evolving concepts in bone-immune interactions in health and disease. Nature Reviews. Immunology. 2019;19(10):626-642
  12. 12. Mariano FS, de Cássia Orlandi Sardi J, Duque C, Höfling JF, Gonçalves RB. The role of immune system in the development of periodontal disease: A brief review. Revista Odonto Ciência. 2010;25(3):300-305
  13. 13. Tyrovola JB, Spyropoulos MN, Makou M, Perrea D. Root resorption and the OPG/RANKL/RANK system: A mini review. Journal of Oral Science. 2008;50(4):367-376
  14. 14. Boyce BF, Xing L. Biology of RANK, RANKL, and osteoprotegerin. Arthritis Research & Therapy. 2007;9(Suppl. 1):S1
  15. 15. Baker PJ, Dixon M, Evans RT, Dufour L, Johnson E, Roopenian DC. CD4(+) T cells and the proinflammatory cytokines gamma interferon and interleukin-6 contribute to alveolar bone loss in mice. Infection and Immunity. 1999;67(6):2804-2809
  16. 16. Vernal R, Dutzan N, Hernandez M, Chandia S, Puente J, Leon R, et al. High expression levels of receptor activator of nuclear factor-kappa B ligand associated with human chronic periodontitis are mainly secreted by CD4+ T lymphocytes. Journal of Periodontology. 2006;77(10):1772-1780
  17. 17. Delima AJ, Karatzas S, Amar S, Graves DT. Inflammation and tissue loss caused by periodontal pathogens is reduced by interleukin-1 antagonists. The Journal of Infectious Diseases. 2002;186(4):511-516
  18. 18. Horwood NJ, Elliott J, Martin TJ, Gillespie MT. IL-12 alone and in synergy with IL-18 inhibits osteoclast formation in vitro. Journal of Immunology. 2001;166(8):4915-4921
  19. 19. Bainbridge B, Verma RK, Eastman C, Yehia B, Rivera M, Moffatt C, et al. Role of Porphyromonas gingivalis phosphoserine phosphatase enzyme SerB in inflammation, immune response, and induction of alveolar bone resorption in rats. Infection and Immunity. 2010;78(11):4560-4569
  20. 20. Ernst CW, Lee JE, Nakanishi T, Karimbux NY, Rezende TM, Stashenko P, et al. Diminished forkhead box P3/CD25 double-positive T regulatory cells are associated with the increased nuclear factor-kappaB ligand (RANKL+) T cells in bone resorption lesion of periodontal disease. Clinical and Experimental Immunology. 2007;148(2):271-280
  21. 21. Glowacki AJ, Yoshizawa S, Jhunjhunwala S, Vieira AE, Garlet GP, Sfeir C, et al. Prevention of inflammation-mediated bone loss in murine and canine periodontal disease via recruitment of regulatory lymphocytes. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(46):18525-18530
  22. 22. Komatsu N, Okamoto K, Sawa S, Nakashima T, Oh-hora M, Kodama T, et al. Pathogenic conversion of Foxp3+ T cells into TH17 cells in autoimmune arthritis. Nature Medicine. 2014;20(1):62-68
  23. 23. Tsukasaki M, Komatsu N, Nagashima K, Nitta T, Pluemsakunthai W, Shukunami C, et al. Host defense against oral microbiota by bone-damaging T cells. Nature Communications. 2018;9(1):701
  24. 24. Kajiya M, Giro G, Taubman MA, Han X, Mayer MP, Kawai T. Role of periodontal pathogenic bacteria in RANKL-mediated bone destruction in periodontal disease. Journal of Oral Microbiology. 2010;2
  25. 25. Komatsu N, Win S, Yan M, Huynh NC, Sawa S, Tsukasaki M, et al. Plasma cells promote osteoclastogenesis and periarticular bone loss in autoimmune arthritis. The Journal of Clinical Investigation. 2021;131(6)
  26. 26. Han X, Kawai T, Eastcott JW, Taubman MA. Bacterial-responsive B lymphocytes induce periodontal bone resorption. Journal of Immunology. 2006;176(1):625-631
  27. 27. Han X, Lin X, Seliger AR, Eastcott J, Kawai T, Taubman MA. Expression of receptor activator of nuclear factor-kappaB ligand by B cells in response to oral bacteria. Oral Microbiology and Immunology. 2009;24(3):190-196
  28. 28. Nguyen TT, Huynh NN, Seubbuk S, Nilmoje T, Wanasuntronwong A, Surarit R. Oxidative stress induced by Porphyromonas gingivalis lysate and nicotine in human periodontal ligament fibroblasts. Odontology. 2019;107(2):133-141
  29. 29. Nguyen TT, Vo DA, Thai TT, Vo TL, Huynh NC. Total oxidant status and total antioxidant capacity in the saliva of patients with chronic periodontitis. Medical and Pharmaceutical Researches. 2020;4(4):21-26
  30. 30. Zhang T, Andrukhov O, Haririan H, Muller-Kern M, Liu S, Liu Z, et al. Total antioxidant capacity and total oxidant status in saliva of periodontitis patients in relation to bacterial load. Frontiers in Cellular and Infection Microbiology. 2015;5:97
  31. 31. Huynh NC, Everts V, Leethanakul C, Pavasant P, Ampornaramveth RS. Rinsing with saline promotes human gingival fibroblast wound healing in vitro. PLoS One. 2016;11(7):e0159843
  32. 32. Belibasakis GN, Bostanci N, Hashim A, Johansson A, Aduse-Opoku J, Curtis MA, et al. Regulation of RANKL and OPG gene expression in human gingival fibroblasts and periodontal ligament cells by Porphyromonas gingivalis: A putative role of the Arg-gingipains. Microbial Pathogenesis. 2007;43(1):46-53
  33. 33. Huynh NC, Everts V, Pavasant P, Ampornaramveth RS. Interleukin-1beta induces human cementoblasts to support osteoclastogenesis. International Journal of Oral Science. 2017;9(12):e5
  34. 34. Shoji-Matsunaga A, Ono T, Hayashi M, Takayanagi H, Moriyama K, Nakashima T. Osteocyte regulation of orthodontic force-mediated tooth movement via RANKL expression. Scientific Reports. 2017;7(1):8753
  35. 35. Silva TA, Garlet GP, Fukada SY, Silva JS, Cunha FQ. Chemokines in oral inflammatory diseases: Apical periodontitis and periodontal disease. Journal of Dental Research. 2007;86(4):306-319
  36. 36. Yu JJ, Ruddy MJ, Wong GC, Sfintescu C, Baker PJ, Smith JB, et al. An essential role for IL-17 in preventing pathogen-initiated bone destruction: Recruitment of neutrophils to inflamed bone requires IL-17 receptor-dependent signals. Blood. 2007;109(9):3794-3802
  37. 37. Dayan S, Stashenko P, Niederman R, Kupper TS. Oral epithelial overexpression of IL-1alpha causes periodontal disease. Journal of Dental Research. 2004;83(10):786-790
  38. 38. Shaker OG, Ghallab NA. IL-17 and IL-11 GCF levels in aggressive and chronic periodontitis patients: Relation to PCR bacterial detection. Mediators of Inflammation. 2012;2012:174764
  39. 39. Gaffen SL. Biology of recently discovered cytokines: Interleukin-17--a unique inflammatory cytokine with roles in bone biology and arthritis. Arthritis Research & Therapy. 2004;6(6):240-247
  40. 40. Garlet GP, Cardoso CR, Campanelli AP, Ferreira BR, Avila-Campos MJ, Cunha FQ, et al. The dual role of p55 tumour necrosis factor-alpha receptor in Actinobacillus actinomycetemcomitans-induced experimental periodontitis: Host protection and tissue destruction. Clinical and Experimental Immunology. 2007;147(1):128-138
  41. 41. Mavrogenis A, Dimitriou R, Parvizi J, Babis GC. Biology of implant osseointegration. Journal of Musculoskeletal & Neuronal Interactions. 2009;9(2):61-71
  42. 42. Mulari M, Qu Q, Härkönen P, Väänänen H. Osteoblast-like cells complete osteoclastic bone resorption and form new mineralized bone matrix in vitro. Calcified Tissue International. 2004;75(3):253-261
  43. 43. Bergomi M, Cugnoni J, Galli M, Botsis J, Belser UC, Wiskott HW. Hydro-mechanical coupling in the periodontal ligament: A porohyperelastic finite element model. Journal of Biomechanics. 2011;44(1):34-38
  44. 44. Yarmolyuk Y. Mechanical Stress Modulates Expression of Toll-Like Receptors in Human PDL. Milwaukee, Wisconsin: Marquette University; 2012
  45. 45. Diercke K, Kohl A, Lux CJ, Erber R. IL-1beta and compressive forces lead to a significant induction of RANKL-expression in primary human cementoblasts. Journal of Orofacial Orthopedics = Fortschritte der Kieferorthopadie: Organ/Official Journal Deutsche Gesellschaft fur Kieferorthopadie. 2012;73(5):397-412
  46. 46. Diercke K, Sen S, Kohl A, Lux CJ, Erber R. Compression-dependent up-regulation of ephrin-A2 in PDL fibroblasts attenuates osteogenesis. Journal of Dental Research. 2011;90(9):1108-1115
  47. 47. Matsuo K, Otaki N. Bone cell interactions through Eph/ephrin: Bone modeling, remodeling and associated diseases. Cell Adhesion & Migration. 2012;6(2):148-156
  48. 48. Kawasaki K, Shimizu N. Effects of low-energy laser irradiation on bone remodeling during experimental tooth movement in rats. Lasers in Surgery and Medicine: The Official Journal of the American Society for Laser Medicine and Surgery. 2000;26(3):282-291
  49. 49. Sun X, Zhu X, Xu C, Ye N, Zhu H. Effects of low energy laser on tooth movement and remodeling of alveolar bone in rabbits. Hua xi kou qiang yi xue za zhi= Huaxi kouqiang yixue zazhi=. West China Journal of Stomatology. 2001;19(5):290-293
  50. 50. Yamaguchi M, Hayashi M, Fujita S, Yoshida T, Utsunomiya T, Yamamoto H, et al. Low-energy laser irradiation facilitates the velocity of tooth movement and the expressions of matrix metalloproteinase-9, cathepsin K, and alpha (v) beta (3) integrin in rats. The European Journal of Orthodontics. 2010;32(2):131-139
  51. 51. Fujita S, Yamaguchi M, Utsunomiya T, Yamamoto H, Kasai K. Low-energy laser stimulates tooth movement velocity via expression of RANK and RANKL. Orthodontics & Craniofacial Research. 2008;11(3):143-155
  52. 52. Cruz DR, Kohara EK, Ribeiro MS, Wetter NU. Effects of low-intensity laser therapy on the orthodontic movement velocity of human teeth: A preliminary study. Lasers in Surgery and Medicine: The Official Journal of the American Society for Laser Medicine and Surgery. 2004;35(2):117-120
  53. 53. Seifi M, Shafeei HA, Daneshdoost S, Mir M. Effects of two types of low-level laser wave lengths (850 and 630 nm) on the orthodontic tooth movements in rabbits. Lasers in Medical Science. 2007;22(4):261-264
  54. 54. Zhu X, Chen Y, Sun X. A study on expression of basic fibroblast growth factors in periodontal tissue following orthodontic tooth movement associated with low power laser irradiation. Hua xi kou qiang yi xue za zhi= Huaxi kouqiang yixue zazhi=. West China Journal of Stomatology. 2002;20(3):166-168

Written By

Nam Cong-Nhat Huynh

Submitted: 28 June 2022 Reviewed: 19 July 2022 Published: 14 August 2022