Open access peer-reviewed chapter

Cytokines’ Involvement in Periodontal Changes

Written By

Petra Surlin, Liliana Foia, Sorina Solomon, Dora Maria Popescu, Dorin Nicolae Gheorghe, Adrian Camen, Maria Alexandra Martu, Anne Marie Rauten, Madalina Olteanu, Allma Pitru, Vasilica Toma, Simona Popa, Mihail Virgil Boldeanu, Silvia Martu and Ion Rogoveanu

Submitted: 25 February 2019 Reviewed: 01 October 2019 Published: 19 August 2020

DOI: 10.5772/intechopen.89999

From the Edited Volume

Cytokines

Edited by Payam Behzadi

Chapter metrics overview

866 Chapter Downloads

View Full Metrics

Abstract

The bacterial challenge on the periodontal tissues triggers an inflammatory reaction, driven by pro-inflammatory cytokines, that eventually leads to the periodontal structures’ damage. The pathogenic mechanisms of this inflammatory reaction are complex and are influenced by the type of host-immune response and certain local and systemic factors. These factors can influence periodontal inflammation, through the action of the various pro-inflammatory cytokines. Periodontal disease and certain systemic conditions can have a mutual association, as the pathogenic mechanisms of these diseases can involve similar molecular and cellular elements. The concept of ‘periodontal medicine’ comprises these pathogenic connections, focusing on the key role that periodontal health has on the general homeostasis and well-being.

Keywords

  • periodontal disease
  • cytokines
  • inflammation
  • systemic conditions
  • associations

1. Introduction

Periodontal disease is defined as an immune, inflammatory disease with a triggering bacterial factor [1]. Its initial phase is characterised by damage to soft gingival tissue (inflammation of gums, gingivitis, plaque accumulation), followed by degradation of the periodontium (chronic inflammation, colonisation by periodontal pathogenic anaerobes) and the surrounding connective tissue matrix of teeth, along with alveolar bone loss, local citrullination, bone resorption and rapid loss of teeth, in the destructive phase [2].

It is considered a multifactorial disease with progressive evolution and pathophysiological mechanisms that involve the association of environmental factors (smoking, stress), microbial factors (several bacterial species, for example, Porphyromonas gingivalis, Treponema denticola, Tannerella forsythia, Aggregatibacter actinomycetemcimtans), and genetic polymorphism (human leukocyte antigen-antigen D, HLA-DR, HLA-DRB1; IL-1, IL-6 and IL-10 gene polymorphism) with the inflammatory and immune response (both innate and adaptive immunity) of the host (Table 1) [3, 4]. These mechanisms will lead to defective host defences, which will contribute to changes in tissue homeostasis, inflammation and bone loss, the main features of periodontal disease (Figure 1) [2].

General functionTypeSpecific functionPeriodontal implication
Pro-inflammatoryIL-1βIncreases production of other pro-inflammatory mediators (PGE2, IL-6); stimulates neutrophil activityKey role in the pathogenic processes; enhances alveolar bone-resorption; fuels the inflammatory reaction
IL-1α‘Alarmin’ for tissue damage and immune system; interacts with TNF-α; induces protease synthesisEnhances alveolar bone-resorption by signalling the presence of bacterial antigens and enhancing the inflammatory response
IL-6Regulates cell growth and differentiation: osteoblasts, B and T lymphocytesImpairs osteoblast growth and function; increases osteoclast formation from monocytes
IL-18Increases neutrophil activity and interferon gamma productionInfluences lymphocyte Th1/Th2 differentiation; interacts with IL-1β
IL-33Activates Th2 and mast cells; stimulates production of IL-5, IL-13 by Th2 cellsInduction of RANKL; induces periodontal damage by stimulation of Th2 cells
TNF-αMajor regulator of immune cells’ activity; involved in the acute phase reactionStimulates damage (by osteoclasts) and prevents repair of periodontal tissues (by fibroblast death); starts IL-1β, PGE2 synthesis
PGE2Increases production of other pro-inflammatory mediators (MMPs); induces feverContributes to bone resorption by increasing osteoclast activity
MMPs (-1, -8, -9)Enzymatic degradation of collagen (and other extra-cellular matrix proteins). Stimulated by IL-1β and TNF-α. Influences immune cell migration and adhesionPeriodontal damage by impairment of collagen type I production and degradation of structural collagen; causes activation of osteoclasts and damage to connective tissue
Anti-inflammatoryIL-1RaInhibits IL-1β, IL-1α activity by preventing cellular signallingLimits alveolar bone-resorption; regulates response to antigens (LPS)
IL-10Decreases cytokine production by immune cells; reduces inflammatory responseDown-regulates periodontal inflammation by reducing cytokine synthesis in immune cells
IL-4Stimulates tissue repair and regulates immunity; regulates differentiation of Th2 cellsDecreases production of Th2 cells, with important implications to periodontal damage
TGF-βEnhances epithelial regeneration/repairStimulates gingival fibroblast activity

Table 1.

Mediators’ role in general and periodontal inflammatory reactions [3].

Figure 1.

The periodontium and its inflammatory reaction in periodontal disease: a—bacterial plaque deposits; b—gingival sulcus; c—inflammatory mediators (interleukins—ILs, matrix-metalloproteinases—MMPs, tissue inhibitors of MMP—TIMPs); d—lymphocytes type B and T; e—polymorphonuclear cells; f—blood vessel; g—macrophage cells; h—osteoclast cells; i—gingival epithelium; j—periodontal ligament; k—gingival connective tissue; and l—alveolar bone.

Pathophysiological processes are explained by the participation of a wide range of locally released soluble factors such as pro-inflammatory cytokines, prostaglandin E2 and reactive oxygen species, inflammatory mediators that can be highlighted by various methods within gingival tissues and within the gingival crevicular fluid (GCF) [5]. The association between elevated TNF-α and IL-6 concentrations and disease activity was also highlighted [6]. It has been shown that IL-1β and interferon-gamma (IFN-γ) have elevated concentrations in active periodontal lesions [7]. IL-12, a key-acting cytokine mediating Th1 differentiation, also implicated in cell-mediated immunity, has been observed to stimulate pro-inflammatory proteins involved in bone resorption [8].

A deficiency in the synthesis and release of anti-inflammatory cytokines can also occur, like type Th2 cytokines (IL-4, IL-5, IL-13), IL-10 and transforming growth factor beta 1 (TGF-β1), which confirms the existence of an imbalance between pro- and anti-inflammatory mechanisms at periodontal level [9]. Along with TNF-α, IL-1β and IL-6, the release of the cytokines IL-8, IL-11, and IL-17 has also been revealed within GCF, which has potent osteoclastogenesis stimulating effects and also reduces osteoprotegerin synthesis in osteoblasts and stromal cells [10].

Periodontal disease can be influenced by certain local factors, such as orthodontic therapy and the existence of coronal or prosthodontic restorations, and by certain systemic diseases, this may interfere with the periodontal inflammatory reaction, by means of pro-inflammatory cytokines. These diseases include diabetes mellitus (type 1 and 2), cardiovascular diseases, rheumatoid arthritis and hepatic and renal conditions. The interactions between periodontal tissues and their pathology and these systemic diseases have been reunited under the concept of ‘periodontal medicine’ [11]. Considering this concept, the chapter aims to exhibit the major implications of cytokines into the pathogenic mechanisms of periodontal disease and its local and systemic influencing factors.

Advertisement

2. Methodology

The design of the chapter has been created in order to reflect the role of cytokines in periodontal pathogenic processes, as both local and systemic risk factors for periodontal disease have been taken into consideration. The information is divided between the influence of local elements—orthodontic and restorative treatments, and systemic ones—diabetes mellitus type 1 and 2, cardiovascular, rheumatic, hepatic and renal diseases. Relevant scientific information has been sourced from the existing scientific literature and structured so as to pursue the established purpose of the chapter.

Advertisement

3. Local influences on the periodontal inflammatory reaction

3.1 Orthodontic influences

The orthodontic dental movement is the consequence of the application of controlled mechanical forces on the teeth [12], the periodontal ligament (PDL) being the one that mediates the task to which the teeth and the alveolar bone are subjected and creates the conditions for the cells to participate in bone remodelling [13]. Orthodontic forces distort the PDL matrix, causing the cellular form and cytoskeleton configuration to change, with neuropeptides release from the afferent nerve terminals [14], while the orthodontic forces can induce a biomolecular-level release of growth factors, prostaglandins and pro-inflammatory cytokines as IL-1, IL-6, IL-8 and TNF-α, that affect alveolar bone remodelling [15]. At present, the molecular mechanisms underlying bone formation, induced by stretching forces, are not fully understood [16]. The initial phase of orthodontic dental movement always involves an aseptic acute inflammatory reaction [17], which lasts for 1–2 days and is predominantly exudative, followed by a chronic, mainly proliferative process [18] and increased release of cytokines [19], such as TNF-α, TNF-β, IL-1, PDGF, INF-γ and RANKL [15, 20].

Inflammatory cytokines are involved in all phases of inflammation during orthodontic treatment [21]; pro-inflammatory and anti-inflammatory cytokines act synergistically or antagonistically on each other [22]:

  • IL1 family: all 3 ligands of the IL-1 family (IL-1α, IL-1β, and IL-1RA) are involved in bone metabolism and orthodontic dental movement [22], as well as IL-1 gene polymorphisms [23]. IL-1β along with TNF-α is a key pro-inflammatory cytokine in acute-phase inflammation [24]. IL-1β gathers leukocytes and activates fibroblasts, endothelial cells, osteoclasts and osteoblasts in order to stimulate bone resorption and to stop bone formation [25]. IL-1β is also a PGE inducer and, along with mechanical stress, synergistically regulates the formation of PGs in periodontal cells [26]. The levels of IL-1β and PGE2 are higher in the tension zones compared to compression zones, which supports the hypothesis that during the initial stage of orthodontic treatment, this cytokine would originate from osteoclasts in response to mechanical stress [27].

  • IL-1RA acts by limiting the inflammatory conditions [28] mediated by IL-1 and bone resorption [22], and therefore positive correlations exist between decreasing IL-1RA levels in GCF and faster bone resorption during orthodontic dental movement and consequently a higher dental movement speed [29].

    The tooth movement speed is influenced by stress and by levels of IL-1RA, IL-1β and IL-1 gene polymorphisms from GCF. These factors provide a better predictive model for the efficiency of dental movement: activity index [AI = experimental (IL-1β/IL-1RA)/control (IL-1β/IL-1RA)], IL-1RA concentration in GCF and IL-1β genotype [29].

  • IL6: IL-6 has a stimulating effect on bone remodelling and osteoclast formation [30] and also in the inflammation associated with orthodontic dental movement [31].

  • IL8: IL-8 has a role in the neutrophils’ recruitment and activation in the presence of inflammation [32], in improving RANKL expression and consequently in increasing the osteoclast production and their activation [33]. Immediately after the application of mechanical forces, IL-8 has an increased level in both pressure and tension zones [34], but later on, the stimulation of IL-8 secretion only continues in the tension zones; this differential regulation probably plays a major role in the initial stage of bone remodelling [34].

  • TNF-α: TNF-α binds with macrophage colony-stimulating factors to induce osteoclast differentiation [35].

  • IFN-γ: IFN-γ increases during late stages of orthodontic dental movement [35], controlling massive osteoclastogenesis [36] related to the increased volume of trabecular bone [37].

The levels of cytokines in the GCF vary with the type and intensity of the applied force, speed of tooth movement, the age of the orthodontic device owner [24] and growth [38]. Equivalent force systems during orthodontic dental movement induce an individualised production of different cytokines [39]. Light continuous forces tend to maintain relatively high levels of IL-1β [29], needed for the continuation of periodontal remodelling, longer periods allowing a reduced frequency of reactivations [38]. A strong force may increase the risk of root resorption and hyalinization of PDL and may also modify the cytokines’ level, causing unwanted tissue reactions and the need for multiple reactivations [40]. The increase of IL-1β’s level in GCF, as a result of the increased applied force, was also associated with intense pain during orthodontic dental movement [38], probably due to the correlation between IL-1β and substance P [41]. Most studies did not detect differences between the levels of cytokines in the tension and pressure zones [29, 34], probably due to the continuous circulation of GCF in the periodontal ligament. Consequently, it can be concluded that GCF cytokine levels cannot be specific indicators of periodontal remodelling in tension and pressure zones [42].

External apical root resorption (EARR), secondary to orthodontic dental movement, is a frequent clinical complication, and treatment variables, environment factors and/or inter-individual genetic variations may give susceptibility or resistance to its occurrence [43]. Among pro-resorptive cytokines, IL-6 promotes osteoclastic function [44] and amplifies the production of fibroblasts from gingival fluid and periodontium [45], having increased levels in patients with severe dental root resorption [46]. IL-7 acts indirectly on osteoclastogenesis by induction of TNF-α [47]. The increase of IL-6, IL-7 and TNF-α level might be a sign of a continuous remodelling of the periodontal tissues during the lag phase of tooth movement and a mechanism of cellular prohibition [48].

Regarding anti-resorptive cytokines, the levels of IL-4 and IFN-γ increase closely following IL-1β’s elevation. This cytokine’s expression type may result as an active combined remodelling of periodontal tissue, during the first stages of dental movement and also of cellular prohibition mechanisms, preventing activation and additional differentiation of osteoclastic cells [48] and suppressing osteoclastogenesis, unlike T cells of RANKL that induce osteoclastogenesis [49]. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is also an anti-resorptive cytokine that prevents bone resorption together with IL-4, IL-10, IL-13, IL-18 and IFN-γ [50].

3.2 Restorative influences

The interplay between periodontology and restorative dentistry exists on many levels and functions, in both directions [51]. For example, interproximal restorations, fixed prosthesis and artificial crowns can be involved in the occurrence and progression of gingival inflammation and periodontal destruction [51]. The presence of dental restorations or other appliance, near or below the gingival margin, which reach deep into the gingival sulcus or within the junctional epithelium, may induce localised inflammation that could lead to future periodontal complications [51].

The most reliable mechanism by which the subgingival margins of restorations lead to gingival inflammation and loss of attachment is the increase of plaque accumulation. In these conditions, the rate flow of gingival fluid will increase, because the gingival fluid protects oral tissues (including the junctional epithelium) against the bacterial invasion, acting as a defensive barrier, within the gingival sulcus [52]. IL-1, prostaglandin E2 (PGE2) and the elements that affect collagen and bone (matrix metalloproteinases (MMPs)) that are detected in gingival fluid can be used as possible indicators for the diagnosis of periodontal disease and its progression [2]. The placement of restorative posts can induce trauma to periodontal structures, mostly in the case of metallic posts and less for the fibre ones, leading to periodontal inflammation [53]. IL-1 family cytokines regulate the activity of other pro-inflammatory cytokines, such as PGE2 and MMPs, their gingival fluid levels being proportionate with the degree of bone loss [10].

The placement of the gingival margins of restorations depends on the position of the decayed portion of the tooth and the extent of caries. The materials used in restorations must not be injurious to periodontal tissues, as they should have highly polished surfaces, the periodontium reacting to the roughness of the material and its accumulation of plaque by inflammation. However, it has been shown that the composition of restorative materials could also initiate periodontal changes, by monocyte activation and changes of the gingival fluid levels of cytokines [54]. It has been observed that periodontal inflammation can occur in areas of the gingival tissue which are adjacent to ceramic, composite or amalgam coronal restorations, even if there are no signs of bacterial plaque accumulation. This may be explained by the stimulation of periodontal neurogenic inflammatory reactions that is caused by these restorative materials. However, these findings are controversial and require further research, as it has been reported that only class V composite fillings can trigger such forms of gingival inflammation, in the areas of their vicinity [55].

Ceramic materials have the lowest plaque retentive capacity, but inner materials of porcelain-fused-to-metal (PFM) crowns can impact periodontal tissues [56]. Certain non-noble metals could have a negative impact on periodontal tissue (Ni-Cr alloy, Co-Cr alloy). It seems that the Au-Pt alloy is least harmful for the periodontium. The spaces between the margins of the restoration, as well as their contour are other contributing factors to gingival inflammation and periodontal destruction, referred to as iatrogenic factors. Restoration margins have been reported as key factors for periodontal health [57]. Another feature referring to periodontal damage is the dental impression that needs to offer a clear image and perspective of the prepared teeth, the neighbouring teeth and the associated gingival tissue. Impressions of tooth preparations with an elastic material that extends subgingivally could damage soft tissues. In order to prevent this, the retraction cord is frequently used during impression taking [58]. Successful restorative and prosthetic treatments require a healthy periodontium, as a start point of therapeutic protocols [51].

Advertisement

4. Systemic pathologic influences on the periodontal inflammatory reaction

4.1 Type 2 diabetes

The relationship between type 2 diabetes mellitus (T2DM) and chronic periodontitis is bidirectional [59]. The deleterious effect of chronic hyperglycemia on the occurrence and progression of periodontitis has been observed in patients suffering from both diseases [59]. Among the factors involved in the pathogenesis of periodontitis in patients with T2DM are the advanced glycation end products, chronic inflammation, altered secretion profile of cytokines, decrease of the immune response to infection and also the exacerbation of bacterial flora at periodontal level [59].

Recent evidence has suggested the role of periodontitis in the pathogenesis of insulin resistance, the adverse effects of periodontitis on glycemic control, and the contribution to the occurrence and progression of chronic diabetic complications [60]. The link between periodontitis and T2DM is potentially explained by the increase of pro-inflammatory cytokines, such as IL-1β, IL-8, IL-6, TNF-α, IL-12, and leptin and decrease of anti-inflammatory cytokines such as IL-4, IL-11, adiponectin and fibroblast growth factor-21 [60, 61].

The assessment of cytokine levels within the gingival crevicular fluid indicated that subjects with T2DM and chronic periodontal disease have significantly higher levels of IL-1β, IL-6 and TNF-α than healthy participants with similar periodontal conditions [59]. The levels of IL-8 within the gingival crevicular fluid and plasma have been evaluated with contradictory results in the context of T2DM and chronic periodontitis, multiple studies reporting that the IL-8 level is higher in patients with T2DM and chronic periodontitis compared to non-diabetic subjects with or without chronic periodontitis [60, 62].

The systemic inflammatory response depends on the ratios of pro-inflammatory to anti-inflammatory cytokines. The patients with T2DM and chronic periodontitis tend to have higher ratios of pro-inflammatory to anti-inflammatory cytokines compared to periodontally and systemically healthy controls [63]. Thus, TNF-α/IL-4, IL-1β/IL-4, IL-23/IL-4, IL-6/IL-4, TNF-α/IL-5, IL-17/IL-5 and IL-6/IL-5 ratios were higher in T2DM patients with chronic periodontitis than in healthy subjects [64].

The literature regarding the systemic plasma cytokine profile’s characteristics in patients with T2DM and chronic periodontitis is not consistent. Some reports indicated lower levels of cytokines, including IL-4, IL-5, IL-6, TNF-α, IL-1β, IL-17, IL-13, IFN-γ, IL-2, IL-23 and IL-12 and higher levels of IL-8 in serum samples of T2DM patients with chronic periodontitis than for the control group [60, 63]. There are other studies showing that the systemic levels of some cytokines, such as IL-4, IL-8, IL-6, IL-10 and TNF-α, did not differ between diabetic and non-diabetic subjects with periodontitis [62, 64].

The improvement of glycemic control after periodontal therapy may be explained by the fact that periodontal treatment relives the periodontal inflammatory reaction and further decreases systemic pro-inflammatory cytokines’ involvement in the pathogenesis of insulin resistance [65]. Reduction of the pro-inflammatory cytokines such as IL-1β and IL-6 in gingival crevicular fluid and improvement of glycemic control after periodontal therapy was reported in T2DM patients [66]. Furthermore, a significant decrease in serum levels of IL-6 and TNF-α and also a reduction of HbA1c in patients with T2DM were reported after periodontal therapy, indicating the impact of periodontal intervention on periodontal and systemic inflammation related to insulin resistance [65, 66]. Recent findings show that reducing periodontal inflammation by periodontal therapy may contribute to an increase of systemic anti-inflammatory cytokines, such as adiponectin and fibroblast growth factor-21 levels, and to a decrease of leptin levels, thus improving insulin sensitivity [65].

As high levels of pro-inflammatory cytokines have been reported at periodontal level, associated with increased risk of destructive effects within the periodontal tissues [67], it was shown that diseases characterised by insulin resistance like T2DM are also associated with the increase of cytokines [68, 69]. Chronic periodontitis may influence systemic cytokines in T2DM. The literature documents the role of cytokines TNF-α, IL-1β, IL-4, IL-6 and IL-10 in chronic periodontitis and T2DM [70].

Pathogenic links between periodontal disease and diabetes involve elevations in IL-1β [71], TNF-α, IL-6, RANKL-b, oxidative stress and Toll-like receptor (TLR) expression. It has been demonstrated that prolific circulatory mediators have higher levels in the association of diabetes with a form of periodontal disease, especially TNF-α, C-reactive protein (CRP) and mediators of oxidative stress, which in turn can affect the control of diabetes. Moreover, complete and correct periodontal treatment can improve serum levels of CRP and TNF-α in patients affected by diabetes [65, 72].

The release of inflammatory mediators that can be detected in GCF stimulates the secretion of metalloproteinases, initiates bone resorption processes and plays an important role in the evolution and prognosis of periodontal disease. Inflammatory cytokine and chemokine levels in GCF decreased after initial periodontal therapy [73]. In periodontitis, concentrations of pro-inflammatory cytokines TNF-α and IL-8 increased not only in periodontal tissues [74] but also in serum samples [62].

4.2 Type 1 diabetes

The existence of a bidirectional relationship between periodontal disease and type one diabetes (T1D) has long been considered [75]. Periodontal breakdown is a pivotal aggravating factor for the health status in subjects with T1D, mainly because this preserves a chronic systemic inflammatory condition, contributing therefore to diabetic complications [75]. In fact, periodontitis has been considered to be the sixth complication of diabetic disease, diabetic metabolic impairment being in turn able to point toward a poor periodontal health status. Being not only a metabolic misbalance, but also tightly associated to an important dysfunction of the immune system, several facets of the systemic immune response, such as antigen challenge or polymorphonuclear leukocyte and T-lymphocyte function are altered in diabetic subjects [76].

Hyperglycemia can lead to immune system disorders; hence, the effects mediated by cytokine alterations in patients with T1D might be significant. IL-1β levels in gingival fluid and those of IL-6 in saliva have been correlated with glycosylated haemoglobin [77]. The changes in the cytokine amount on the systemic level are crucial in the pathogenesis of diabetes and can condition the islet cell turnover and apoptosis, with subsequent disease progression toward devastating complications, as macro- and microvascular modifications [78].

Regarding the involvement of cytokines in the immune-inflammatory response in subjects with T1D and periodontal breakdown, the literature highlights the enhancement of IL-1β, IL-6 and prostaglandin E2 in the gingival fluid of these patients, compared to systemically healthy subjects, with comparable periodontal alteration [79, 80]. The results on experimental murine models of induced diabetes include data on a broader range of mediators: IFN (interferon), chemokines such as macrophage inhibiting protein (MIP-2) and monocyte chemo-attractant protein (MCP-1), most likely mediated via TNF-α [81].

The importance of TNF for enhancing the immune response, generated by bacterial plaque challenge, in T1D and T2D, has been experimentally shown for murine models with chronic periodontitis, but without a clear relation between the TNF-α levels found in oral tissues and those found in oral fluids of T2D periodontal subjects [81]. Along with these pro-inflammatory mediators, research has also been carried upon chemokines’ performance, growth factors and soluble adhesion molecules, which possess immune-regulatory capacity [79]. The simultaneous action of multiple mediators in evaluating oral immune response in periodontal patients with T1D has also been assessed [82].

Monocytes can have a hyper-inflammatory phenotype in patients with T1D, and these cells are susceptible to the action of lipopolysaccharides (LPS) of the periodontal bacteria, and respond through generation of increased amounts of IL-1β, TNF-α and PGE2, compared to non-diabetic subjects [79]. This inflammatory phenotype of monocytes represents one of the relevant links between periodontal pathogenesis and diabetes mellitus [79].

Research on other cell populations involved in mediating the immune response advocates that T cells accumulation around insulin-sensitive cells is important in metabolic changes correlated to diabetes, via their capacity to moderate macrophage activity [83]. There are a number of distinct subclasses of T cells, with remarkable plasticity, their role being strongly correlated to the local cytokine amount. However, although important in maintaining the Th1/Th2 cell balance in the pathogenesis and progression of periodontitis, data on the underlying mechanisms and their role in homeostasis of periodontal status in patients with diabetes are still to be elucidated [76].

Gathering the whole data related to the modulation of locally expressed mediators, the T1D-periodontitis association would be of particular relevance in the curative management plan, through conduction of long-term studies. Further research is necessary to better understand the cytokine expression in periodontal disease and type 1 diabetes.

4.3 Cardiovascular diseases

Cardiovascular disease (CVD) constitutes an extensive cluster of conditions that deter the physiological function of the heart and/or blood vessels and includes (a) coronary heart disease—angina/myocardial infarction; (b) ischaemic cerebrovascular disease—transient ischaemic attack—TIA and (c) peripheral vascular disease. In both cardiovascular disease and periodontal disease, a systemic inflammatory overload is present in the organism, which can be exacerbated by factors such as smoking, obesity or diabetes mellitus that eventually leads to an altered dishomeostatic status [84]. Due to the ulceration and inflammation of subgingival epithelial layer, oral pathogens and their by-products can access the blood stream, increasing the risk and aggravating the evolution of any pre-existing heart disease [85].

Atherosclerosis is commenced by deterioration of the endothelial tissue of vessels. Following impairment, endothelial pro-inflammatory signals drive not only the expression of adhesion molecules such as E-selectin, ICAM-1, P-selectin and VCAM-1, but also of IL-8 and thrombin that act as chemoattractants and determine an upsurge in the aggregation of platelets and migration of leukocytes [86]. Furthermore, these pro-inflammatory signals trigger the proliferation of smooth muscle cells and the apoptosis of endothelial cells. In this context of complex interactions, leukocytes migrate to the injured site and release additional pro-inflammatory cytokines (IL-1α and β, IL-6, IL-17, IL-22 and TNF-α), reactive oxygen species, as well as proteinases that break down the extracellular matrix of the endothelium [87].

High concentrations of low-density lipoproteins (LDL) in plasma accumulate in the aortic wall beneath the intima layer of the endothelium and are oxidated into oxLDL. Increased blood pressure activates endothelial cells which advocate the activation of adhesion molecules that favour the migration of monocytes. In the aortic wall, these cells transform into macrophages that absorb the oxidated LDL and develop into foam cells filled with lipids. This circumstance further promotes the dispersion of pro-inflammatory molecules which stimulate the invasion and activation of supplementary inflammatory cells and facilitate their confinement in the plaque, precipitating subsequent build-up of inflammation factors [88].

Periodontal microorganisms can alter the mentioned mechanisms by virtue of a direct interaction (for example by the invasion of endothelial, smooth muscle cells, leukocytes and platelets), but also an indirect interplay by stimulating the release of paracrine factors that eventually affect normal cellular function [89]. In addition to direct invasion, microorganisms can release products into the circulation and induce pro-atherogenic responses in endothelial cells [90]. Research has highlighted that vesicles pertaining to the outer membrane such as gingipains from P. gingivalis and free soluble components deriving from A. actinomycetemcomitans cause irritation to endothelial cells and promote inflammation [91]. Effusion of pro-inflammatory cytokines (IL-1, IL-6 and TNF-α) and other chemokines from the affected periodontal tissues leads to the generation of acute phase proteins (C reactive protein, fibrinogen, amyloid A, etc.) by the liver [92]. As a result of this injury, the activation of the adaptive immune system occurs as a typical response to chronic bacteraemia. Pathogen-associated molecular patterns (PAMPs) of periodontal pathogens determine the production of antibodies and elicit a cross-response between endothelial cells and the altered LDL to exacerbate the migration of lipids into cells inside the blood vessel walls [93]. Antigen-presenting cells, for example, dendritic cells and effector T lymphocytes, bear a substantial part in the generation of proatherogenic cytokines, for example, IL-20, IL-18 and IFN-γ, and are consequently also relevant in the progression of atherosclerotic plaques. A noteworthy role among the proatherogenic cytokines has been assigned to IL-12 due to the fact that when this certain molecule was absent early lesion development was inhibited. However, late progression was unaffected [94].

Considering the recent epidemiological, experimental and clinical evidence presented in the literature on this subject matter [86], the issue of an interrelationship between periodontal disease and cardiovascular disease is clearly supported. In spite this, the exact nature of this relationship, whether it is a direct or causal one, remains to be edified through interventional and longitudinal studies.

4.4 Rheumatoid arthritis

Rheumatoid arthritis (RA) is considered to be an autoimmune, progressive, inflammatory and chronic disorder, in which the human immune system reacts in an erroneous manner to the articular structures. This response is characterised by an inflammatory immune cell infiltrate (activation of innate immunity, followed by the emergence of adaptive immune responses) in the early phase, followed by a destructive phase characterised by degenerative phenomena, such as denaturation of normal synovial structures by hyperproliferation, reactive fibro-vascular proliferation, bone destruction and cartilage degradation. This explains the occurrence of swelling and pain within and around joints [95].

Over the past two decades, RA has been described as a disease model in which various pathophysiological mechanisms have been studied to better explain the inflammatory process, but also the modality of involvement of the human immune system, through two types of specific responses [95]. These research projects culminated with the introduction into patients’ treatment regimens of the chimeric anti-TNF-α monoclonal antibodies (infliximab, 75% human and 25% mouse peptide sequences) [96], a recombinant human TNF receptor (p75)–Fc fusion protein (etanercept) and biological therapy with fully human anti-TNF-α monoclonal antibodies [97].

Major cytokines, such as TNF-α and interleukins (IL-17 and IL-1β), function by promoting inflammatory responses, causing inflammation of the synovium and inducing cartilage degradation. Other soluble mediators, such as cytokines released by the Th2 lymphocyte subpopulation (the most frequently studied and for which statistically significant results were obtained, would be IL-4, IL-10, IL-13) are mainly anti-inflammatory molecules [95, 98]. In addition, IL-13 could have potential clinical importance because it can suppress both secretion and actions of IL-17 [98].

Over the past two decades, the existence of links or associations between RA and periodontitis has been investigated [99]. RA could be a triggering risk factor for periodontitis as there is a high incidence of RA in patients with periodontal disease, and RA patients have more chances to experience moderate to severe periodontal changes, as compared to healthy subjects. It has also been found that calcifications and soft tissue injuries have the same characteristics, leading to the conclusion that the destructive inflammatory chronic lesions are similar [100].

New theories have highlighted that periodontal disease is a risk factor for RA, with several similarities existing between RA and periodontitis [101, 102, 103]:

  • both diseases are multifactorial, chronic immune-inflammatory diseases with progressive evolution;

  • pathophysiological mechanisms involve the association of environmental factors, microbial factors, genetic susceptibility (it was found that the HLA-DRB1 subtype is associated with both fast progressive periodontitis as well as RA);

  • locally, the cellular composition of the inflammatory reactions is similar, it includes both the T and B subtypes of lymphocyte populations, but also the cells involved in the activation of lymphocytes such as dendritic cells;

  • for both diseases, it was found that the changes occurring in early phases, as well as those in destructive phases are characterised by degenerative phenomena and periodontal degradation, but also the chronic systemic inflammation is determined and mediated by the interruption of balance between the pro-inflammatory and the anti-inflammatory cytokines; and

  • change in connective tissue and bone homeostasis is irreversible, consisting in the deterioration of collagen-rich structures, a process in which they intensively participate and have collagenolytic effects as matrix metalloproteinases and other enzymes (elastase, bacterial cysteine proteases, enzymes associated with neutrophils).

4.5 Liver diseases

One of the most damaging hepatic diseases is chronic hepatitis C (CHC), occurring after the infection with the hepatitis C virus (HCV), which replicates within hepatic and peripheral blood cells [104]. It is considered that worldwide, more than 200 million persons are affected by this disease, making it a major public health concern [105]. As the hepatic inflammation progresses and becomes chronic, healthy liver tissue is replaced by fibrotic tissues, which is unable to perform normal hepatic functions, leading to cirrhosis [106]. During chronic hepatic inflammation important cytokine profile changes can be observed in affected patients, with increased levels of pro-inflammatory markers, such as TNF-α, being found in their serum samples [107].

Cytokine levels, measured in either gingival fluid or serum samples, can be used to assess the progression and severity of both periodontal and hepatic disease [108]. In periodontal disease, the secretion rate of gingival fluid increases, as well as its content in pro-inflammatory cytokines [109]. The levels of these cytokines can be determined, as an indirect indicator of the disease’s severity and progression rate [110]. Being easy to sample, the gingival fluid has allowed extensive research on its pro-inflammatory cytokine content [111]. Similarly, the evolution of chronic hepatitis C can be monitored by the assessment of some cytokine’s levels (such as IL-18 and IL-33) in the serum samples of affected patients [112, 113]. Moreover, the same pro-inflammatory cytokines, such as IL-1, Il-6 and interferon-gamma, can express elevated levels during chronic hepatic inflammation (as in chronic hepatitis C), as well as during chronic periodontal inflammation (as in chronic periodontal disease) [114]. The similar profiles of these cytokines suggest that the two inflammatory reactions could be driven by the same pro-inflammatory markers, endorsing extensive research on their possible common pathogenic mechanisms [108].

Another frequently encountered hepatic condition, particularly within developed regions, is non-alcoholic fatty liver disease (NAFLD). Affecting almost 24% of the global population [115], the disease is mainly caused by genetic or behavioural factors, such as a misbalanced diet, rich in lipids and sugars, lack of physical activity or can occur during other systemic conditions like obesity and diabetes mellitus [116]. The accumulation of fat leads to hepatic steatosis, which, in time, can develop life-threatening complications, as liver cirrhosis or hepatic cancer [117]. NAFLD has been associated with metabolic disorders, as insulin resistance [118], as the adipocytes of the fatty tissue will produce increased levels of TNF-α [119]. This cytokine can alter cellular sensitivity to insulin, therefore decreasing normal glucose metabolism. As a result, serum glucose will rise, increasing the risk of diabetes mellitus [118].

TNF-α is also intensively involved in periodontal inflammation. This cytokine has been shown to express increased levels in the gingival fluid of periodontal patients [120] and has similar pro-inflammatory mechanisms to IL-1β [121]. Its main damaging role during periodontal inflammation is the disruption of the normal reparatory function of fibroblasts and the stimulation of bone-resorption osteoclasts [122]. TNF-α may represent a key cytokine that links the pathogenic mechanisms that are common between periodontal disease and insulin resistance, insulin resistance and NAFLD, and consequently between periodontal disease and NAFLD [123].

The relationship between periodontal disease and hepatic conditions could be considered as functioning into two directions. Firstly, periodontal disease can impact the development and outcome of hepatic conditions. The main explanation of the root cause of the association between periodontal disease and hepatic conditions seems to be the bacterial challenge that leads to periodontal inflammation [124]. Pre-existing hepatic conditions, as chronic hepatitis, NAFLD or cirrhosis can be aggravated by the impact of bacterial attack on the periodontal tissues [125]. Important periodontal pathogens, like Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans stimulate the synthesis of pro-inflammatory cytokines by periodontal cells [126]. These cytokines, including TNF-α and IL-1 family ones, will be carried by the vascular system and reach the liver. Here, they can have damaging effects on the hepatocytes, if there is a pre-existing hepatic condition, by adding to the distress of the already impaired hepatic tissue [127]. Consequently, the liver functions will be more difficult to perform and the conditions will aggravate, as a result of a bacterial periodontal challenge [128].

Conversely, hepatic conditions can impact the evolution and manifestations of periodontal disease, by means of increased cytokine production [129]. Various liver diseases, as NAFLD, CHC and liver cirrhosis, trigger the increased production of pro-inflammatory cytokines, including TNF-α, IL-1 family, IL-6, which also have a proven active role in the promoting of the periodontal inflammatory reaction [130]. These cytokines, originating from the affected hepatic tissue, enter the blood stream and reach the periodontal tissues. When a periodontal bacterial challenge occurs, they contribute to the exacerbation of the inflammatory reaction, inflicting damage on the periodontal tissues, together with the periodontal-originating pro-inflammatory cytokines. Therefore, an exaggerated inflammatory response is triggered, causing important loss of periodontal structures in patients who also suffer from chronic liver diseases [131, 132].

4.6 Renal diseases

Chronic kidney disease (CKD) is an official public health concern, with 10–12% of the population affected in terms of mortality and morbidity [133, 134]. CKD is characterised by the use of certain markers that indicate the degree of kidney malfunction, notably the glomerular filtration rate (GFR), which indicates the kidney’s functioning efficiency. CKD is diagnosed when multiple standard criteria are met, including a GFR lower than 60 mL/min, albumin levels higher than 30 mg/g of creatinine and the existence of morphological kidney changes. When the GFR drops below 15–20 mL/min, it can be considered that end-stage renal disease has occurred [135, 136].

Various harmful stimuli are triggers for inflammation, the physiological protecting mechanism of the body. In CKD, as in several other chronic debilitating disorders, inflammation becomes maladaptive, uncontrolled and persistent. In this group of patients, a majority of the patients with minimum Stage 3 CKD have increased levels of C-reactive protein (CRP) [137], this prevalence being even higher either in final stage CKD or in dialysis patients [138]. For the evaluation of inflammatory state in clinical practice, a series of specific markers are used. One of the most important inflammation indicators is the CRP, which can also be found in its high sensitive form (hs-CRP) in elevated levels in the serum samples of patients with chronic renal failure, along other pro-inflammatory cytokines [139]. Moreover, another pro-inflammatory cytokine, IL-6, could be a reliable indicator of the risk of cardiovascular diseases and mortality in subjects with end stage renal disease (ESRD) [140].

Malnutrition, a severe consequence of CKD, inflicts important changes in most ESRD patients, in terms of anthropometric and serologic aspects, most of which have an irreversible character, even with proper nutritional supplementation. In addition, malnutrition also comprises a chronic inflammatory reaction, driven by pro-inflammatory cytokines (IL-1, IL-6, TNF-α, IFN-γ, etc). This immune response can accelerate the muscular protein catabolism, on the one hand, by elevating the hepatic synthesis of positive acute phase proteins and, on the other hand, by suppressing the production of negative acute phase proteins [141].

ESRD and haemodialysis (HD) itself lead to an inflammatory status, influenced by numerous factors. The main factor of morbidity and mortality in dialysis patients is considered chronic inflammation, a major determinant of ‘dialysis syndrome’. Inflammation in dialysed patients is characterised by enhanced production of CRP, TNF-α, IL-6, IL-2 and chemokines, such as IL-8, and it may vary over time and during this process [142].

The bacteria of the subgingival biofilm can reach the systemic blood circulatory system, causing lesions to the arterial endothelium and triggering a series of pathogenic events that can eventually lead to atherosclerosis [85]. This mechanism can explain certain pathogenic connections between local and systemic conditions. These connections may have a bidirectional nature, deriving from the similar local and systemic inflammatory reactions that these conditions manifest [143]. For instance, a renal inflammatory response (glomerulonephritis) can be triggered by various acute and chronic infections. This is supported by the increased prevalence of periodontal pathology in patients with renal diseases, suggesting the high relevance of the periodontal pathology in the onset of renal inflammatory responses [144].

Recently, research has focused on the contribution of inflammation, determined by periodontitis, to the overall inflammatory systemic burden in patients with CKD and dialysis. In this context, plasma levels of certain inflammatory cytokines may be relevant to malnutrition, morbidity and mortality of these patients, as well as to their quality of life [145]. Individuals with ESRD experience reduced quality of life and many of them also associate low oral health [145].

Advertisement

5. Conclusion

Through the intense implication of cytokines, the periodontal inflammatory reaction can exhibit a variety of clinical manifestations, in terms of onset, evolution, treatment and prognosis. The local and systemic factors, which can influence the development of periodontal inflammation, often have molecular and cellular implications, and cytokines become the means through which different pathologies can have mutual impact. The cytokine standpoint on periodontal disease and its connective systemic conditions offers wide and promising perspectives on further developments of more precise methods of diagnosis and more efficient therapeutic protocols.

Advertisement

Conflict of interest

None.

References

  1. 1. Scott DA, Krauss J. Neutrophils in periodontal inflammation. In: Kinane DF, Mombelli A, editors. Periodontal Disease. Vol. 15. Basel: Karger Publishers; 2012. pp. 56-83
  2. 2. Cekici A, Kantarci A, Hasturk H, Van Dyke TE. Inflammatory and immune pathways in the pathogenesis of periodontal disease. Periodontology 2000. 2014;64(1):57-80
  3. 3. Preshaw PM. Periodontal disease pathogenesis. In: Takei HH, Klokkevold PR, Carranza FA, editors. Carranza’s Clinical Periodontology. 13th ed. Philadelphia: Elsevier Saunders; 2018. pp. 89-111
  4. 4. Tribble GD, Lamont RJ. Bacterial invasion of epithelial cells and spreading in periodontal tissue. Periodontology 2000. 2010;52(1):68-83
  5. 5. Hernández M, Dutzan N, García-Sesnich J, Abusleme L, Dezerega A, Silva N, et al. Host-pathogen interactions in progressive chronic periodontitis. Journal of Dental Research. 2011;90(10):1164-1170
  6. 6. Machado V, Mesquita MF, Bernardo MA, Casal E, Proença L, Mendes JJ. IL-6 and TNF-α salivary levels according to the periodontal status in Portuguese pregnant women. PeerJ. 2018;6:e4710
  7. 7. Dutzan N, Gamonal J, Silva A, Sanz M, Vernal R. Over-expression of forkhead box P3 and its association with receptor activator of nuclear factor-kappa B ligand, interleukin (IL)-17, IL-10 and transforming growth factor-beta during the progression of chronic periodontitis. Journal of Clinical Periodontology. 2009;36:396-403
  8. 8. Issaranggun Na Ayuthaya B, Everts V, Pavasant P. The immunopathogenic and immunomodulatory effects of interleukin-12 in periodontal disease. European Journal of Oral Sciences. 2018;126:75-83
  9. 9. Chen D, Zhang TL, Wang X. Association between polymorphisms in interleukins 4 and 13 genes and chronic periodontitis in a Han Chinese population. Biomed Research International. 2016;2016:8389020
  10. 10. 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
  11. 11. Pizzo G, Guiglia R, Russo L, Campisi G. Dentistry and internal medicine: From the focal infection theory to the periodontal medicine concept. European Journal Of Internal Medicine. 2010;21(6):496-502
  12. 12. Tsuge A, Noda K, Nakamura Y. Early tissue reaction in the tension zone of PDL during orthodontic tooth movement. Archives of Oral Biology. 2016;65:17-25
  13. 13. Liu O, Xu J, Ding G, Liu D, Fan Z, Zhang C, et al. Periodontal ligament stem cells regulate B lymphocyte function via programmed cell death protein 1. Stem Cells. 2013;31:1371-1382
  14. 14. Jose JA, Somaiah S, Muddaiah S, Shetty B, Reddy G, Roopa S. A comparative evaluation of interleukin 1 beta and prostaglandin E2 with and without low-level laser therapy during en masse retraction. Contemporary Clinical Dentistry. 2018;9(2):267-275
  15. 15. Meeran NA. Biological response at the cellular level within the periodontal ligament on application of orthodontic force—An update. Journal of Orthodontic Science. 2012;1(1):2-10
  16. 16. Mao Y, Wang L, Zhu Y, Liu Y, Dai H, Zhou J, et al. Tension force-induced bone formation in orthodontic tooth movement via modulation of the GSK-3β/β-catenin signaling pathway. Journal of Molecular Histology. 2018;49(1):75-84
  17. 17. Wise GE, King GJ. Mechanisms of tooth eruption and orthodontic tooth movement. Journal of Dental Research. 2008;87(5):414-434
  18. 18. Krishnan V, Davidovitch Z. Cellular, molecular, and tissue-level reactions to orthodontic force. American Journal of Orthodontics and Dentofacial Orthopedics. 2006;129:469.e1-469.e32
  19. 19. Dudic A, Kiliaridis S, Mombelli A, Giannopoulou C. Composition changes in gingival crevicular fluid during orthodontic tooth movement: Comparisons between tension and compression sides. Europrean Journal of Oral Sciences. 2006;114:416-422
  20. 20. Andrade I, Silva TA, Silva GAB, Teixeira AL, Teixeira MM. The role of tumor necrosis factor type 1 in orthodontic tooth movement. Journal of Dental Research. 2007;86:1089-1094
  21. 21. Basaran G, Ozer T, Kaya FA, Hamamci O. Interleukins 2, 6, and 8 levels in human gingival sulcus during orthodontic treatment. American Journal of Orthodontics and Dentofacial Orthopedics. 2006;130(1):7.e1-7.e6
  22. 22. Rody WJ Jr, Wijegunasinghe M, Holliday LS, McHugh KP, Wallet SM. Immunoassay analysis of proteins in gingival crevicular fluid samples from resorbing teeth. Angle Orthodontist. 2016;86(2):187-192
  23. 23. Li Y, Li M, Tan L, Huang S, Zhao L, Tang T, et al. Analysis of time-course gene expression profiles of a periodontal ligament tissue model under compression. Archives of Oral Biology. 2013;58(5):511-522
  24. 24. Luppanapornlarp S, Iida J. Orthodontic force, tooth movement, and interleukin-1β. Hokkaido Journal of Dental Science. 2017;38:20-27
  25. 25. Davidovitch Z. Cell biology associated with orthodontic tooth movement. In: Berkovitz BK, Moxham BJ, Newman HN, editors. The Periodontal Ligament in Health and Disease. St Louis: Mosby-Wolfe; 1995
  26. 26. Lee TY, Lee KJ, Baik HS. Expression of IL-1beta, MMP-9 and TIMP-1 on the pressure side of gingiva under orthodontic loading. Angle Orthodontist. 2009;79:733-739
  27. 27. Allgayer S, Macedo de Menezes L, Roennau Lemos RM. Cytokines in crevicular fluid and orthodontic force: A systematic review. Revista Odonto Ciência. 2017;32(2):104-110
  28. 28. de Oliveira Chami V, Nunes L, Capelli J Jr. Expression of cytokines in gingival crevicular fluid associated with tooth movement induced by aligners: A pilot study. Dental Press Journal of Orthodontics. 2018;23(5):41-46
  29. 29. Iwasaki LR, Crouch LD, Tutor A, Gibson S, Hukmani N, Marx DB, et al. Tooth movement and cytokines in gingival crevicular fluid and whole blood in growing and adult subjects. American Journal of Orthodontics and Dentofacial Orthopedics. 2005;128(4):483-491
  30. 30. Anand Kumar A, Saravanan K, Kohila K, Sathesh Kumar S. Biomarkers in orthodontic tooth movement. Review article. Dental Science. 2015;7(6):325-330
  31. 31. Franchimont N, Wertz S, Malaise M. Interleukin-6: An osteotropic factor influencing bone formation. Bone. 2005;37(5):601-606
  32. 32. Baggiolini M, Walz A, Kunkel SL. Neutrophil-activating peptide-1/IL-8, a novel cytokine that activates neutrophils. Journal of Clinical Investigation. 1989;84:1045-1049
  33. 33. Baumrind S, Korn EL, Boyd RL. Apical root resorption in orthodontically treated adults. American Journal of Orthodontics and Dentofacial Orthopedics. 1996;110:311-320
  34. 34. Tuncer BB, Ozmeriç N, Tuncer C, Teoman I, Cakilci B, Yücel A, et al. Levels of interleukin-8 during tooth movement. Angle Orthodontist. 2005;75:631-636
  35. 35. Alhashimi N, Frithiof L, Brudvik P, Bakhiet M. Orthodontic tooth movement and de novo synthesis of proinflammatory cytokines. American Journal of Orthodontics and Dentofacial Orthopedics. 2001;119:307-312
  36. 36. Kohara H, Kitaura H, Yoshimatsu M, Fujimura Y, Morita Y, Eguchi T, et al. Inhibitory effect of interferon-γ on experimental tooth movement in mice. Journal of Interferon & Cytokine Research. 2012;32:426-431
  37. 37. Mermut S, Bengi AO, Akin E, Kurcku M, Karacay S. Effects of interferon-gamma on bone remodeling during experimental tooth movement. Angle Orthodontist. 2007;77:135-141
  38. 38. Kapoor P, Kharbanda OP, Monga N, Miglani R, Kapila S. Effect of orthodontic forces on cytokine and receptor levels in gingival crevicular fluid: A systematic review. Progress in Orthodontics. 2014;15:65
  39. 39. Madureira DF, da Silva JM, Teixeira AL, Abreu MH, Pretti H, Lages EM, et al. Cytokine measurements in gingival crevicular fluid and periodontal ligament: Are they correlated? American Journal of Orthodontics and Dentofacial Orthopedics. 2015;148:293-301
  40. 40. Maltha JC, van Leeuwen EJ, Dijkman G, Kuijpers-Jagtman AM. Incidence and severity of root resorption in orthodontically moved premolars in dogs. Orthodontics & Craniofacial Research. 2004;7(2):115-121
  41. 41. Yamaguchi M, Yoshii M, Kasai K. Relationship between substance P and interleukin-1beta in gingival crevicular fluid during orthodontic tooth movement in adults. European Journal of Orthodontics. 2006;28(3):241-246
  42. 42. Al-Ghurabi BH, Mohammed-Salih HS, Ghazi A, Saloom H. Evaluation of salivary levels of proinflammatory cytokines (IL-1α, IL-8 and GM-CSF) in adult orthodontic patients. Journal of Dental and Medical Sciences (IOSR-JDMS). 2014;13:75-78
  43. 43. Nieto-Nieto N, Solano JE, Yañez-Vico R. External apical root resorption concurrent with orthodontic forces: The genetic influence. Acta Odontologica Scandinavica. 2017;75(4):280-287
  44. 44. Kim N, Kadono Y, Takami M, Lee J, Lee SH, Okada F, et al. Osteoclast differentiation independent of the TRANCE-RANK-TRAF6 axis. Journal of Experimental Medicine. 2005;202:589-595
  45. 45. Barbieri G, Solano P, Alarcon JA, Vernal R, Rios-Lugo J, Sanz M, et al. Biochemical markers of bone metabolism in gingival crevicular fluid during early orthodontic tooth movement. Angle Orthodontist. 2013;83:63-69
  46. 46. Kunii R, Yamaguchi M. Role of interleukin-6 in orthodontically induced inflammatory root resorption in humans. Korean Journal of Orthodontics. 2013;43:294-301
  47. 47. Toraldo G, Roggia C, Qian WP, Pacifici R, Weitzmann MN. IL-7 induces bone loss in vivo by induction of receptor activator of RANKL and TNFα from T cells. Proceedings of the National Academy of Sciences. 2003;100:125-130
  48. 48. Ahuja R, Almuzian M, Khan A, Pascovici D, Dalci O, Ali Darendeliler M. A preliminary investigation of short-term cytokine expression in gingival crevicular fluid secondary to high-level orthodontic forces and the associated root resorption: Case series analytical study. Progress in Orthodontics. 2017;18:23
  49. 49. Takayanagi H, Sato K, Takaoka A, Taniguchi T. Interplay between interferon and other cytokine systems in bone metabolism. Immunology Reviews. 2005;208:181-193
  50. 50. Walsh MC, Kim N, Kadono Y, Rho J, Lee SY, Lorenzo J, et al. Osteoimmunology: Interplay between the immune system and bone metabolism. Annual Review of Immunology. 2006;24:33-63
  51. 51. Syed S, Kumuda MN, Veenadharini G, Sachidananda C, Ambica SW. Iatrogenic damage to periodontium by restorative treatment procedures: An overview. Open Dental Journal. 2015;9:217-222
  52. 52. Bergman B, Hugoson A, Olsson CO. Periodontal and prosthetic conditions in patients treated with removable partial dentures and artificial crowns. Alongitudinal two-year study. Acta Odontologica Scandinavica. 1971;29(6):621-638
  53. 53. Zhang ZH, Zhang XH, Zhang Y, Fang M. Detection of inflammatory factor and protease content as well as oxidative progress level in gingival crevicular fluid after fiber post and metal post repair. Journal of Hainan Medical University. 2016;22(14):148-151
  54. 54. Neslihan C, Seda A, Mehmet AG, Nilgun S. The effect of restorative materials on cytokines in gingival crevicular fluid. Archives of Oral Biology. 2017;84:139-144
  55. 55. Willershausen B, Kottgen C, Ernest CP. The influence of restorative materials on marginals gingiva. European Journal of Medical Reasearch. 2001;6(10):433-439
  56. 56. Kosyfaki P, del Pilar Pinilla Martin M, Strub JR. Relathionship between crowns and the periodontium: A literature update. Quintessence International. 2010;41(2):109-126
  57. 57. Dimova M, Doseva V. Changes in correlations between cervical crowns edge and marginal gingiva in fixed prosthodontics. Journal of IMAB—Annual Proceeding (Scientific Papers). 2006;12(2)
  58. 58. Harish PV, Sonila AJ, Sirajuddin S, Gundapaneni V, Chungkham S. Iatrogenic damage to the periodontium caused by fixed prsthodontic treatment procedures. Open Dental Journal. 2015;9:217-222
  59. 59. Atieh MA, Faggion CM Jr, Seymour GJ. Cytokines in patients with type 2 diabetes and chronic periodontitis: A systematic review and meta-analysis. Diabetes Research and Clinical Practice. 2014;104(2):e38-e45
  60. 60. Borilova Linhartova P, Poskerova H, Tomandlova M, Bartova J, Kankova K, Fassmann A, et al. Interleukin-1 gene variability and plasma levels in Czech patients with chronic periodontitis and diabetes mellitus. International Journal of Dentistry. 2019;2019:6802349
  61. 61. Acharya AB, Satyanarayan A, Thakur SL. Status of association studies linking diabetes mellitus and periodontal disease in India. International Journal of Diabetes in Developing Countries. 2010;30(2):69-74
  62. 62. Longo PL, Artese HP, Rabelo MS, Kawamoto D, Foz AM, Romito GA, et al. Serum levels of inflammatory markers in type 2 diabetes patients with chronic periodontitis. Journal of Applied Oral Science. 2014;22(2):103-108
  63. 63. Miranda TS, Heluy SL, Cruz DF, da Silva HDP, Feres M, Figueiredo LC, et al. The ratios of pro-inflammatory to anti-inflammatory cytokines in the serum of chronic periodontitis patients with and without type 2 diabetes and/or smoking habit. Clinical Oral Investigations. 2019;23(2):641-650
  64. 64. Acharya AB, Thakur S, Muddapur MV, Kulkarni RD. Cytokine ratios in chronic periodontitis and type 2 diabetes mellitus. Diabetes & Metabolic Syndrome. 2017;11(4):277-278
  65. 65. Wang S, Liu J, Zhang J, Lin J, Yang S, Yao J, et al. Glycemic control and adipokines after periodontal therapy in patients with type 2 diabetes and chronic periodontitis. Brazilian Oral Research. 2017;31:e90
  66. 66. Koçak E, Sağlam M, Kayış SA, Dündar N, Kebapçılar L, Loos BG, et al. Nonsurgical periodontal therapy with/without diode laser modulates metabolic control of type 2 diabetics with periodontitis: A randomized clinical trial. Lasers in Medical Science. 2016;31(2):343-353
  67. 67. Garlet GP. Destructive and protective roles of cytokines in periodontitis: A re-appraisal from host defense and tissue destruction viewpoints. Journal of Dental Research. 2010;89:1349-1363
  68. 68. King GL. The role of inflammatory cytokines in diabetes and its complications. Journal of Periodontology. 2008;79:1527-1534
  69. 69. Moldawer LL. Biology of proinflammatory cytokines and their antagonists. The Journal of Critical Care Medicine. 1994;2:S3-S7
  70. 70. Anirudh BA, Thakurb S, Muddapurc MV, Kulkarnid RD. Cytokine ratios in chronic periodontitis and type 2 diabetes mellitus. Diabetes & Metabolic Syndrome: Clinical Research & Reviews. 2017;11:277-278
  71. 71. Popescu D, Gheorghe D, Boldeanu V, Rica A. Glycemic control and interleukin-1β in periodontal patients with type 1 and 2 diabetes in a group of subjects in south-west Romania. Revista de Chimie. 2017;68(12):3002-3005
  72. 72. Polak D, Shapira L. An update of the evidence for pathogenic mechanisms that may link periodontitis and diabetes. Journal of Clinical Periodontolology. 2018;45(2):150-166
  73. 73. Thunell DH, Tymkiw KD, Johnson JK, Joly S, Burnell KK, Cavanaugh JE, et al. A multiplex immunoassay demonstrates reductions in gingival crevicular fluid cytokines following initial periodontal therapy. Journal of Periodontal Research. 2010;45(1):148-152
  74. 74. Noh MK, Jung M, Kim SH. Assessment of IL-6, IL-8 and TNF-α levels in the gingival tissue of patients with periodontitis. Experimental and Therapeutic Medicine. 2013;6(3):847-851
  75. 75. Taylor GW. Bidirectional interrelationships between diabetes and periodontal diseases: An epidemiologic perspective. Annals of Periodontology. 2001;6(1):99-112
  76. 76. Huang J, Xiao Y, Xu A, Zhou Z. Neutrophils in type 1 diabetes. Journal of Diabetes Investigation. 2016;7(5):652-663
  77. 77. Costa PP, Trevisan GL, Macedo GO, et al. Salivary interleukin-6, matrix metalloproteinase-8, and osteoprotegerin in patients with periodontitis and diabetes. Journal of Periodontology. 2010;81(3):384-391
  78. 78. Tomita T. Apoptosis of pancreatic β-cells in Type 1 diabetes. Bosnian Journal of Basic Medical Sciences. 2017;17(3):183-193
  79. 79. Salvi GE, Franco LM, Braun TM, Lee A, Rutger Persson G, Lang NP, et al. Pro-inflammatory biomarkers during experimental gingivitis in patients with type 1 diabetes mellitus: A proof-of-concept study. Journal of Clinical Periodontology. 2010;37(1):9-16
  80. 80. Salvi GE, Yalda B, Collins JG, Jones BH, Smith FW, Arnold RR, et al. Inflammatory mediator response as a potential risk marker for periodontal diseases in insulin-dependent diabetes mellitus patients. Journal of Periodontology. 1997;68:127-135
  81. 81. Pacios S, Kang J, Galicia J, Gluck K, Patel H, Ovaydi-Mandel A, et al. Diabetes aggravates periodontitis by limiting repair through enhanced inflammation. FASEB Journal. 2012;26(4):1423-1430
  82. 82. Duque C, João MF, Camargo GA, et al. Microbiological, lipid and immunological profiles in children with gingivitis and type 1 diabetes mellitus. Journal of Applied Oral Science. 2017;25(2):217-226
  83. 83. Lauterbach MA, Wunderlich FT. Macrophage function in obesity-induced inflammation and insulin resistance. Pflügers Archiv. 2017;469(3):385-396
  84. 84. Schenkein HA, Loos BG. Inflammatory mechanisms linking periodontal diseases to cardiovascular diseases. Journal of Clinical Periodontology. 2013;40:S51-S69
  85. 85. Nguyen CM, Kim JW, Quan VH, Nguyen BH, Tran SD. Periodontal associations in cardiovascular diseases: The latest evidence and understanding. Journal of Oral Biology and Craniofacial Research. 2015;5(3):203-206
  86. 86. Berlin-Broner Y, Febbraio M, Levin L. Association between apical periodontitis and cardiovascular diseases: A systematic review of the literature. International Endodontic Journal. 2017;50(9):847-859
  87. 87. El Kholy K, Genco RJ, Van Dyke TE. Oral infections and cardiovascular disease. Trends in Endocrinology & Metabolism. 2015;26(6):315-321
  88. 88. Fatkhullina AR, Peshkova IO, Koltsova EK. The role of cytokines in the development of atherosclerosis. Biochemistry (Moscow). 2016;81(11):1358-1370
  89. 89. Ziebolz D, Jahn C, Pegel J, Semper-Pinnecke E, Mausberg RF, Waldmann-Beushausen R, et al. Periodontal bacteria DNA findings in human cardiac tissue—Is there a link of periodontitis to heart valve disease? International Journal of Cardiology. 2018;251:74-79
  90. 90. Mozos I, Malainer C, Horbańczuk J, Gug C, Stoian D, Luca CT, et al. Inflammatory markers for arterial stiffness in cardiovascular diseases. Frontiers in Immunology. 2017;8:1058
  91. 91. Chistiakov DA, Orekhov AN, Bobryshev YV. Links between atherosclerotic and periodontal disease. Experimental and Molecular Pathology. 2016;100(1):220-235
  92. 92. Amar S, Al-Hashemi J. Periodontal innate immune mechanisms relevant to atherosclerosis. In: Gavins NEF, Karen Y, editors. Vascular Responses to Pathogens. Cambridge: Elsevier; 2016. pp. 75-85
  93. 93. Nguyen TT, Wu KY, Leclerc M, Pham HM, Tran SD. Cardiovascular diseases and periodontal disease. Current Oral Health Reports. 2018;5(1):13-18
  94. 94. Hansson GK, Hermansson A. The immune system in atherosclerosis. Nature Immunology. 2011;12(3):204
  95. 95. Choy E. Understanding the dynamics: Pathways involved in the pathogenesis of rheumatoid arthritis. Rheumatology (Oxford, England). 2012;51(5):3-11
  96. 96. Kempeni J. Update on D2E7: A fully human anti-tumour necrosis factor α monoclonal antibody. Annals of the Rheumatic Diseases. 2000;59(S1):144-145
  97. 97. Barrera P, Joosten LA, den Broeder AA, van de Putte LB, vanRiel PL, van den Berg WB, et al. Effects of treatment with a fullyhuman anti-tumour necrosis factor α monoclonal antibody on the local and systemic homeostasis of interleukin 1 and TNF α in patients with rheumatoid arthritis. Annals of the Rheumatic Diseases. 2001;60:660-669
  98. 98. Spadaro A, Rinaldi T, Riccieri V, Taccari E, Valesini G. Interleukin-13 in autoimmune rheumatic diseases: Relationship with the autoantibody profile. Clinical and Experimental Rheumatology. 2002;20(2):213-216
  99. 99. Silosi I, Cojocaru M, Foia L, Boldeanu MV, Petrescu F, Surlin P, et al. Significance of circulating and crevicular matrix metalloproteinase-9 in rheumatoid arthritis-chronic periodontitis association. Journal of Immunology Research. 2015;2015:218060
  100. 100. Koziel J, Mydel P, Potempa J. The link between periodontal disease and rheumatoid arthritis: An updated review. Current Rheumatology Reports. 2014;16:408
  101. 101. Kaur S, White S, Bartold PM. Periodontal disease and rheumatoid arthritis: A systematic review. Journal of Dental Research. 2013;92(5):399-408
  102. 102. Ogrendik M. Rheumatoid arthritis is linked to oral bacteria: Etiological association. Modern Rheumatology. 2009;19:453-456
  103. 103. Detert J, Pischon N, Burmester GR. The association between rheumatoid arthritis and periodontal disease. Arthritis Research & Therapy. 2010;218(1-7):12
  104. 104. Sarhan MA, Pham TN, Chen AY, Michalak TI. Hepatitis C virus infection of human T lymphocytes is mediated by CD5. Journal of Virology. 2012;86(7):3723-3735
  105. 105. Chen SL, Morgan TR. The natural history of hepatitis C virus (HCV) infection. International Journal of Medical Sciences. 2006;3(2):47
  106. 106. Bataller R, Brenner DA. Liver fibrosis. The Journal of Clinical Investigation. 2005;115(2):209-218
  107. 107. Capone F, Guerriero E, Colonna G, Maio P, Mangia A, Castello G, et al. Cytokinome profile evaluation in patients with hepatitis C virus infection. World Journal of Gastroenterology. 2014;20(28):9261-9269
  108. 108. Gheorghe DN, Foia L, Toma V, Surdu A, Herascu E, Popescu DM, et al. Hepatitis C infection and periodontal disease: Is there a common immunological link? Journal of Immunology Research. 2018;2018:8720101
  109. 109. Barros SP, Williams R, Offenbacher S, Morelli T. Gingival crevicular fluid as a source of biomarkers for periodontitis. Periodontology 2000. 2016;70(1):53-64
  110. 110. Akram Z, Abduljabbar T, Hassan A, Ibrahim M, Javed F, Vohra F. Cytokine profile in chronic periodontitis patients with and without obesity: A systematic review and meta-analysis. Disease Markers. 2016;2016:PMC5067315
  111. 111. Majeed Z, Philip K, Alabsi AM, Pushparajan S, Swaminathan D. Identification of gingival crevicular fluid sampling, analytical methods, and oral biomarkers for the diagnosis and monitoring of periodontal diseases: A systematic review. Disease Markers. 2016;2016:1804727
  112. 112. Sharma A, Chakraborti A, Das A, Dhiman RK, Chawla Y. Elevation of interleukin-18 in chronic hepatitis C: Implications for hepatitis C virus pathogenesis. Immunology. 2009;128(1pt2):e514-e522
  113. 113. Wang J, Zhao P, Guo H, Sun X, Jiang Z, Xu L, et al. Serum IL-33 levels are associated with liver damage in patients with chronic hepatitis C. Mediators of Inflammation. 2012;2012:819636. 7 p
  114. 114. Nishitsuji H, Funami K, Shimizu Y, Ujino S, Sugiyama K, Seya T, et al. Hepatitis C virus infection induces inflammatory cytokines and chemokines mediated by the cross talk between hepatocytes and stellate cells. Journal of Virology. 2013;87(14):8169-8178
  115. 115. Younossi Z, Anstee QM, Marietti M, Hardy T, Henry L, Eslam M, et al. Global burden of NAFLD and NASH: Trends, predictions, risk factors and prevention. Nature Reviews Gastroenterology & Hepatology. 2018;15(1):11
  116. 116. Bedogni G, Miglioli L, Masutti F, Tiribelli C, Marchesini G, Bellentani S. Prevalence of and risk factors for nonalcoholic fatty liver disease: The Dionysos nutrition and liver study. Hepatology. 2005;42(1):44-52
  117. 117. Bellentani S. The epidemiology of non-alcoholic fatty liver disease. Liver International. 2017;37:81-84
  118. 118. Abdelmalek MF, Diehl AM. Nonalcoholic fatty liver disease as a complication of insulin resistance. Medical Clinics of North America. 2007;91(6):1125-1149
  119. 119. Seo YY, Cho YK, Bae JC, Seo MH, Park SE, Rhee EJ, et al. Tumor necrosis factor-α as a predictor for the development of nonalcoholic fatty liver disease: A 4-year follow-up study. Endocrinology and Metabolism. 2013;28(1):41-45
  120. 120. Singh P, Gupta ND, Bey A, Khan S. Salivary TNF-alpha: A potential marker of periodontal destruction. Journal of Indian Society of Periodontology. 2014;18(3):306-310
  121. 121. Stadler AF, Angst PD, Arce RM, Gomes SC, Oppermann RV, Susin C. Gingival crevicular fluid levels of cytokines/chemokines in chronic periodontitis: A meta-analysis. Journal of Clinical Periodontology. 2016;43(9):727-745
  122. 122. Algate K, Haynes DR, Bartold PM, Crotti TN, Cantley MD. The effects of tumour necrosis factor-α on bone cells involved in periodontal alveolar bone loss; osteoclasts, osteoblasts and osteocytes. Journal of Periodontal Research. 2016;51(5):549-566
  123. 123. Arsenie C, Sandulescu DL, Popescu DM, Gheorghe DN, Martu A, Foia L, et al. Periodontal changes and the non-alcoholic fatty liver disease. International Journal of Medical Dentistry. 2018;22(3):280-287
  124. 124. Han P, Sun D, Yang J. Interaction between periodontitis and liver diseases. Biomedical Reports. 2016;5(3):267-276
  125. 125. Kuraji R, Ito H, Fujita M, Ishiguro H, Hashimoto S, Numabe Y. Porphyromonas gingivalis induced periodontitis exacerbates progression of non-alcoholic steatohepatitis in rats. Clinical and Experimental Dental Research. 2016;2(3):216-225
  126. 126. Duarte PM, Bastos MF, Fermiano D, Rabelo CC, Perez-Chaparro PJ, Figueiredo LC, et al. Do subjects with aggressive and chronic periodontitis exhibit a different cytokine/chemokine profile in the gingival crevicular fluid? A systematic review. Journal of Periodontal Research. 2015;50(1):18-27
  127. 127. Goral V, Atayan Y, Kaplan A. The relation between pathogenesis of liver cirrhosis, hepatic encephalopathy and serum cytokine levels: What is the role of tumor necrosis factor α? Hepato-Gastroenterology. 2011;58(107-108):943-948
  128. 128. Alazawi W, Bernabe E, Tai D, Janicki T, Kemos P, Samsuddin S, et al. Periodontitis is associated with significant hepatic fibrosis in patients with non-alcoholic fatty liver disease. PLoS One. 2017;12(12):e0185902
  129. 129. Costantini S, Capone F, Guerriero E, Maio P, Colonna G, Castello G. Serum cytokine levels as putative prognostic markers in the progression of chronic HCV hepatitis to cirrhosis. European Cytokine Network. 2010;21(4):251-256
  130. 130. Cardoso EM, Reis C, Manzanares-Céspedes MC. Chronic periodontitis, inflammatory cytokines, and interrelationship with other chronic diseases. Postgraduate Medicine. 2018;130(1):98-104
  131. 131. Gheorghe DN, Camen A, Foia L, Solomon S, Toma V, Mateescu OG, et al. Histologic and immunohistochemical assessment of gingival Tissue's changes induced by periodontal disease in association with chronic hepatitis C. Medical-Surgical Journal-Revista Medico-Chirurgicala. 2018;122(4):789-797
  132. 132. Șurlin P, Camen A, Stratul SI, Roman A, Gheorghe DN, Herăscu E, et al. Optical coherence tomography assessment of gingival epithelium inflammatory status in periodontal—Systemic affected patients. Annals of Anatomy-Anatomischer Anzeiger. 2018;219:51-56
  133. 133. Bokhari SA, Khan AA. Growing burden of noncommunicable diseases: The contributory role of oral diseases, Eastern Mediterranean Region perspective. East Mediterranean Health Journal. 2009;15(4):1011-1020
  134. 134. Levin A, Tonelli M, Bonventre J. Global kidney health 2017 and beyond: A roadmap for closing gaps in care, research, and policy. Lancet. 2017;390:1888-1917
  135. 135. Seikaly MG, Loleh S, Rosenblum A, Browne R. Validation of the Center for Medicare and Medicaid Services algorithm for eligibility for dialysis. Pediatric Nephrology. 2004;19(8):893-897
  136. 136. Neild GH. Life expectancy with chronic kidney disease: An educational review. Pediatric Nephrology. 2017;32(2):243-248
  137. 137. Eustace JA, Astor B, Muntner PM, et al. Prevalence of acidosis and inflammation and their association with low serumalbumin in chronic kidney disease. Kidney International. 2004;65:1031-1040
  138. 138. Carrero JJ, Stenvinkel P. Inflammation in end-stage renal disease—What have we learned in 10 years? Seminars in Dialysis. 2010;23:498-509
  139. 139. Brito F, Ameida S, Figueredo CM, et al. Extent and severity of chronic periodontitis in chronic kidney disease patients. Journal of Periodontology Research. 2012;47:426-430
  140. 140. Spoto B, Mattace-Raso F, Sijbrands EL. Association of IL-6 and a functional polymorphism in the IL-6 gene with cardiovascular events in patients with CKD. Clinical Journal of American Society of Nephrology. 2015;10(2):232-240
  141. 141. Lam WKC. Inflammation, cytokines and chemokines in chronic kidney disease. EJIFCC. 2009;20(1):12-20
  142. 142. Lima SM, Otoni A, Sabino Ade P. Inflammation, neoangiogenesis and fibrosis in peritoneal dialysis. Clinica Chimica Acta. 2013;421:46-50
  143. 143. Sincar C, Solomon S, Rudnic I, Martu I, Martu S. The efects of periodontal therapy on biochemical inflammatory markers on renal dysfunction patiens. Romanian Journal of Oral Rehabilitation. 2013;5(1):71-76
  144. 144. Sincar C, Solomon S, Rudnic I, Martu S. Evaluation of odonto-periodontal parameters in patients with chronic renal insufficiency treated by hemodialysis Int. International Journal of Medical Dentistry. 2012;5(2):214-219
  145. 145. Veisa G, Tasmoc A, Nistor I, Segall L, Siriopol D, Solomon SM, et al. The impact of periodontal disease on physical and psychological domains in long-term hemodialysis patients: A cross-sectional study. International Urology and Nephrology. 2017;49(7):1261-1266

Written By

Petra Surlin, Liliana Foia, Sorina Solomon, Dora Maria Popescu, Dorin Nicolae Gheorghe, Adrian Camen, Maria Alexandra Martu, Anne Marie Rauten, Madalina Olteanu, Allma Pitru, Vasilica Toma, Simona Popa, Mihail Virgil Boldeanu, Silvia Martu and Ion Rogoveanu

Submitted: 25 February 2019 Reviewed: 01 October 2019 Published: 19 August 2020