GET THE APP

T-cell Responses Involved in the Predisposition to Periodontal Di
Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

+44 1223 790975

Review Article - (2013) Volume 0, Issue 0

T-cell Responses Involved in the Predisposition to Periodontal Disease: Lessons from Immunogenetic Studies of Leprosy

Hideki Ohyama1*, Nahoko Kato-Kogoe2, Kazu Takeuchi-Hatanaka3, Koji Yamanegi1, Naoko Yamada1, Keiji Nakasho1, Sho Matsushita4 and Nobuyuki Terada1
1Department of Pathology, Hyogo College of Medicine, Nishinomiya, Japan
2Department of Pharmaceutical Health Care, Himeji Dokkyo University, Himeji, Japan
3Department of Pathophysiology/Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
4Department of Allergy and Immunology, Saitama Medical University, Moroyama, Japan
*Corresponding Author: Dr. Hideki Ohyama, D.D.S., Ph.D., Department of Pathology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya 663-8501, Japan, Tel: +81-798-45-6432, Fax: +81-798-45-6431 Email:

Abstract

Periodontitis, which involves loss of periodontal attachment and resorption of alveolar bone, is initially caused by infection with many kinds of anaerobic, gram-negative bacteria forming a subgingival biofilm. To prevent bacterial invasion, host defense mechanisms need to recruit many kinds of immunoregulatory cells, including helper T (Th) cells which play a central immune-regulatory role against periodontal infection. Similar to many infectious diseases, susceptibility to periodontal disease is partially determined by individual differences in Th cell responsiveness, especially cytokine production, against periodontopathic pathogens. Susceptibility to periodontitis has been associated with gene polymorphisms of several cytokines such as interleukin (IL)-2, IL-4, IL-6 and IL-10, but these correlations are predominantly weak due to their multifactorial nature. Distinct from these studies, we performed immunogenetic studies to investigate associations between periodontal disease susceptibility and hereditary cellmediated immune responses. In these studies, leprosy patients were used as a human model to understand the susceptibility to periodontitis, as leprosy is considered to be an infectious disease whose pathogenesis is regulated by diverse individually-inherited Th1/Th2 immune responses against the bacterial pathogen. In addition to the results of these studies, the discovery of a distinct Th17 lineage helps us to explain that disease susceptibility to periodontitis appears to be predominantly associated with the IL-23/IL-17 pathway. Therefore, individuals whose immunogenetic background is characterized as having low IL-12/interferon-γ activity may have a tendency to skew their immune system toward the IL-23/IL-17 pathway in periodontal lesions, which results in a predisposition to periodontal diseases. These studies help us to understand the complex immunological factors underlying susceptibility to periodontitis.

Keywords: Periodontitis; Leprosy; Immunogenetics; Disease susceptibility; Th1/Th2/Th17

Introduction

Periodontal disease, which involves loss of periodontal attachment and resorption of alveolar bone, is initially caused by infection with many kinds of anaerobic, gram-negative bacteria forming a subgingival biofilm [1]. To prevent this bacterial invasion, host defense functions are required to recruit many kinds of immunoregulatory cells. Periodontal disease is broadly classified into two types of diseasegingivitis and periodontitis. Gingivitis is clinically characterized by gingival inflammation limited to the gingival tissue, and this stage of the disease is a reversible condition. However, once the inflammatory process of gingivitis extends into the periodontal ligament and alveolar bone with accompanying tissue destruction, this stage of periodontal disease is called periodontitis, which is no longer a reversible condition. The destructive process is caused by the production of many kinds of proinflammatory and osteotropic cytokines such as interleukin (IL)-1, IL-6 and tumor necrosis factor (TNF)-α and destructive mediators such as matrix metalloproteinases and cathepsins from the immunoregulatory cells that accumulate and become activated in periodontal lesions.

It has been widely accepted that gingivitis and periodontitis form a continuum of periodontal disease conditions. However, several epidemiological studies have demonstrated that not all cases of gingivitis progress to periodontitis [2,3], which is explained by the involvement of many inherited and environmental factors influencing the susceptibility to periodontitis. Periodontitis itself is subdivided into two main types: chronic periodontitis (CP), and aggressive periodontitis (AgP). CP is a common form of the disease that is prevalent among adults and seniors with poor oral hygiene. AgP is characterized by an early-onset of the disease with severe and rapid loss of periodontal attachment and alveolar bone destruction, and is found to be aggregated in some families, suggesting that genetic factors are important in the development of AgP, as compared with CP [4-7]. Thus, many studies have tried to identify the important genes responsible for disease susceptibility, however, such genes are yet to be determined [8].

Previously, it has been reported that the proband-wise concordance rate of periodontal disease is higher in monozygotic than dizygotic twin pairs [9]. This etiological study of a large number of twins from the population-based Virginia Twin Registry has been regarded as one of the studies showing evidence implicating genetic factors in the susceptibility to periodontal disease. Subsequently, extraordinary progress in molecular biological techniques has permitted us to estimate gene polymorphisms in patients with various clinical types of periodontal disease. Additionally we can identify important genes responsible for the disease by studies targeting genes encoding: 1) cytokines and their receptors; 2) human leukocyte antigens (HLA); 3) Fcγ receptors; 4) Toll-like receptors; and 5) other kinds of molecules regulating bone metabolism, such as vitamin D [10,11]. Many of these epidemiological studies have been performed in patients with AgP since these patients are regarded as susceptible individuals with regard to their genetic factors. Linkage analysis in CP patients has also revealed that severe CP is associated with polymorphisms of the genes regulating immune-cell functions, suggesting that several genetic factors are involved in the pathogenesis of the disease by affecting immune cell activity.

Individual differences in immune responses against periodontopathic bacteria, especially T cell immune responses, are regarded as one of the crucial factors affecting susceptibility to periodontal disease. In particular, cytokines produced by CD4+ helper T (Th) cells play an important role in host defense against periodontal infection. The determination of naïve CD4+ T cells to differentiate into Th1/Th2 cells greatly influences the effectiveness of subsequent immune responses against pathogens, and the progression of the disease [12-15]. Since impairment in Th1/Th2 balance, leading to conditions conducive to periodontal disease, is likely to be dependent on genetic factors, immune responses against various antigens derived from periodontopathic bacteria have been estimated and compared among individuals with different immunogenetic backgrounds to determine important genetic factors responsible for the disease. Nonetheless, it has been postulated that the degree of correlation between each genetic factor regulating immunological responses against periodontopathic bacteria and susceptibility to the disease is not very strong, since there is a great deal of complexity in the immunological responses involved in the pathogenesis of periodontal disease. To overcome this issue, we adopted leprosy patients as a human model to understand the susceptibility to periodontal diseases, as leprosy is considered an infectious disease whose pathogenesis is regulated by diverse individually-inherited Th1/Th2 immune responses.

This article reviews the genetic factors responsible for the susceptibility to periodontal disease with a focus on the cytokines involved in T cell responses, including our immunogenetic studies of periodontal disease and leprosy.

Imunogenetics of Leprosy

Leprosy, a chronic disease caused by infection with Mycobacterium leprae (M. leprae), shows a wide spectrum of clinical features [16]. Tuberculoid leprosy (T-lep) is at one end of the spectrum and lepromatous leprosy (L-lep) is at the other end. T-lep patients show high levels of cell-mediated immune (CMI) responses against M. leprae, which results in resistance to infection with less severe clinical manifestations. On the other hand, L-lep patients show very low CMI responses against the pathogen as well as the progressive form of the disease. This clinical spectrum of leprosy is explained by hereditary differences, and hence genetic background, among individuals in their antigen-specific CMI responses against pathogens. The results of epidemiological studies of the occurrence and form of leprosy among twins are useful in evaluating the involvement of genetic factors. Chakravartti et al. conducted an epidemiological study showing that the agreement rate of leprosy was significantly higher among monozygotic twins than dizygotic twins [17]. This suggests considerable involvement of genetic factors in the susceptibility to leprosy. Therefore, investigation of genetic factors that are common among patients with each type of leprosy will prove invaluable for understanding the mechanisms underlying the occurrence and form of leprosy.

There has already been extensive research on the relationships of various factors involved with both leprosy susceptibility and type. A large number of studies of candidate genes conferring susceptibility to leprosy have been performed. These have revealed many kinds of polymorphisms in genes regulating possible immunological events involved in leprosy, such as: 1) HLA genes; 2) pattern recognition receptors including toll-like receptor and NOD2 genes; 3) the mannose receptor gene; 4) the NRAMP1 (SLC11A1) gene; 5) Parkin and Parkin co-regulated (PARK2/PACRG) genes; 6) the vitamin D receptor gene; and 6) cytokine genes and their receptor genes including TNF- α, IL- 12, IL-23, interferon (IFN)-γ and IL-10 [18,19]. This review focuses on the cytokines and their receptors associated with Th1 responses, especially, IL-12 and IL-12 receptors (IL-12R).

Immunogenetic Studies of CMI Responses in Patients with Leprosy

Immunogenetic research has intensively focused on gene polymorphisms of cytokines associated with the regulation of Th1 responses, since examining individual differences in CMI responses against M. leprae may provide a clue as to the pathogenesis of leprosy.

IL-12 is primarily produced from antigen presenting cells, such as monocytes/macrophages and dendritic cells, and acts on naïve T cells. IL-12 is well-known as a potential inducer of IFN-γ production from T cells, which results in skewing its responses towards Th1. The CMI response against M. leprae varies in each individual, since gene expression levels and the protein structures of cytokines and their receptors involved in the IL-12/IFN-γ production system may be genetically restricted, which results in differences in leprosy susceptibility and type. The gene polymorphism of IL12B, which encodes the p40 subunit of IL-12, is likely to be associated with a susceptibility to leprosy. However, the consequences differ among studies of different ethnic groups and other mycobacterial infectious diseases [20-22]. Interestingly, an epidemiological study of a Mexican population revealed that low expression of the IL-12p40 genotype was frequently detected in L-lep patients whereas serum concentration of p40 protein was significantly higher in L-lep patients when compared with healthy subjects, suggesting the possibility that low expression of IL-12R might be implicated in the pathogenesis of L-lep leprosy [21].

IL-12R is a heterodimer consisting of β1 and β2 chains. β1 and β2 chains are involved equally in the binding to human IL-12 molecules, but differences in the expression of IL-12R β2 chains play a central role in the induction of Th1/Th2 cell differentiation. Since gene expression of IL-12R β2 molecules has been found to be significantly lower in lesions of L-lep patients, as compared to T-lep patients [23], we investigated gene polymorphisms in the transcriptional regulatory region of IL12RB2 in Japanese subjects with a history of leprosy. In this recent study, we found that several single nucleotide polymorphisms (SNPs) in the 5’ flanking region of IL12RB2, including -1035A>G, -1023A>G, -650delG and -464A>G, whose frequencies are significantly different among the different clinical types of leprosy, are associated with congenital low expression levels of IL-12R β2 molecules [24] and low IFN-γ production from T cells [25]. Thus, it is possible that these SNPs are not only associated with the establishment of certain clinical types of leprosy, but are also associated with susceptibility to other infectious diseases, whose pathogenesis is greatly aggravated by an impaired Th1/ Th2 balance against pathogens. Indeed, these SNPs could be one of the possible hereditary factors that determine differences in the intensities of CMI responses among individuals.

In analysis that focused on the CA repeat sequence in the IFN-γ gene (IFNG) as a microsatellite marker in Brazilian subjects, polymorphism was significantly more frequent in leprosy patients [26]. In addition, polymorphism of a promoter region that controls the productivity of IL-10, which is a suppressive cytokine in the IL-12/IFN-γ production system, was studied in Brazilians. Differences in polymorphisms and haplotypes at positions -3575, -2849, -2763, and -819 were found to have a possible effect on susceptibility to leprosy [27,28]. These gene polymorphisms, which determine the productivity and responsiveness of cytokines involved in cellular immune responses, are considered to be important genetic factors that may be essential to understanding individual variation in leprosy susceptibility and type.

Th1/Th2 Paradigm in the Pathogenesis of Periodontal Diseases and its Immunogenetic Study

There has long been controversy as to whether the pathogenesis of periodontal disease is explained exclusively by the Th1/Th2 paradigm. Many previous studies which estimated the differences in gene expression profiles of the Th1/Th2 cytokines in periodontal lesions have provided clues for the development of possible hypotheses concerning this issue. Previous studies have demonstrated that Th2 cells are abundant in periodontal lesions [29,30], whereas several other reports have shown that the cytokine profiles produced from cells in periodontal lesions are consistent with those of Th1 or Th0 cells, but not Th2 cells [13,31]. Other studies have suggested that the production of both Th1- and Th2-related cytokines or Th1-dominant cytokine production is induced in response to periodontal micro-organisms and their components [32-34]. Thus, the argument about which immune response, Th1 or Th2, is more dominant, still continues.

In general, Th1 cells are more frequently detected in the early stage of periodontal lesions whereas Th2 cells predominate in the later and advanced period of the disease, suggesting that Th1 cells are associated with a protective response against bacterial infection while Th2 cells have a role in the destruction and progression of periodontal lesions [35-37]. Thus, the predominant Th subset accumulating in the periodontal lesion would change along with the development of the stage of the disease. However, it is difficult to understand whether Th1/ Th2 skewing of the Th cells accumulating in periodontal lesions is a result of the progression of the disease, or causes the formation and/ or progression of the disease. An immunogenetic approach focusing on the relationship between Th1/Th2 polarization and susceptibility to disease allows us to address this issue.

When investigating other infectious diseases, several studies have used animal models, such as the Th1/Th2 genetic background mouse (C57BL/6 vs. BALB/c strains) or knock-out mice, which is a powerful approach to clarify which subtype of Th cell is implicated in the pathogenesis of a particular disease [38]. In humans, another approach to evaluate the susceptibility to disease is by comparing clinical parameters between individuals with or without SNPs located in the genes related to the regulation of Th1/Th2 immune responses. In studies using individuals with periodontitis, gene polymorphisms of the promoter regions of IL2 [39] IL4 [40-42], IL6 [43] and IL10 [44-47] have been examined, with respect to the establishment of several types of periodontal disease. Distinct from these studies, we used leprosy patients, as human models, to understand the susceptibility to periodontal disease, as leprosy is considered to be an infectious disease whose pathogenesis is regulated by an individually-inherited Th1/Th2 immune response against the pathogen.

In our assessment of the clinical features of periodontal disease in leprosy patients, L-lep patients show more severe periodontitis than T-lep patients or age-matched control subjects, and have lower humoral immune responses against the periodontopathic bacteria, Porphyromonas gingivalis [48]. We also previously demonstrated that the frequencies of SNPs in the 5’ flanking region of IL12RB2 including -1035A>G, -1023A>G, -650delG, and -464A>G, which are frequently detected in L-lep patients, are significantly higher in AgP patients, compared with CP patients and healthy controls [49]. Interestingly, when we defined ‘haplotype 1’ as the haplotype consisting of -1035A, -1023A, -650G, and -464A (i.e. without these SNPs), the frequency of individuals carrying haplotype 1 is apparently lower in AgP patients, compared with L-lep patients (Figure 1). Additionally, IgG levels against several periodontal bacteria are significantly higher in patients carrying IL12RB2 SNPs than in patients without the SNPs, suggesting the possibility that the humoral immune responses against periodontal bacteria in patients with IL12RB2 SNPs are hyper-activated as a result of the low CMI response against periodontopathic bacteria. Thus, it is likely that the immune system in patients carrying IL12RB2 SNPs is skewed toward Th2 responses and produces higher amounts of immunoglobulins after infection with periodontal bacteria. Based on the results of these studies, we proposed that low CMI responses or high humoral responses against periodontopathic bacterial infection might constitute a genetic factor that influences susceptibility to this disease. Interestingly, it has also been reported by others that peripheral blood mononuclear cells (PBMCs) in adult periodontitis patients produce low levels of Th1 cytokines, including IFN-γ, in response to mitogenic stimulation [15,50].

clinical-cellular-immunology-leprosy-patients

Figure 1: Frequency (%) of haplotype 1 in groups of leprosy patients, periodontitis patients and healthy controls.
Haplotype frequency was calculated based on the assumption that each group would be in accordance with Hardy-Weinberg’s equilibrium as the subjects were selected from the Japanese Wajin population of mainland Japan (Honshu). The clinical forms of leprosy and periodontitis were subdivided into lepromatous (L-lep) and tuberculoid (T-lep) leprosy, and aggressive (AgP) and chronic (CP) periodontitis, respectively. A p-value was calculated using the StatView statistical software program (Abacus Concepts, Berkeley, CA), by comparing the frequency of haplotype 1 among the groups.

In contrast, several studies have established that cytokines associated with Th1 cells, especially IFN-γ, play central roles in the progression of inflammatory responses and bone resorption in rodent periodontal disease models [38,51-53]. Since IFN-γ has a strong inhibitory effect on receptor activator of NF-κB ligand (RANKL)- associated osteoclastogenesis due to degradation of tumor necrosis factor receptor-associated factor 6 from osteoclast precursor cells [54], Th1 cells may be associated with resistance against disease. Meanwhile, another study using a general mouse model demonstrates that the net effect of IFN-γ is skewed toward bone resorption due to the greater secretion of osteoclastogenic factors such as RANKL and TNF-α from activated T cells and proinflammatory cytokines, whereas IFN-γ actually inhibits osteoclast formation [55]. These studies support the relevance of Th1 cells in alveolar bone resorption in periodontitis and the progression of the disease. However, it is likely that the net balance of the two opposing effects of IFN-γ on osteoclastogenesis may vary among different animal species and/or among different strains and individuals according to their different genetic backgrounds.

Th cell Paradigm Shift from Th1/Th2 toward Th1/Th2/ Th17

As mentioned above, there is controversy as to whether periodontal pathogenesis can be explained within the limitations of the Th1/Th2 paradigm. However, a third distinct Th cell lineage, Th17, has been identified, and is composed of IL-17-producing T cells which expand in the presence of IL-23 following the priming of IL-6 and TGF-β in naïve T cells [56,57]. IL-23 is a cytokine belonging to the IL-12 family which is secreted as a heterodimer composed of a p40 subunit, identical to the p40 subunit of IL-12, and a unique p19 subunit, similar to the p35 subunit of IL-12 [58]. The ligands of these molecules, IL-12Rβ1 and IL-23 receptor (IL-23R), are expressed on the surface of Th17 cells, while IL-12Rβ1 and β2 molecules are expressed on the surface of Th1 cells. The Th17 cell lineage is thought to play an important role in the pathogenesis of cell-mediated tissue damage caused by either autoimmunity or immune responses against microbial infection [59].

The IL-23 and Th17 pathways have recently been implicated in the pathogenesis of rheumatoid arthritis (RA) [60,61], a disease classically regarded as a good model for periodontal tissue destruction [62]. To date, a number of studies have reported the involvement of IL-17 and Th17 cells in the pathogenesis of RA [63]. Since the Th17 cell lineage has a high capacity to induce osteoclastogenesis caused by IL-17-mediated induction of RANKL on osteoblastic cells [64], expansion of the Th17 lineage, which is augmented by the IL-23/IL-17 pathway, is considered to play an important role in inflammatory tissue destruction in RA. Moreover, a recent study revealed that osteoclastogenesis by IL-17 was induced from monocytes alone in the absence of osteoblasts, resulting from TNF-β production and constitutive expression of RANKL on monocytes by IL-17 [65]. Based on these RA research reports, a number of studies have also been performed implicating IL-17 and Th17 cells in the pathogenesis of periodontal disease. IL-17 and Th17-related cytokine production is elevated in the inflamed tissues and gingival cervical fluids (GCFs) of diseased sites in periodontitis [66-71]. Th17 cells are also highly detected in diseased tissues from periodontitis patients [72]. Our previous study demonstrated that gene expressions of IL23R and IL17A, but not IL12RB2 and IFNG, were elevated in periodontal lesions compared to periodontally healthy sites which were almost free from apparent inflammation and tissue destruction, if any, with only mild gingivitis apparent [70]. In addition, IL17A expression especially, appears to be frequently detected at the sites adjacent to bone destruction [70]. These findings suggest that the IL-23/IL-17 pathway, rather than the IL-12/IFN-γ pathway, is stimulated in periodontal lesions involving tissue and alveolar bone destruction.

Sufficient IL-17 production, but not IFN-γ, is detected in the synovial tissues and fluids of RA patients, even though a number of T cells accumulate and are activated there [73,74]. Since the neutralization of IL-17 in a murine collagen-induced arthritis model reduces joint inflammation, cartilage destruction and bone erosion, IL-17 is considered to be one of the possible cytokines playing an important role in the pathogenesis of RA [75]. Neutralizing IL-23 activity ameliorates synovial tissue inflammation and bone destruction in a rat collagen-induced arthritis model, suggesting that IL-23 is also involved in the progression of the disease by inducing IL-17 production [76]. Actually, the ratio of IL-17 to IFN-γ production from activated human T cells is elevated by IL-23 at 1-10 ng/mL in cultured PBMCs [63]. Thus, these RA results suggest that the predominance of the IL-23/IL-17 pathway over the IL-12/IFN-γ pathway in periodontal lesions may be explained by our previous results, in which the relative expression level was higher for IL23A than IL12A at each site in each patient, whereas the expression levels of both IL-23 and IL-12 were also higher in periodontal lesions than in control sites. While it is controversial whether differentiation of the Th1/Th2/Th17 lineage could be regulated by the local environmental cytokine profiles of the periodontal lesions, local IL-23 production may induce the expansion of the Th17 cell lineage in periodontal tissues, since IL-23 acts on memory Th cells primed by IL-6 and TGF-β, but not naïve T cells, to induce proliferation and terminal differentiation [77].

Meanwhile, an IL-17 receptor A (IL17RA) knockout mouse study, using oral infection with Porphyromonas gingivalis, showed enhancement of alveolar bone loss in the knockout mice [78]. This result contradicts the RA studies but is explained by decreased chemokine expression levels in the knockout mice, resulting in a reduction in neutrophils migrating into the alveolar bone of periodontal lesions. This mouse model suggests that IL-17 plays an important role in host defense functions by the recruitment of neutrophils into the periodontal lesions. Inherited dysfunctions of neutrophils such as chemotaxis, phagocytosis and adhesiveness have been well studied to show an association with susceptibility to AgP [79].

In periodontal lesions, Lester et al. described that gingival concentrations of IL-12 and IFN-γ were significantly lower at severe clinical attachment loss sites than at moderate clinical attachment loss sites, and that the gingival concentration of IL-12 and IFN-γ was negatively correlated with IL-23, whereas IL-17 showed a positive correlation with IL-23 [69]. Actually, IFN-γ strongly inhibits the differentiation of T cells into Th17 cells [56]. A recent study has revealed that IFN-γ regulates macrophages to attenuate Th17 responses through inhibition of IL-23 production [80]. On the basis of these studies, Th17 development is likely restricted by IFN-γ production. However, as mentioned above, it is unclear whether Th cell differentiation could be affected by the cytokine profile of the local environment of periodontal lesions, but not the secondary lymphoid tissues. Another recent study demonstrated that IL-21 production is enhanced in GCF and periodontal tissues of CP patients, which induces Th17 differentiation in periodontal lesions [81,82]; however, the effect of local IL-21 production on Th17 polarization is still unclear. Meanwhile, a report by Zhao et al. shows that the GCF level of IFN-γ does not significantly change after periodontal treatment [71], which provides corroborating evidence for our result of no difference between periodontal lesions and periodontally healthy sites [70]. Taken together, these studies suggest that susceptibility to periodontitis appears to be principally and directly associated with the IL-23/IL-17 pathway rather than the IL- 12/IFN-γ pathway. Moreover, from an immunogenetic viewpoint, we can speculate that individuals with low IL-12/IFN-γ activity, including those who possess genetic factors such as gene polymorphisms in IL12RB2, discovered from our immunogenetic studies in leprosy patients, may have a tendency to skew their immune system toward the IL-23/IL-17 pathway in periodontal lesions, which results in a predisposition to periodontal diseases.

IL-22 has been implicated in protection from cell damage, enhancement of regeneration, and the repair process [83-85]. In keratinocytes, IL-22 plays a role in host defense responses by inducing the production of various antimicrobial proteins including β-defensins [86]. Th17 cells are reported as one of the most predominant subset lineages that produce IL-22 [87]. However, IL-22 expression levels remain low even though the IL-23-induced Th17 pathway is stimulated in inflammatory periodontal lesions, suggesting that the IL-17-producing Th cells that accumulate in these lesions have no potential to produce IL-22 [88]. Indeed, recent criteria for classifying functional T-cell subsets place Th cells into several subsets according to their production of IL-17, IFN-γ, and IL-22. These subsets include Th cells producing only IL-17, but not IL-22, Th cells producing only IL-22 (Th22 cells) [89], and Th cells producing both IL-17 and IFN-γ (Th17/Th1 cells) [90]. This Th17/Th1 cell lineage is suggested to be implicated in the pathogenesis of periodontal diseases [71]. Since IL- 22 plays a role in host defense responses by inducing the production of various antimicrobial proteins and enhancing the mineralized matrix-forming activity of periodontal ligament cells [88], we speculate that low or no expression of IL-22 could be implicated in protracted inflammation of periodontal disease with severe tissue destruction. It is unclear why Th cell clones, having the potential to produce IL- 22 in response to periodontal pathogens, are frequently detected in PBMCs, even though IL-22 expression is low in each individual (our personal communication). The involvement of IL-22 is also supported by a recent study suggesting that IL-22 deficiency may be involved in chronic inflammatory disorders such as acne inversa [91].

Concluding Remarks

Elucidating the pathogenesis of periodontal diseases is extremely complicated because numerous biological, genetic, and environmental factors are involved. In this paper, we have reviewed the implication for Th cell responses in periodontal biology, especially focusing on the immune system regulated by cytokines, from an immunogenetic viewpoint along with our previous studies. Although T cell responses against periodontopathic bacteria play central roles in the host defensive functions to eliminate foreign pathogens, they also coincidentally induce destructive reactions in periodontal tissues. These two opposing T cell responses create a so-called ‘double-edged sword’ in the pathogenesis of periodontal disease. It is very important to analyze these responses, both the protective and destructive roles. Several studies have provided results implicating Th1 cells in tissue destruction in periodontal disease, and we agree with this issue to a certain extent since there are some reports showing similar mechanisms in other diseases. However, we and several other researchers think that Th1 responses contribute greatly to protective function against microbial infections. Meanwhile, many studies have similarly provided evidence that Th17 cells are involved in the tissue destruction in periodontitis lesions. However, given there is an immunological difference between healthy humans and mice in the presence of IL-17-producing cells in their PBMCs [92], it is controversial whether Th1 cells play a central role in periodontal tissue destruction in humans, since the studies implicating Th1 cells in bone destruction were performed using a Th1-slanted C57/BL6 mouse model [38,51-53]. Moreover, it is important to consider that the net effect of various immune responses on clinical manifestations such as bone resorption in periodontal lesions would vary between different animal species, such as humans and mice. Therefore, it may be valuable to evaluate the net effect of Th cell responses on the pathogenesis of periodontal disease in individuals with infectious diseases, such as leprosy, whose pathogenesis is regulated by an individually-inherited Th1/Th2 immune responsiveness against infected pathogens.

Acknowledgement

This study was supported, in part, by a Grant-in-Aid for Scientific Research (C) (No. 20592443, No. 20599020, No. 23593076 and No. 23593077) from the Japan Society for the Promotion of Science and a Strategic Program Grant for Research Infrastructure Development in Private Institutes.

References

  1. Tatakis DN, Trombelli L (2004) Modulation of clinical expression of plaque-induced gingivitis. I. Background review and rationale. J Clin Periodontol 31: 229-238.
  2. Trombelli L (2004) Susceptibility to gingivitis: a way to predict periodontal disease? Oral Health Prev Dent 2 Suppl 1: 265-269.
  3. Butler JH (1969) A familial pattern of juvenile periodontitis (periodontosis). J Periodontol 40: 115-118.
  4. Saxén L (1980) Heredity of juvenile periodontitis. J Clin Periodontol 7: 276-288.
  5. Saxén L, Nevanlinna HR (1984) Autosomal recessive inheritance of juvenile periodontitis: test of a hypothesis. Clin Genet 25: 332-335.
  6. Risom LL, Suzuki JB, Boughman JA (1985) Juvenile periodontitis (periodontosis) and inheritance patterns. Dent Hyg (Chic) 59: 65-70.
  7. Nares S (2003) The genetic relationship to periodontal disease. Periodontol 2000 32: 36-49.
  8. Corey LA, Nance WE, Hofstede P, Schenkein HA (1993) Self-reported periodontal disease in a Virginia twin population. J Periodontol 64: 1205-1208.
  9. Vijayalakshmi R, Geetha A, Ramakrishnan T, Emmadi P (2010) Genetic polymorphisms in periodontal diseases: an overview. Indian J Dent Res 21: 568-574.
  10. Zhang J, Sun X, Xiao L, Xie C, Xuan D, et al. (2011) Gene polymorphisms and periodontitis. Periodontol 2000 56: 102-124.
  11. Seymour GJ, Gemmell E, Reinhardt RA, Eastcott J, Taubman MA (1993) Immunopathogenesis of chronic inflammatory periodontal disease: cellular and molecular mechanisms. J Periodontal Res 28: 478-486.
  12. Ebersole JL, Taubman MA (1994) The protective nature of host responses in periodontal diseases. Periodontol 2000 5: 112-141.
  13. Dennison DK, Van Dyke TE (1997) The acute inflammatory response and the role of phagocytic cells in periodontal health and disease. Periodontol 2000 14: 54-78.
  14. Bártová J, Krátká-Opatrná Z, Procházková J, Krejsa O, Dusková J, et al. (2000) Th1 and Th2 cytokine profile in patients with early onset periodontitis and their healthy siblings. Mediators Inflamm 9: 115-120.
  15. Ridley DS, Jopling WH (1966) Classification of leprosy according to immunity. A five-group system. Int J Lepr Other Mycobact Dis 34: 255-273.
  16. Chakravartti MR, Vogel F (1973) A twin study on leprosy. Top Hum Genet 1: 1-123.
  17. Goulart LR, Goulart IM (2009) Leprosy pathogenetic background: a review and lessons from other mycobacterial diseases. Arch Dermatol Res 301: 123-137.
  18. Cardoso CC, Pereira AC, de Sales Marques C, Moraes MO (2011) Leprosy susceptibility: genetic variations regulate innate and adaptive immunity, and disease outcome. Future Microbiol 6: 533-549.
  19. Morahan G, Kaur G, Singh M, Rapthap CC, Kumar N, et al. (2007) Association of variants in the IL12B gene with leprosy and tuberculosis. Tissue Antigens 1: 234-236.
  20. Alvarado-Navarro A, Montoya-Buelna M, Muñoz-Valle JF, López-Roa RI, Guillén-Vargas C, et al. (2008) The 3'UTR 1188 A/C polymorphism in the interleukin-12p40 gene (IL-12B) is associated with lepromatous leprosy in the west of Mexico. Immunol Lett 118: 148-151.
  21. Tso HW, Lau YL, Tam CM, Wong HS, Chiang AK (2004) Associations between IL12B polymorphisms and tuberculosis in the Hong Kong Chinese population. J Infect Dis 190: 913-919.
  22. Kim J, Uyemura K, Van Dyke MK, Legaspi AJ, Rea TH, et al. (2001) A role for IL-12 receptor expression and signal transduction in host defense in leprosy. J Immunol 167: 779-786.
  23. Ohyama H, Ogata K, Takeuchi K, Namisato M, Fukutomi Y, et al. (2005) Polymorphism of the 5' flanking region of the IL-12 receptor beta2 gene partially determines the clinical types of leprosy through impaired transcriptional activity. J Clin Pathol 58: 740-743.
  24. Ohyama H, Kato-Kogoe N, Nishimura F, Takeuchi-Hatanaka K, Matsushita S, et al. (2008) Differential effects of polymorphisms in the 5' flanking region of IL12RB2 on NK- and T-cell activity. J Interferon Cytokine Res 28: 563-569.
  25. Reynard MP, Turner D, Junqueira-Kipnis AP, Ramos de Souza M, Moreno C, et al. (2003) Allele frequencies for an interferon-gamma microsatellite in a population of Brazilian leprosy patients. Eur J Immunogenet 30: 149-151.
  26. Santos AR, Suffys PN, Vanderborght PR, Moraes MO, Vieira LM, et al. (2002) Role of tumor necrosis factor-alpha and interleukin-10 promoter gene polymorphisms in leprosy. J Infect Dis 186: 1687-1691.
  27. Moraes MO, Pacheco AG, Schonkeren JJ, Vanderborght PR, Nery JA, et al. (2004) Interleukin-10 promoter single-nucleotide polymorphisms as markers for disease susceptibility and disease severity in leprosy. Genes Immun 5: 592-595.
  28. Aoyagi T, Sugawara-Aoyagi M, Yamazaki K, Hara K (1995) Interleukin 4 (IL-4) and IL-6-producing memory T-cells in peripheral blood and gingival tissue in periodontitis patients with high serum antibody titers to Porphyromonas gingivalis. Oral Microbiol Immunol 10: 304-310.
  29. Tokoro Y, Matsuki Y, Yamamoto T, Suzuki T, Hara K (1997) Relevance of local Th2-type cytokine mRNA expression in immunocompetent infiltrates in inflamed gingival tissue to periodontal diseases. Clin Exp Immunol 107: 166-174.
  30. Roberts FA, McCaffery KA, Michalek SM (1997) Profile of cytokine mRNA expression in chronic adult periodontitis. J Dent Res 76: 1833-1839.
  31. Gemmell E, Kjeldsen M, Yamazaki K, Nakajima, Aldred MJ, et al. (1995) Cytokine profiles of Porphyromonas gingivalis-reactive T lymphocyte line and clones derived from P. gingivalis-infected subjects. Oral Dis 1: 139-146.
  32. Salvi GE, Brown CE, Fujihashi K, Kiyono H, Smith FW, et al. (1998) Inflammatory mediators of the terminal dentition in adult and early onset periodontitis. J Periodontal Res 33: 212-225.
  33. Kato N, Ohyama H, Nishimura F, Matsushita S, Takashiba S, et al. (2005) Role of helper T cells in the humoral immune responses against 53-kDa outer membrane protein from Porphyromonas gingivalis. Oral Microbiol Immunol 20: 112-117.
  34. Garlet GP, Cardoso CR, Silva TA, Ferreira BR, Avila-Campos MJ, et al. (2006) Cytokine pattern determines the progression of experimental periodontal disease induced by Actinobacillus actinomycetemcomitans through the modulation of MMPs, RANKL, and their physiological inhibitors. Oral Microbiol Immunol 21: 12-20.
  35. Gaffen SL, Hajishengallis G (2008) A new inflammatory cytokine on the block: re-thinking periodontal disease and the Th1/Th2 paradigm in the context of Th17 cells and IL-17. J Dent Res 87: 817-828.
  36. Garlet GP (2010) Destructive and protective roles of cytokines in periodontitis: a re-appraisal from host defense and tissue destruction viewpoints. J Dent Res 89: 1349-1363.
  37. Baker PJ, Dixon M, Evans RT, Dufour L, Johnson E, et al. (1999) CD4(+) T cells and the proinflammatory cytokines gamma interferon and interleukin-6 contribute to alveolar bone loss in mice. Infect Immun 67: 2804-2809.
  38. Scarel-Caminaga RM, Trevilatto PC, Souza AP, Brito RB, Line SR (2002) Investigation of an IL-2 polymorphism in patients with different levels of chronic periodontitis. J Clin Periodontol 29: 587-591.
  39. Michel J, Gonzáles JR, Wunderlich D, Diete A, Herrmann JM, et al. (2001) Interleukin-4 polymorphisms in early onset periodontitis. J Clin Periodontol 28: 483-488.
  40. Kang BY, Choi YK, Choi WH, Kim KT, Choi SS, et al. (2003) Two polymorphisms of interleukin-4 gene in Korean adult periodontitis. Arch Pharm Res 26: 482-486.
  41. Gonzales JR, Kobayashi T, Michel J, Mann M, Yoshie H, et al. (2004) Interleukin-4 gene polymorphisms in Japanese and Caucasian patients with aggressive periodontitis. J Clin Periodontol 31: 384-389.
  42. Nibali L, D'Aiuto F, Donos N, Griffiths GS, Parkar M, et al. (2009) Association between periodontitis and common variants in the promoter of the interleukin-6 gene. Cytokine 45: 50-54.
  43. Kinane DF, Hodge P, Eskdale J, Ellis R, Gallagher G (1999) Analysis of genetic polymorphisms at the interleukin-10 and tumour necrosis factor loci in early-onset periodontitis. J Periodontal Res 34: 379-386.
  44. Yamazaki K, Tabeta K, Nakajima T, Ohsawa Y, Ueki K, et al. (2001) Interleukin-10 gene promoter polymorphism in Japanese patients with adult and early-onset periodontitis. J Clin Periodontol 28: 828-832.
  45. Berglundh T, Donati M, Hahn-Zoric M, Hanson LA, Padyukov L (2003) Association of the -1087 IL 10 gene polymorphism with severe chronic periodontitis in Swedish Caucasians. J Clin Periodontol 30: 249-254.
  46. Cullinan MP, Westerman B, Hamlet SM, Palmer JE, Faddy MJ, et al. (2008) Progression of periodontal disease and interleukin-10 gene polymorphism. J Periodontal Res 43: 328-333.
  47. Ohyama H, Hongyo H, Shimizu N, Shimizu Y, Nishimura F, et al. (2010) Clinical and immunological assessment of periodontal disease in Japanese leprosy patients. Jpn J Infect Dis 63: 427-432.
  48. Takeuchi-Hatanaka K, Ohyama H, Nishimura F, Kato-Kogoe N, Soga Y, et al. (2008) Polymorphisms in the 5' flanking region of IL12RB2 are associated with susceptibility to periodontal diseases in the Japanese population. J Clin Periodontol 35: 317-323.
  49. Sigusch B, Klinger G, Glockmann E, Simon HU (1998) Early-onset and adult periodontitis associated with abnormal cytokine production by activated T lymphocytes. J Periodontol 69: 1098-1104.
  50. Teng YT, Mahamed D, Singh B (2005) Gamma interferon positively modulates Actinobacillus actinomycetemcomitans-specific RANKL+ CD4+ Th-cell-mediated alveolar bone destruction in vivo. Infect Immun 73: 3453-3461.
  51. Stashenko P, Gonçalves RB, Lipkin B, Ficarelli A, Sasaki H, et al. (2007) Th1 immune response promotes severe bone resorption caused by Porphyromonas gingivalis. Am J Pathol 170: 203-213.
  52. Garlet GP, Cardoso CR, Campanelli AP, Garlet TP, Avila-Campos MJ, et al. (2008) The essential role of IFN-gamma in the control of lethal Aggregatibacter actinomycetemcomitans infection in mice. Microbes Infect 10: 489-496.
  53. Takayanagi H, Ogasawara K, Hida S, Chiba T, Murata S, et al. (2000) T-cell-mediated regulation of osteoclastogenesis by signalling cross-talk between RANKL and IFN-gamma. Nature 408: 600-605.
  54. Gao Y, Grassi F, Ryan MR, Terauchi M, Page K, et al. (2007) IFN-gamma stimulates osteoclast formation and bone loss in vivo via antigen-driven T cell activation. J Clin Invest 117: 122-132.
  55. Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, et al. (2005) Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol 6: 1123-1132.
  56. Harrington LE, Mangan PR, Weaver CT (2006) Expanding the effector CD4 T-cell repertoire: the Th17 lineage. Curr Opin Immunol 18: 349-356.
  57. Oppmann B, Lesley R, Blom B, Timans JC, Xu Y, et al. (2000) Novel p19 protein engages IL-12p40 to form a cytokine, IL-23, with biological activities similar as well as distinct from IL-12. Immunity 13: 715-725.
  58. Steinman L (2007) A brief history of T(H)17, the first major revision in the T(H)1/T(H)2 hypothesis of T cell-mediated tissue damage. Nat Med 13: 139-145.
  59. Nakae S, Saijo S, Horai R, Sudo K, Mori S, et al. (2003) IL-17 production from activated T cells is required for the spontaneous development of destructive arthritis in mice deficient in IL-1 receptor antagonist. Proc Natl Acad Sci U S A 100: 5986-5990.
  60. Hirota K, Hashimoto M, Yoshitomi H, Tanaka S, Nomura T, et al. (2007) T cell self-reactivity forms a cytokine milieu for spontaneous development of IL-17+ Th cells that cause autoimmune arthritis. J Exp Med 204: 41-47.
  61. Snyderman R, McCarty GA (1982) Analogous mechanism of tissue destruction in rheumatoid arthritis and periodontal disease. In: Genco RJ, Mergenhagen SE (eds) Host-parasite interactions in periodontal diseases. American Society for Microbiology, Washington DC, pp. 354-362.
  62. Kotake S, Yago T, Kawamoto M, Nanke Y (2012) Role of osteoclasts and interleukin-17 in the pathogenesis of rheumatoid arthritis: crucial 'human osteoclastology'. J Bone Miner Metab 30: 125-135.
  63. Sato K, Suematsu A, Okamoto K, Yamaguchi A, Morishita Y, et al. (2006) Th17 functions as an osteoclastogenic helper T cell subset that links T cell activation and bone destruction. J Exp Med 203: 2673-2682.
  64. Yago T, Nanke Y, Ichikawa N, Kobashigawa T, Mogi M, et al. (2009) IL-17 induces osteoclastogenesis from human monocytes alone in the absence of osteoblasts, which is potently inhibited by anti-TNF-alpha antibody: a novel mechanism of osteoclastogenesis by IL-17. J Cell Biochem 108: 947-955.
  65. Takahashi K, Azuma T, Motohira H, Kinane DF, Kitetsu S (2005) The potential role of interleukin-17 in the immunopathology of periodontal disease. J Clin Periodontol 32: 369-374.
  66. Vernal R, Dutzan N, Chaparro A, Puente J, Antonieta Valenzuela M, et al. (2005) Levels of interleukin-17 in gingival crevicular fluid and in supernatants of cellular cultures of gingival tissue from patients with chronic periodontitis. J Clin Periodontol 32: 383-389.
  67. Honda T, Domon H, Okui T, Kajita K, Amanuma R, et al. (2006) Balance of inflammatory response in stable gingivitis and progressive periodontitis lesions. Clin Exp Immunol 144: 35-40.
  68. Lester SR, Bain JL, Johnson RB, Serio FG (2007) Gingival concentrations of interleukin-23 and -17 at healthy sites and at sites of clinical attachment loss. J Periodontol 78: 1545-1550.
  69. Ohyama H, Kato-Kogoe N, Kuhara A, Nishimura F, Nakasho K, et al. (2009) The involvement of IL-23 and the Th17 pathway in periodontitis. J Dent Res 88: 633-638.
  70. Zhao L, Zhou Y, Xu Y, Sun Y, Li L, et al. (2011) Effect of non-surgical periodontal therapy on the levels of Th17/Th1/Th2 cytokines and their transcription factors in Chinese chronic periodontitis patients. J Clin Periodontol 38: 509-516.
  71. Cardoso CR, Garlet GP, Crippa GE, Rosa AL, Júnior WM, et al. (2009) Evidence of the presence of T helper type 17 cells in chronic lesions of human periodontal disease. Oral Microbiol Immunol 24: 1-6.
  72. Fox DA (1997) The role of T cells in the immunopathogenesis of rheumatoid arthritis: new perspectives. Arthritis Rheum 40: 598-609.
  73. Kotake S, Udagawa N, Takahashi N, Matsuzaki K, Itoh K, et al. (1999) IL-17 in synovial fluids from patients with rheumatoid arthritis is a potent stimulator of osteoclastogenesis. J Clin Invest 103: 1345-1352.
  74. Lubberts E, Koenders MI, Oppers-Walgreen B, van den Bersselaar L, Coenen-de Roo CJ, et al. (2004) Treatment with a neutralizing anti-murine interleukin-17 antibody after the onset of collagen-induced arthritis reduces joint inflammation, cartilage destruction, and bone erosion. Arthritis Rheum 50: 650-659.
  75. Yago T, Nanke Y, Kawamoto M, Furuya T, Kobashigawa T, et al. (2007) IL-23 induces human osteoclastogenesis via IL-17 in vitro, and anti-IL-23 antibody attenuates collagen-induced arthritis in rats. Arthritis Res Ther 9: R96.
  76. McGeachy MJ, Chen Y, Tato CM, Laurence A, Joyce-Shaikh B, et al. (2009) The interleukin 23 receptor is essential for the terminal differentiation of interleukin 17-producing effector T helper cells in vivo. Nat Immunol 10: 314-324.
  77. Yu JJ, Ruddy MJ, Wong GC, Sfintescu C, Baker PJ, et al. (2007) 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 109: 3794-3802.
  78. Genco RJ (1996) Current view of risk factors for periodontal diseases. J Periodontol 67: 1041-1049.
  79. Sheikh SZ, Matsuoka K, Kobayashi T, Li F, Rubinas T, et al. (2010) Cutting edge: IFN-gamma is a negative regulator of IL-23 in murine macrophages and experimental colitis. J Immunol 184: 4069-4073.
  80. Dutzan N, Rivas C, García-Sesnich J, Henríquez L, Rivera O, et al. (2011) Levels of interleukin-21 in patients with untreated chronic periodontitis. J Periodontol 82: 1483-1489.
  81. Dutzan N, Vernal R, Vaque JP, García-Sesnich J, Hernandez M, et al. (2012) Interleukin-21 expression and its association with proinflammatory cytokines in untreated chronic periodontitis patients. J Periodontol 83: 948-954.
  82. Wolk K, Sabat R (2006) Interleukin-22: a novel T- and NK-cell derived cytokine that regulates the biology of tissue cells. Cytokine Growth Factor Rev 17: 367-380.
  83. Eyerich S, Eyerich K, Cavani A, Schmidt-Weber C (2010) IL-17 and IL-22: siblings, not twins. Trends Immunol 31: 354-361.
  84. Sonnenberg GF, Fouser LA, Artis D (2011) Border patrol: regulation of immunity, inflammation and tissue homeostasis at barrier surfaces by IL-22. Nat Immunol 12: 383-390.
  85. Wolk K, Kunz S, Witte E, Friedrich M, Asadullah K, et al. (2004) IL-22 increases the innate immunity of tissues. Immunity 21: 241-254.
  86. Liang SC, Tan XY, Luxenberg DP, Karim R, Dunussi-Joannopoulos K, et al. (2006) Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J Exp Med 203: 2271-2279.
  87. Kato-Kogoe N, Nishioka T, Kawabe M, Kataoka F, Yamanegi K, et al. (2012) The promotional effect of IL-22 on mineralization activity of periodontal ligament cells. Cytokine 59: 41-48.
  88. Duhen T, Geiger R, Jarrossay D, Lanzavecchia A, Sallusto F (2009) Production of interleukin 22 but not interleukin 17 by a subset of human skin-homing memory T cells. Nat Immunol 10: 857-863.
  89. Annunziato F, Cosmi L, Liotta F, Maggi E, Romagnani S (2009) Human Th17 cells: are they different from murine Th17 cells? Eur J Immunol 39: 637-640.
  90. Wolk K, Warszawska K, Hoeflich C, Witte E, Schneider-Burrus S, et al. (2011) Deficiency of IL-22 contributes to a chronic inflammatory disease: pathogenetic mechanisms in acne inversa. J Immunol 186: 1228-1239.
  91. Amadi-Obi A, Yu CR, Liu X, Mahdi RM, Clarke GL, et al. (2007) TH17 cells contribute to uveitis and scleritis and are expanded by IL-2 and inhibited by IL-27/STAT1. Nat Med 13: 711-718.
Citation: Ohyama H, Kato-Kogoe N, Takeuchi-Hatanaka K, Yamanegi K, Yamada N, et al. (2012) T-cell Responses Involved in the Predisposition to Periodontal Disease: Lessons from Immunogenetic Studies of Leprosy. J Clin Cell Immunol S1:005.

Copyright: © 2012 Ohyama H, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top