9
Overlapping Protective and Destructive Regulatory Pathways in Apical Periodontitis Ildik o J. M arton, MD, LDS, PhD, DSc,* and Csongor Kiss, MD, PhD, DSc Abstract Introduction: Protective and destructive immunoreac- tions take place simultaneously in apical periodontitis. However, the same reactions defending the periapical area from infection-derived damage may also result in host tissue injury. Methods: The inflammatory reaction of the periapical tissues is self-limited. Regeneration of the injured tooth-supporting structures may follow elim- ination of the causative microbial irritation. Results: Recent experimental and clinical observations have identified important interplay between positive and negative regulatory pathways. A network of stimulatory and inhibitory feedback loops may influence the inten- sity of the defense and inflammatory responses and the balance between bone resorption and regeneration, resulting in lesion expansion or healing of apical peri- odontitis. Conclusions: We critically discuss research data on regulatory mechanisms that control the activity of host effector cells and signaling molecules during in- teractions with pathogenic microbes. (J Endod 2014;40:155–163) Key Words Apical periodontitis, bone resorption, cytokines, immunity, inflammation, stem cells A pical periodontitis, an inflammatory response within the periapical tissues, most frequently results from polymicrobial infection of pulpal origin. The disease may take an acute or chronic course. Chronic apical periodontitis may present as dental granuloma or periradicular cyst (1). Based on clinical and experimental reports from the 80s and 90s, we have proposed that protective and destructive immunoreac- tions take place simultaneously in chronic periapical lesions (2). However, ‘‘destruc- tive’’ and ‘‘protective’’ terms need to be carefully defined because the same reactions responsible for host defense by the elimination of invading microorganisms may also result in tissue injury. Moreover, tissue injury can be prevented by the inhibition of anti- microbial immunoinflammatory reactions, albeit at the price of overwhelming infection (3, 4). Here, we review the results of recent animal and human studies and laboratory experiments that contribute to a better understanding of the periapical inflammatory processes. Furthermore, we analyze regulatory mechanisms that control the activity of effector cells and molecules. These mechanisms can influence the fine-tuned balance between protection from apical infection, lesion progression, and repair. Table 1 sum- marizes the major subclasses of immune cells playing an important role in apical peri- odontitis, their major actions, and examples of major released mediators. Regulation of the Initial Inammatory Cell Assembly Proliferative activity is restricted to epithelial cells (ECs) in periapical lesions (5, 6). A robust influx of leukocytes is initiated by the interaction between microorganisms of the infected root canal and the periodontal ligament (PDL) (7–10). PDL cells constitutively express low levels of adhesion molecules and chemokines. They provide a weak stimulus for recruiting and activating leukocytes, thereby maintaining the homeostasis of healthy periapical tissues (11–14). Pathogenic bacteria and bacterial molecules released from the infected root canal stimulate PDL cells to up-regulate a broad set of inflammatory mediators. These include chemoattractant molecules, which stimulate the influx of inflammatory cells to the lesion site (7, 9, 15, 16). Human PDL fibroblasts (PDLFs) express toll-like receptor (TLR)-related molecules (ie, TLR2, TLR4, MD-2, and MyD88) and CD14. Using the CD14/TLR pathway, PDLFs challenged with lipopolysaccharide (LPS) and Staphylo- coccus epidermidis peptidoglycan initiate an interleukin (IL)-8/CXCL8 response, the key signal for polymorphonuclear (PMN) leukocyte chemoattraction (17) (Fig. 1). As shown in cultured cells, TLR receptors may stimulate inflammation independently of the CD14 molecule. The synthetic peptidoglycan analog muramyl dipeptide induces a positive regulatory loop via nucleotide-binding oligomerization domain-like receptors in apical periodontitis (17–19). Porphyromonas gingivalis and LPS extracted from P. gingivalis and from Escherichia coli increase gene expression of both pro- and anti- inflammatory cytokines in human PDLF cell cultures. The former ones are represented by tumor necrosis factor alpha (TNF-a), IL-6, monocyte chemoattractant protein (MCP)-1/CCL2, the chemoattractant ‘‘regulated upon activation, normal T-cell ex- pressed and secreted’’ (RANTES)/CCL5, and stromal-derived factor 1 and the latter ones by transforming growth factor-beta (TGF-b). The pattern and intensity of cytokine gene expression by PDLFs shows marked variation both between different individuals and depending on the source of the stimulatory agent (20–22). In lesion-derived hu- man PDLF extracts, 54% of the genes investigated with an inflammatory gene array were significantly up-regulated compared with healthy PDLF extracts (23). Herpesviruses including Epstein-Barr virus, cytomegalovirus, and human herpes- virus 6, but not human herpesvirus 8, contribute to the etiopathogenesis of human apical From the *Department of Restorative Dentistry, Faculty of Dentistry and Department of Pediatric Hematology-Oncology, Institute of Pediatrics, Faculty of Medicine, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary. Address requests for reprints to Dr Ildik o J. M arton, Medical and Health Center, University of Debrecen, 98 Nagyerdei Krt, Debrecen H-4032, Hungary. E-mail address: marton.ildiko@ dental.unideb.hu 0099-2399/$ - see front matter Copyright ª 2014 American Association of Endodontists. http://dx.doi.org/10.1016/j.joen.2013.10.036 Review Article JOE Volume 40, Number 2, February 2014 Protective and Destructive Regulatory Pathways 155

Overlapping Protective and Destructive Regulatory Pathways in Apical Periodontitis

  • Upload
    csongor

  • View
    215

  • Download
    1

Embed Size (px)

Citation preview

Page 1: Overlapping Protective and Destructive Regulatory Pathways in Apical Periodontitis

Review Article

Overlapping Protective and Destructive Regulatory Pathwaysin Apical PeriodontitisIldik�o J. M�arton, MD, LDS, PhD, DSc,* and Csongor Kiss, MD, PhD, DSc†

Abstract

Introduction: Protective and destructive immunoreac-tions take place simultaneously in apical periodontitis.However, the same reactions defending the periapicalarea from infection-derived damage may also result inhost tissue injury. Methods: The inflammatory reactionof the periapical tissues is self-limited. Regeneration ofthe injured tooth-supporting structures may follow elim-ination of the causative microbial irritation. Results:Recent experimental and clinical observations haveidentified important interplay between positive andnegative regulatory pathways. A network of stimulatoryand inhibitory feedback loops may influence the inten-sity of the defense and inflammatory responses andthe balance between bone resorption and regeneration,resulting in lesion expansion or healing of apical peri-odontitis. Conclusions: We critically discuss researchdata on regulatory mechanisms that control the activityof host effector cells and signaling molecules during in-teractions with pathogenic microbes. (J Endod2014;40:155–163)

Key WordsApical periodontitis, bone resorption, cytokines, immunity,inflammation, stem cells

From the *Department of Restorative Dentistry, Faculty ofDentistry and †Department of Pediatric Hematology-Oncology,Institute of Pediatrics, Faculty of Medicine, Medical and HealthScience Center, University of Debrecen, Debrecen, Hungary.

Address requests for reprints to Dr Ildik�o J. M�arton, Medicaland Health Center, University of Debrecen, 98 Nagyerdei Krt,Debrecen H-4032, Hungary. E-mail address: [email protected]/$ - see front matter

Copyright ª 2014 American Association of Endodontists.http://dx.doi.org/10.1016/j.joen.2013.10.036

JOE — Volume 40, Number 2, February 2014

Apical periodontitis, an inflammatory response within the periapical tissues, mostfrequently results from polymicrobial infection of pulpal origin. The disease may

take an acute or chronic course. Chronic apical periodontitis may present as dentalgranuloma or periradicular cyst (1). Based on clinical and experimental reportsfrom the 80s and 90s, we have proposed that protective and destructive immunoreac-tions take place simultaneously in chronic periapical lesions (2). However, ‘‘destruc-tive’’ and ‘‘protective’’ terms need to be carefully defined because the same reactionsresponsible for host defense by the elimination of invading microorganisms may alsoresult in tissue injury. Moreover, tissue injury can be prevented by the inhibition of anti-microbial immunoinflammatory reactions, albeit at the price of overwhelming infection(3, 4). Here, we review the results of recent animal and human studies and laboratoryexperiments that contribute to a better understanding of the periapical inflammatoryprocesses. Furthermore, we analyze regulatory mechanisms that control the activityof effector cells andmolecules. These mechanisms can influence the fine-tuned balancebetween protection from apical infection, lesion progression, and repair. Table 1 sum-marizes the major subclasses of immune cells playing an important role in apical peri-odontitis, their major actions, and examples of major released mediators.

Regulation of the Initial Inflammatory Cell AssemblyProliferative activity is restricted to epithelial cells (ECs) in periapical lesions

(5, 6). A robust influx of leukocytes is initiated by the interaction betweenmicroorganisms of the infected root canal and the periodontal ligament (PDL)(7–10). PDL cells constitutively express low levels of adhesion molecules andchemokines. They provide a weak stimulus for recruiting and activating leukocytes,thereby maintaining the homeostasis of healthy periapical tissues (11–14).Pathogenic bacteria and bacterial molecules released from the infected root canalstimulate PDL cells to up-regulate a broad set of inflammatory mediators. These includechemoattractant molecules, which stimulate the influx of inflammatory cells to thelesion site (7, 9, 15, 16). Human PDL fibroblasts (PDLFs) express toll-like receptor(TLR)-related molecules (ie, TLR2, TLR4, MD-2, and MyD88) and CD14. Using theCD14/TLR pathway, PDLFs challenged with lipopolysaccharide (LPS) and Staphylo-coccus epidermidis peptidoglycan initiate an interleukin (IL)-8/CXCL8 response, thekey signal for polymorphonuclear (PMN) leukocyte chemoattraction (17) (Fig. 1).As shown in cultured cells, TLR receptors may stimulate inflammation independentlyof the CD14 molecule. The synthetic peptidoglycan analog muramyl dipeptide inducesa positive regulatory loop via nucleotide-binding oligomerization domain-like receptorsin apical periodontitis (17–19). Porphyromonas gingivalis and LPS extracted from P.gingivalis and from Escherichia coli increase gene expression of both pro- and anti-inflammatory cytokines in human PDLF cell cultures. The former ones are representedby tumor necrosis factor alpha (TNF-a), IL-6, monocyte chemoattractant protein(MCP)-1/CCL2, the chemoattractant ‘‘regulated upon activation, normal T-cell ex-pressed and secreted’’ (RANTES)/CCL5, and stromal-derived factor 1 and the latterones by transforming growth factor-beta (TGF-b). The pattern and intensity of cytokinegene expression by PDLFs shows marked variation both between different individualsand depending on the source of the stimulatory agent (20–22). In lesion-derived hu-man PDLF extracts, 54% of the genes investigated with an inflammatory gene array weresignificantly up-regulated compared with healthy PDLF extracts (23).

Herpesviruses including Epstein-Barr virus, cytomegalovirus, and human herpes-virus 6, but not human herpesvirus 8, contribute to the etiopathogenesis of human apical

Protective and Destructive Regulatory Pathways 155

Page 2: Overlapping Protective and Destructive Regulatory Pathways in Apical Periodontitis

TABLE 1. Major Subclasses of Immune Cells and Their Major Actions

Subclasses of immune cells Major actions Major released mediators

Antigen presenting cells (APC) Uptake and processing of antigens anddisplaying them to T cells

Depending on the subtype of APC (seebelow)

B cells/lymphocytes Formation of antigen-specific antibodies;performing as APCs

Fully activated B cells (plasma B cells/plasma cells) secrete antibodies

DCs Function as professional APCs, innaterecognition of microbes by TLR,regulate other immune cells includingT and B cells

mDCs produce TNF-a and cytokines of theIL-1 and IL-12 families; pDCs producetype I interferons

LCs mDCs of the mucosa and skin Similar to other mDCsMacrophages (together with monocytes,

macrophages are called MNPs)Develop from monocytes; function as

professional phagocytes, and APCs;release toxic compounds that destroymicrobes and innocent bystander hostcells; regulate immune cells

Cytokines: IFN-b, IFN-g, IL-1a, IL-1b, IL-6,IL-10, IL-15, IL-18, migration inhibitoryfactor, TGF-b TNF-a; chemokines:CCL-2CCL-3,CCL-4, CCL-5, CCL-22, IL-8,macrophage inflammatory proteins;reactive oxygen and nitrogenintermediates; eicosanoids; proteases

Mast cells Anaphylactic type of allergic reactions;nonspecific antimicrobial defense;wound healing; immunoregulation

Histamine, serotonine; eicosanoids;cytokines: IL-1b, IL-3, IL-4, IL-5, IL-13,GM-CSF, SCF; chemokines: CCL-2, CCL-5,eotaxins

MOs Phagocytosis, antigen presentation, giverise to mDCs and macrophages

Similar to mDCs and macrophages

NK cells Cytotoxic lymphocytes of innate immunity,contribute to self- tolerance andimmune memory

Cytotoxins: perforin, cytotoxins: perforin,chemokines CCL3, CCL4, and CCL5;cytokines IFN-g, GM-CSF, and TNF-a

NKT cells CD1d-restricted T cells; recognize lipidsand glycolipids; rapid release of solubleregulatory mediators

Cytotoxins as NK cells; IL-2, IL-4, IL-13,IL-17, IL-21, IFN-g, GM-CSF, TNF-a

PMNs Of the 3 types of PMNs (basophilic,eosinophilic, and neutrophilic),neutrophils play a major role in apicalperiodontitis: provide first line ofdefense against pathogens byphagocytosis; attract and stimulatefurther PMNs, MOs, and macrophages;destroy periapical tissue components

Reactive oxygen and nitrogenintermediates; proteins withantimicrobial activities: acid hidrolases,defensins, lactoferrin, lysozyme;proteases destroying microbes and hosttissues; eicosanoids; cytokines andchemokines: IL-1b, TNF-a, IL-8, andCXCL-10

T cells/lymphocytes Participate in cell-mediated immunereactions as effectors and regulatorycells

Depending on the subtype of T cells

Cytotoxic T cells (CD8-positive T cells) MHC class I–restricted cells, destruction ofinfected cells

Cytotoxins: NK cells; cytokines: IFN-g, IL-2,GM-CSF; TNF-a; chemokines: CCL-3,CCL-4, CCL-5

Memory T cells (CD45RO-positive cells) Rapid expansion to effector T cells uponantigen re-exposure

IFN-g, IL-4, IL-17

Helper T cells (Th) (CD4-positive T cells) MHC class I–restricted cells;promotematuration of B cells, activation of Tcellsand macrophages; differentiate intoseveral subtypes, which producestimulatory and inhibitory cytokines

Activated Th cells: IL-2; subset of activatedTh cells: IL-17; Th1 cytokines: IFN-g,TGF-b; Th2 cytokines: IL-4, IL-5, IL-6,IL-10, IL-13; Th17 cytokines: IL-1, IL-6,IL-17, TNF-a

Regulatory T cells (Treg) (CD4+/CD25hi/Foxp3+ cells)

Suppress activation of the immune systemby secreting regulatory cytokines and bycell-to-cell contacts

TGF-b and IL-10

APCs, antigen-presenting cells; DCs, dendritic cells; GM-CSF, granulocyte-macrophage colony-stimulating factor; IFN-b, interferon beta; IFN-g, interferon gamma; IL, interleukin; LCs, Langehans cells;

MDCs, myeloid dendritic cells; MNPs, mononuclear phagocytes; MOs, monocytes; NK, natural killer; NKT, natural killer T; pDCs, plasmacytoid dendritic cells; PMN, polymorphonuclear leukocyte;

TLR, Toll-like receptor; TNF-a, tumor necrosis factor alpha.

Review Article

periodontitis by stimulating an increased production of early inflamma-tory cytokines and chemokines (24–27). Synergistic interactionsbetween herpesviruses and endodontopathogenic bacteria mayexacerbate the severity of the inflammatory response within theperiradicular area (28–30). An aggressive type of pathologic pictureand a symptomatic manifestation often characterize herpesvirus-productive periapical infection.

In vivo and in vitro studies showed that the production of chemo-attractant molecules can be induced by irritation from dental materials,implants, and orthodontic tooth movement (31–33). Dentin proteins

156 M�arton and Kiss

can augment the migration of neutrophil leukocytes by the inductionof KC/CXCL1 and MIP-2/CXCL2 in mice (34). Sensory neuropeptidesubstance P, which is released during painful stimuli, has been shownto induce the expression of MIP-3a/CCL20 in immortalized PDLF cells.Concomitantly, substance SP stimulated the expression of the stressenzyme heme oxygenase 1 as a down-regulatory signal (35).

The first wave of chemokines released from pulpal and PDL cellsreach the blood vessels, increasing their permeability and selectively at-tracting white blood cells to the site of inflammation (8, 16, 36).Chemokines bind to the molecules of the extracellular matrix within

JOE — Volume 40, Number 2, February 2014

Page 3: Overlapping Protective and Destructive Regulatory Pathways in Apical Periodontitis

Figure 1. A schematic representation of the regulation of the initial inflammatory cell responses in the periapical area. The interaction of LPS and TLRs expressedon PDLFs initiates the production of proinflammatory cytokines and chemokines IL-8/CXCL8, MCP-1/CCL2, RANTES/CCL5, pro-IL-1b, IL-6, stromal-derived factor 1,and TNF-a as well as the immunoregulatory compounds TGF-b, IL-1Ra, and sTNF-aR. Proinflammatory mediators induce vasodilation and attract PMNs and MNPsto the periapical area. Invading bacteria, their byproducts, and the first wave of inflammatory mediators activate phagocytes. These cells produce a broad set ofsoluble mediators, which destroy pathogenic microbes, injure tooth-supporting tissues, and attract further cells of the innate and adaptive immune system. Incontrast, immunoregulatory compounds attenuate the intensity of inflammatory reactions and host tissue destruction (see text for references).

Review Article

the lesion providing a permanent attraction stimulus for migrating cells(37). Cells of the adaptive immune system migrate with a lag period of2–3 days after the initial infiltration of the periradicular area by neutro-phils and macrophages in experimental animals with exposed root ca-nals (38). In parallel with lesion size expansion after days 21–56 ofpulp exposure, protective and destructive mechanisms reach a dynamicequilibrium (39). Analysis of the expression profile of a 30,000 genedata set in a rodent model of apical periodontitis revealed a networkof interrelated regulatory loops. The differential expression of 2 mono-cyte-/macrophage-derived cytokines, IL-1a and IL-1b, was describedduring the inflammatory and healing phases. This observation suggeststhat IL-1b may be involved in reactions contributing to cellular diversi-fication of the early lesion and T-lymphocyte–dependent antibody pro-duction in later phases, whereas IL-1a may promote healing (40).Similarly, in human primary endodontic infections with apical peri-odontitis, individual cytokines were shown to correlate with lesionscharacterized by different clinical and radiographic appearances. IL-1b and TNF-a correlated with the presence of exudate during thecourse of root canal treatment. IL-6 exhibited a negative correlationwith lesion size (41).

Participation of Cellular Components inPeriapical Protective and Destructive Reactions

Mononuclear phagocytes (MNPs) are among the initially recruitedcells in early apical periodontitis. These cells participate both in protec-tive and destructive reactions. MNPs directly eliminate microbes

JOE — Volume 40, Number 2, February 2014

invading from the infected root canal and initiate antigen-specific re-sponses by processing and presenting antigens to T lymphocytes.They contribute to the establishment of the lesion by destroying healthyperiapical tissues (4, 42–44). Exudative macrophages express a varietyof chemokine receptors. Multiple chemokines, including MCP-1/CCL2,MIP-1a/CCL3, and RANTES/CCL5, and IL-8/CXCL8, the main neutrophilchemoattractant, may mediate MNP migration to the periapical lesion(36). The expression of these ligand-receptor pairs at the RNA and pro-tein levels has been reported in experimental and human periapical in-flammatory lesions (37, 40, 45–47). Stimulated MNPs produce a broadset of soluble molecules that attract further cells to the lesion and up- ordown-regulate their activities (Fig. 1). Advanced immunophenotypingmethods and ex vivo functional assays have revealed important detailsof the cross-talk between antigen-presenting cells (APCs) and T lym-phocytes (48, 49). In clinical samples, CD14+/HLA-DR+ activatedmacrophages made up about 17% of the mononuclear cells (MNCs)isolated from periapical lesions (50). Langerhans-type dendritic cells(DCs) made up 12%–15% of the total DC population and were foundto be associated with CD3+ T-lymphocyte infiltration and EC prolifera-tion (44, 51, 52). Interestingly, the predominant subpopulation oflesion-associated APCs was represented by HLA-DR+/CD19+ B lym-phocytes (44, 50).

Investigations of experimental rodent models and human samplesin the 90s suggested that T-helper type 1 (Th1) lymphocytes character-ized early forms of apical periodontitis. This suggests a role for Th1 cellsin the initiation and expansion of lesions. The abundance of B lympho-cytes, plasma cells, CD8+ cytotoxic/suppressor T cells, and Th2 cells in

Protective and Destructive Regulatory Pathways 157

Page 4: Overlapping Protective and Destructive Regulatory Pathways in Apical Periodontitis

Review Article

late periapical granulation tissues suggests that these cells participate inlesion stabilization and healing (53–56). However, recent clinical andexperimental findings indicate that the inhibition of helper T-lymphocyte function can contribute both to the attenuation andexacerbation of local inflammation and tissue destruction in apicalperiodontitis (57–59). The secretion of 2 anti-inflammatory cytokines,TGF-b and IL-10, showed a positive correlation with the ratio of CD8+ Tlymphocytes in large-sized periapical lesions and in lesions predomi-nated by B lymphocytes and plasma cells (‘‘B-type’’ lesions). These ob-servations suggest the importance of immunosuppressive reactions inthe late phase of lesion development in human apical periodontitis(44, 49). The overexpression of Th1-type (CCR1, CCR5, and CXCR3)and Th2-type (CCR2 and CCR3) chemokine receptors in individual hu-man periapical lesions compared with healthy tissue indicates theimportance of simultaneous action of both types of responses in diseasepathogenesis (45, 47). The frequency of Th1 cells consistentlyexceeded that of Th2 cells in every lesion type. Th1-type cytokines(IL-2, interferon gamma [IFN-g], and IL-12) were identified in100% of lesions, whereas Th2-type cytokines (IL-4 and IL-5) were de-tected only in a restricted number of cell culture supernatants from hu-man apical periodontitis lesions (43, 44). Mast cells, consistentlypresent in periapical granulation tissue, may contribute to Th2-type po-larization (60, 61). Two recently described T-helper cellsubpopulations, T-helper cell type 17 (Th17) and regulatory Tlymphocytes (CD4+/CD25hi/Foxp3+ ‘‘Treg’’ cells), were shown tocontribute to the cellular networking in advanced apical periodontitislesions (38, 46, 62, 63).

ECs are present in approximately 50% of periapical lesions. Their3-dimensional strands may merge to form true periradicular cysts andperiapical pocket or ‘‘bay’’ cysts. ECs have been suggested to perpetuatethe periapical inflammatory process (5, 64, 65). Epithelializedperiapical lesions are characterized by the production of high levelsof IL-6 and granulocyte-macrophage colony-stimulating factor (GM-CSF), and the presence of ECs was correlated with clinical symptomsin patients with chronic apical periodontitis (9, 65, 66). Naturalkiller cells (CD56/57+/CD3-) and natural killer T cells (CD56/57+/CD3+) make up approximately 1% of the granulation tissue. Theyhave been suggested to promote proapoptotic pathways for ECswithin the lesion. In cooperation with plasmacytoid DCs, theyproduce type I interferons with antiproliferatory and antiviralactivities (43, 67, 68).

Lesion expansion is associated with angioneogenesis in chronicinflammatory diseases. Vascular endothelial growth factors (VEGF)and their receptors (VEGF-R) play a critical role in angiogenesis. Theexpression of VEGFs and VEGF-Rs, in particular VEGF-A, -C, and –D,as well as VEGFR-2 and -3, has been shown immunohistochemicallyin human apical periodontitis of chronic nature. In addition to locali-zation on blood vessels, VEGFs and VGEF-Rs were also identified on neu-trophils, macrophages, and T and B lymphocytes. Up-regulateddownstream mediators of VEGF-mediated angiogenic activity wereshown in these samples by quantitative real-time polymerase chain re-action and compared with healthy PDL samples (69, 70).

Network of Regulatory Loopsin Chronic Apical Periodontitis

Results of immunophenotyping, gene expression analysis, andfunctional investigations indicate a key regulatory role for monocytesand DCs in the induction of antigen-specific T-cell responses. Moreover,these cells are essential for the polarization of the T-helper immune re-sponses toward Th1, Th2, Th17, and Treg pathways in advanced apicalperiodontitis. DCs andmacrophages differentially produce Th1 and Th2

158 M�arton and Kiss

cytokines. The former cells represent the major source of IL-12 and ofIL-23. DCs, cocultured in vitro with allogeneic T cells, secrete highlevels of IFN-g and stimulate the proliferation of CD4+ lymphocytes.A close association between CD83+ DCs and CD3+ T cells in humansamples has been observed with confocal laser scanning microscopy(42). In lymphocyte-rich areas, up-regulated expression of HLA-DRa-chain, CD83, and CD86 in DCs and CD28 in T lymphocytes, respec-tively, was observed (48). These observations suggest that DCs presentin human periapical lesions are able to process microbial antigens, acti-vate na€ıve T cells, support both Th1 and Th17 reactions, and are essen-tial for granuloma formation (71–73) (Fig. 2).

Macrophages may mediate protective and destructive stimuli at thesame time. Macrophages serve as APCs for effector T cells and producematuration factors for DCs, proinflammatory cytokines, arachidonicacid derivatives, and reactive oxygen intermediates stimulating boneresorption. Monocytes down-regulate the production of IFN-g in DC/CD4+ cell cocultures. Macrophages produce IL-10 in cooperationwith Th2 and Treg cells and IL-27 in cooperation with Treg cells(71) (Fig. 2). These 2 cytokines were shown to exert potent immuno-regulatory activities, which restrict untoward proinflammatory re-sponses and bone resorption in human periapical lesions (49, 72, 73).

IL-1b, produced by activated macrophages, is a primordial cyto-kine of the inflammatory phase in experimental periodontitis, mediatinglesion expansion (40, 74, 75). High levels of IL-1b, caused bypolymorphic alleles of the encoding gene alone or in combinationwith polymorphisms of genes encoding for IL-6 and TNF-a, are associ-ated with symptomatic periapical abscesses and root canal treatmentfailure (76, 77). Negative feedback mechanisms prevent IL-1b–induced severe destruction of periodontal tissues. In human samples,IL-1b and IL-6 induce suppression of cytokine signaling-3 proteinexpression, thereby diminishing bone resorption through achemokine-dependent mechanism. Moreover, IL-1b directly inhibitshuman osteoclast precursors (78, 79).

Th17 cells are present in all phases of induced rat periapical lesions,and their numbers increase during lesion expansion (63). IL-17–positivecells have been shown in human dental granulomas and radicular cystswhere they correlated with the presence of sinus tracts. The number ofTh17 cells was higher in periapical lesions than in control samples(46). IL-17 was invariably detected in lesion-derived MNC culture super-natants, and its level was significantly higher in symptomatic lesions thanin asymptomatic ones. The ability of IL-17 to induce neutrophil attractionand the activation of the soluble mediators GM-CSF, IL-1, IL-6, IL-8/CXCL8, and GROa/CXCL1 were shown to correlate positively with the ratioof neutrophils in periapical lesions (44, 80). Interestingly, completeabrogation of IL-17 responses, as shown in IL-17 receptor (IL-17RA)-deficient mice, resulted in enhanced periodontal bone destructioninduced by P. gingivalis. This indicates a nonredundant role for IL-17in mediating host defense via neutrophil mobilization (81).

TGF-b is a potent down-regulatory factor for IL-17–induced in-flammatory responses although the two cytokines do not exert mutualinhibitory effects (82). The major source of TGF-b is CD4+/CD25hi/Foxp3+ Treg cells, making up 30%–50% of all activated T-helper lym-phocytes (CD4+/CD25+ cells) in human periapical lesions (62, 83,84). The antiproliferative activity of lesion-derived MNCs on autologousand allogeneic peripheral blood-derived MNCs depended on the pres-ence of IL-10 and Tregs within the cell suspension (62). Clinical andexperimental observations suggest that the balance between Th17and Treg cells is critical to the outcomes of periapical infection in termsof ‘‘destruction’’ (lesion expansion with or without symptoms) or ‘‘pro-tection’’ (healing). The Th17/Treg ratio may influence the intensity ofthe inflammatory response, the efficacy of local and systemic antimicro-bial defense reactions, and the activity of processes resulting in bone

JOE — Volume 40, Number 2, February 2014

Page 5: Overlapping Protective and Destructive Regulatory Pathways in Apical Periodontitis

Figure 2. The balance of Th17 and Treg cells regulates the efficacy of antimicrobial defense reactions, the intensity of destructive inflammatory responses, and therepair processes. Th17 lymphocytes produce the master proinflammatory cytokine IL-17, which, by inducing the secretion of GM-CSF, IL-1, IL-6, IL-8/CXCL8, andGROa/CXCL1 and the expression of RANKL, results in the recruitment of PMNs and MNPs and enhances osteoclastogenesis. The most potent inducers of Th17differentiation and IL-17 expression are IL-1b and IL-23, which are produced mainly by DCs activated by bacterial molecules and proinflammatory cytokines.CD4+/CD25hi/Foxp3+ Treg cells express TGF-b and IL-10. The latter cytokine, together with IL-27 produced by Treg cells in cooperation with monocyte-derived APCs, suppresses IL-8/CXCL8 release by inhibiting Th17 lymphocytes. TGF-b is considered as the major ‘‘protective’’ factor in apical periodontitis. Itdown-regulates IL-17–induced inflammatory responses. However, in the presence of monocyte-derived APCs, IL-2 and IL-15 Treg cells can transdifferentiateinto Th17 lymphocytes, probably through a small subset of double-positive IL-17+/Foxp3+ cells (see text for references).

Review Article

resorption or regeneration. Treg and Th17 cells, characterized by areciprocally interrelated differentiation program, may not only inhibitbut also support the differentiation and expansion of one and another.TGF-b, considered as one of the major ‘‘protective’’ factors in apicalperiodontitis, may drive Th17 polarization of na€ıve CD4 T-lymphocytesin the presence of IL-1b or IL-6 (85, 86). Moreover, human Treg cellscan transdifferentiate into IL-17–producing cells in the presence ofAPCs and exogenous IL-2 and IL-15 (87). The plasticity of the T-celllineage may depend in part on the presence of a small subset of doublepositive T-helper lymphocytes coexpressing ‘‘lineage-restricted mole-cules’’ (eg, IL-17 and Foxp3). These partially differentiated T cellsmay retain their capability to become IL-17–producing cells (Fig. 2).Such cells have been observed in human inflammatory lesions,including apical periodontitis. They have been suggested to play atwin role in antimicrobial defense and restricting the intensity of inflam-matory responses (44, 88).

Molecular Regulation of Effector MechanismsThe first step in lesion development is the destruction of extracel-

lular matrix by serine proteases and matrix metalloproteinases (MMPs)secreted from the lysosomal granules of phagocytes. In experimental ratperiradicular lesions, the number of elastase-expressing neutrophilgranulocytes andmacrophages increased up to 28 days after pulp expo-sure. Cells expressing the secretory leukocyte protease inhibitoremerged after day 28 in parallel with the start of lesion shrinkage(89, 90). PDLFs represent additional sources of MMPs, which areusually secreted as latent proenzymes and become activated by otherhuman endopeptidases, bacterial proteases, and reactive oxygenintermediates. MMP synthesis and release are tightly controlled by

JOE — Volume 40, Number 2, February 2014

the local cytokine milieu, endotoxins, and cell-to-cell contacts(91–93). The presence of a broad set of MMPs at the messengerRNA and protein levels has been widely observed in inflamed but nothealthy periradicular tissues. This applies to both patients andexperimental animals (10, 39, 94–98). Significantly higher numbersof gram-negative bacteria and elevated MMP-9 expression have beenobserved in patients with symptomatic periapical lesions whencompared with asymptomatic patients (99). The kinetics of MMP up-and down-regulation in experimental apical periodontitis models isassociated with lesion expansion and stabilization and is in accordwith a dominant role for IL-1 in early expanding periapical lesionsand the immunoregulatory actions of TGF-b during later stages (39,91, 100). Polymorphisms of MMP2 and MMP3 genes, encoding forMMP-2 and -3, were reported to be associated with an increased riskof developing periapical lesions as well as predicting healing responsesin patients with advanced caries (101).

Effector molecules produced by phagocytes may be key therapeu-tic targets in periapical disease. In addition to the well-established bene-ficial effects of Ca(OH)2 for pulp capping in humans, this root canaldressing was shown to decrease the number of MMP-positive cells inexperimental apical periodontitis (2, 10). In contrast, systemicantioxidant therapy failed to prevent lesion development or todecrease lesion size of induced apical periodontitis in a rat model oftype 2 diabetes (102).

The equilibriumbetween bone formation and resorption in the peri-apical region relies on the well-recognized interactions between osteo-blasts and osteoclasts involving the stimulatory action of receptoractivator of nuclear factor kappa-B (RANK)-RANK ligand (RANKL). Themajor negative regulator of the RANKL-RANK pathway is the soluble decoyreceptor osteoprotegerin (OPG) (103) (Fig. 3). In addition to

Protective and Destructive Regulatory Pathways 159

Page 6: Overlapping Protective and Destructive Regulatory Pathways in Apical Periodontitis

Figure 3. The equilibrium between bone formation and resorption is regulated by the interaction between osteoblasts and osteoclasts and their precursors.Membrane-bound RANKL expressed in osteoblasts stimulates the differentiation of osteoclast precursors and the bone-resorbing activity of mature osteoclaststhrough interaction with its receptor RANK. The soluble form of RANK, OPG, inhibits RANKL-RANK interaction. RANKL-RANK signaling activates nuclear factor-kB. Osteoclasts are descendants of the bone marrow–derived monocyte/macrophage lineage and are characterized by the expression of tartarate-resistant acidphosphatase (TRAP). The osteoclast releases hydrogen ions (H+) through the action of carbonic anhydrase, resulting in a low pH environment in the bone-resorbing lacuna. Bicarbonate (HCO3

�)-chloride (Cl�) ion exchange restores the intracellular pH of this cell. The expression of RANKL and OPG is reciprocallyregulated by IL-1, IL-6, IL-11, IL-17, M-CSF, GM-CSF, and TNF-a, which induce bone resorption. sRANKL is an additional stimulator of bone resorption. LPS maycontribute to the differentiation of osteoclast precursors. In contrast, IFN-g and IL-10 decrease the RANKL-OPG expression ratio and attenuate bone resorption. Inthe bone formation process, osteoblasts differentiate into osteocytes.

Review Article

osteoblasts, monocytes, DCs, fibroblasts, and endothelial and epithelialcells, PMNs and activated T lymphocytes express RANKL in apical peri-odontitis lesions from patients and experimental animals. The widespreadmanifestation of RANKL may explain the characteristic shift toward bonedestruction (104–106). Expression levels of both RANKL and OPG inhuman dental granuloma samples were higher than in healthy PDLcells, indicating an elevated bone turnover in apical periodontitisversus noninflamed periapical tissues. The ratios of RANKL and OPGexpression exhibited a heterogeneous pattern suggesting that somelesions were in the stage of active bone resorption, whereas otherswere at the stable chronic or healing stages (105, 107). The numbersof osteoclasts and the RANKL:OPG expression ratios show parallelincreases in expanding experimental periapical lesions (79, 108, 109).TNF-a, IL-1b, IL-6, and IL-17 reciprocally regulate the expression ofRANKL and OPG favoring RANKL expression (110, 111). TNF-a, IL-1a,and macrophage migration inhibitory factor induce further osteoclaststimulating molecules such as IL-6, IL-11, IL-17, M-CSF, RANKL, andprostaglandin E2 in human dental pulp cells and induced rat periapicallesions (112, 113). Periodontopathogenic bacteria and their constituentsinduce RANKL andOPG expression in PDLFs and dental pulp cells directlyor by up-regulating osteolytic cytokines, such as TNF-a and IL-1b(114–116) (Fig. 3).

In contrast, some other cytokines, including IFN-g, decrease theRANKL:OPG expression ratio, thereby attenuating bone resorption(110). The osteoprotective role of IFN-g, together with IL-10, intercel-lular adhesion molecule-1 (ICAM-1), and CCR5, was confirmed inmice with genetic ablations of the genes encoding for these factors(117) (Fig. 3). Experimental observations indicate that direct cross-talk between IFN-g and the RANKL-RANK signaling pathway and the

160 M�arton and Kiss

inhibition of Th17 polarization by IFN-gmay contribute to bone protec-tion (118, 119). Other bone-protective mechanisms do not directlyinvolve the RANKL-RANK signaling pathway. In experimental rat apicalperiodontitis, the expression of the non–DNA-binding intracellular coac-tivator was inversely related to osteoblast activity and lesion progression(120). In a similar experimental model, the cholesterol-lowering agentsimvastatin suppressed bone resorption and decreased the expression ofthe angiogenic cysteine-rich 61 protein. This was carried out by enhance-ment of the transcription factor Forkhead/winged helix box protein O3a(FoxO3a). These in vivo results have been corroborated in a murineosteoblast cell line (121).

Regulation of Repair MechanismsTissue destruction and bone loss is self-limiting in most cases of

apical periodontitis. Induced apical periodontitis in rats showed adecrease in the number of infiltrating osteoclasts accompanied bylesion size stabilization after 21 days after pulp exposure. Thus, a spon-taneously established new equilibrium between root canal pathogensand anti-infective defense mechanisms may prevent tissue destructionand initiate remodeling (122). Stem cells able to generate dentin-likestructures and odontoblast-like cells were first reported in the dentalpulp in 2000 (123). A growing number of publications have since re-ported several stem cell populations of dental origin (124–126).Because pulp-derived stem cells of teeth affected by apical periodontitiswill be increasingly compromised by inflammation, stem cell recruit-ment from the PDL of neighboring teeth may be favored (127). Underappropriate conditions, PDL-derived stem cells may be suitable forengineering bone tissue, PDL, and cementum simultaneously

JOE — Volume 40, Number 2, February 2014

Page 7: Overlapping Protective and Destructive Regulatory Pathways in Apical Periodontitis

Review Article

(125, 128). The remaining granulation tissue after endodontictreatment may provide the stimulus required for stem cellrecruitment, proliferation, and differentiation. Regenerative responsesare mediated by a variety of growth factors, including TGF-b andother members of the TGF-b superfamily (125, 129–131).

ConclusionInflammatory reactions in the periapical tissues in response to

endodontic infection are self-limiting in the majority of cases. Both mi-crobial influx from the infected root canal and excessive destruction oftooth-supporting structures are controlled for extended periods of timeby complex interactions between immunoinflammatory cells and solu-ble mediators. The permanent shift between anti- and proinflammatoryprocesses influences the balance between tissue destruction and regen-eration at different stages of apical periodontitis. This dynamic equilib-rium between protection from infection and host tissue injury andrepair processes is driven by overlapping networks of pleiotropicand redundant regulatory mechanisms. Recent advances have high-lighted important new aspects of overlapping ‘‘destructive’’ and ‘‘protec-tive’’ reactions in apical periodontitis. Focus on cellular and moleculartargets may allow the development and translation of novel therapeutictools tomoderate periapical tissue destruction and stimulate supportingtissue regeneration.

AcknowledgmentsThe authors thank Dr Anthony J. Smith, Birmingham, UK, and

Dr Gunnar Bergenholz, Gothenburg, Sweden, for their criticalreading of this manuscript and Erika S�ari for graphical work.

Supported by the T�AMOP 4.2.1./B-09/1/KONV-2010-0007project. The project is cofinanced by the European Union and theEuropean Social Fund and the OTKA (Hungarian Scientific ResearchFund) K 105034 grant.

The authors deny any conflicts of interest related to thisstudy.

References1. Abbott PV. Classification, diagnosis and clinical manifestations of apical periodon-

titis. Endod Top 2004;8:36–54.2. M�arton IJ, Kiss C. Protective and destructive immune reactions in apical periodon-

titis. Oral Microbiol Immunol 2000;15:139–50.3. Garlet GP. Destructive and protective roles of cytokines in periodontitis: a re-

appraisal from host defense and tissue destruction viewpoints. J Dent Res 2010;89:1349–63.

4. Puliti M, von Hunolstein C, Bistoni F, et al. Role of macrophages in experimentalgroup B streptococcal arthritis. Cell Microbiol 2002;4:691–700.

5. Nickolaychuk B, McNicol A, Gilchrist J, et al. Evidence for a role of mitogen-activated protein kinases in proliferating and differentiating odontogenic epitheliaof inflammatory and developmental cysts. Oral Surg Oral Med Oral Pathol Oral Ra-diol Endod 2002;93:720–9.

6. Takahashi K, McDonald DG, Murayama Y, et al. Cell synthesis, proliferation andapoptosis in human dental periapical lesions analysed by in situ hybridisationand immunohistochemistry. Oral Dis 1999;5:313–20.

7. J€onsson D, Nebel D, Bratthall G, et al. The human periodontal ligament cell: afibroblast-like cell acting as an immune cell. J Periodont Res 2011;46:153–7.

8. Kabashima H, Nagata K, Maeda K, et al. Involvment of substance P, mast cells, TNF-alpha and ICAM-1 in the infiltration of inflammatory cells in human periapicalgranulomas. J Oral Pathol Med 2002;31:175–80.

9. Ohshima M, Yamaguchi Y, Micke P, et al. In vitro characterization of the cytokineprofile of the epithelial cell rests of Malassez. J Periodontol 2008;79:912–9.

10. Paula-Silva FW, da Silva LA, Kapila YL. Matrix metalloproteinase expression in teethwith apical periodontitis is differentially modulated by the modality of root canaltreatment. J Endod 2010;36:231–7.

11. Darveau RP. The oral microbial consortium’s interaction with the periodontalinnate defense system. DNA Cell Biol 2009;28:389–95.

JOE — Volume 40, Number 2, February 2014

12. Han DC, Huang GT, Lin LM, et al. Expression of MHC class II, CD70, CD80, CD86and pro-inflammatory cytokines is differentially regulated in oral epithelial cellsfollowing bacterial challenge. Oral Microbiol Immunol 2003;18:350–8.

13. HuangGT,KimD,LeeJK,etal.Interleukin-8andintercellularadhesionmolecule1regu-lationinoralepithelialcellsbyselectedperiodontalbacteria:multipleeffectsofPorphyr-omonasgingivalisviaantagonisticmechansims. Infect Immun2001;69:1364–72.

14. Liu F, Abiko Y, Nishimura M, et al. Expression of inflammatory cytokines and beta-defensin 1 mRNAs in porcine epithelial rests of Malassez in vitro. Med ElectronMicrosc 2001;34:174–8.

15. Lukic A, Vojvodic D, Majstorovic D, et al. Production of interleukin-8 in vitro bymononuclear cells isolated from human periapical lesions. Oral Microbiol Immu-nol 2006;21:296–300.

16. Takeichi O, Hama S, Iwata K, et al. Confocal immunolocalization of VE-cadherin-and CXC chemokine-expressing endothelial cells in periapical granulomas. Int En-dod J 2008;41:401–7.

17. Hatakeyama J, Tamai R, Sugiyama A, et al. Contrasting responses of human gingivaland periodontal ligament fibroblasts to bacterial cell-surface components throughthe CD14/Toll-like receptor system. Oral Microbiol Immunol 2003;18:14–23.

18. Creagh EM, O’Neill LA. TLRs, NLRs and RLRs: a trinity of pathogen sensors that co-operate in innate immunity. Trends Immunol 2006;27:352–7.

19. Strober W, Murray PJ, Kitani A, et al. Signalling pathways and molecular interac-tions of NOD1 and NOD2. Nat Rev Immunol 2006;6:9–20.

20. Morandini AC, Sipert CR, Gasparoto TH, et al. Differential production of macro-phage inflammatory protein-1alpha, stromal-derived factor-1, and IL-6 by humancultured periodontal ligament and gingival fibroblasts challenged with lipopolysac-charide from P. gingivalis. J Periodontol 2010;81:310–7.

21. Scheres N, Laine ML, de Vries TJ, et al. Gingival and periodontal ligament fibro-blasts differ in their inflammatory response to viable Porphyromonas gingivalis.J Periodontal Res 2010;45:262–70.

22. Yamaji Y, Kubota T, Sasaguri K, et al. Inflammatory cytokine gene expression inhuman periodontal ligament fibroblasts stimulated with bacterial lipopolysaccha-rides. Infect Immun 1995;63:3576–81.

23. El-Awady AR, Messer RL, Gamal AY, et al. Periodontal ligament fibroblasts sustaindestructive immune modulators of chronic periodontitis. J Periodontol 2010;81:1324–35.

24. Hern�adi K, Szalm�as A, Mogyor�osi R, et al. Prevalence and activity of Epstein-Barrvirus and human cytomegalovirus in symptomatic and asymptomatic apical peri-odontitis lesions. J Endod 2010;36:1485–9.

25. Li H, Chen V, Chen Y, et al. Herpesviruses in endodontic pathoses: association ofEpstein-Barr virus with irreversible pulpitis and apical periodontitis. J Endod2009;35:23–9.

26. Slots J, Sabeti M, Simon JH. Herpesviruses in periapical pathosis: an etiopa-thogenic relationship? Oral Surg Oral Med Oral Pathol Oral Radiol Endod2003;96:327–31.

27. Hern�adi K,Gy€ongy€osi E,M�esz�aros B, et al. Elevated tumor necrosis factor-alpha expres-sion in periapical lesions infected by Epstein-Barr virus. J Endod 2013;39:456–60.

28. Sabeti M, Simon JH, Nowzari H, et al. Cytomegalovirus and Epstein-Barr virusactive infection in periapical lesions of teeth with intact crowns. J Endod2003;29:321–3.

29. Sabeti M, Valles Y, Nowzari H, et al. Cytomegalovirus and Epstein-Barr virus DNAtranscription in endodontic symptomatic lesions. Oral Microbiol Immunol 2003;18:104–8.

30. Slots J. Herpesviral-bacterial interactions in periodontal diseases. Periodontol2000 2010;52:117–40.

31. Berglundh T, Zitzmann NU, Donati M. Are peri-implantitis lesions different fromperiodontitis lesions? J Clin Periodontol 2011;38:188–202.

32. Garlet TP, Coelho U, Repeke CE, et al. Differential expression of osteoblast andosteoclast chemmoatractants in compression and tension sides during orthodonticmovement. Cytokine 2008;42:330–5.

33. Schmaltz G, Schweikl H, Hiller KA. Release of prostaglandin E2, IL-6 and IL-8 fromhuman oral epithelial culture models after exposure to compounds of dental ma-terials. Eur J Oral Sci 2000;108:442–8.

34. Silva TA, Lara VS, Silva JS, et al. Dentin sialoprotein and phosphoprotein induceneutrophil recruitment: a mechanism dependent on IL-1beta, TNF-beta, andCXC chemokines. Calcif Tissue Int 2004;74:532–41.

35. Lee SK, Pi SH, Kim SH, et al. Substance P regulates macrophage inflammatory pro-tein 3alpha/chemokine C-C ligand 20 (CCL20) with heme oxygenase-1 in humanperiodontal ligament cells. Clin Exp Immunol 2007;150:567–75.

36. Silva TA, Garlet GP, Fukada SY, et al. Chemokines in oral inflammatory diseases:apical periodontitis and periodontal disease. J Dent Res 2007;86:306–19.

37. M�arton IJ, Rot A, Schwarzinger E, et al. Differential in situ distribution ofinterleukin-8, monocyte chemoattractant protein-1 and Rantes in human chronicperiapical granuloma. Oral Microbiol Immunol 2000;15:63–5.

38. AlShwaimi E, Purcell P, Kawai T, et al. Regulatory T cells in mouse periapical le-sions. J Endod 2009;35:1229–33.

Protective and Destructive Regulatory Pathways 161

Page 8: Overlapping Protective and Destructive Regulatory Pathways in Apical Periodontitis

Review Article

39. Corotti MV, Zambuzzi WF, Paiva KB, et al. Immunolocalization of matrix

metalloproteinases-2 and -9 during apical periodontitis development. Arch OralBiol 2009;54:764–71.

40. Martinez ZR, Naruishi K, Yamashiro K, et al. Gene profiles during root canal treat-ment in experimental rat periapical lesions. J Endod 2007;33:936–43.

41. Martinho FC, Chiesa WM, Leite FR, et al. Correlation between clinical/radiographicfeatures and inflammatory cytokine networks produced by macrophages stimu-lated with endodontic content. J Endod 2012;38:740–5.

42. Chae P, Im M, Gibson F, et al. Mice lacking monocyte chemoattractant protein 1have enhanced susceptibility to an interstitial polymicrobial infection due toimpaired monocyte recruitment. Infect Immun 2002;70:3164–9.

43. Colic M, Lukic A, Vucevic D, et al. Correlation between phenotypic characteristicsof mononuclear cells isolated from human periapical lesions and their in vitroproduction of Th1 and Th2 cytokines. Arch Oral Biol 2006;51:1120–30.

44. Colic M, Gazivoda D, Vucevic D, et al. Proinflammatory and immunoregulatorymechanisms in periapical lesions. Mol Immunol 2009;47:101–13.

45. Kabashima H, Yoneda M, Nagata K, et al. The presence of chemokine rceptor (CCR5,CXCR3, CCR3)-positive cells and chemokine (MCP1, MIP-1alpha, MIP-1beta, IP-10)-positive cells in human periapical granulomas. Cytokine 2001;16:62–6.

46. Marcal JRB, Samuel RO, Fernandes D, et al. T-helper cell type 17/regulatory T-cellimmunoregulatory balance in human radicular cysts and periapical granulomas.J Endod 2010;36:995–9.

47. Silva TA, Garlet GP, Lara VS, et al. Differential expresion of chemokines and chemo-kine receptors in inflammatory periapical diseases. Oral Microbiol Immunol2005;20:310–6.

48. Kaneko T, Okiji T, Kaneko R, et al. H. Antigen-presenting cells in human radiculargranulomas. J Dent Res 2008;87:553–7.

49. Gazivoda D, Dzopalic T, Bozic B, et al. Production of proinflammatory and immu-noregulatory cytokines by inflammatory cells from periapical lesions in culture.J Oral Pathol Med 2009;38:605–11.

50. Lukic A, Vasilijic S, Majstorovic I, et al. Characterization of antigen-presenting cellsin human apical periodontitis lesions by flow cytometry and immunocytochem-istry. Int Endod J 2006;39:626–36.

51. Carrillo C, Penarrocha M, Penarrocha M, et al. Immunohistochemical study ofLangerhans cells in periapical lesions: correlation with inflammatory cell infiltra-tion and epithelial cell proliferation. Med Oral Patol Oral Cir Bucal 2010;15:e335–9.

52. Suzuki T, Kumamoto H, Ooya K, et al. Immunohistochemical analysis of CD1a-labeled Langerhans cells in human dental periapical inflammatory lesions—cor-relation with inflammatory cells and epithelial cells. Oral Dis 2001;7:336–43.

53. Akamine A, Hashiguchi I, Toriya Y, et al. Immunohistochemical examination on thelocalization of macrophages and plasma cells in induced rat periapical lesions.Endod Dent Traumatol 1994;10:121–8.

54. Kawashima N, Stashenko P. Expression of bone-resorptive and regulatory cyto-kines in murine priapical inflammation. Arch Oral Biol 1999;44:55–66.

55. Stashenko P, Wang CY, Tani-Ishii N, et al. Pathogenesis of induced rat periapicallesions. Oral Surg Oral Med Oral Pathol 1994;78:494–502.

56. Rodini CO, Lara VS. Study of the expression of CD68+ macrophages and CD8+ Tcells in human granulomas and periapical cysts. Oral Surg Oral Med Oral PatholOral Radiol Endod 2001;92:221–7.

57. de Brito LC, da Rosa MA, Lopes VS, et al. Brazilian HIV-infected population: assess-ment of the needs of endodontic treatment in the post-highly active antiretroviraltherapy era. J Endod 2009;35:1178–81.

58. Kawahara T, Murakami S, Noiri Y, et al. Effects of cyclosporin-A-induced immuno-suppression on periapical lesions in rats. J Dent Res 2004;83:683–7.

59. Yamasaki M, Morimoto T, Tsuji M, et al. Role of IL-2 and helper T-lymphocytes inlimiting periapical pathosis. J Endod 2006;32:24–9.

60. Drazic R, Sopta J, Minic AJ. Mast cells in periapical lesions: potential role in theirpathogenesis. J Oral Pathol Med 2010;39:257–62.

61. de Oliveira Rodini C, Batista AC, Lara VS. Comparative immunohistochemical studyof the presence of mast cells in apical granulomas and periapical cysts: possiblerole of mast cells in the course of human periapical lesions. Oral Surg Oral MedOral Pathol Oral Radiol Endod 2004;97:59–63.

62. Colic M, Gazivoda D, Vucevic D, et al. Regulatory T-cells in periapical lesions.J Dent Res 2009;88:997–1002.

63. Xiong H, Wei L, Peng B. Immunohistochemical localization of IL-17 in induced ratperiapical lesions. J Endod 2009;35:216–20.

64. Nair PNR. Non-microbial etiology: periapical cysts sustain post-treatment apicalperiodontitis. Endod Top 2003;6:96–113.

65. Lin LM, Huang GT, Rosenberg PA. Proliferation of epithelial cell rests, formation ofapical cysts, and regression of apical cysts after periapical wound healing. J Endod2007;33:908–16.

66. Radics T, Kiss C, Tar I, et al. Interleukin-6 and granulocyte-macrophage colony-stimulating factor in apical periodontitis: correlation with clinical and histologicfindings of the involved teeth. Oral Microbiol Immunol 2003;18:9–13.

162 M�arton and Kiss

67. Moreira PR, Santos DF, Martins RD, et al. CD57+ cells in radicular cys. Int Endod J2000;33:99–102.

68. Saboia-Dantas CJ, Coutrin de Toledo LF, Siqueira JF Jr, et al. Natural killer cells andalterations in collagen density: signs of periradicular herpesvirus infection? ClinOral Investig 2008;12:129–35.

69. Bletsa A, Virtej A, Berggreen E. Vascular endothelial growth factors and receptorsare up-regulated during development of apical periodontitis. J Endod 2012;38:628–35.

70. Virtej A, Loes SS, Berggreen E, et al. Localization and signaling patterns of vascularendothelial growth factors and receptors in human periapical lesions. J Endod2013;39:605–11.

71. Colic M, Gazivoda D, Vasilijic S, et al. Production of IL-10 and IL-12 by antigen-presenting cells in periapical lesions. J Oral Pathol Med 2010;39:690–6.

72. Colic M, Gazivoda D, Majstorovic I, et al. Immunomodulatory activity of IL-27 inhuman periapical lesions. J Dent Res 2009;88:1142–7.

73. Walker KF, Lappin DF, Takahashi K, et al. Cytokine expression in periapical gran-ulation tissue as assessed by immunohistochemistry. Eur J Oral Sci 2000;108:195–201.

74. Nakae S, Asano M, Horai R, et al. Interleukin-1beta, but not interleukin-1alpha, isrequired for T-cell-dependent antibody production. Immunology 2001;104:402–9.

75. Bloemen V, Schoenmaker T, de Vries TJ, et al. Direct cell-cell contact betweenperiodontal ligament fibroblasts and osteoclasts precursors synergistically in-creases the expression of genes related to osteoclastogenesis. J Cell Physiol2010;222:565–73.

76. de S�a AR, Moreira PR, Xavier GM, et al. Association of CD14, IL1B, IL6, IL10 andTNFA functional gene polymorphisms with symptomatic dental abscesses. Int En-dod J 2007;40:563–72.

77. Morsani JM, Aminoshariae A, Han YW, et al. Genetic predisposition to persistentapical periodontitis. J Endod 2011;37:455–9.

78. Fukushima A, Kajiya H, Izumi T, et al. Pro-inflammatory cytokines induce suppres-sor of cytokine signaling-3 in human periodontal ligament cells. J Endod 2010;36:1004–8.

79. Lee B, Kim TH, Jun JB, et al. Direct inhibition of human RANK+ osteoclasts pre-cursors identifies a homeostatic function of IL-1beta. J Immunol 2010;185:5926–34.

80. Yu JJ, Gaffen SL. Interleukin-17: a novel inflammatory cytokine that bridges innateand adaptive immunity. Front Biosci 2008;13:170–7.

81. Yu JJ, Ruddy MJ, Wong GC, et al. An essential role for IL-17 in preventing pathogen-initiated bone destruction: recruitment of neutrophils to inflamed bone requiresIL-17 receptor-dependent signals. Blood 2007;109:3794–802.

82. Zhu J, Paul WE. CD4 T cells: fates, functions, and faults. Blood 2008;112:1557–69.83. Couper KN, Blount DG, Riley EM. IL-10: the master regulator of immunity to infec-

tion. J Immunol 2008;180:5771–7.84. Xu L, Xu W, Wen Z, et al. In situ prior proliferation of CD4+ CCR6+ regulatory T

cells facilitated by TGF-b secreting DCs is crucial for their enrichment and sup-pression in tumor immunity. PLoS One 2011;6:e20282.

85. Veldhoen M, Hocking RJ, Atkins CJ, et al. TGFbeta in the context of an inflammatorycytokine milieu supports de novo differentiation of IL-17-producing T cells. Immu-nity 2006;24:179–89.

86. Xu L, Kitani A, Fuss I, et al. Cutting edge: regulatory T cells induce CD4+CD25-Foxp3- T cells or are self-induced to become Th17 cells in the absence of exog-enous TGF-beta. J Immunol 2007;178:6725–9.

87. Koenen HJ, Smeets RL, Vink PM, et al. Human CD25highFoxp3pos regulatory Tcells differentiate into IL-17-producing cells. Blood 2008;112:2340–52.

88. Jia X, Hu M, Wang C, et al. Coordinated gene expression of Th17- and Treg-associated molecules correlated with resolution of the monophasic experimentalautoimmune uveitis. Mol Vis 2011;17:1493–507.

89. Morimoto T, Yamasaki M, Nakata T, et al. The expression of macrophage andneutrophil elastases in rat periradicular lesions. J Endod 2008;34:1072–6.

90. Tsuji M, Yamasaki M, Amano K, et al. Histochemical localization of neutral prote-ases released during development of rat periradicular lesion. Arch Oral Biol 2009;54:1128–35.

91. Chang YC, Yang SF, Lai CC, et al. Regulation of matrix metalloproteinase productionby cytokines, pharmacological agents and periodontal pathogens in human peri-odontal ligament fibroblast cultures. J Periodontal Res 2002;37:196–203.

92. Hern�andez M, Gamonal J, Tervahaltiala T, et al. Associations between matrixmetalloproteinase-8 and -14 and myeloperoxidase in gingival crevicular fluidfrom subjects with progressive chronic periodontitis; a longitudinal study.J Periodontol 2010;81:1644–52.

93. Webster NL, Crowe SM. Matrix metalloproteinases, their production by monocytesand macrophages and their potential role in HIV-related diseases. J Leukoc Biol2006;80:1052–66.

94. Shin SJ, Lee JI, Baek SH, et al. Tissue levels of matrix metaloproteinases in pulp andperiapical lesions. J Endod 2002;28:313–5.

JOE — Volume 40, Number 2, February 2014

Page 9: Overlapping Protective and Destructive Regulatory Pathways in Apical Periodontitis

Review Article

95. Leonardi R, Caltabiano R, Loreto C. Collagenase-3 (MMP-13) is expressed in peri-

apical lesions: an immunohistochemical study. Int Endod J 2005;38:297–301.96. Buzoglu HD, Unal H, Ulger C, et al. The zymographic evaluation of gelatinase

(MMP-2 and -9) levels in acute and chronic periapical abscesses. Oral SurgOral Med Oral Pathol Oral Radiol Endod 2009;108:e121–6.

97. Carneiro E, Menezes R, Garlet GP, et al. Expression analysis of matrixmetalloproteinase-9 in epithelialized and nonepithelialized apical periodontitis le-sions. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2009;107:127–32.

98. de Paula-Silva FW, D’Silva NJ, da Silva LA, et al. High matrix metalloproteinase ac-tivity is a hallmark of periapical granulomas. J Endod 2009;35:1234–42.

99. Ahmed GM, El-Baz AA, Hashem AA, Shalaan AK. Expression levels of matrixmetalloproteinase-9 and gram-negative bacteria in symptomatic and asymptomaticperiapical lesions. J Endod 2013;39:444–8.

100. Chang YC, Yang SF, Hsieh YS. Regulation of matrix metalloproteinase-2 productionby cytokines and pharmacological agents in human pulp cell cultures. J Endod2001;27:679–82.

101. Menezes-Silva R, Khaliq S, Deeley K, et al. Genetic susceptibility to periapical dis-ease: conditional contribution of MMP2 and MMP3 genes to the development ofperiapical lesions and healing response. J Endod 2012;38:604–7.

102. Wolle CF, Zollmann LA, Bairros PO, et al. Outcome of periapical lesions in a ratmodel of type 2 diabetes: refractoriness to systemic antioxidant therapy.J Endod 2013;39:643–7.

103. Graves DT, Oates T, Garlet GP. Review of osteoimmunology and the host responsein endodontic and periodontal lesions. J Oral Microbiol 2011;3:5304.

104. Menezes R, Bramante CM, da Silva Pavia KB, et al. Receptor activator NFkappaB-ligand and osteoprotegerin protein expression in human periapical cysts and gran-ulomas. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2006;102:404–9.

105. Vernal R, Dezerga A, Dutzan N, et al. RANKL in human periapical granuloma:possible involvement in periapical bone destruction. Oral Dis 2006;12:283–9.

106. Zhang C, Yang L, Peng B. Critical role of NFATc1 in periapical lesions. Int Endod J2010;43:109–14.

107. Menezes R, Garlet TP, Letra A, et al. Differential patterns of receptor activator ofnuclear factor kappa B ligand/osteoprotegein expression in human periapicalgranulomas: possible association with progressive or stable nature of the lesions.J Endod 2008;34:932–8.

108. Kawashima N, Suzuki N, Yang G, et al. Kinetics of RANKL, RANK and OPG expres-sions in experimentally induced rat periapical lesions. Oral Surg Oral Med OralPathol Oral Radiol Endod 2007;103:707–11.

109. Wang L, Peng B. Correlation between platelet-derived growth factor B chain andbone resorption in rat periapical lesions. J Endod 2007;33:709–11.

110. Nakashima T, Kobayashi Y, Yamasaki S, et al. Protein expression and functionaldifference of membrane-bound and soluble receptor activator of NF-kappaBligand: Modulation of the expression by osteotropic factors and cytokines. Bio-chem Biophys Res Commun 2000;275:768–75.

111. Valverde P, Kawai T, Taubman MA. Potassium channel-blockers as therapeuticagents to interfere with bone resorption of periodontal disease. J Dent Res2005;84:488–99.

112. Kim YS, Min KS, Lee HD, et al. Effect of cytosolic phospholipase A2 on proinflam-matory cytokine-induced bone resorptive genes including receptor activator of nu-clear factor kappa B ligand in human dental pulp cells. J Endod 2010;36:636–41.

JOE — Volume 40, Number 2, February 2014

113. Liu L, Peng B. The expression of macrophage migration inhibitory factor is corre-lated with receptor activator of nuclear factor kappa B ligand in induced rat peri-apical lesions. J Endod 2013;39:984–9.

114. Wada N, Maeda H, Yoshimine Y, et al. Lipopolysaccharide stimulates expression ofosteoprotegerin and receptor activator of NF-kappa B ligand in periodontal liga-ment fibroblasts through the induction of interleukin-1 beta and tumor necrosisfactor-alpha. Bone 2004;35:629–35.

115. Kajiya M, Giro G, Taubman MA, et al. Role of periodontal pathogenic bacteria inRANKL-mediated bone destruction in periodontal disease. J Oral Microbiol2010;2:5532.

116. Belibasakis GN, Meier A, Guggenheim B, et al. Oral biofilm challenge regulates theRANKL-OPG system in periodontal ligament and dental pulp cells. Microb Pathog2011;50:6–11.

117. De Rossi A, Rocha LB, Rossi MA. Interferon-gamma, interleukin-10, intercellularadhesion molecule-1, and chemokine receptor 5, but not interleukin-4, attenuatethe development of periapical lesions. J Endod 2008;34:31–8.

118. Takayanagi H, Ogasawara K, Hida S, et al. T-cell-mediated regulation of osteoclas-togenesis by signalling cross-talk between RANKL and IFN-gamma. Nature 2000;408:600–5.

119. Harrington LE, Hatton RD, Mangan PR, et al. Interleukin 17-producing CD4+effector T cells develop via lineage distinct from the T helper type 1 and 2 lineages.Nat Immunol 2005;6:1123–32.

120. Lee YL, Lin SK, Hong CY, et al. Major histocompatibility complex class II transac-tivator inhibits cysteine-rich 61 expression in osteoblastic cells and its implicationin the pathogenesis of periapical lesions. J Endod 2010;36:1021–5.

121. Lin LD, Lin SK, Chao YL, et al. Simvastatin suppresses osteoblastic expression ofCyr61 and progression of apical periodontitis through enhancement of the tran-scription factor Forkhead/winged helix box protein O3a. J Endod 2013;39:619–25.

122. Wang L, Zhang R, Xiong H, et al. The involvement of platelet-derived growth factor-A in the course of apical periodontitis. Int Endod J 2011;44:65–71.

123. Gronthos S, Mankani M, Brahim J, et al. Postnatal human dental pulp stem cells(DPSCs) in vitro and in vivo. Proc Natl Acad Sci U S A 2000;97:13625–30.

124. Miura M, Gronthos S, Zhao M, et al. SHED: stem cells from human exfoliated de-ciduous teeth. Proc Natl Acad Sci U S A 2003;100:5807–12.

125. Saber SE. Tissue engineering in endodontics. J Oral Sci 2009;51:495–507.126. Sonoyama W, Liu Y, Yamaza T, et al. Characterization of the apical papilla and its

residing stem cells from human immature permanent teeth: a pilot study. J Endod2008;34:166–71.

127. Seo BM, Miura M, Gronthos S, et al. Investigation of multipotent postnatal stemcells from human periodontal ligament. Lancet 2004;364:149–55.

128. Lin L, Chen MY, Ricucci D, et al. Guided tissue regeneration in periapical surgery.J Endod 2010;36:618–25.

129. Fujii S, Maeda H, Tomokyo A, et al. Effects of TGF-b1 on the proliferation and dif-ferentiation of human periodontal ligament cells and a human periodontal liga-ment stem/progenitor cell line. Cell Tissue Res 2010;342:233–42.

130. Lin Z, Navarro VP, Kempeinen KM, et al. LMP1 regulates periodontal ligament pro-genitor cell proliferation and differentiation. Bone 2010;47:55–64.

131. Sant’Ana AC, Marques MM, Barroso TE, et al. Effects of TGF-beta1, PDGF-BB, andIGF-1 on the rate of proliferation and adhesion of a periodontal ligament cell line-age in vitro. J Periodontol 2007;78:2007–17.

Protective and Destructive Regulatory Pathways 163