You are on page 1of 16

Endodontic Topics 2004, 8, 88–103 Copyright r Blackwell Munksgaard

All rights reserved ENDODONTIC TOPICS 2004


1601-1538

Cell-to-cell interactions
CSONGOR KISS

The narrow anatomic periodontal space between the root surface and the alveolar bone is relatively poor in cells.
However, a broad range of noxious stimuli, most frequently root canal infection, will result in the accumulation of a
large number of inflammatory cells in the periapical region of the affected tooth/root. The encounter between
invading microorganisms and host cells as well as the interactions of resident and immigrating cells will determine
the course of the inflammatory events that may take place. This review attempts to summarize current knowledge
on how cellular interactions may contribute to dynamic equilibrium between protective, self-maintaining,
propagating, destructive and downregulating events in apical periodontitis.

Introduction Chronic apical periodontitis represents a dynamic


balance between exogenous irritative agents, usually the
Apical periodontitis is an inflammatory response of the root canal microbiota and their by-products, and host
periapical area to irritative stimuli from the root canal defense mechanisms, where the latter are not able to
space, most frequently root canal infection. Depending destroy and completely eliminate the pathogenic factors
on the intensity and duration of the initiating event, but, by forming a histological as well as functional
periapical inflammatory lesions may develop either in barrier, effectively prevent from their further invasion
an acute or a chronic direction. The histopathological (4). The chronic lesion, termed periapical granuloma, is
appearance of the lesion correlates with the clinical made up by mononuclear and polymorphonuclear
course of apical periodontitis (1). In acute apical leukocytes and fibrovascular elements. The histological
periodontitis lesions, vasodilatation, vascular conges- appearance of periapical granuloma represents every
tion, edema, extravasation of macrophages and neu- momentum of the periradicular inflammatory process
trophil leukocytes involve the periapical ligament. from the acute incipient response to the burned out
Lamina dura disruption and limited bone resorption end-stage lesion. Neighboring histological structures,
of the neighboring spongiosa can be noticed. As a i.e. necrotic, exudative, granulomatous and fib-
result of the first line of host defense, the spreading of rous zones penetrate into the surrounding alveolar
microorganisms from the infected root canal is limited. bone in an onion leaf-like fashion starting from
Occasionally, virulent root canal microbes may gain the apical foramen (Fig. 1). According to the pre-
access through the apical foramen to establish a film on dominant zone(s), lesions can be classified as exudative,
the external root surface or by forming nests within the granulomatous, granulofibrotic and fibrotic granulo-
previously healthy periapex. In response, an abundant mas (5, 6).
number of neutrophil leukocytes accumulate and the In about 50% of periapical granulomas, epithelial
incipient acute lesion develops into primary periapical strands originating from the epithelial rests of Malassez
abscess (2). Acute exacerbation of a pre-existing can be identified (7, 8). Coalescing epithelial strands
chronic lesion results in a similar histological picture, enclose a hollow space forming radicular cysts in about
leading to the development of secondary abscess. Both 6–55% of all chronic periapical lesions. The cavity of pocket
primary abscess formation and episodes of exacerba- or bay cyst communicates with the root canal. In contrast,
tion are associated with significant bone loss and to the true cyst is independent of the root canal system. Even
some apical root resorption giving space for the in cysts, the lumen is almost always embedded into the
development of a chronic lesion (3). inflammatory granulation tissue (1, 2, 9).

88
Cell-to-cell interactions

Fig. 1. Schematic presentation of periapical granuloma.


Necrotic, exudative, granulomatous and fibrotic zones of
the periapical inflammatory lesion form an onion leaf-like
structure around of the tooth apex.

Apical periodontitis – dynamic chemokines (Fig. 2A). These regulatory glycoproteins


equilibrium between protective and provide a week stimulus for attracting and activating
destructive mechanisms macrophages and polymorphonuclear leukocytes (11,
12). This low-level activity of innate immunity has been
The periapical inflammatory cell infiltrate in chronic apical suggested to play a key role in maintaining clinically
periodontitis consists of virtually every component of the healthy periodontal and periapical tissues (13, 14).
specific and non-specific branches of the immune system
(1, 2). Accumulating immune-inflammatory cells effec-
tively kill microorganisms preventing tissues surrounding Early inflammatory events in response to
the tooth apex from excessive bacterial invasion. However, root canal infection
anti-infective effector mechanisms are not restricted to In root canal infection, as soon as the bacterial front
protect the host but also destroy normal tissue components penetrates the tooth apex, cell wall or soluble compo-
resulting in bone resorption and ultimately, tooth loss (3, nents of endodontopathic bacteria may upregulate the
4, 9). The lesion has the potential to compromise general expression of first-line proinflammatory cytokines, such
health, as discussed by I. Marton elsewhere in this issue. as interleukin (IL)-1 and tumor necrosis factor (TNF)-
Although the periapical inflammation is fuelled by the a, cell adhesion molecules and chemokines inducing a
constant release of microorganisms and their by-products robust influx of inflammatory cells into the periodontal
from the root canal space, autocrine and paracrine loops of ligament (Fig. 2B). Indeed in human chronic periapical
stimulation may contribute to the promotion and main- granuloma, our group has observed differential in situ
tenance of the lesion (3). According to some investigators, distribution of IL-8, monocyte chemoattractant pro-
true cysts may be considered to be autonomous lesions tein-1 (MCP-1) and Rantes (Fig. 3). IL-8 was found
once initiated as they may not respond successfully to primarily in the cytoplasm of the Malassez epithelial
proper endodontic treatment (10, 1). cells, MCP-1 immunoreactivity was confined to en-
dothelial cells (15). Subsequently, a second wave of
inflammatory mediators, produced by the stimulated
Initiation of the apical periodontitis resident and newly recruited cells, may amplify,
lesion – interaction between resident propagate and prolong the expression of cell adhesion
host cells and root canal microbes molecules and chemokine genes (16, 17).

Contribution of innate immunity to The role of neuropeptides in eliciting


maintain a healthy periapex periapical inflammation
Epithelial and endothelial cells within the periodontal In cases of non-infectious pulp necrosis root canal and
ligament surrounding the healthy root apex constitu- periapical inflammation can be elicited by sensory
tively express low levels of cell adhesion molecules and neuropeptides that are released from nociceptive nerve

89
Kiss

endings by a variety of noxious stimuli (18, 19). mandibular molar (21). In periapical granuloma
Neuropeptides induce the production of proinflamma- samples, CGRP and substance P (SP) immunoreactivity
tory cytokines, such as TNF-a and IL-1 that propagate was confined to nerve fibers in close proximity to blood
protective and destructive inflammatory changes (20). vessels in areas associated with the collection of
In an elegant model of periodontitis, contralateral inflammatory cells, including TNF-a-containing mast
increase in calcitonin gene-related peptide (CGRP) and cells. The vascular endothelial cells stained positively for
CGRP messenger RNA (mRNA) expression accom- E-selectin and intercellular adhesion molecule-1
panied by inflammation and bone loss was demon- (ICAM-1). In contrast, clinically healthy periapical
strated in rats having received intragingival injection of ligment contained only a few macrophages and
a lipopolysaccharide (LPS) preparation at the first right lymphocytes. No immunostaining for CGRP, SP,

90
Cell-to-cell interactions

Fig. 3. Differential expression of chemokines in human periapical granuloma. (A) Expression of interleukin-8 as red
staining in the cytoplasm of the epithelial cells of Malassez. Alkaline phosphatase anti-alkaline phosphatase (APAAP)
reaction, ! 250 (15). (B) MCP-1-specific monoclonal antibody labels the surface of small blood vessels (red staining)
penetrating the granulomatous zone (APAAP reaction, ! 250) (15).

TNF-a, E-selectin and ICAM-1 was detected (22, 23). sites of bacterial invasion, mainly to the immediate
Indeed stimuli mediated by sympathetic nerve fibers vicinity of the apical foramen (1). A variety of partially
were shown to increase TNF-a production in experi- overlapping signals is involved in this process. The
mental pulp infections and periapical lesions of recruitment of leukocytes requires them to transiently
unilaterally sympathectomized rats (24). In addition attach to endothelial cells, migrate through them and
to the potential inflammatory mediator function in then to interact with cells and substrates in the
sterile lesions, neuropeptides were demonstrated to co- extravascular tissue. This cascade is governed by a set
operate with LPS from endodontopathic bacteria (25). of cell adhesion molecules, such as selectins and
integrins and the receptors for them (26). The driving
The exudative phase – robust force, the chemotactic signal can be provided by
recruitment of phagocytes regulated complement components of the alternative pathway
of complement activation and, more significantly, by
by redundant signals
chemokines (27, 28). Pleiotropic (IL-1, TNF), lineage-
The initial interaction between root canal microbes and restricted (granulocyte macrophage-colony stimulating
resident host cells may set the stage for mounting a factor (GM-CSF)) and lineage-specific (granulocyte
robust and rapid protective response reaction by colony-stimulating factor and monocyte colony-stimu-
attracting a large number of phagocytic cells, mono- lating factor) cytokines activate these infiltrating
cyte/macrophages and neutrophil leukocytes, to the leukocytes to stimulate their effector functions that

3
Fig. 2. Schematic presentation of the evolution of the periapical inflammatory response. (A) Epithelial and endothelial
cells within the periodontal ligament surrounding the healthy root apex constitutively express low levels of cell adhesion
molecules and chemokines providing a week stimulus for attracting and activating macrophages and polymorphonuclear
leukocytes. Low-level activity of innate immunity plays a key role in maintaining clinically healthy periodontal and
periapical tissues. (B) Root canal infection results in pulp necrosis. Endodontopathic bacteria can be found within the
root canal, at the apical foramen and within the lesion per se. Bacteria and their by-products upregulate the expression of
cell adhesion molecules, chemokines and proinflammatory cytokines attracting an abundant number of neutrophil
leukocytes and monocyte-macrophages that represent the first line of defense. (C) Antigen-presenting cells (dendritic
cells and macrophages) present bacterial antigens to CD41 T-helper 1 (Th1) lymphocytes via the antigen-specific T-cell
receptor in conjunction with major histocompatibility complex Class II molecules. Primed Th1 cells produce interferon-
c and interleukin-2. The former cytokine stimulates monocyte-macrophage cells establishing a positive loop of
stimulation. The latter cytokine promotes the proliferation of further antigen-specific Th1 cells and other activated
lymphocytes resulting in granulation tissue formation. (D) In established periapical granulomas Th2 cells outnumber
Th1 cells resulting in the stimulation of B-lymphocytes and plasma cells producing immunoglobulins and in a change of
the CD4/CD8 ratio in favor of the latter cells. The cytokine milieu provided by Th2 cells and CD81 lymphocytes can
stimulate the proliferation of epithelial cells and may contribute to the healing of the lesion.

91
Kiss

contribute to both protective and self-destructive The role of chemokines


events.
The complex process of chemotaxis, i.e. the directed
migration of phagocytes from within the capillary bed,
through the endothelial cells, across the interstitium,
between epithelial cells, into the site of infection may
Induction of the exudative phase in require sequential interactions with different chemo-
transgenic mice kines expressed by different local cells or attached to
matrix components (15, 34, 35). Chemokines of the a
Much of our understanding stems from the investiga-
(CXC) family have been shown essential to leukocyte
tion of transgenic mice. In these animals, the gene for
migration induced by bacteria or LPS (36, 37). In vitro
either the ligand or the receptor participating in
incubation of neutrophil leukocytes with TNF-a and
leukocyte recruitment and function has been disrupted
GM-CSF was demonstrated to upregulate receptors for
rendering the animals deficient in functions regulated
and responsiveness to CC chemokines, members of the
by the given ligand–receptor interaction. Probably as a
chemokine b family (38, 39).
result of the overlap in signals that attract leukocytes to
sites of infection, it has been proven that almost all
ligand–receptor pairs involved in the process can be Role of nuclear transcription factors
bypassed by proper functioning of unperturbed signal
pathways (16). For example, the dental pulp of mice The overlapping stimulatory effects exerted by ligand–
with functional deletions of receptors to IL-1, TNF, or receptor pairs involved in leukocyte migration and
both was inoculated with anaerobic pathogens. Poly- chemotaxis may converge to the nuclear transcription
morphonuclear and mononuclear phagocyte recruit- factor NF-kB regulating the expression of the genes for
ment occurred to the greatest extent in double negative many cell adhesion molecules and chemokines and
mice, and to a lesser extent in IL-1 receptor (IL-1R) or their receptors (40). Investigation of transgenic mice,
TNF receptor (TNFR) mice, and the least in wild-type deficient in TNF and in RelA, a 65 kDa subunit of NF-
mice (29). This study confirmed that despite being kB suggested that RelA may promote the coordinated
involved in the upregulation of cell adhesion molecules expression of adhesion molecules and chemokines
and chemokines influencing the migration of mono- essential to leukocyte infiltration in response to
cytes and neutrophil leukocytes, IL-1/IL-1R- as well as Gram-negative bacteria and LPS (41).
TNF/TNFR-independent pathways for phagocyte
recruitment can be induced by endodontopathic
Infiltrating phagocytes represent the
bacteria (30, 31).
dominant cell population of the exudative
phase
Once reaching at the infected periapical space, the role
of leukocytes is clearly protective. The infiltrating
Role of integrins and their ligands
nature of these cells has been firmly established.
ICAM-1, a ligand for the heterodimer integrin Neutrophil leukocytes are virtually absent in the
molecule CD11/CD18 seems to be essential for healthy periodontal ligament therefore, they must be
neutrophil recruitment induced by Gram-negative attracted from the bone marrow via the circulating
bacterial stimuli (32). Both CD11/CD18-dependent blood in case of root canal and concomitant periapical
and -independent pathways for neutrophil emigration infection. Most neutrophil leukocytes have been shown
have been identified suggesting that Gram-negative metabolically inactive or dead in human periapical
bacteria induce predominantly CD11/CD18-depen- lesions indicating their short-lived effector cell nature
dent emigration, whereas Gram-positive bacteria in- (42). Newly recruited macrophages clearly outnumber
duce predominantly CD11/CD18-independent resident macrophages as demonstrated by immunohis-
emigration (16). Additional b1 integrins, very late tochemistry, in situ hybridization and ultrastructural
antigen (VLA)-5 and VLA-6 were suggested to analysis. Actively phagocytozing macrophages of in-
contribute to LPS-induced neutrophil migration (33). filtrating phenotype were found in great numbers just

92
Cell-to-cell interactions

beneath the periapical abscess in experimentally in- Table 1. Host regulatory factors induced by endo-
duced rattine periapical lesion (43). The presence of dontopathic microorganisms and their by-products
large, cytophagocytic macrophages containing apopto-
tic neutrophil leukocytes characterized human periapi- Cell adhesion molecules and their ligands
cal lesions. Although macrophages present in the ICAM-1
human lesion actively transcribe and translate genes as
CD11/18
indicated by detecting polyadenosine RNA and ribo-
somal RNA, they exhibit only a very low level of LFA-1
proliferative activity (42). VLA-5, VLA-6

CD29, CD49
Protective and destructive functions of Chemokines and their receptors
infiltrating phagocytes
IL-8
Inhibiting leukocyte recruitment or activation either by
KC
disrupting regulatory signals mediated by IL-1, TNF,
IL-6 and their receptors or by depressing bone marrow Macrophage inflammatory protein-2

function have been shown to result in a more wide- MCP-1


spread local bacterial infiltration, even in generalized Rantes
infections (17, 29, 44). Although local tissue damage
has been connected with the release of reactive oxygen Proinflammatory cytokines, their receptors and antago-
nists
and nitrogen radicals and matrix metalloproteases
during phagocytosis, interestingly however, osteolytic GM-CSF

periapical lesions were larger in IL-1R-, TNFR- and in IL-1


IL-6-deficient transgenic mice (29, 44–46). In turn,
IL-1R
pharmacological enhancement of the number and
activity of circulating neutrophils and monocytes IL-1Ra

resulted in the reduction of periapical bone and soft IL-6


tissue lost in mice (47). These observations suggest that IFN-c
the more effective is the local control of infection, the
TNF
less tissue damage occur during periapical lesion
initiation. Reactive oxygen and nitrogen intermediates and arachi-
donic acid metabolites

Superoxide radical
Regulatory molecules derived from Hydrogen peroxide
endodontopathic bacteria participate
Leucotrienes
in the initiation and progression of
Prostaglandins
the periapical inflammatory process
Nuclear transcription factors
Bacteria do not need necessarily emigrate from the
infected root canal. Among their soluble products a NF-jB
number of compounds has been defined that may elicit IE2-protein
and sustain a vigorous inflammatory response in the
ICAM-1, intercellular adhesion molecule 1; LFA,
periapical region. In addition to attract phagocytes, lymphocyte function-associated antigen; VLA, very late
representing the first line of defense, directly or by antigen; IL, interleukin; KC, member of the chemokine
inducing the expression of proinflammatory cytokines family; MCP, monocyte chemoattractant protein; Rantes,
and cell adhesion molecules by resident host cells, member of the chemokine family; GM-CSF, granulocyte
macrophage-colony stimulating factor; IFN, interferon;
bacterial molecules can stimulate the specific immune TNF, tumor necrosis factor; IE, immediate early.
responses (Table 1). The appearance of lymphocytes
and plasma cells will transform the early periapical

93
Kiss

lesion into periapical granuloma. LPSs represent Table 2. Factors influencing the biological activities
probably the most potent, and best-studied bacterial of lipopolysaccharides (LPS)
factors. In addition, some more recently characterized
microbial molecules may contribute to the pathome- Availability of cell surface or soluble CD14 antigen and
chanism of apical periodontitis. LPS-binding protein

Target tissue specificity

The role of LPSs in periapical inflammation Constitutively determined hyper/hyporesponsive pheno-


type
LPS present in and released from cell walls of Gram-
negative bacteria are probably the best-characterized The source of LPS
components of endodontopathic bacteria to induce
Induction of agonistic and antagonistic cytokines and cell
cytokines and other inflammatory compounds. Most adhesion molecules
studies have used peripheral blood monocytes and
fibroblasts as target cells. LPSs isolated from Actino-
bacillus actinomycetemcomitans, Fusobacterium nucle-
atum, Porphyromonas gingivalis and Prevotella greater amounts of IL-1b and TNF-a than those
intermedia, for example, have been shown to induce obtained from normal individuals (53, 54). In murine
the production of an array of proinflammatory and monocytes, an impaired ability to respond to LPS has
chemotactic cytokines including GM-CSF, interferon been attributed to a mutation in the gene that encodes
(IFN)-g, IL-1a and -b, IL-6, IL-8, MCP-1 and TNF-a Toll-like receptor 4 (55, 56). In humans, polymorph-
(48). Local IL-1, IL-6 and GM-CSF production was isms, most often single nucleotide polymorphisms of
repeatedly detected in human dental pulp and periapi- genes encoding for cytokines and enzymes influencing
cal lesions (49–52). Expression of IL-6 mRNA was the function of immune cells have been shown to alter
confirmed in immortalized oral keratinocytes upon the intensity of immune reactions and may be
challenge with LPS from endodontopathic bacteria by responsible for the hypo/hyperresponsive pattern of
RT-PCR (14). The broad concentration range of monocytes and macrophages (57–59).
activity and the colorful biological spectrum of LPS
depend on a number of factors (Table 2).
The source of LPS
LPS derived from A. actinomycetemcomitans, Actino-
The availability of CD14 antigen bacillus israeli and F. nucleatum were shown to
The availability of CD14 antigen and LPS-binding upregulate ICAM-1, IL-8 and MHC Class II antigens
protein are absolute requirements of LPS–receptor in oral epithelial cell lines, whereas P. gingivalis LPS
binding and signal translation (48). CD14 can be downregulated the former two molecules. In contrast
present either on the surface of the targeted cell, or as a to the strong and moderate stimulation by F. nucle-
soluble compound, which in a complex with LPS and atum and A. israeli, respectively, LPS from Actinoba-
LPS-binding protein stimulates the target cell. cillus viscosus did not increase constitutive expression of
MHC Class II molecules. The inability of certain
endodontopathic bacteria to induce MHC Class II
Target tissue specificity expression or their potential to inhibit IL-8 accumula-
LPS preparations extracted from the same endodonto- tion may result in a decreased level of inflammatory
pathic bacteria were shown to induce cytokine secre- cellular infiltrate thereby contributing to bacterial
tion in oral but not in skin-derived epithelial cells (14). invasion (13, 14, 60).

Constitutive differences Induction of agonistic and antagonistic


cytokines and cell adhesion molecules
Recently, a hyperinflammatory phenotype has been
defined in humans. Peripheral blood monocytes, F. nucleatum-derived LPS were shown to induce MHC
derived from such subjects, produce 3- to 10-fold Class II, ICAM-1, IL-1 and IL-6 molecules and

94
Cell-to-cell interactions

suppressed the manifestation of co-stimulatory mole- sponding LPS preparations. SAM prepared from A.
cules CD70 and CD80/86 in oral epithelial cells (14). actinomycetemcomitans, Eikenella corrodens and P.
In the lack of a signal via CD80/86 molecules, T-cell gingivalis was shown to directly stimulate IL-6 synth-
activation will be impaired despite proper antigen esis of human gingival fibroblasts without affecting
presentation through MHC Class II antigens and transcription of IL-1 and TNF genes that induce the
despite ICAM-1 binding to its receptor, lymphocyte expression of IL-6 by positive loops of stimulation
function-associated antigen-1 (61). Moreover, LPS upon challenge with LPS (67).
may induce simultaneous expression of IL-1 and its
antagonist IL-1Ra (48). Thus, LPS-induced differen-
tial expression of key regulatory molecules in resident Heat shock proteins
root canal and periradicular cells may result either in a A 64 kDa protein component of A. actinomycetemco-
powerful initiation of the inflammatory lesion or in a mitans SAM with potent osteolytic activity has been
state of unresponsiveness contributing to the charac- identified as a homologue of the 60 kDa Escherichia coli
teristic long-term quiescent periods observed in many GroEL protein, member of the bacterial heat shock
chronic periapical lesions. protein (Hsp) family (68). Hsp64 exerted a powerful
proliferative stimulus on porcine periodontal ligament
The role of other bacterial compounds in epithelial cells in low concentrations but proved
periapical inflammation cytotoxic for immortalized skin epithelial cells in large
concentrations (69). Bacterial Hsp’s, frequently pre-
There are a number of further cell components of sented as immunodominant antigens in mammals, have
endodontopathic bacteria which are able to initiate a significant homology with human Hsp’s and cross-
inflammation by inducing inflammatory cytokines react in vitro with anti-human Hsp antibodies (70, 71).
(48). Among these, a few compounds deserve parti- Antibodies to Hsp’s of endodontopathic bacteria may
cular interest. cross-react with human Hsp’s exposed in an injured
root canal or periapical tissue, promoting the initial
Fimbrial protein extracts inflammatory response (72).

Fimbrial protein extracts from P. gingivalis were


demonstrated to increase IL-1a and IL-1b mRNS An emerging role of viruses in the
expression even in macrophages from an ‘LPS-unre-
periapical inflammatory process
sponsive’ mouse strain (62). In addition to IL-1a and
IL-1b, the fimbrial protein extract of this microorgan- A growing attention has been focused recently on two
ism was demonstrated to stimulate the production of members of human herpesviruses, Cytomegalovirus
further proinflammatory cytokines, TNF-a, IL-6 and (CMV) and Ebstein–Barr virus (EBV) playing an
GM-CSF, with bone-resorptive activities and two etiopathogenic role in the development of root canal
chemokines, IL-8 and KC (63, 64). The latter two and periapical lesions. The research group at the
chemotactic cytokines play a role in driving neutrophil University of Southern California has repeatedly
emigration from blood and in promoting endothelial confirmed the association between CMV and EBV
and epithelial cell proliferation (28, 65, 66). A 12-mer infection and symptomatic periapical pathosis (73, 74).
amino acid sequence between residues 69 and 80 CMV and EBV infections induce an array of inflam-
connected with the biological activities of the native matory and chemotactic cytokines that stimulate the
fimbrial protein has been identified, opening a pathway expression of further activation and cell adhesion
for developing targeted therapy (63). molecules, recruit inflammatory effector cells to the
site of infection and contribute to bone resorption.
Increase in the expression of IL-1b gene is particularly
Surface-associated material
abundant as IE2 protein, one of the immediate early
The biological activity of easily solubilized components (IE) CMV gene products, has been shown to provide
of certain Gram-negative bacteria, termed surface- robust transactivation of the IL-1b gene without
associated material (SAM), may exceed that of corre- requiring the involvement of the far-upstream enhancer

95
Kiss

of the gene (75). These effects may become especially and secondary immune responses, respectively and
powerful in synergy with LPS (76). MHC Class II-positive macrophages eliciting a sec-
ondary immune response (43). A similar distribution
and morphology of macrophages labeled with anti-
Granulation tissue formation – fine- CD14, anti-CD68 and anti-human leukocyte antigen-
tuned cellular interactions establish a DR monoclonal antibodies was observed in human
balance between protective, periapical lesions (Fig. 4) (80, 81). Characterizing the
destructive and reparative reactions isoforms of leukocyte common antigen CD45, naive
(CD45RA and CD45RC) and memory (CD45RO and
If the invading microorganisms cannot be completely CD45RB) T-lymphocytes were found in almost equal
eliminated in course of the anti-infective reactions of numbers within the granulation tissue confirming the
the exudative phase and the irritative effects of soluble possibility, extrapolating from the results of the rattine
bacterial by-products and viruses persist, the lesion model, that both primary and secondary T-cell
becomes chronic and a characteristic, complex granula- responses can be initiated in human periapical lesions
tion tissue will be established. Granulomatous inflam- (82).
mation is a protective host response built up to limit
periapical tissue damage by localizing root canal
T-helper cell subpopulations
microbiota and the inflammatory response. The initia-
tion and formation of granulomas is largely dependent According to their cytokine expression pattern, CD41
on the interaction of antigen-presenting cells with T-cells can be classified as T-helper 1 (Th1) cells that
CD41 T-lymphocytes (77, 78). Antigen-presenting produce and secrete IL-2 and IFN-g and Th2 cells that
cells activate CD41 T-lymphocytes through their produce and secrete IL-4, IL-5, IL-6, IL-10 and IL-13.
antigen-specific receptors (TCR) in conjunction with Positive loops of stimulations exist between macro-
MHC Class II molecules (79). phages and Th1 cells as well as within the Th1 and Th2
subpopulations, respectively. In contrast, Th1 and Th2
cells exert a mutually inhibitory effect on each other.
Interaction of antigen-presenting cells and Th1-type cytokines augment cytotoxic T-cell func-
T-lymphocytes tions, stimulate the expression of proinflammatory and
The research group from the Tokyo Medical and bone resorptive molecules in other cell types, whereas
Dental University has performed a detailed study of Th2-type cytokines participate in B-lymphocyte stimu-
antigen-presenting cells in experimentally induced lation to mount a humoral immune response and in
periapical lesions in the rat (43). Early lesions,
established after 28 days of unsealed pulp exposure
were studied. In contrast to MHC Class II-negative,
actively phagocytozing macrophages accumulating in
the vicinity of periapical abscess, MHC Class II-positive
macrophages were found evenly interspersed among
other cell types within the cell-rich granulomatous
zone. The various appearances of these permanently
immigrating cells may mirror the heterogeneity the
stage of differentiation at the time of observation.
MHC Class II-positive dendritic cells (DCs) were also
noticed in great numbers, mostly located in the outer,
fibrotic border of the lesion. Both MHC Class II-
positive macrophages and DC established cell-to-cell
contacts with MHC Class II-negative lymphocytes. Fig. 4. Distribution of CD141 macrophages in the
granulomatous zone of periapical granuloma. The
These results indicate that local antigen presentation
antigen–antibody reaction was visualized by the avidin–
takes place in periapical lesions with DC being able to biotin–peroxidase complex method resulting in a brown
activate both naive and memory T-cells during primary staining of CD141 macrophages, ! 250 (80).

96
Cell-to-cell interactions

dampening the inflammatory reactions (83). As ex-


pected, Th1-lymphocytes prevail in early, expanding
periapical lesions (84, 85). As a result of positive loops
of stimulation, the number of Th1 cells increases
rapidly within the lesion (Fig. 2C).

The origin of T-lymphocytes in


granulomatous inflammatory lesions
The origin of these cells has been characterized
precisely in the schistosoma-induced granuloma mod-
Fig. 5. Distribution of major histocompatibility complex
el. In this model, granulomatous inflammatory lesions
Class II-expressing cells in the granulomatous zone of
in the livers of mice were induced by the parasite periapical granuloma. Small, lymphocyte-like and larger
Schistosoma mansoni. The very diverse TCR repertoire mononuclear cells resembling macrophages display a
of the individual granulomas in the liver of the same circumferential brown staining using the avidin–biotin–
peroxidase complex (ABC) method, ! 250 (80).
infestated mouse indicate that most of the T-cells are
recruited to the lesions and are activated systemically.
At the same time, nucleotide sequence analyses of
Humoral immune response in human
individually sized products from the complementarity-
periapical granuloma lesions
determining region 3 (CDR3) of the TCR gene from In more advanced lesions, Th2 cells slowly outnumber
single granuloma indicate that a fraction of T-cells Th1 lymphocytes. Recently, bacterial Hsp’s were
expand locally at the lesion site. Furthermore, it has shown to contribute to switching the Th1 response
been shown, that non-schistosoma-specific T-cells towards Th2 (89). The resulting change in the cytokine
home to the granuloma if they are activated, resulting milieu is accompanied by a steady increase in the
in a change of the original Th1 vs. Th2 and CD41 vs. number of B-lymphocytes and plasma cells present in
CD81 balance (86). The low level of S-phase prolifer- the lesion (Fig. 6) (51, 90). The functional significance
ating and cycling lymphocytes, assessed by in situ of local humoral immunity in periapical lesions is
hybridization using a histone probe and by Ki-67 underscored by the observation that both T/B-null
immunostaining in contrast to their transcriptional and RAG-2 SCID and B-cell-deficient mice, but not T-cell-
translational activity support the view that a similar or C5-deficient mice, have increased susceptibility to
scenery may be operative in the human periapical lesion
(42).

T-lymphocyte activation markers in human


periapical granuloma lesions
Similar to the schistosoma-induced granulomas, T-
lymphocytes express activation markers, such as IL-2
receptor a-chain (CD25) and MHC Class II molecules
in expanding periapical lesions, implying their active
participation in anti-infective and proinflammatory
reactions (Fig. 5) (80, 87). This hypothesis was
confirmed recently by demonstrating that cyclosporine Fig. 6. Distribution of B-lymphocytes and plasma cells in
A treatment resulted in a dose-dependent decrease in the granulomatous zone of periapical granuloma.
lesion size accompanied by a significant reduction in Numerous plasma cells and B-lymphocytes reacting with
the mixed heavy chain (immunoglobulin (Ig)A1IgG1
the number of CD251 cells in rat molars exposed to the
IgM) anti-serum in the granulomatous zone. Reactive
oral environment when compared with non-treated cells appear in brown with the peroxidase anti-peroxidase
controls (88). method. ! 160 (114).

97
Kiss

the development of disseminating anaerobic infections lial rests, investigators at the University of Manitoba
following experimental root canal infection with a proposed a model, in which LPS and proinflammatory
mixture of anaerobic pathogens (91). cytokines stimulated proliferation and differentiation
of the epithelial rests of Malassez by means of MAP
kinase activation (97).
Changing Th/suppressor ratio
In contrast to early, expanding periapical granulomas,
CD81 cytotoxic/suppressor T-lymphocytes usually out- Mechanism of bone resorption in
number CD41 helper/inducer T-cells in advanced periapical lesions
lesions contributing either to a more effective elimination
of infected and destructed cells or to the suppression of The most prominent destructive event connected with
the intensity of inflammatory reactions, or both (3, 78). both periapical granuloma formation and cyst develop-
ment is the resorption of alveolar bone, although the rate
of bone loss is not as high as in the exudative phase(s),
Possible mechanism of cyst i.e. during initiation and exacerbation(s) of apical
periodontitis lesions. The effector cells of this degen-
development
erative process are osteoclasts. However, osteoblasts
The CD41 Th2/CD81-rich nature is even more regulate the dynamic equilibrium of bone remodeling.
obvious in periradicular cysts that were demonstrated The signaling is mediated by the interaction of a recently
to develop late during the course of experimentally discovered cell membrane-bound ligand–receptor pair,
induced periapical inflammation (92, 93). The factors members of the TNFR and ligand families (98).
that initiate and promote the proliferation of the Osteoblasts express receptor activator of NF-kB ligand
epithelial rests of Malassez to form either pocket or (RANKL) that binds to its receptor, RANK expressed on
true radicular cysts, are not well understood. Antigen- the surface of osteoclast precursor cells. As a result of
presenting cells and CD81 cytotoxic/suppressor T- RANKL–RANK binding, osteoclast precursors differ-
lymphocytes have been shown repeatedly to be entiate into mature osteoclasts, mature osteoclasts
associated with pre-existing and newly formed epithe- become activated resulting in bone resorption. Osteo-
lium (81, 94). It has been suggested that the local blasts also express and secrete the soluble form of
cytokine milieu provided by the neighboring macro- RANKL, osteoprotegerin (OPG) which, by acting as a
phages and CD81 cells may contribute to epithelial decoy receptor, inhibits RANKL–RANK interaction and
proliferation. Epithelial cells, indeed, have only been thus bone resorption. Reciprocal gene expression of
shown to be in the phase of active proliferation in RANKL and OPG has been shown to play a key role in
human periapical lesions (42). Another group of the coupling of osteoblast–osteoclast interaction (99).
investigators observed that intensive immunostaining Humoral regulatory molecules of bone resorption and
for Hsp27 was in coincidence with the presence of local formation, such as hormones and cytokines exert their
infiltration of immune cells suggesting that Hsp’s may effects largely by influencing RANKL–RANK interac-
contribute to epithelial proliferation (95). In contrast, tion directly or by changing the ratio of RANKL/OPG
CD57 antigen-positive cells may play a role in reciprocal gene expression (100). Prostaglandins appear
degeneration of the epithelial lining of late radicular to act even more indirectly as permissive cofactors of
cysts as they have been shown to be present in atrophic bone-resorptive cytokines (101, 102).
radicular cysts in a significantly greater cell density than Results from the extensively studied rat periapical
in radicular cysts with hyperplastic epithelium (96). granuloma model indicate that IL-1a is involved in the
More recently, a role for mitogen-activated protein bone destruction induced by bacterial root canal
(MAP) kinases has been suggested in promoting infection (103). In human periapical lesions, IL-1b is
epithelial proliferation and differentiation. Based on the predominant bone-resorptive cytokine (104).
the finding that two ubiquitous MAP kinases, ERK1 Bone-resorbing activity of radicular and periapical
and ERK2 were overexpressed in differentiated and CMV infection has also been connected with the ability
highly proliferative epithelial cells, as compared with of the virus to stimulate IL-1 release (75, 76). Other
those containing non-differentiated, quiescent epithe- cytokines that have been implicated in alveolar bone

98
Cell-to-cell interactions

loss are TNFa and IL-6, IL-18 (76, 105). As ‘bone- pathic bacteria, that, in interaction with resident host
resorptive’ cytokines may alter the balance in favor of cells, recruit large number of phagocytes, representing
bone formation by their ability to stimulate osteoblasts the first line of host defense. Subsequent appearance of
and their precursors on the one hand, and taking the lymphocytes and plasma cells will transform the early
overlapping nature of the effects induced by them, it is periapical lesion into periapical granuloma. The char-
probably not surprising that disrupting the function of acteristic lesion would not develop in the absence of
one of these cytokines did not result in a reduced bone permanent release of bacteria and their by-products from
loss in IL-1R-, TNFR- and in IL-6-deficient transgenic the infected root canal. On the other hand, the formation
mice exposed to root canal infection (29, 44). of a granulation tissue, walled off by a fibrotic capsule
Bacterial enzymes, LPS and other cell-wall compo- towards the neighboring healthy tissues is entirely
nents may be involved directly in bone degradation as dependent on the presence and proper function of host
demonstrated under experimental conditions (106– cells and regulatory molecules. Inhibiting leukocyte
108). However, endodontopathic bacteria only rarely recruitment or activation either by disrupting regulatory
establish themselves outside of the external root canal signals mediated by host-derived cell-bound or soluble
surface in granulomatous periapical lesions (109). regulatory compounds or by depleting the sources of the
Moreover, the concentration of LPS to directly elicit infiltrating cells result in a widespread local bacterial
bone resorption is in the range of 10 " 3 g/L, whereas as infiltration, even in generalized infections. Thus, a
tiny concentration of LPS as 10 " 9 g/L was demon- complex, fine-tuned interaction between microorgan-
strated to induce the release of endogenous bone isms, resident host cells and infiltrating leukocytes,
resorptive cytokines and prostaglandins (110, 111). recruited by them will determine the dynamic equili-
That is, the probability for an indirect involvement of brium between protective, destructive and reparative
endodontopathic microorganisms in the process of events in apical periodontitis. Much of our under-
alveolar bone loss in established periapical lesions is 106 standing of this process comes from experimental models
times as much as their direct role. and represents extrapolations based on investigations of
similar responses occurring in different tissues and body
parts of humans and other mammalian species. The
Healing of the periapical lesions elucidation of the exact role of cell-to-cell interactions in
human apical periodontitis requires further research
Having eliminated successfully the initiating root canal
utilizing recent advances of molecular biologic methods.
infection by proper endodontic and restorative treat-
ment, repairing mechanisms exerted by the granulation
tissue may prevail over tissue destructive events. With Acknowledgements
the exception of a not exactly characterized portion of
true radicular cysts, which may take an autonomous Microphotographs were taken in collaboration with Drs
character, the majority of periapical lesions improve Ildikó J. Márton, Balazs Dezsö, Zoltan Nemes and Antal Rot.
significantly over an extended period of time suggesting The help of Dr Edit Bárdi in preparing computer graphics is
that even radicular cysts in general are treatable by kindly acknowledged. This work was supported by grants of
conventional endodontics (112, 113). The lesion may the National Research Development Fund (OTKA) Nos.
heal completely by remineralization as indicated by the T038307 and T046588, and of the Health Science Council
disappearance of the former radiolucent area. Finally, the (ETT) of the Hungarian Ministry of Health No. 225/2003.
lesion may develop into a fibrotic periapical granuloma,
or periapical scar containing only a few cells, mostly
fibroblasts and lacking an inflammatory character. References
1. Beer R, Baumann MA, Kim S. Endodontology. In:
Rateitschak KH, Wolf HF, eds. Color Atlas of Dental
Concluding remarks Medicine. Stuttgart: Thieme, 2000: 26–34.
2. Happonen RP, Bergenholtz G. Apical periodontitis. In:
Constant irritation from the root canal space will result in
Bergenholtz G, H!rsted-Bindslev P, Reit C, eds.
an inflammatory response within the periapical area. Textbook of Endodontology. Oxford: Blackwell, 2003:
Noxious stimuli are most often provided by endodonto- 130–144.

99
Kiss

3. Márton IJ, Kiss C. Protective and destructive immune 18. Bjurholm A, Kreicbergs A, Brodin E, Schultzberg M.
reactions in apical periodontitis. Oral Microbiol Im- Substance P- and CGRP-immunoreactive nerves in
munol 2000: 15: 139–150. bone. Peptides 1988: 9: 165–171.
4. Nair PNR. Apical periodontitis: a dynamic encounter 19. Hargreaves KM, Swift JQ, Roszkoeski MT, Bowles W,
between root canal infection and host response. Perio- Garry MG, Jackson DL. Pharmacology of peripheral
dontology 2000 1997: 13: 121–148. neuropeptide and inflammatory mediator release. Oral
5. Morse DR. Immunologic aspects of pulpal–periapical Surg Oral Med Oral Pathol Oral Radiol Endod 1994:
diseases. A review. Oral Surg Oral Med Oral Pathol Oral 78: 503–510.
Radiol Endod 1977: 43: 436–451. 20. Lotz M, Vaughan JH, Carson DA. Effect of neuropep-
6. Yanagisawa S. Pathologic study of periapical lesions. I. tides on production of inflammatory cytokines by
Periapical granulomas: clinical, histopathologic and human monocytes. Science 1988: 241: 1218–1221.
immunohistopathologic studies. J Oral Pathol 1980: 21. Dumitrescu AL, Abd El-Aleem S, Morales-Aza B,
9: 288–300. Donaldson LF. A model of periodontitis in the rat:
7. Nair PNR, Pajarola G, Schroeder HE. Types and effect of lipopolysaccharide on bone resorption, osteo-
incidence of human periapical lesions obtained with clast activity, and local peptidergic innervation. J Clin
extracted teeth. Oral Surg Oral Med Oral Pathol Oral Periodontol 2004: 31: 596–603.
Radiol Endod 1996: 81: 93–102. 22. Kabashima H, Nagata K, Maeda K, Iijima T. Involve-
8. Seltzer S, Soltanoff W, Bender IB. Epithelial prolifera- ment of substance P, mast cells, TNF-a and ICAM-1 in
tion in periapical lesions. Oral Surg Oral Med Oral the infiltratory cells in human periapical granulomas.
Pathol Oral Radiol Endod 1969: 27: 110–121. J Oral Pathol Med 2002: 31: 175–180.
9. Rotstein I, Simon JHS. Diagnosis, prognosis and 23. Byers MR, Taylor PE, Khayat BG, Kimberly CL. Effects
decision-making in the treatment of combined perio- of injury and inflammation on pulpal and periapical
dontal–andodontic lesions. Periodontol 2000 2004: 34: nerves. J Endod 1990: 16: 78–84.
165–203. 24. Bletsa A, Heyeraas KJ, Haug SR, Berggreen E. IL-1
10. Nair PNR. New perspectives on radicular cysts: do they alpha and TNF-alpha expression in rat periapical lesions
heal? Int Endod J 1998: 31: 155–160. and dental pulp after unilateral sympathectomy. Neu-
11. Huang GT, Kim D, Lee JK, Kuramitsu HK, Haake SK. roimmunomodulation 2004: 11: 376–384.
Interleukin-8 and intercellular adhesion molecule 1 25. Azuma H, Kido J, Ikedo D, Kataoka M, Nagata T.
regulation in oral epithelial cells by selected periodontal Substance P enhances the inhibition of osteoblastic cell
bacteria: multiple effects of Porphyromonas gingivalis differentiation induced by lipopolysaccharide from
via antagonistic mechansims. Infect Immunol 2001: 69: Porphyromonas gingivalis. J Periodontol 2004: 75:
1364–1372. 974–981.
12. Liu F, Abiko Y, Nishimura M, Kusano K, Shi S, Kaku T. 26. Carlos TM, Harlan JM. Leukocyte–endothelial adhe-
Expression of inflammatory cytokines and beta- sion molecules. Blood 1994: 84: 2068–2101.
defensin 1 mRNAs in porcine epithelial rests of 27. Muller-Eberhard HJ. Complement. Annu Rev Biochem
Malassez in vitro. Med Electron Microsc 2001: 34: 1975: 44: 697–724.
174–178. 28. Luster AD. Chemokines – chemotactic cytokines that
13. Darveau RP, Belton CM, Reife RA, Lamont RJ. Local mediate inflammation. N Engl J Med 1998: 338: 436–
chemokine paralysis, a novel pathogenic mechanism for 445.
Porphyromonas gingivalis. Infect Immun 1998: 66: 29. Chen CP, Hertzberg M, Jiang YL, Graves DT.
1660–1665. Interleukin-1 and tumor necrosis factor receptor
14. Han DC, Huang GTJ, Lin LM, Warner NA, Gim JS, signaling is not required for bacteria-induced osteo-
Jewett A. Expression of MHC class II, CD70, CD80, clastogenesis and bone loss but is essential for protect-
CD68 and pro-inflammatory cytokines is differentially ing the host from a mixed anaerobic infection. Am J
regulated in oral epithelial cells following bacterial Pathol 1999: 155: 2145–2152.
challenge. Oral Microbiol Immunol 2003: 18: 350– 30. Bevilaqua MP, Nelson RM, Mannor G, Cecconi O.
358. Endothelial–leukocyte adhesion molecules in human
15. Marton IJ, Rot A, Schwarzinger E, Szakáll S, Radics T, disease. Annu Rev Med 1994: 45: 361–378.
Vályi-Nagy I, Kiss C. Differential in situ distribution of 31. Cutolo M, Sulli A, Barone A, Seriolo B, Accardo S.
interleukin-8, monocyte chemoattractant protein-1 Macrophages, synovial tissue and rheumatoid arthritis.
and Rantes in human chronic periapical granuloma. Clin Exp Rheumatol 1993: 11: 331–339.
Oral Microbiol Immunol 2000: 15: 63–65. 32. Qin L, Quinlan WM, Doyle NA, Graham L, Sligh JE,
16. Mizgerd JP. Molecular mechanisms of neutrophil Takei F, Beaudet AL, Doerschuk CM. The roles of
recruitment elicited by bacteria in the lungs. Semin CD11/CD18 and ICAM-1 in acute Pseudomonas
Immunol 2002: 14: 123–132. aeruginosa-induced pneumonia in mice. J Immunol
17. Puliti M, von Hunolstein C, Bistoni F, Castronari R, 1996: 157: 5016–5021.
Orefici G, Tissi L. Role of macrophages in experimental 33. Ridger VC, Wagner BE, Wallace WAH, Hellewell PG.
group B streptococcal arthritis. Cell Microbiol 2002: 4: Differential effects of CD18, CD29, and CD49
691–699. integrin subunit inhibition on neutrophil migration in

100
Cell-to-cell interactions

pulmonary inflammation. J Immunol 2001: 166: ponse modifier PGG glucan. J Dent Res 1995: 74:
3484–3490. 323–330.
34. Middleton J, Neil S, Wintle J, Clark-Lewis I, Moore H, 48. Wilson M, Reddi K, Henderson B. Cytokine-inducing
Lam C, Auer M, Hub E, Rot A. Transcytosis and components of periodontopathogenic bacteria. J Per-
surface presentation of IL-8 by venular endothelial iodontal Res 1996: 31: 393–407.
cells. Cell 1997: 91: 385–395. 49. Lim GC, Torabinejad M, Kettering J, Linkhardt TA,
35. Kuschert GS, Coulin F, Power CA, Proudfoot AE, Finkelman RD. Interleukin 1-beta in symptomatic and
Hubbard RE, Hoogewerf AJ, Wells TN. Glycosami- asymptomatic human periradicular lesions. J Endod
noglycans interact selectively with chemokines and 1994: 20: 225–227.
modulate receptor binding and cellular responses. 50. Barkhordar RA, Hayashi C, Hussain MZ. Detection of
Biochemistry 1999: 38: 12959–12968. interleukin-6 in human dental pulp and periapical
36. Frevert CW, Huang S, Danaee H, Paulauskis JD, lesions. Endod Dent Traumatol 1999: 15: 26–27.
Kobzik I. Functional characterization of the rat 51. Walker KF, Lappin DF, Takahashi K, Hope J, Macdo-
chemokine KC and its importance in neutrophil nald DG, Kinane DF. Cytokine expression in periapical
recruitment in a rat model of pulmonary inflammation. granulation tissue as assessed by immunohistochemis-
J Immunol 1995: 154: 335–344. try. Eur J Oral Sci 2000: 108: 195–201.
37. Schmal H, Shanley TP, Jones ML, Friedl HP, Ward PA. 52. Radics T, Kiss C, Tar I, Márton IJ. Interleukin-6 and
Role for macrophage inflammatory protein-2 in lipo- granulocyte-macrophage colony-stimulating factor in
polysaccharide-induced lung injury in rats. J Immunol apical periodontitis: correlation with clinical and
1996: 156: 1963–1972. histologic findings of the involved teeth. Oral Microbiol
38. Yamashiro S, Wang JM, Yang D, Gong WH, Kamohara Immunol 2003: 18: 9–13.
H, Yoshimura T. Expression of CCR6 and CD83 by 53. Beck J, Garcia R, Heiss G, Vokonas PS, Offenbacher S.
cytokine-activated human neutrophils. Blood 2000: 96: Periodontal disease and cardiovascular disease. J Perio-
3958–3963. dontol 1996: 67: 1123–1137.
39. Cheng SS, Lai JJ, Lukacs NW, Kunkel SL. Granulocyte- 54. Hernichel-Gorbach E, Kornman K, Hole SC. Host
macrophage colony-stimulating factor up-regulates responses in patients with generalized refractory
CCR1 in human neutrophils. J Immunol 2001: 166: periodontitis. J Periodontol 1994: 65: 8–16.
1178–1184. 55. Bramanti TE, Wong GG, Weintraub ST, Holt SC.
40. Pahl HL. Activators and target genes of Rel/NF-kB Chemical characterization and biological properties of
transcription factors. Oncogene 1999: 18: 6853–6866. lipopolysaccharide from Bacteroides gingivalis strains
41. Alcamo EA, Mizgerd JP, Horwitz BH, Bronson R, Beg W50, W83 and ATCC 33277. Oral Microbiol Immunol
AA, Scott M, Doerschuk CM, Hynes RO, Baltimore D. 1989: 4: 183–192.
Targeted mutation of tumor necrosis factor 1 rescues 56. Hou L, Sasaki H, Stashenko P. Toll-like receptor 4-
RelA-deficient mouse and reveals a critical role for NF- deficient mice have reduced bone destruction following
kB in leukocyte recruitment. J Immunol 2001: 167: mixed anaerobic infection. Infect Immun 2000: 68:
1592–1600. 4681–4687.
42. Takahashi K, MacDonald DG, Murayama Y, Kinane 57. Costerousse O, Allegrini J, Lopez M, Alhenc-Gelas F.
DF. Cell synthesis, proliferation and apoptosis in Angiotensin I-converting enzyme in human circulating
human dental periapical lesions analysed by in situ mononuclear cells: genetic polymorphism of expression
hybridisation and immunohistochemistry. Oral Dis in T-lymphocytes. Biochem J 1993: 290: 33–40.
1999: 5: 313–320. 58. Cavet J, Dickinson AM, Norden J, Taylor PR, Jackson
43. Kaneko T, Okiji T, Kan L, Takagi M, Suda H. GH, Middleton PG. Interferon-gamma and interleu-
Ultrastructural analysis of MHC Class II molecule- kin-6 gene polymorphisms associate with graft-versus-
expressing cells in experimentally induced periapical host disease in HLA-matched sibling bone marrow
lesions in the rat. J Endod 2001: 27: 337–342. transplantation. Blood 2001: 98: 1594–1600.
44. Huang GTJ, Do M, Wingard M, Park JS, Chugal N. 59. D’Aiuto F, Parkar M, Andreou G, Suvan J, Brett PM,
Effect of interleukin-6 deficiency on the formation of Ready D, Tonetti MS. Periodontitis and systemic
periapical lesions after pulp exposure in mice. Oral Surg inflammation: control of the local infection is associated
Oral Med Oral Pathol Oral Radiol Endod 2001: 92: with a reduction in serum inflammatory markers. J Dent
83–88. Res 2004: 83: 156–160.
45. Waddington RJ, Moseley R, Embery G. Reactive 60. Huang GT, Kinder Haake S, Kim JW, Park NH.
oxygen species: a potential role in the pathogenesis of Differential expression of interleukin-8 and intercellular
periodontal diseases. Oral Dis 2000: 6: 138–151. adhesion molecule-1 by human gingival epithelial cells
46. Birkedal-Hansen H. Role of matrix metalloproteinases in response to Actinobacillus actinomycetemcomitans or
in human periodontal diseases. J Periodontol 1993: 64: Porphyromonas gingivalis infection. Oral Microbiol
474–484. Immunol 1998: 13: 301–309.
47. Stashenko P, Wang CY, Riley E, Wu Y, Ostroff G, 61. Lechler R, Chai JG, Marelli-Berg F, Lombardi G. The
Niederman R. Reduction of infection-stimulated contributions of T-cell anergy to peripheral T-cell
periapical bone resorption by the biological res- tolerance. Immunology 2001: 103: 262–269.

101
Kiss

62. Hanazawa S, Murakami Y, Hirose K. Bacteroides periapical pathosis. Oral Microbiol Immunol 2003: 18:
(Porphyromonas) gingivalis fimbriae activate mouse 327–328.
peritoneal macrophages and induce gene expression 75. Wara-aswapati N, Boch JA, Auron PE. Activation of
and production of interleukin-1. Infect Immun 1991: interleukin1b gene transcription by human cytomega-
59: 1972–1977. lovirus: molecular mechanisms and relevance to perio-
63. Ogawa T, Kusumoto Y, Uchida H, Nagashima S, Ogo dontitis. Oral Microlbiol Immunol 2003: 18: 67–71.
H, Hamada S. Immunobiological activities of synthetic 76. Slots J, Sabeti M, Simon JH. Herpesviruses in periapical
peptide segments of fimbrial protein from Porphyromo- pathosis: an etiopathogenic relationship? Oral Surg
nas gingivalis. Biochem Biophys Res Comm 1991: 180: Oral Med Oral Pathol Oral Radiol Endod 2003: 96:
1335–1341. 327–331.
64. Hanazawa S, Murakami Y, Takeshita A, Kitami H, Ohta 77. Scott P, Pearce E, Cheever AW, Coffman RL, Sher A.
K, Amano S, Kitano S. Porphyromonas gingivalis Role of cytokines and CD41 T-cell subsets in the
fimbriae induce expression of the neutrophil chemo- regulation of parasite immunity and disease. Immunol
tactic factor KC gene of mouse peritoneal macrophages; Rev 1989: 112: 161–182.
role of protein kinase C. Infect Immun 1992: 60: 78. Stashenko P, Yu SM. T helper and T suppressor cell
1544–1549. reversal during the development of induced rat
65. Koch AE, Polverini PJ, Kunkel SL, Harlow LA, periapical lesions. J Dent Res 1989: 68: 830–834.
DiPietro LA, Elner VM, Elner SG, Streiter RM. 79. Reinherz EL, Schlossman SF. The differentiation and
Interleukin-8 as a macrophage-derived mediator of function of human T lymphocytes. Cell 1980: 19: 821–
angiogenesis. Science 1992: 258: 1798–1801. 827.
66. Tuschil A, Lam C, Halsberger A, Lindley I. Interleukin- 80. Marton IJ, Dezs B, Radics T, Kiss C Distribution of
8 stimulates calcium transients and promotes epidermal interleukin-2 receptor a-chain and cells expressing
cell proliferation. Invest Dermatol 1992: 99: 294–298. major histocompatibility complex class II antigen in
67. Reddi K, Wilson M, Nair S, Poole S, Henderson B. chronic human periapical lesions. Oral Microbiol Im-
Comparison of the pro-inflammatory cytokine-stimu- munol 1998: 13: 259–262.
lating activity of the surface-associated proteins of 81. Rodini CO, Lara VS. Study of the expression of CD681
periodontopathic bacteria. J Periodontal Res 1996: macrophages and CD81 T cells in human granulomas
31: 120–130. and periapical cysts. Oral Surg Oral Med Oral Pathol
68. Kirby AC, Meghi S, Nair S, White P, Reddi K, Nishihara Oral Radiol Endod 2001: 92: 221–227.
T, Nakashima K, Willis AC, Sim R, Wilson M, 82. Tasman F, Er N, Atac A, Dagdeviren A, Kendir B.
Henderson B. The potent bone resorbing mediator of CD45/isotypes expression in the immune cells of
Actinobacillus actinomycetemcomitans is homologous human periapical lesions. J Endod 2000: 26: 166–168.
to the molecular chaperone GroEL. J Clin Invest 1995: 83. Mosmann TR, Coffman RL. TH1 and TH2 cells:
96: 1185–1194. different pattern of lymphokine secretion leads to
69. Goulhen F, Hafezi A, Uitto VJ, Nakamura R, Grenier different functional properties. Annu Rev Immunol
D, Mayrand D. Subcellular localization and cytotoxic 1989: 7: 145–173.
activity of the GroEL-like protein isolated from 84. Kabashima H, Nagata K, Maeda K, Iijima T. Interferon-
Actinobacillus actinomycetemcomitans. Infect Immun g-producing cells and inducible nitric oxide synthase-
1998: 66: 5307–5313. producing cells in periapical granulomas. J Oral Pathol
70. Maeda H, Miyamoto M, Hongyo H, Nagai A, Kusihara Med 1998: 27: 95–100.
H, Muryama Y. Heat shock protein60 (GroEL) from 85. Kawashima N, Stashenko P. Expression of bone-
Porphyromonas gingivalis: molecular cloning and se- resorptive and regulatory cytokines in murine periapical
quence analysis of its gene and purification of the inflammation. Arch Oral Biol 1999: 44: 55–66.
recombinant protein. FEMS Microbiol Lett 1994: 124: 86. Hogan LH, Wang M, Suresh M, Co DO, Wienstock JV,
121–122. Sandor M. CD41 TCR repertoire heterogeneity in
71. Kadri R, Devine D, Ashraf W. Purification and Schistosoma mansoni-induced granulomas. J Immunol
functional analysis of the DnaK homologue from 2002: 169: 6386–6393.
Prevotella intermedia OMZ 326. FEMS Microbiol Lett 87. Suzuki N, Okiji T, Suda H. Enhanced expression of
1998: 167: 63–68. activation-associated molecules on macrophages of
72. Scannapieco FA, Genco RJ. Association of periodontal heterogeneous populations in expanding periapical
infections with atherosclerotic and pulmonary diseases. lesions in rat molars. Arch Oral Biol 1999: 44: 67–79.
J Periodontal Res 1999: 34: 340–345. 88. Kawahara T, Murakami S, Noiri Y, Ehara A, Takemura
73. Sabeti M, Valles Y, Nowzari H, Simon JH, Kermani- N, Furukawa S, Ebisu S. Effects of cyclosporin-A-
Arab V, Slots J. Cytomegalovirus and Epstein– induced immunosuppression on periapical lesions in
Barr virus DNA transcription in endodontic sympto- rat. J Dent Res 2004: 83: 683–687.
matic lesions. Oral Microbiol Immunol 2003: 18: 89. Polla BS, Baladi S, Fuller K, Rook G. Presence of hsp65
104–108. in bacterial extracts (OM-89): a possible mediator
74. Sabeti M, Simon JH, Slots J. Cytomegalovirus and of orally-induced tolerance? Experientia 1995: 51:
Epstein–Barr virus are associated with symptomatic 775–779.

102
Cell-to-cell interactions

90. Babal P, Soler P, Brozman M, Jakubovsky J, Beyly M, N, Gillespie MT, Martin TJ, Suda T. IL-17 in synovial
Basset F. In situ characterization of cells in periapical gran- fluids from patients with rheumatoid arthritis is a potent
uloma by monoclonal antibodies. Oral Surg Oral Med stimulator of osteoclastogenesis. J Clin Invest 1999:
Oral Pathol Oral Radiol Endod 1987: 64: 348–352. 103: 1345–1352.
91. Hou L, Sasakj H, Stashenko P. B-cell deficiency 103. Wang CY, Stashenko P. The role of interleukin-1a in the
predisposes mice to disseminating anaerobic infections: pathogenesis of periapical bone destruction in a rat model
protection by passive antibody transfer. Infect Immun system. Oral Microbiol Immunol 1993: 8: 50–56.
2000: 68: 5645–5651. 104. Wang CY, Stashenko P. Characterization of bone-
92. Valderhaug J. Experimentally induced periapical in- resorbing activity in human periapical lesions. J Endod
flammation in permanent and primary teeth of mon- 1993: 19: 107–111.
keys. Thesis, University of Oslo, Norway, 1974. 105. Formigli L, Orlandini S, Tonelli P, Giannelli M, Martini
93. Cury VCF, Sette PS, da Silva JV, de Araujo VC, Gomez M, Brandi ML, Bergamini M, Orlandini GE. Osteolytic
RS. Immunohistochemical study of apical periodontal processes in human radicular cysts: morphological and
cysts. J Endod 1998: 24: 36–37. biochemical results. J Oral Pathol Med 1995: 24: 416–
94. Liapatas S, Nakou M, Rontogianni D. Inflammatory 420.
infiltrate of chronic periradicular lesions: an immuno- 106. Haapasalo M, Ranta K, Shah H. Black-pigmented
histochemical study. Int Endod J 2003: 36: 464–471. Bacteroides ssp. in human apical periodontitis. Infect
95. Leonardi R, Villari L, Caltabiano M, Travali S. Heat Immun 1986: 53: 149–153.
shock protein 27 expression in the epithelium of 107. Hausmann E, Weinfeld N, Miller WA. Effects of
periapical lesions. J Endod 2001: 27: 89–92. lipopolysaccharides on bone resorption in tissue cul-
96. Moreira PR, Santos DFM, Martins RD, Gomez RS. ture. Calcif Tissue 1972: 9: 272–282.
CD571 cells in radicular cyst. Int Endod J 2000: 33: 108. Schoenfeld SE, Greening AB, Glick DH, Frank AL,
99–102. Simon JH, Herles SM. Endotoxic activity in periapical
97. Nickolaychuk B, McNicol A, Gilchrist J, Birek C. lesions. Oral Surg Oral Med Oral Pathol Oral Radiol
Evidence for a role of mitogen-activated protein kinases Endod 1982: 53: 82–87.
in proliferating and differentiating odontogenic epithe- 109. Nair PNR. Light and electron microscopic studies of
lia of inflammatory and developmental cysts. Oral Surg root canal flora and periapical lesions. J Endod 1987:
Oral Med Oral Pathol Oral Radiol Endod 2002: 93: 13: 29–39.
720–729. 110. Beuscher HU, Fallon RJ, Colten JR. Macrophage
98. Suda T, Takahashi N, Udagawa N, Jimi E, Gillespie MT, membrane interleukin 1 regulates the expression of
Martin TJ. Modulation of osteoclast differentiation and acute phase proteins in human hepatoma Hep 3B cells.
function by new members of the tumor necrosis factor J Immunol 1987: 139: 1896–1901.
receptor and ligand families. Endocr Rev 1999: 20: 111. Tatakis DN, Schneeberger G, Dziak R. Recombinant
345–357. interleukin-1 stimulates prostaglandin E2 production
99. Nagai M, Sato N. Reciprocal gene expression of by osteoblastic cells: synergy with parathyreoid hor-
osteogenesis inhibitory factor and osteoclast differen- mone. Calcif Tissue Int 1988: 42: 358–362.
tiation factor regulates osteoclast formation. Biochem 112. Kerekes K, Tronstad L. Long-term results of endodon-
Biophys Res Commun 1999: 257: 719–723. tic treatment performed with standardized techniques.
100. Fukagawa M, Kazama JJ, Kurokawa K. Renal osteodys- J Endod 1979: 5: 83–90.
trophy and secondary hyperparathyroidism. Nephrol 113. Ørstavik D. Time-course and risk analyses of the
Dial Transplant 2002: 17: 2–5. development and healing of chronic apical periodontitis
101. Stashenko P, Wang CY, Tani-Ishii N, Yu SM. Pathogen- in man. Int Endod J 1996: 29: 150–155.
esis of induced rat oeriapical lesions. Oral Surg Oral Med 114. Marton I, Nemes Z, Harmati S. Quantitative signifi-
Oral Pathol Oral Radiol Endod 1994: 78: 494–502. cance of IgE producing plasma cells and tissue
102. Kotake S, Udagawa N, Takahashi N, Matsuzaki K, Itoh distribution of mast cells in apical periodontitis. Oral
K, Ishiyama S, Ishiyama S, Saito S, Inoue K, Kamatani Microbiol Immunol 1990: 5: 46–48.

103

You might also like