You are on page 1of 9

Acta Odontologica Scandinavica

ISSN: 0001-6357 (Print) 1502-3850 (Online) Journal homepage: https://www.tandfonline.com/loi/iode20

Inflammatory profile of chronic apical


periodontitis: a literature review

Paulo Henrique Braz-Silva, Mariana Lobo Bergamini, Andressa Pinto


Mardegan, Catharina Simioni De Rosa, Bengt Hasseus & Peter Jonasson

To cite this article: Paulo Henrique Braz-Silva, Mariana Lobo Bergamini, Andressa Pinto
Mardegan, Catharina Simioni De Rosa, Bengt Hasseus & Peter Jonasson (2019) Inflammatory
profile of chronic apical periodontitis: a literature review, Acta Odontologica Scandinavica, 77:3,
173-180, DOI: 10.1080/00016357.2018.1521005

To link to this article: https://doi.org/10.1080/00016357.2018.1521005

© 2018 The Author(s). Published by Informa


UK Limited, trading as Taylor & Francis
Group.

Published online: 26 Dec 2018.

Submit your article to this journal

Article views: 13546

View related articles

View Crossmark data

Citing articles: 70 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=iode20
ACTA ODONTOLOGICA SCANDINAVICA
2019, VOL. 77, NO. 3, 173–180
https://doi.org/10.1080/00016357.2018.1521005

REVIEW ARTICLE

Inflammatory profile of chronic apical periodontitis: a literature review


Paulo Henrique Braz-Silvaa,b , Mariana Lobo Bergaminia, Andressa Pinto Mardegana,
Catharina Simioni De Rosaa, Bengt Hasseusc and Peter Jonassond
a
Division of General Pathology, Department of Stomatology, School of Dentistry, University of Sao Paulo, Sao Paulo, Brazil; bLaboratory of
Virology, Institute of Tropical Medicine of Sao Paulo, University of Sao Paulo, Sao Paulo, Brazil; cDepartment of Oral Medicine and
Pathology, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; dDepartment of
Endodontology, Institute of Odontology The Sahlgrenska Academy University of Gothenburg, Gothenburg, Sweden

ABSTRACT ARTICLE HISTORY


Apical periodontitis caused by root canal infection is the most frequent pathological lesion in the Received 20 April 2018
jaws, mainly manifested as periapical granulomas and cysts. Understanding of the formation and pro- Revised 5 August 2018
gression of apical periodontitis as well as the identification of inflammatory biomarkers can help Accepted 4 September 2018
increase the knowledge of pathogenic mechanisms, improve the diagnosis and provide support for
KEYWORDS
different therapeutic strategies. The objective of the present article is to review inflammatory bio- Apical periodontitis;
markers such as cytokines, chemokines, inflammatory cells, neuropeptides, RANK/RANKL/OPG system endodontic inflammation;
and other inflammatory markers and to relate these systems to the development and progression of inflammation mediators;
pathological conditions related to apical periodontitis. osteoprotegerin;
RANK ligand

Introduction It is known that microbial antigens from the root


canal infection are capable to stimulate both specific and
Apical periodontitis (AP) is a prevalent infectious disease
non-specific immune responses in periapical tissue [14,15].
worldwide and is characterised by an inflammatory response
The inflammatory cell infiltrate in granulomas and radicular
and bone destruction in the periapical tissues caused by
cysts is primarily mononuclear cells. The interaction between
microbial infection in the dental pulp [1–3]. AP is the most
cells, cytokines and other inflammatory elements present in
frequent inflammatory lesion related to teeth in the jaws.
AP, including their specific functions, is not completely eluci-
The jaws are unique in comparison with other bones in the
dated [16–19].
body as the presence of teeth creates a direct pathway
Studies have demonstrated that macrophages, mastcells,
towards the bone marrow, with no epithelial barriers against
T cells and neutrophils participate in the formation of AP,
infectious and inflammatory agents if the dental pulp
also including cytokines, chemokines and RANK/RANKL/OPG
becomes necrotic and infected. Hence, tissue- and immuno-
(Receptor Activator of Nuclear Factor kappa B ligand/osteo-
logical responses are fundamental for protecting from the
protegerin) system [6,19,20]. The great amount and interac-
spread of infectious agents to other locations. Due to the
tions of various inflammatory stimuli can influence and alter
state of the infection in the root canal, acute or chronic
the state and progression of the disease [21–23].
inflammatory reaction can develop. In AP, bone destruction
The objective of this article is to review inflammatory bio-
is caused by both microbial infection and immune response
markers related to the development and progression of
as part of the defence reaction [1–3].
chronic AP, which can help increase the knowledge of patho-
Histologically, AP is classified as abscess, granuloma or
genic mechanisms. A computerized literature search was
radicular cyst [4–6]. Periapical abscess reflects a formation of
conducted on the PubMed database for studies evaluating
pus as a consequence of a shift in cellular dynamics in
the human and animal inflammatory response in the periapi-
response to an acute infection, whereas periapical granulomas
cal tissue published from 1965 to 2018. This resulted in 87
consist of granulation tissue with inflammatory cells, fibro-
included articles that were read in full text.
blasts and well-developed fibrous capsule. The granuloma
may eventually evolve into radicular cysts when epithelial
rests of Malassez, located in the periodontal tissue, are stimu-
lated by the immunological response to proliferate [7–13].

CONTACT Peter Jonasson Peter.Jonasson@odontologi.gu.se Department of Endodontology, Institute of Odontology, The Sahlgrenska Academy, University
of Gothenburg, Medicinaregatan 12, Gothenburg, 41390, Sweden
ß 2018 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License (http://creativecommons.org/licenses/by-nc-nd/4.
0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited, and is not altered, transformed, or built upon in
any way.
174 P. H. BRAZ-SILVA ET AL.

Review regulation of inflammatory processes through activation and


differentiation of osteoclasts, activation and proliferation of
Infection and inflammatory response fibroblasts, production of collagen and neo-vascularisa-
AP is essentially an inflammatory disease of microbial aeti- tion [12,19,33].
ology. Knowledge of microbial location, organisation and T cells are abundant in AP, but the activation and func-
virulence factors within the root canal system is important tion of these cells are yet not well understood. Several regu-
for understanding the disease process. Although fungi, latory circuits are activated by T cells, influencing not only
archaea and viruses have been found in association with AP, the immune system, but also the interaction with other cells,
bacteria are the main microbial aetiological agents. oral epithelium and oral bacteria. T helper 1 (Th1) and
Microbiota are found in highly organised and complex enti- T helper 2 (Th2) cell responses modulate the expression of
ties, known as multi-species biofilms mainly located inside IL-1, which according to IhanHren and Ihan [34], might be
the root canal. In specific circumstances, microorganisms can the main mediators of bone resorption and activator of mac-
overcome the defence barrier and even establish an extra- rophages in the context of AP. A larger population of acti-
radicular infection [24,25]. vated T cells was found in granulomas compared to cysts,
There is a clear evidence that microbial interaction with a Th1/Th2 ratio being similar between cysts and granu-
plays an important role for the pathogenesis of AP [25,26]. lomas [34,35].
Microorganisms may cause direct tissue damage and modu- According to some studies, Th1 and Th2 response pat-
late the immunological response by secretion of products, terns occur concomitantly [6,34]. It is believed that the Th1
including enzymes, exotoxins and metabolic end-products immune response is involved in both lesion progression and
[27–29]. Based on the variation in root-canal infection with bone destruction, whereas Th2 involves immunosuppressive
different virulence factors, the immunological response caus- mechanisms which are important in the repair process,
ing AP will vary over time. occurring in the more advanced stages of lesion develop-
ment [35]. This is demonstrated in experimental models in
which absence of Th1 cytokines does not alter significantly
Inflammatory cells the lesion development, whereas the lack of Th2 cytokines
Distinct sub-populations of inflammatory cells have been results in an increase in lesion size [14,36]. This concept,
described in AP. Neutrophils and their interaction with micro- however, needs to be reviewed as recent findings indicate a
organisms are of particular importance in the acute phase as pattern characterised by the presence of IL-17, a cytokine dif-
first line of defence and for progression of AP, causing tissue ferent from the traditional Th1 and Th2 lines [34].
damage and chemotaxis. Mast cells and macrophages are The predominance of Th1 response in granulomas and
related to important components of the inflammatory infil- Th2 in cysts is demonstrated by authors who suggest
trate of chronic AP, although one cannot know exactly how that both immune responses can be suppressed by regula-
these cells are associated with the production of Interleukin- tory T cells (Tregs) through mechanisms depending on cell
6 (IL-6) and other inflammatory mediators. A study on the contact and/or production of IL-10 and TGF-b [36,37]. An
distribution of these cells in periapical cysts showed that increase in the expression of IL-10 and FoxP3 can be
mast cells have the capacity to degranulate and produce sev- observed in granulomas compared to cysts, which suggests
eral inflammatogenic substances, such as vasoactive media- that Treg cell population is present to a higher extent in the
tors [19]. The release of these substances is directly related former [36]. Francisconi et al. [37] suggest that the use of
to inflammatory events and bone resorption, also influencing Treg chemoattractants could be a promising strategy for clin-
other cells of the immune system [19]. ical management of AP, since the migration of these cells
Studies have compared the presence of mast cells in peri- switches active lesion into inactivity phenotypes [37].
apical granulomas and cysts, confirming that these cells are There are still studies reporting a predominance of Th1
found in great amount in the latter. In addition, it was response in lesions mainly mediated by T cells, whereas
observed that mast cells are widely found in sub-epithelial lesions mediated by B cells and/or plasma cells exhibit a Th2
regions rather than in the deep regions of the cystic capsule. response [15]. Some authors suggest that T cells would be
These results would explain the growth trend of the cysts as involved in the beginning and development of the lesion,
these cells account for the release of inflammatory media- whereas B cells would in turn be involved in the process of
tors, such as tumour necrosis factor a (TNF-a), interleukin-1b repair [38].
(IL-1b), macrophage inflammatory protein-1a (MIP-1a) and New subsets of T-helper cells (e.g. Th9, Th17 and Th22)
monocyte chemoattractant protein-1 (MCP-1). Mast cell have been recently discovered, including their prototypic
degranulation may cause inflammatory and vascular changes, cytokines IL-9 and IL-22. It was found that the expression of
thus contributing to the development and expansion of the IL-9 and IL-22 is predominant in inactive lesions, suggesting
cysts [30–32]. the existence of a possible protective role of these cytokines
Macrophages, in turn, are capable to produce pro- and in the pathogenesis of AP [39]. Aranha et al. [39] demon-
anti-inflammatory substances which act on the development strated the over-expression of IL-9 in inactive granulomas
and repair of these lesions by secreting soluble IL-1a, TNF-a, and a negative correlation with TNF-a, IFN-c and IL-17 in the
IL-6, TGF-b (Transforming growth factor beta) and prosta- lesions [39]. Studies with experimental models of AP in mice
glandins. These cytokines play a role in the initiation and also showed that IL-9 expression seems to help in the
ACTA ODONTOLOGICA SCANDINAVICA 175

control of the lesion as no differences were observed in its differentiation of Treg and Th17. Therefore, TGF-b drives the
extension. However, further studies are needed to determine immune response into a suppressive mode by inducing and
the immunosuppressive role of IL-9 and IL-22 in AP recruiting Tregs in order to restrain the inflammatory reac-
[23,39,40]. Oseko et al. [41] showed that IL-17 knockout mice tion, whereas low numbers of Tregs drive the immune
are resistant to the development of experimental AP, indicat- response into Th17 in order to stimulate the inflammatory
ing a role for Th17 cells and their cytokine production in the process [12].
inflammatory process [41]. IL-6 is an important cytokine in bone re-modelling and
Langerhans cells, a subset of dendritic cells, are found in activation and differentiation of immune cells and osteo-
AP and seem to be strongly related to the chemotaxis of T clasts, with macrophages being the main sources in periapi-
cells with an epithelial proliferative potential. An increase in cal cysts [19,20,45,46]. High levels of IL-6 in AP seem to be
the density of Langerhans cells may be one of the factors correlated with symptomatic and active lesions [45]. IL-6 is
associated with the development of lesions with more an important pro-inflammatory biomarker and some recent
intense inflammatory infiltrates. Discrepant results were studies aimed to correlate the expression in AP (as a reser-
found in studies aimed at detecting these cells in periapical voir of inflammatory cytokines) with blood levels in order to
cysts and granulomas. This fact may be attributed to the verify the relationship of these lesions with systemic inflam-
apoptosis process the dendritic cells undergo after presenta-
matory conditions, showing very conflicting results [3,47–49].
tion of an antigen, tissue repair process characterised by
Analysis of the number of macrophages and TNF-a levels
lower number of these cells, maturation profile of these cells
in cyst capsules showed a positive correlation with capsule
and difference in the methods used by authors [42–44].
thickness. In fact, the expression of TNF-a was correlated
with the amount of macrophages in cystic walls, since they
Cytokines are the main source of TNF-a. A relationship between
amount of macrophages in tissues surrounding the cyst and
Cytokines are proteins secreted by cells into injured tissues
high expression of TNF-a was also determined, thus indicat-
in response to microbial agents and other injuries through
ing a correlation between production of this cytokine and
recruitment of leucocytes [21]. Together with prostaglandins,
tissue vascularisation in cysts as well as between angiogen-
cytokines participate in the initiation and regulation of
esis and inflammation [50].
inflammatory processes through activation and differenti-
ation of osteoblasts, activation and proliferation of fibro-
blasts, production of collagen and neo-vascularisation [21].
IL-17 and TGF-b are two important cytokines found in
apical periodontitis, being secreted mainly by Th17 cells and Chemokines
Tregs, respectively. They seem to interact with each other, Chemokines represent a family of small proteins (8-10kD)
thus characterising the action of pro-inflammatory and which are associated with migration and activation of leuko-
immunoregulatory cytokines. These cytokines have opposite cytes and selectins, with the latter accounting for the adhe-
effects, but with no mutual inhibition. While TGF-b is a
sion of inflammatory cells to the vessel walls in different
potent immune-modulating cytokine, IL-17 is capable to
inflammatory cells [13]. Silva et al. [6] demonstrated the
reactivate the inflammatory process by stimulating the pro-
expression of chemokines receptors (i.e. CCR1, CCR2, CCR3,
duction of IL-8 [38,43], meaning that re-acutisation of chronic
CCR5, CXCR1, CXCR3) in both periapical granulomas and
apical periodontitis is closely related to higher levels of IL-17
cysts. These receptors are found in Th1 and Th2 cells as well
and increased infiltration of leukocytes [12,13].
as in monocytes and neutrophils [1,3–5], with Th1 cells
Higher levels of IL-17 were observed in patients with fistu-
expressing predominantly chemokine receptors CCR5 and
lae and exhibiting mixed inflammatory infiltrate, which in
turn promotes exacerbation of the inflammatory response CXCR3, Th2 cells expressing mainly CCR4 and CCR3, and
and increases the number of neutrophils and bone resorp- monocytes/macrophages expressing CCR2 and CCR5 [34].
tion [13,14]. IL-17 seems to attract neutrophils and might High levels of chemokine CXCL12/SDF-1 are found in peri-
also induce the production of RANK-L by activating osteo- apical inflammatory lesions, with CD117þ mast cells being
clasts, involved in the bone resorption present in AP [19]. the main source expressing this chemokine in this type of
Nevertheless, there are studies attributing a protective role lesion. It is suggested that CXCL12/SDF-1 plays an important
to IL-17 in the bone resorption process as a result of the role in the destruction of the periapical tissue, probably
control this cytokine exerts in the expression of chemokines inducing infiltration by immune cells, especially mast
and recruitment of neutrophils. Therefore, the oppositional cells [51].
influence of IL-17 on the regulation of neutrophils would High levels of chemokines RANTES, IP-10 and MCP-1,
outweigh its potential of causing bone destruction [34]. including chemokine receptors CCR3, CCR5, CXCR1, CXCR3 in
High levels of TGF-b may be important for the resolution periapical cysts, may be related to a possible evolution of
of AP, since this factor not only inhibits bone resorption and granulomas and cysts. However, the mechanisms involved
promotes tissue remodelling and repair by stimulating syn- are poorly understood, suggesting a possible involvement of
thesis of collagen, neo-vascularisation and proliferation of cytokines in the proliferation of epithelial rests of
fibroblasts, but it also seems to be involved in the Malassez [6].
176 P. H. BRAZ-SILVA ET AL.

RANK/RANKL/OPG system stable as OPG competes with RANKL by preventing its


binding to RANK, thus impeding the osteolytic activ-
Odontogenic cysts are lesions characterised by local
ity [64].
bone destruction, in both development and inflammatory
The complex process of bone resorption involves other
groups. The RANK/RANKL/OPG system is an important
bone metabolism-related factors rather than RANKL and OPG
bone metabolism regulator which acts on differentiation and
mediators, such as TNF-a and macrophage colony-stimulat-
activity of osteoclasts. RANK is a transmembrane protein – a
ing factor, showing that bone metabolism can be directly
member of the tumour necrosis factor receptor family – and
associated with the inflammatory conditions [46,60]. An
expressed mainly by macrophages, pre-osteoclastic mono-
nuclear cells, T- and B cells, dendritic cells and fibroblasts. immune-regulatory role of RANKL has been recently
RANKL (Receptor Activator of Nuclear Factor kappa B ligand) described, mediated by the induction of Treg development
is a cytokine similar to TNF-a and acts as a ligand of RANK and resulting in an antigen-specific immunologic hypo-
and osteoprotegerin (OPG) [52,53]. Activation of RANK by responsiveness, with arrest of the periapical lesion progres-
RANKL results in its interaction with TNF-associated recep- sion in animal model [65].
tors, activation of nuclear factor kB (NF-kB) and protein c-Fos, Armada et al. [66] analysed the expression and distribu-
all being related to the maturation of osteoclasts through tion of RANK, RANKL and OPG in periradicular cysts and
the increased expression of specific genes [52,54]. On the found higher expressions of RANK and RANKL in the con-
other hand, the OPG soluble receptor, also produced by nective tissue during presence of chronic inflammatory
osteoclasts, is capable to block the interaction RANK/RANKL infiltrate compared to mixed inflammatory infiltrate.
by binding to RANKL and preventing osteoclastic differenti- Another study investigated the possible association
ation and activation, thus reducing the bone resorp- between expressions of RANKL and OPG and inflammatory
tion [53,55,56]. infiltrate in the chronic AP, demonstrating that RANKL
Imbalance in this system is found in some diseases, such expression was increased in chronic inflammatory infiltrate
as rheumatoid arthritis and osteoporosis [56]. High levels of containing lymphocytes (T and B cells) and macrophages
RANKL have been associated with an increase in osteoclastic [67]. These findings are corroborated by the fact that other
activity, thus favouring bone resorption, whereas higher lev- cells rather than osteoblasts, such as fibroblasts and lym-
els of OPG exert an inhibitory effect on the survival of osteo- phocytes T and B, express a greater amount of
clasts, thus reducing the bone resorption capacity [36,57]. RANKL [66,67].
This fact is already well established in the literature as sam-
ples of AP were shown to have higher levels of RANKL and
OPG than the levels found in healthy tissues [23,58,59]. Neuropeptides
The comparison between expressions of RANKL and OPG Neuropeptides are conceptualised as neurotransmitters of
in odontogenic lesions with biologically different behaviours peptides or neuromodulators [68] as they determine their
(e.g. radicular cysts, dentigerous cysts, solid ameloblastomas synthesis and release from neurons and have biological
and keratocysts) shows a higher immune-detection of RANKL actions mediated by extracellular receptors on target cells
as well as a lower immune-detection of OPG in the latter
[69]. The involvement of afferent neuron fibres in peripheral
two lesions compared to the former two, which is compat-
inflammatory processes has been studied for more than a
ible to the more aggressive behaviour of ameloblastomas
century, and the excitation of these fibres is known to lead
and keratocysts [60]. Despite these expected findings, there
to vasodilatation and subsequent oedema [68,69].
are studies reporting a higher number of OPG þ than
The term ‘neurogenic inflammation’ describes the compo-
RANKL þ cells in keratocystic capsules [61] and lining epithe-
nent of the inflammatory process triggered by an appropri-
lium of dentigerous and radicular cysts [57]. This demon-
ate stimulus applied to peripheral neurons, resulting in the
strates that discrepant results may be found by different
release of neuropeptides that alter multiple processes,
authors, probably due to differences in their methods and
also to the developmental stage of the lesion [6,57]. including vascular permeability, hypersensitivity and vasodila-
In addition, it was observed that the expression of tation at the injury site [68]. From these findings, studies
RANKL/OPG is not significantly different between symptom- have focussed on the role of neuropeptides, including sub-
atic and asymptomatic AP, but a positive correlation was stance P (SP), vasoactive intestinal polypeptide (VIP), neuro-
found between number of bacteria and levels of OPG in peptide Y (NPY), calcitonin gene-related peptide (CGRP) and
symptomatic AP [62,63]. Santos et al. [64] investigated the neurokinin A (NKA) [68,69].
participation of RANKL, OPG, IL-33, cathepsin K and TNF-a Neuropeptides have been associated with the develop-
in radicular cysts and periapical granulomas, demonstrating ment of AP due to their abundant innervation. Studies dem-
that there was a higher immune-expression of these mole- onstrate that VIP is related to growth and maturation
cules in connective tissue cells (capsule of radicular cysts), processes of the lesion, since it was associated with bone
which indicates that RANKL, OPG, IL-33, cathepsin K and resorption and regulation of osteoclast functions [68,70]. SP
TNF-a stimulate the osteolytic activity. Differently from array in immune cells located in periradicular granulomas
radicular cysts, however, levels of OPG were found to be has also been reported, with SP being expressed cytoplas-
rather lower compared to RANKL and cathepsin K in peri- matically in macrophages in acute and chronic inflammatory
apical granulomas, suggesting that these lesions are more lesions [68,71].
ACTA ODONTOLOGICA SCANDINAVICA 177

Experimental studies support the involvement of CGRP in the expression of its target-genes, thus influencing various
bone remodelling. Reduction of active osteoclasts in induced cell signalling pathways [79]. The profile of miRNA expression
periapical lesions was observed when the density of the can be considered as a diagnostic and prognostic marker in
nerve fibres of the immune-reactive CGRP reached its peak, a number of conditions, including odontogenic tumours [81].
suggesting a possible role for CGRP in inhibiting reabsorp- Yue et al. [79] assessed the expression of miRNAs (miR-
tion [68,72,73]. CGRP receptors have been found in osteo- 29b, 106b, 125b, 143, 155 and 198) associated with inflam-
blasts, providing evidence of the nervous systems influence mation involving AP lesions and human periodontal ligament
on bone metabolism [68]. fibroblasts (HPDLFs), showing that all of them were signifi-
cantly up-regulated in tissues of asymptomatic AP. On the
other hand, miR-29b, 106b, 125b and 198 were significantly
Other inflammatory markers reduced in acute inflammation involving HPDLFs, whereas
The cell protection mechanism involves the expression of a miR-143 and 155 suffered no change, thus suggesting that
polypeptide family called ‘heat-shock’ proteins (HSPs) [74], miRNA expressions associated with inflammation were differ-
which play a protective role against harmful environmental ent between AP lesions and HPDLFs.
conditions and pathogens. HSPs are characteristically Reactive oxygen species (ROS) are unstable, extremely-
induced by stress signals, such as inflammatory mediators, reactive molecules capable to transform other molecules
high temperature, reduced oxygen supply, and infectious they collide with. ROS are generated in great amount during
agents, playing important roles in the doubling and trans- oxidative stress (i.e. an imbalance between oxidant anti-oxi-
location of polypeptides through the cell’s membrane [75]. dant agents), a condition in which proteins, carbohydrates,
According to their molecular weight, HSPs are sub-divided lipids and nucleic acids are affected [82]. The production of
into the following groups: HSPH (HSP110), HSPC (HSP90), ROS is an important defence mechanism against pathogen
HSPA (HSP70), DNAJ (HSP40), HSPD (HSP60) and HSPB invasion, with close involvement of bone resorption in the
(HSP27) [74,75]. case of endodontic lesions [82–87].
Goodman et al. [74] compared the expression of 44 HSP During the process of endodontic infection, the binding
genes in human periapical granulomas and cell culture with of bacterial motifs to Toll-like receptors (TLRs) on the surface
healthy periodontal ligament tissue (control) in macrophages of phagocytes leads to the beginning of phagocytosis,
with and without LPS treatment, revealing that members of inducement of humoral and cellular responses (mediated by
the families of genes HSP27 (HSPB1), HSP40 (HSPA6), HSP70 lymphocytes B and T, respectively), ROS synthesis and conse-
(DNAJC3) and HSP110 (HSPA4) were significantly over- quent production of inflammatory mediators with cytocines
expressed in periapical granulomas, especially in active ones and metalloproteinases (MMPs) [82,83]. The increased expres-
compared to controls, as well as in LPS-treated cells. HSPA-4, sion of ROS-related markers has been associated with the
a member of the HSP70 family, showed higher expression in pathogenesis of periapical periodontitis [82–86] at both local
inactive granulomas. These findings suggest that HSPs can and systemic levels [83,85–87].
have modulatory functions during the development of peri- Dezerega et al. [83] demonstrated that lesions of asymp-
apical lesion and that different heat-shock genes/proteins tomatic AP are characterised by a pro-oxidative profile, with
may play different roles in this process [74]. consequent increase in the expressions of MMP-2 and MMP-
Nitric oxide (NO) is also an important inflammatory medi- 9 compared to healthy tissue. This same difference in profile
ator involved in the AP. NO is an omnipresent free radical was demonstrated in the gingival crevicular fluid, thus show-
produced by several cells through a family of enzymes col- ing the potential use of these biomarkers as a possible diag-
lectively known as NO synthases (NOSs) [76]. Although the nostic tool.
role of NO in AP is unknown, studies have shown that NO The bone metabolism regulation is also altered in the oxi-
modulates the levels of pro-inflammatory cytokine, such as dative stress environment [84]. In a recent study, Jakovljevic
IFN-c and TNF-a during the pathogenesis of AP [77]. et al. [84] suggested that periapical periodontitis is character-
Cintra et al. [77] assessed the serum levels of TNF-a, IFN-c, ised by increased levels of oxidative stress markers, such as
IL-6, IL-17, IL-23 and NO in rats with AP regarding one tooth 8-hydroxydeoxy guanosine (8-OHdG), oxidised glutathione
or multiple teeth. The findings showed an increase in the (GSSG) and bone resorption regulators (RANKL and OPG),
serum levels of IL-6, IL-17, IL-23 and TNF-a in rats with AP compared to healthy tissue – a factor which might explain
involving multiple teeth. On the other hand, the serum levels the process of extensive bone resorption in these lesions.
of NO were decreased in rats with AP involving either one
tooth or multiple teeth. The increase in the serum levels of
Conclusion
pro-inflammatory cytokines in rats with AP corroborates the
hypothesis that endodontic infections affect negatively the Understanding the formation process of apical periodontitis,
systemic health. as well as the inflammatory biomarkers associated with its
Micro-RNAs (miRNAs) are small non-coding RNAs account- development, is important for evaluation of pathogenesis,
ing for the regulation of gene expression at the post-tran- diagnosis and development of therapeutic strategies for AP.
scriptional level [78–80]. They act as controllers of several Although the inflammatory profile of these lesions is still
biological activities, including differentiation, proliferation poorly understood, it can be concluded that this formation
and apoptosis [78]. Changes in the miRNA expression affect process is dynamic and that different inflammatory cells and
178 P. H. BRAZ-SILVA ET AL.

their by-products are involved in the process. Nevertheless, [15] Kawashima N, Stashenko P. Expression of bone-resorptive and
further studies are needed to understand how cellular and regulatory cytokines in murine periapical inflammation. Arch Oral
Biol. 1999;44:55–66.
molecular events contribute to the development of AP.
[16] Takahashi K. Microbiological, pathological, inflammatory,
immunological and molecular biological aspects of periradicular
disease. Int Endod J. 1998;31:311–325.
Disclosure statement [17] Stashenko P, Teles R, D’Souza R. Periapical inflammatory
The authors have stated explicitly that there are no conflicts of interest responses and their modulation. Crit Rev Oral Biol Med. 1998;9:
regarding this article. 498–521.
[18] Graunaite I, Lodiene G, Maciulskiene V. Pathogenesis of apical
periodontitis: a literature review. JOMR. 2011;2:1–15.
Funding [19] Bracks IV, Armada L, Gonçalves LS, et al. Distribution of mast cells
and macrophages and expression of interleukin-6 in periapical
This study was supported by grants from Sao Paulo Research cysts. J Endod. 2014;40:63–68.
Foundation – FAPESP, Sao Paulo, Brazil [grant no. 2015/07727-9] and [20] Huang GT, Do M, Wingard M, et al. Effect of interleukin-6 defi-
from Coordenacao de Aperfeicoamento de Pessoal de Nivel Superior - ciency on the formation of periapical lesions after pulp exposure
Brasil (CAPES) - Finance Code 001. in mice. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2001;
92:83–88.
[21] Araujo-Pires AC, Francisconi CF, Biguetti CC, et al. Simultaneous
ORCID analysis of T helper subsets (Th1, Th2, Th9, Th17, Th22, Tfh, Tr1
and Tregs) markers expression in periapical lesions reveals mul-
Paulo Henrique Braz-Silva http://orcid.org/0000-0002-1842-9521 tiple cytokine clusters accountable for lesions activity and inactiv-
Peter Jonasson http://orcid.org/0000-0002-7541-983X ity status. J Appl Oral Sci. 2014;22:336–346.
[22] Garlet GP. Destructive and protective roles of cytokines in peri-
odontitis: a re-appraisal from host defense and tissue destruction
viewpoints. J Dent Res. 2010;89:1349–1363.
References [23] Menezes R, Garlet GP, Letra A, et al. Differential patterns of recep-
tor activator of nuclear factor Kappa B ligand/osteoprotegerin
[1] Kakehashi S, Stanley HR, Fitzgerald RJ. The effects of surgical expression in human periapical granulomas: possible association
exposures of dental pulps in germ-free and conventional labora- with progressive or stable nature of the lesions. J Endod. 2008;
tory rats. Oral Surg Oral Med Oral Pathol. 1965;20:340–349. 34:932–938.
[2] Mo €ller AJR, Fabricius L, Dahlen G, et al. Influence on periapical [24] Sunde PT, Tronstad L, Eribe ER, et al. Assessment of periradicular
tissues of indigenous oral bacteria and necrotic pulp tissue in microbiota by DNA-DNA hybridization. Endod Dent Traumatol.
monkeys. Eur J Oral Sci. 1981;89:475–484. 2000;16:191–196.
[3] Azuma MM, Samuel RO, Gomes-Filho JE, et al. The role of IL-6 on [25] Nair PN. On the causes of persistent apical periodontitis: a review.
apical periodontitis: a systematic review. Int Endod J. 2014;47: Int Endod J. 2006;39:249–281.
615–621. [26] Sundqvist GK, Eckerbom MI, Larsson AP, et al. Capacity of anaer-
[4] Bhaskar SN. Oral surgery-oral pathology conference no. 17, obic bacteria from necrotic dental pulps to induce purulent infec-
Walter Reed Army Medical Center Periapical lesions – types, inci- tions. Infect Immun. 1979;25:685–693.
dence, and clinical features. Oral Surg Oral Med Oral Pathol Oral [27] Siqueira JF Jr. Endodontic infections: concepts, paradigms, and
Radiol. 1966;21:657–671. perspectives. Oral Surg Oral Med Oral Pathol Oral Radiol Endod.
[5] Koivisto T, Bowles WR, Rohrer M. Frequency and distribution of 2002;94:281–293.
radiolucent jaw lesions: a retrospective analysis of 9,723 cases. [28] Henderson B, Poole S, Wilson M. Bacterial modulins: a novel class
J Endod. 2012;38:729–732. of virulence factors which cause host tissue pathology by induc-
[6] Silva TA, Garlet GP, Lara VS, et al. (2005) Differential expression of ing cytokine synthesis. Microbiol Rev. 1996;60:316–341.
chemokines and chemokine receptors in inflammatory periapical [29] Takahama A Jr, Ro ^ças IN, Faustino ISP. Association between bac-
diseases. Oral Microbiol Immunol. 2005;20:310–316. teria occurring in the apical canal system and expression of
[7] Nair PN. Pathogenesis of apical periodontitis and the causes of bone-resorbing mediators and matrix metalloproteinases in apical
endodontic failures. Crit Rev Oral Biol Med. 2004;15:348–381. periodontitis. Int Endod J. 2018;51:738–746.
[8] Ten Cate AR. The epithelial cell rests of Malassez and the genesis [30] Nogueira EFC, Farias EGF, Silva LB, et al. Analysis of the presence
and location of mast cells in periapical cysts and periapical gran-
of the dental cyst. Oral Surg Oral Med Oral Pathol. 1972;34:
ulomas. RGO, Rev Ga uch Odontol. 2016;64:376–381.
956–964.
[31] Rodini CO, Batista AC, Lara VS. Comparative immunohistochemi-
[9] Valderhaug J. A histologic study of experimentally induced
cal study of the presence of mast cells in apical granulomas and
radicular cysts. Int J Oral Surg. 1972;1:137–147.
periapical cysts: Possible role of mast cells in the course of
[10] Harris M, Toller P. The pathogenesis of dental cysts. Br Med Bull.
human periapical lesions. Oral Surg Oral Med Oral Pathol Oral
1975;31:159–163.
Radiol Endod. 2004;97:59–63.
[11] Shear M. (1985) Cysts of the jaws: recent advances. J Oral Pathol.
[32] Patidar KA, Parwani RN, Wanjari SP, et al. Mast cells in human
1985;14:43–59.
odontogenic cysts. Biotech Histochem. 2012;87:397–402.
[12] Andrade AL, Nonaka CF, Gordo n-Nun~ez MA, et al.
[33] Marton IJ, Kiss C. Overlapping protective and destructive regula-
Immunoexpression of interleukin 17, transforming growth factor tory pathways in apical periodontitis. J Endod. 2014;40:155–163.
b1, and forkhead box P3 in periapical granulomas, radicular cysts, [34] Ihan Hren N, Ihan A. T lymphocyte activation and cytokine
and residual radicular cysts. J Endod. 2013;39:990–994. expression in periapical granulomas and radicular cysts. Arch Oral
[13] Marçal JR, Samuel RO, Fernandes D, et al. T-helper cell type 17/ Biol. 2009;54:156–161.
regulatory T-cell immunoregulatory balance in human radicular [35] de Carvalho Fraga CA, Alves LR, de Sousa AA, et al. Th1 and Th2-
cysts and periapical granulomas. J Endod. 2010;36:995–999. like protein balance in human inflammatory radicular cysts and
[14] Colic M, Gazivoda D, Vucevic D, et al. Proinflammatory and periapical granulomas. J Endod. 2013;39:453–455.
immunoregulatory mechanisms in periapical lesions. Mol [36] Fukada SY, Silva TA, Garlet GP, et al. Factors involved in the T
Immunol. 2009;47:101–113. helper type 1 and type 2 cell commitment and osteoclast
ACTA ODONTOLOGICA SCANDINAVICA 179

regulation in inflammatory apical diseases. Oral Microbiol [58] Fan R, Sun B, Zhang CF, et al. Receptor activator of nuclear factor
Immunol. 2009;24:25–31. kappa B ligand and osteoprotegerin expression in chronic apical
[37] Francisconi CF, Vieira AE, Biguetti CC, et al. Characterization of periodontitis: possible association with inflammatory cells. Chin
the protective role of regulatory T cells in experimental periapical Med J. 2011;124:2162–2166.
lesion development and their chemoattraction manipulation as a [59] Estrela C, Decurcio DA, Silva JA, et al. Immune-inflammatory cell
therapeutic tool. J Endod. 2016;42:120–126. profile and receptor activator of nuclear factor Kappa B ligand/
[38]  c M, Lukic A, Vucevic D, et al. Correlation between pheno-
Coli osteoprotegerin expression in persistent apical periodontitis after
typic characteristics of mononuclear cells isolated from human root canal retreatment failure. J Endod. 2016;42:439–446.
periapical lesions and their in vitro production of Th1 and Th2 [60] Matos FR, Moraes M, Neves EBS, et al. Immunohistochemical
cytokines. Arch Oral Biol. 2006;51:1120–1130. detection of receptor activator nuclear jB ligand and osteoprote-
[39] Aranha AM, Repeke CE, Garlet TP, et al. Evidence supporting a gerin in odontogenic cysts and tumors. J Oral Maxillofac Surg.
protective role for Th9 and Th22 cytokines in human and experi- 2013;71:1886–1892.
mental periapical lesions. J Endod. 2013;39:83–87. [61] Silva TA, Batista AC, Mendonça EF, et al. Comparative expression
[40] Garlet GP, Horwat R, Ray HLJ, et al. Expression analysis of wound of RANK, RANKL and OPG in keratocystic odontogenic tumors,
healing genes in human periapical granulomas of progressive ameloblastomas and dentigerous cysts. Oral Surg Oral Med Oral
and stable nature. J Endod. 2012;38:185–190. Pathol Oral Radiol Endod. 2008;105:333–341.
[41] Oseko F, Yamamoto T, Akamatsu Y, et al. IL-17 is involved in [62] Carneiro E, Parolin AB, Wichnieski C, et al. Expression levels of
bone resorption in mouse periapical lesions. Microbiol Immunol. the receptor activator of NF-jB ligand and osteoprotegerin and
2009;53:287–294. the number of gram-negative bacteria in symptomatic
[42] Carrillo C, ~arrocha
Pen M, ~arrocha
Pen M, et al. and asymptomatic periapical lesions. Arch Oral Biol. 2017;73:
Immunohistochemical study of Langerhans cells in periapical 166–171.
lesions; correla-tion with inflammatory cell infiltration and epithe- [63] Salinas-Mun ~oz M, Garrido-Flores M, Baeza M, et al. Bone resorp-
lial cell proliferation. Med Oral Patol Oral Cir Bucal. 2010;15: tive activity in symptomatic and asymptomatic apical lesions of
335–339. endodontic origin. Clin Oral Invest. 2017;21:2613–2618.
[43] Naufel AO, Aguiar MCF, Madeira FM, et al. Treg and Th17 cells in [64] Santos SCLT, Couto LA, Fonseca JM, et al. Participation of osteo-
inflammatory periapical disease: a systematic review. Braz Oral clastogenic factors in immunopathogenesis of human chronic
Res. 2017;18:1–14. periapical lesions. J Oral Pathol Med. 2017;46:846–852.
[44] Suzuki T, Kumamoto H, Ooya K, et al. Immunohistochemical ana- [65] Silva I, Branco JC. Rank/Rankl/opg: literature review. Acta
lysis of CD1a-labeled Langerhans cells in human dental periapical Reumatol Port. 2011;36:209–218.
inflammatory lesions – correlation with inflammatory cells and [66] Francisconi CF, Vieira AE, Azevedo MCS, et al. RANKL triggers
epithelial cells. Oral Dis. 2001;7:336–343. treg-mediated immunoregulation in inflammatory osteolysis.
[45] Santos LCS, Ramos EAG, Gurgel CAS, et al. Immunohistochemical J Dent Res. 2018;97:917–927.
detection of Langerhans cells in dental granulomas and radicular [67] Armada L, Marotta PS, Pires FR, et al. Expression and distribution
cysts. J Mol Hist. 2007;38:201–205. of receptor activator of nuclear factor Kappa B, receptor activator
[46] Jakovljevic A, Knezevic A, Karalic D, et al. Pro-inflammatory cyto- of nuclear Factor Kappa B ligand, and osteoprotegerin in perira-
kine levels in human apical periodontitis: Correlation with clinical dicular cysts. J Endod. 2015;41:1281–1287.
and histological findings. Aust Endod J. 2015;41:72–77. [68] Fan R, Sun B, Zhang CF, et al. Receptor activator of nuclear factor
[47] Zhang J, Huang X, Lu B, et al. Can apical periodontitis affect kappa B ligand and osteoprotegerin expression in chronic apical
serum levels of CRP, IL-2, and IL-6 as well as induce pathological periodontitis: possible association with inflammatory cells. Chin
changes in remote organs?. Clin Oral Invest. 2016;20:1617–1624. Med J (Engl). 2011;124:2162–2166.
[48] Vidal F, Fontes TV, Marques TVF, et al. Association between apical [69] Caviedes-Bucheli J, Mun ~oz HR, Azuero-Holguın MM, et al.
periodontitis lesions and plasmatic levels of C-reactive protein, Neuropeptides in dental pulp: the silent protagonists. J Endod.
interleukin 6 and fibronegen in hypertensive patients. Int Endod J. 2008;34:773–788.
2016;49:1107–1115. [70] Lundy FT, Linden GJ. Neuropeptides and neurogenic mechanisms
[49] Garrido M, Dezerega A, Bordagaray MJ, et al. C-reactive protein in oral and periodontal inflammation. Crit Rev Oral Biol Med.
expression is up-regulated in apical lesions of endodontic origin 2004;15:82–98.
in association with interleukin-6. J Endod. 2015;41:464–469. [71] Lundberg P, Lie A, Bjurholm A, et al. Vasoactive intestinal peptide
[50] Jurisic V, Terzic T, Colic S, et al. The concentration of TNF-alpha regulates osteoclast activity via specific binding sites on both
correlate with number of inflammatory cells and degree of vascu- osteoclasts and osteoblasts. Bone 2000;27:803–810.
larization in radicular cysts. Oral Dis. 2008;14:600–605. [72] Tuncer LI, Alaçam T, Oral B. (2004) Substance P expression is ele-
[51] Cavalla F, Reyes M, Vernal R, et al. High Levels of CXC Ligand 12/ vated in inflamed human periradicular tissue. J Endod. 2004;30:
Stromal cell-derived factor 1 in apical lesions of endodontic ori- 329–332.
gin associated with mast cell infiltration. J Endod. 2013;39: [73] Irie K, Hara-Irie F, Ozawa H, et al. Calcitonin gene-related peptide
1234–1239. (CGRP)-containing nerve fibers in bone tissue and their involve-
[52] Boyle WJ, Simonet WS, Lacey DL. Osteoclast differentiation and ment in bone remodeling. Microsc Res Tech. 2002;58:85–90.
activation. Nature. 2003;423:337–342. [74] Konttinen YT, Imai S, Suda A. Neuropeptides and the puzzle of
[53] Khosla S. Minireview: the OPG/RANKL/RANK system. bone remodeling. State of the art. Acta Orthop Scand. 1996;67:
Endocrinology. 2001;142:5050–5055. 632–639.
[54] Wada T, Nakashima T, Hiroshi N, et al. RANKL-RANK signaling in [75] Goodman SC, Letra A, Dorn S, et al. Expression of heat shock pro-
osteoclastogenesis and bone disease. Trends Mol Med. 2006;12: teins in periapical granulomas. J Endod. 2014;40:830–836.
17–25. [76] Bambirra WJr, Maciel KF, Thebit MM, et al. Assessment of apical
[55] Boyce BF, Xing L. Functions of RANKL/RANK/OPG in bone model- expression of alpha-2 integrin, heat shock protein, and proinflam-
ing and remodeling. Arch Biochem Biophys. 2008;473:139–146. matory and immunoregulatory cytokines in response to endo-
[56] Belibasakis GN, Rechenberg DK, Zehnder M. The receptor activa- dontic infection. J Endod. 2015;41:1085–1090.
tor of NF-jB ligand-osteoprotegerin system in pulpal and periapi- [77] Silva MJ, Sousa LM, Lara VP, et al. The role of iNOS and PHOX in
cal disease. Int Endod J. 2013;46:99–111. periapical bone resorption. J Dent Res. 2011;90:495–500.
[57] Moraes M, Lucena HF, Azevedo PR, et al. Comparative immuno- [78] Cintra LTA, Samuel RO, Azuma MM, et al. Multiple apical peri-
histochemical expression of RANK, RANKL and OPG in radicular odontitis influences serum levels of cytokines and nitric oxide.
and dentigerous cysts. Arch Oral Biol. 2011;56:1256–1263. J Endod. 2016;42:747–751.
180 P. H. BRAZ-SILVA ET AL.

[79] Yue J, Wang P, Hong Q, et al. MicroRNA-335-5p plays dual roles [84] Dezerega A, Madrid S, Mundi V, et al. Pro-oxidant status and
in periapical lesions by complex regulation pathways. J Endod. matrix metalloproteinases in apical lesions and gingival crevicular
2017;43:1323–1328. fluid as potential biomarkers for asymptomatic apical periodon-
[80] Yue J, Song D, Lu W, et al. Expression profiles of inflammation- titis and endodontic treatment response. J Inflamm. 2012;9:8.
associated microRNAs in periapical lesions and human periodon- [85] Jakovljevic A, Andric M, Nikolic N, et al. Levels of oxidative stress
tal ligament fibroblasts inflammation. J Endod. 2016;42: biomarkers and bone resorption regulators in apical periodontitis
1773–1778. lesions infected by Epstein-Barr virus. Int Endod J. 2018;51:
[81] Chan LT, Zhong S, Naqvi AR, et al. MicroRNAs: new insights into 593–604.
the pathogenesis of endodontic periapical disease. J Endod. [86] Vengerfeldt V, M€andar R, Saag M, et al. Oxidative stress in
2013;39:1498–1503. patients with endodontic pathologies. J Pain Res. 2017;10:
[82] Setien-Olarra A, Marichalar-Mendia X, Bediaga NG, et al. 2031–2040.
MicroRNAs expression profile in solid and unicystic ameloblasto- [87] Inchingolo F, Marrelli M, Annibali S, et al. Influence of endodontic
mas. PLoS One. 2017;2012:e0186841. treatment on systemic oxidative stress. Int J Med Sci. 2014;11:
[83] Hernandez-Rıos P, Pussinen PJ, Vernal R, et al. Oxidative stress in 1–6.
the local and systemic events of apical periodontitis. Front
Physiol. 2017;8:869.

You might also like