Sie sind auf Seite 1von 27

Inflammatory Pathways of Bone

Resorption in Periodontitis
6
Franco Cavalla, Claudia C. Biguetti,
Thiago P. Garlet, Ana Paula F. Trombone,
and Gustavo P. Garlet

6.1 Introduction determine the best course of action and to pro-


vide the best possible care. In spite of the aston-
The most common periodontal diseases in ishing amount of time and resources devoted to
humans are periodontitis and gingivitis. These unveil the intricate details of periodontitis’ patho-
diseases share dental biofilm as their etiologic genesis invested in the last four decades, we are
factor and periodontal inflammation as their main still far from a comprehensive explanatory model,
feature. Although the diagnosis, treatment, and and, more importantly, of new therapeutic tools
prognosis of the vast majority of the periodontitis to take better care of the most susceptible patients.
cases pose no special challenges to the trained That said, new developments in the field of osteo-
periodontist, there are extreme clinical pheno- immunology that will be discussed later in this
types at both ends of the susceptibility chain that chapter provide us with an appealing pathway to
are disturbingly difficult to identify with the clas- follow in our search for novel diagnostic tools,
sical periodontal diagnose tools. When a patient therapies, and clinical interventions.
presents a differential clinical phenotype, partic-
ularly of extreme susceptibility to alveolar bone
destruction, the periodontist often found himself/ 6.2  eneralities of Inflammation
G
herself lacking of the conceptual framework to
in Periodontitis
Pathogenesis
F. Cavalla
Periodontitis’ hallmark feature is the inflamma-
OSTEOimmunolgy Lab, Department of Biological
Sciences, School of Dentistry of Bauru, University of tory resorption of tooth-supporting alveolar bone
Sao Paulo (FOB/USP), Bauru, Brazil due to uncontrolled host response to periodontal
Departamento de Odontología Conservadora, infection. Even though the periodontal infection
Facultad de Odontología de la, Universidad de Chile, is essential to trigger the host’s immune and
Santiago, Chile inflammatory response, the destructive events
C.C. Biguetti • T.P. Garlet • G.P. Garlet (*) that lead to the irreversible disease phenotype of
OSTEOimmunolgy Lab, Department of Biological periodontitis are the result of the uncoupling of
Sciences, School of Dentistry of Bauru, University of
the soft and mineralized tissue turnover mecha-
Sao Paulo (FOB/USP), Bauru, Brazil
e-mail: garletgp@usp.br nisms caused by the persistence of a chronic and
exacerbated inflammatory immune response.
A.P.F. Trombone
Universidade do Sagrado Coração (USC), Although the primary role of the immune sys-
Bauru, Brazil tem is to provide the defense of the host against

© Springer International Publishing AG 2018 59


N. Bostanci, G.N. Belibasakis (eds.), Pathogenesis of Periodontal Diseases,
DOI 10.1007/978-3-319-53737-5_6
60 F. Cavalla et al.

potentially hazardous microorganisms, the per- but we are still only scraping the surface of the
sistence of immune responses due to their inabil- pathogenic mechanisms that tilt the balance from
ity to eliminate the microbial antigens causes the health to disease in the periodontal environment.
alterations in the tissue’s metabolism that eventu- From a clinician’s perspective, the understand-
ally results in its irreversible destruction. The ing of the pathogenic mechanisms of periodontal
anatomic particularities of periodontium and the disease is an invaluable asset in the establishment
capacity of periodontal bacteria to form a biofilm of a successful treatment plan, general and indi-
structure favor the establishment of a long-last- vidual teeth prognosis, and follow-­up strategies.
ing and non-resolving chronic inflammation that In addition, the knowledge of the general mecha-
acquires a destructive nature. The bacterial bio- nisms of periodontitis’ pathogenesis helps the
film adheres to the tooth surface and confers a clinician to identify putative susceptible subjects
series of advantages to the microbes, including and implement individualized therapeutic mea-
the isolation and protection from host defensive sures to better their long-­term prognosis. Least,
mechanism, ultimately impairing the eradication but not less important, is the perspective of the
of the infectious focus. development of new therapeutic approaches using
It is noteworthy that in periodontal health the the knowledge gained from the applied research,
immune and inflammatory response is still pres- which include the appealing possibility of using
ent. While the exact nature of the host response immune modulatory drugs. These drugs will be
associated with periodontal health remains to be specifically targeted to the periodontal tissues and
established, such state is considered an equilib- aimed to abolish the deleterious effects of inflam-
rium state where a low intensity response is mation in the alveolar bone without dampening
enough to limit the invasion and multiplication of the ability of the host to deal with potentially
periodontal microorganisms without inflicting hazardous periodontal microorganisms. As pre-
damage to the host tissues as a collateral damage. viously stated, although periodontitis is an infec-
This balance is finely tuned and dependent on a tion, the pathologic tissue changes characteristic
series of active modulatory processes, which can of the disease (particularly bone resorption) are a
be tilted in each direction (exacerbated suppres- consequence of the sustained inflammation of the
sion or activation) by intrinsic or environmental periodontal tissues.
factors. The active suppression of the immune Inflammation can be defined as a protective
response, without complete abolition of its infec- response intended to eliminate injurious stimuli
tion control mechanisms, is supposed to be the that causes tissue damage, as well as removing
cornerstone in the maintenance of stable healthy the injured tissue resulting from the original
periodontal tissues. insult. Inflammation can be triggered by physical
In this scenario, this chapter focuses in the role or chemical trauma or by foreign bodies, includ-
of the inflammatory and immunological events that ing microbes. Even though inflammation is
lead to the pathologic destruction of the alveolar intended to eliminate harmful stimuli, the inflam-
bone, and consequently of the periodontal attach- matory reaction has the potential to cause wide-
ment during the progression of periodontitis. spread tissue damage, since the same mechanisms
intended to kill infecting microbes and clear the
tissue from injured and dead cells have the poten-
6.2.1 F
 rom Health to Inflammatory tial to damage normal tissue.
Immune Response: How It is fundamental to understand that the distinc-
Homeostasis Evolves to tion between inflammation and immune response
Disease is only didactic and that both processes not only
occur simultaneously, but also are functionally
After more than four decades of incessant research integrated. Indeed, the main purpose of inflamma-
in the immune aspects of the pathogenesis of tion is to facilitate the access of immune cells and
periodontitis many lessons have been learned, defense molecules from the circulation to the
6  Inflammatory Pathways of Bone Resorption in Periodontitis 61

damaged or infected tissues. Thus, most inflam- Conceptually, inflammation is categorized in


matory events are related to vascular and perivas- two types: acute and chronic. Acute inflammation
cular events, including increased blood flow, is by definition a short duration response, ranging
alterations on the vasculature to facilitate immune from minutes to a few days, and it is character-
cells transmigration, and increased extracellular ized by profuse exudate and the predominance of
matrix metabolism. These molecular and cellular neutrophilic leukocyte infiltration. With the per-
processes translate into the clinical signs of sistence of the inflammatory stimuli, acute
inflammation (a.k.a. “cardinal signs”), as classi- inflammation evolves to chronic inflammation is
cally described by Rudolph Carl Virchow in the a long-lasting response, ranging from days to
late nineteenth century: redness, heat, swelling, years, characterized by the tissue infiltration of
pain, and loss of function. In the particular case of mixed populations of leukocytes and monocytes/
periodontal diseases, these first inflammatory and macrophages following specific chemotactic gra-
immune phenomena translate in the clinical entity dients, and tissue degeneration including vascu-
known as gingivitis. lar proliferation, parenchymal involution, and
In the sequence, we will approach inflamma- fibrosis [7]. The events of acute and chronic
tion and immune response as intermingled and inflammation are analog and complementary to
mutually dependent occurrences in order to bet- events of innate and adaptive response. The first
ter picture the actual cellular and molecular phe- vascular and molecular occurrences of inflamma-
nomena that lie beneath the clinical signs and tion favor the migration of the cell populations’
symptoms of periodontitis, which ultimately characteristic of the innate immunity, namely
explain the pathologic irreversible destruction of neutrophils and macrophages, which possess
alveolar bone. We will focus in bone destruction, very effective but unspecific microbe clearance
since it is the defining occurrence of periodonti- machineries. When innate immune responses
tis, even though the same inflammatory immune cannot clear the invading microorganisms, the
events depicted in this chapter could be consid- inflammation becomes chronic and the character-
ered as responsible for the destruction of the istic effector cells of the adaptive immune
remaining of the tooth-support apparatus. response invade the tissue, predominantly acti-
In normal healthy conditions, the gingival tis- vated lymphocytes with highly selective and spe-
sues are capable of coping with the presence of cific microbe killing capabilities [2, 8]. As in any
bacteria due to various innate immune defense biological process, these rigid definitions do not
mechanisms, among them: the flushing action of capture the changing dynamics of inflammation
gingival crevicular fluid and saliva, the rapid epi- and immune response, where long-lasting chronic
thelial turnover, the secretion of immune active inflammatory processes are periodically broken
peptides, a permanent influx of innate immune up by acute inflammatory bursts [9, 10].
response cells to the periodontal tissue and the The inflammatory immune response is trig-
transmigration of neutrophils into the sulcus, and gered by the interaction of resident cells with the
the action of saliva agglutinins and antibodies, bacterial biofilm attached to the tooth surface. The
which are supposed to limit the potential of the particularities of the periodontal anatomy and
bacterial biofilm to grow and invade [1–4]. Indeed, periodontal tissue architecture are intimately asso-
in a distinctive fashion in comparison with other ciated with the natural course and p­ athophysiology
body tissues, in the healthy periodontium exist a of the inflammatory processes that leads to peri-
constant subclinical inflammation. When the bal- odontitis [5, 11]. Bacterial biofilm attaches to the
ance between the infection control mechanism and tooth surface, making impossible for the immune
the subgingival biofilm is lost, the mandatory first system to eradicate the infecting microorganisms
step into the pathologic chain of events leading to efficiently. The attached biofilm acts as a perma-
periodontitis is gingival inflammation, an acute nent reservoir of microorganism and their prod-
inflammatory response of the gingiva to the micro- ucts, perpetuating the insult to the periodontal
bial insult that is clinically evident [5, 6]. tissues [12].
62 F. Cavalla et al.

The junctional epithelium is the first peri- family (TLR), composed of at least nine isoforms
odontal structure to face the bacterial challenge. named TLR1 to TLR9. The signaling pathway is
This is a highly specialized and unique tissue, initiated by the binding of the pathogen-derived
characterized by the capability to attach directly ligands to membrane-bounded TLRs, leading to
to the mineralized surface of the tooth by the for- the dimerization of the receptor. The dimeriza-
mation of an extremely organized structure tion of the TLRs triggers the recruitment of vari-
known as the internal base membrane. The junc- ous protein kinases in the cytoplasmic end of the
tional epithelium is more permeable than most receptors, ultimately causing the activation of
epitheliums, mainly because of the incomplete proinflammatory transcription factors (such as
maturation of its supra basal layer, by its unusu- NFκB and AP-1) [22, 23].
ally high proliferation rate, by the relatively Following the initial stimuli, classic inflam-
scarce presence of desmosome junctions between matory pathways are deflagrated, where the
cells, and by the absence of keratohyalin [13, 14]. cyclooxygenase pathway oxygenates arachidonic
After surpassing the epithelial barrier, infecting acid producing a variety of proinflammatory
microorganisms gain access to the subjacent gin- molecules, such as prostaglandin E-2 (PGE-2),
gival connective tissue, being gingival fibroblasts prostacyclin, and leukotriene A-4. The enzyme
the predominant cell type in this compartment. cyclooxygenase 1 (COX-1) is constitutively
Despite the fact that fibroblast are not profes- expressed, while COX-2 is induced under proin-
sional immune cells and have a limited output of flammatory conditions and responsible for the
signaling molecules available, such cell type can amplification of the inflammation [7, 24]. Several
be also active in the triggering of initial inflam- in vivo studies have demonstrated that the inhibi-
matory events [15]. tion of COX-2 activity significantly reduces the
Extensive evidence indicates that the first alveolar bone loss in experimental periodontitis
immune modulatory events in this process are [25, 26]. In parallel with the activation of classic
orchestrated by the keratinocytes of the junc- mediators of the vascular events of inflammation,
tional epithelia and fibroblast of the periodontal TLR recognition of microbial ligands results in
connective tissue [16–20]. These cells are capa- the secretion of important molecular mediators of
ble of “sensing” the environment and differen- inflammation, including inflammatory cytokines
tially react to diverse bacterial antigens and other (such as TNFα and IL-1β) and chemokines (such
signals, responding with the secretion of media- as CXCL8/IL-8, CCL2, CCL3, and CCL5),
tors that will mediate the vascular and cellular which along the mediators that target the blood
events of inflammation. Indeed, the resident peri- vessels, will orchestrate the inflammatory cell
odontal cells (epithelial cells and gingival fibro- influx into the inflaming tissue [1, 27].
blast) directly interact with the microbes or its At this step, to make room for the profuse
products produce and secrete molecular signals inflammatory cell infiltration the perivascular
to trigger inflammation and chemoattract immune extracellular matrix is degraded by enzymes such
cells [14, 15]. The host cells recognize the as matrix metalloproteinases (MMPs), which also
microbes by the interaction of pathogen-­ will degrade the extracellular matrix along the
associated molecular patterns (PAMPs) with way of the cells from the vessel until the inflam-
PAMP-receptors constitutively expressed in the matory focus, being this way determined by gra-
cell membrane of most cells. Those PAMPs are dients of chemoattractant molecules [4, 28].
signature molecules preferentially associated In this environment, it is mandatory to con-
with pathogens and absent from host’s cells, such sider that gingival fibroblasts are responsible
as lipid polysaccharide (LPS), a main component for the periodontal tissue turnover, doubling as
of Gram-negative cell wall, and lipoteichoic acid, the main source of collagen fibers and of MMPs
a major constituent of the cell wall of Gram-­ within the connective tissue. However, under
positive bacteria [21]. The archetypal membrane-­ the influence of the copious inflammatory sig-
bound PAMP-receptors are the Toll-like receptor nals, gingival fibroblast and periodontal liga-
6  Inflammatory Pathways of Bone Resorption in Periodontitis 63

ment fibroblasts are directly responsible for the activated by the interaction of their own TLRs
destruction and disorganization of the fibrous with PAMPs. After their activation, the neutro-
component of the extracellular matrix of peri- phils produce and secrete molecular mediators to
odontal tissue by increasing the local production amplify the inflammation; also, their phagocytic
and activity of MMPs [29]. While the extracellu- activity is enhanced, as well as the production of
lar matrix degradation has a physiological value, reactive oxygen species (ROS) used to kill the
since it is necessary to facilitate the transmigra- phagocyted microorganism. These ROS affect
tion and permanence of immune cells within the the oxidative status of the periodontal tissue,
tissue during the immune response, the problem activating and sustaining a pervasive inflamma-
arises when the inflammatory/immune response tory response that amplifies the initial signaling,
turn out to be indefinite and the tissue homeo- and when uncontrolled promotes the spreading
stasis becomes permanently affected, leading to of the inflammation to the surrounding tissues
the pathological irreversible destruction of teeth- [15, 35, 36].
supportive tissues. While neutrophils exert a very important anti-­
At this stage, the first clinical signs of gingi- infective role in the periodontal environment, it is
vitis become evident. Namely, redness, swelling also important to consider that most of the peri-
and loss of texture of the free gingiva, provoked odontal microorganisms have evolved complex
bleeding (at the clinical examination with a blunt and elegant strategies of immune evasion and
instrument or during normal oral hygiene pro- have the capacity to use the immune/inflamma-
cedures), and eventual suppuration. When the tory response to their own benefit, modulating the
inflammation persists, the disease progresses into periodontal microenvironment to adjust it to their
periodontitis, characterized by the widespread specific metabolic needs [37]. For example, the
destruction of the extracellular matrix and the recognized periodontopathogen Porphyromonas
collagenous fibers that compose the connec- gingivalis is capable of downregulating the initial
tive attachment apparatus, the apical migration immune response of periodontium resident cells,
of the junctional epithelium, and the pathologi- completely abolishing the secretion of the neutro-
cal resorption of the alveolar bone and radicu- phil-attractant chemokine IL-8/CXCL8 by a gin-
lar cementum. In the periodontal examination, gipain-dependent mechanism [19, 38], facilitating
these histopathological changes are evidenced as its survival and growth within the periodontal
increased probing depth usually accompanied by connective tissue. Interestingly, despite the initial
all the clinical signs of gingivitis [30]. dampening of inflammatory cell migration over
time will ultimately result in an exacerbated, but
inefficient in microbial control terms, host
6.2.2 T
 he Parallels Between Acute response. Indeed, as previously mentioned, in the
Inflammation and Innate development of periodontitis, infecting microor-
Immune Response ganism resists eradication in part due to the par-
ticular anatomy of the periodontal sulcus/pocket
At the early host response phase, the predomi- and in part due to their proprietary virulence and
nant immune cell type in the periodontium is the evasion ­mechanism along the additional protec-
polymorph nuclear neutrophil [6, 31]. Even in tion conferred by the biofilm structure, eliciting a
healthy conditions, a stable influx of neutrophils sustained chronic inflammatory response.
(3 × 105 cells/min) permeates the connective tis- At this point, neutrophils may have or have
sue in transit to the sulcus though the junctional not succeeded in eliminating the agent that elic-
epithelium, attracted by a constant gradient of ited the inflammatory response. If not, additional
IL-8/CXCL8 permanently secreted from kerati- cell types with different antimicrobial strategies
nocytes and gingival fibroblast in response to the and weapons may be able to achieve the task, but
recognition of PAMPs by TLRs [32–34]. Once even if neutrophils are successful in eliminating
inside the tissue, neutrophil leukocytes become infecting agents, additional cell recruitment is
64 F. Cavalla et al.

required to repair the damage (even if minimal) In summary, the acute inflammatory response
inflicted at the response site. To achieve this in the early stages of periodontitis development
objective, a special polarized subpopulation of begins when bacteria and their products gain
macrophages with regulatory and reparative access to the gingival connective tissue through-
functions may be recruited to the damaged tissue. out the junctional epithelium, whose loose inter-
This distinctive subpopulation of macrophages cellular junctions do not provide an impermeable
possesses the capacity to potentiate the repair, physical barrier. Proinflammatory molecular sig-
and in some instances, the fibrosis of the tissue as nals emanated from the epithelial and connective
required [39]. tissue trigger the first inflammatory events,
Additionally, monocytes also play a relevant increasing the blood flow and permeability of the
role during the early stages of the acute inflam- subepithelial gingival plexus and recruiting large
matory process and initial innate immune amounts of leukocytes to the site, particularly
response, and readily infiltrate the periodontal neutrophils and macrophages.
tissue in the first hours post infection [12, 40]. In the coming sections we will describe the
Monocytes are attracted from the circulation to chronic inflammatory events that later will be
the periodontal tissues following gradients of responsible for the bulk of the tissue destruction
chemokines, such as monocyte chemoattractant in periodontitis. Briefly, we can summarize the
protein 1 (MCP-1, a.k.a. CCL2), which is the process as a continued response driven by the
master regulator of monocyte/macrophage mobi- impossibility of eliminating the microbes exclu-
lization [41–43]. Some indirect evidence links sively with the recruitment of neutrophils and
the infiltration of macrophages into the periodon- macrophages. In turn, other leukocyte subsets are
tium with the severity of the periodontitis, since recruited and join the inflammatory infiltrate, and
MCP-1/CCL2 levels appear augmented in the the host response acquires a chronic nature that
gingival crevicular fluid of periodontitis patients will result in the dampening of the normal
and its levels seem to correlate with the severity homeostatic balance of the periodontal tissues.
of the disease [44].
The macrophages are able to phagocyte the
invading microorganism into phagocytic vesicles 6.2.3 Osteoimmunology
(a.k.a. phagosomes), but require additional cyto- and the Molecular Connection
kine signaling to promote the fusion of phago-
somes and lysosomes, where the microbicidal Indeed, when the response becomes chronic,
mechanisms take place to kill effectively the adaptive immune cells invade the tissue and the
ingested microbes. The principal molecular inflammatory reaction becomes firmly estab-
mechanisms of microbe killing and digestion are lished, flooding the periodontium with additional
the conversion of molecular oxygen into reactive bioactive proinflammatory molecular signals
oxygen species (ROS), the production of highly (cytokines, chemokines, enzymes, ROS, bacte-
reactive nitrogen species, such as nitric oxide rial products and metabolites, etc.) [1, 2, 4, 28].
(NO), and the action of several proteolytic The accumulation of these molecular signals in
enzymes [45, 46]. It is noteworthy that all these the tissue facilitates the spreading of the inflam-
microbicidal mechanisms, when exacerbated and mation to the underlying bone and tamper with
uncontrolled, are partially responsible for the the bone homeostasis signaling system, tilting
amplification of the inflammatory response and the balance of bone metabolism favoring resorp-
the degradation of the host’s tissues. Indeed, acti- tion over formation. More than four decades ago,
vated macrophages are an important cellular Page and Schroeder postulated that the presence
source of MMPs and greatly contribute to the of bacterial plaque closer than 2.5 mm from the
intensification of the degradation of the collage- alveolar bone could trigger its inflammatory
nous matrix in the connective periodontal tissue resorption [31, 49]. Nowadays we know that the
[47, 48]. pathological mechanism underlying this observa-
6  Inflammatory Pathways of Bone Resorption in Periodontitis 65

tion is the disruption of the balance of bone through calcium-dependent activation of the tran-
metabolism by inflammatory and immune molec- scription of NFATc1 gene [55]. The membrane-
ular mediators, which mark the transition from bound form is characteristically expressed in the
gingivitis to periodontitis. surface of osteoblasts (a.k.a. RANKL isoform 1
The understanding of the molecular basis of and 2) and the soluble form (a.k.a. RANKL iso-
the interplay between inflammatory immune form 3) is the secreted product of various cell
responses and the bone tissue is the keystone of types, including B-cells and T-cells [56, 57].
“osteoimmunology,” a new and evolving field RANKL is the master activator of osteoclasts
that studies the shared components and mecha- and the molecular signal directly responsible
nisms between the immune and bone systems. To for bone resorption. RANKL interacts with its
understand such connection, we must remember cognate receptor RANK in the surface of osteo-
that bone is a highly specialized mineralized con- clast and osteoclasts’ precursors, triggering their
nective tissue, characterized by constant renewal recruitment to the bone surface, cell fusion, and
dependent on the coupling of bone formative and activation. The secreted form of RANKL is the
resorptive processes. This dynamic behavior of molecular signal that couples immune response
bone enhances its adaptive capacity to functional and bone metabolism. Numerous animal model
demands and increases its healing potential after experiments have demonstrated that alveolar
an injury. The balance between bone resorption bone resorption could be prevented by the selec-
and apposition is governed by a unified molecu- tive inhibition of the RANKL/RANK axis [58,
lar signaling system and is dependent on the 59]. Osteoprotegerin (OPG) is a soluble protein
effector functions of specialized bone cells. upregulated in inflammatory conditions, with the
The modulatory processes leading to bone capacity to block RANKL’s biological functions
resorption in periodontitis are very complex and by competitive inhibition, acting as a decoy recep-
intricate, and will be described along this entire tor, limiting the availability of RANKL able to
chapter, including the interaction of different spe- bind to RANK. The quotient or ratio of RANKL
cies of bacteria and their subproducts with ele- to OPG determines if at any given moment the
ments of the innate and adaptive immune systems conditions are favorable for bone apposition or
[50–52]. Fortunately, despite the astonishing bone resorption. A high ratio of RANKL/OPG
complexity of the input signals, the bone has a creates the conditions favorable to bone resorp-
very straightforward transduction system and a tion, while a low RANKL/OPG ratio favors bone
limited number of molecular signals and cells are apposition [60, 61]. During bacteria-induced
directly involved in controlling the balance inflammation in experimental periodontitis, it has
between bone apposition and bone resorption. been demonstrated a net increase in the RANKL/
This is a common finding among complex bio- OPG ratio, leading to osteoclast genesis and bone
logical systems, which are capable of “sensing” a resorption [62]. Analog evidence is also available
vast variety of inputs, but where the effector in human periodontitis [28], and in the closely
mechanisms are governed by a much more lim- related condition of pathologic bone resorption
ited number of “key effector signals.” In the spe- of periapical bone as a consequence of end-
cific case of alveolar bone, the system that odontic infection [63, 64]. Indeed, the key event
controls the bone metabolism balance comprises in the inflammatory alveolar bone resorption in
the RANKL/OPG/RANK triad, secreted and rec- periodontitis is the manifold increase in the tis-
ognized by the specialized bone effector cells sue levels of RANKL unaccompanied by an
osteoblasts and osteoclasts [53, 54]. equivalent increase in OPG levels. The resultant
RANKL (receptor activator of nuclear factor augmented RANKL/OPG ratio drives the recruit-
κB ligand) is a cytokine, member of the TNF fam- ment of osteoclast’s monocyte precursors, their
ily that can be membrane bounded or secreted, fusion, and later activation. The uncoupling of
and stimulates osteoclasts’ differentiation, cell bone metabolism due to the imbalance between
fusion, and activation leading to bone resorption bone apposition and resorption is the result of the
66 F. Cavalla et al.

uncontrolled interplay of the immune system and adaptive immunity cells. Indeed, after the initial
the bone metabolism throughout their common acute inflammation and innate immune response
molecular mediators. had taken place, the selective migration of spe-
Recently, it was reported that RANKL levels cific T lymphocyte subsets drives the transition to
in gingival crevicular were increased fourfold in the adaptive immune response [1, 4]. These two
chronic periodontitis patients compared to processes are analogous and in a certain way cor-
healthy controls, while OPG levels were equiva- respondent to the acute and chronic phases of the
lent in both groups, causing an increased inflammatory response.
RANKL/OPG ratio in the patient’s group. In this second stage of the immune response,
Interestingly, RANKL/OPG ratios levels were T CD4+ lymphocytes (a.k.a. T helper or Th) play
unaffected by the periodontal treatment and a critical role in orchestrating the host’s response.
remained augmented for at least 6 weeks post- T helper lymphocytes secrete bioactive signaling
treatment, despite the notorious clinical improve- molecules, namely cytokines, as their main effec-
ment and normalization of inflammatory tor mechanism, fine-tuning almost every aspect
parameters of the subjects [65]. This could be the of the inflammatory/immune response.
reflection of a long-lasting unbalance of bone Depending on a series of environmental and
metabolism, spanning beyond the resolution of host’s intrinsic factors, Th lymphocytes could
the inflammation in the immediate posttreatment differentiate into distinct subtypes or lineages,
period. Alternatively, this could be the result of each one with a characteristic subset of cytokines
an inherent predisposition to increased secretion in its secretion pattern. The differential lineage
of RANKL, which could be the underlying commitment depends of the predominant cyto-
molecular basis for increased susceptibility to kine present in the environment during the pro-
bone resorption. cess of antigen presentation by antigen-presenting
cells (APC) in the regional lymph node. During
this process, APC capture antigens in the periph-
6.2.4 Chronic Inflammation eral tissues and migrate to the regional lymph
and Adaptive Immune node, where they encounter a naïve T cell with a
Response in Periodontitis specific receptor for the antigen. After the spe-
cific interaction of APC-T CD4+ cell, the naïve
The activation of adaptive immunity has a great lymphocyte becomes activated and committed,
influence in the bone loss associated with peri- suffers clonal expansion, and massively migrates
odontitis, since numerous evidence points to B into the peripheral tissue to perform their effector
and T lymphocytes as the main cellular sources functions. It is during this stage that the different
of soluble RANKL during periodontal inflamma- lineages of Th lymphocytes emerge and differen-
tion [66]. Experimental evidence from SCID tiate, dictating the curse of the following steps of
mice (Severe Combined Immune Deficient, lack- the disease [1, 10].
ing both T and B lymphocytes), demonstrated the The relative predominance of a subpopulation
importance of adaptive immunity effector cells in of Th lymphocytes over the rest determines that
bone loss in periodontitis. When SCID mice the immune response variates from a controlled
where challenged with Porphyromonas gingiva- self-contained process with low potential to
lis, they demonstrated significantly lesser bone destroy the periodontal tissues to an exaggerated
resorption than the control wild-type mice [67]. reaction with high destructive potential. The
As previously mentioned, with the impossibil- knowledge of the existence and regulatory func-
ity of clearing the insulting microbes by means of tions of the different Th subpopulations has been
the recruitment of neutrophils and macrophages, one of the most dramatic revolutions in the
other leukocyte subsets are subsequently enrolled immunology field of the past three decades and
to join the inflammatory infiltrate, and the host has affected profoundly our understanding of the
response acquires a chronic nature and mobilizes immune processes underling the clinical course
6  Inflammatory Pathways of Bone Resorption in Periodontitis 67

of periodontal disease beyond the classical pro- sion potential of periodontal pathogens.
versus anti-inflammatory perspective [1, 8, 68]. Experimental evidence in IFN-γ deficient mice
In the late eighties, the first two Th lympho- demonstrates that although they develop a less
cyte subtypes where described first in mice and severe phenotype of alveolar bone destruction
then in humans and named Th1 and Th2 [69, 70]. following the infection with Aggregatibacter
Th1 cells were characterized as responsible to actinomycetemcomitans, they are also more
mediate the immune responses against intracellu- prone to suffer widespread infection, with lethal
lar pathogens. Naïve Th cells committed to Th1 consequences in some cases. The IFN-γ deficient
lineage when the antigen presentation occurred in mice demonstrated reduced levels of many
an IL-12 enriched environment, and the stabiliza- inflammatory cytokines and chemokines, as well
tion of the Th1 phenotype was dependent on the as significantly reduced numbers of infiltrating
transcription of the key transcription factor T-bet macrophage and markers of macrophage activa-
[71]. The characteristic cytokine product of Th1 tion in the periodontal tissue [78].
committed lymphocytes is IFN-γ, which is the The Th1 lineage is also responsible for the
cytokine responsible for the classical pathway of secretion of various cytokines that support, main-
macrophage’s activation, and fundamentally tain, and amplify the inflammatory response,
involved in the stimulation of the eradication of directly or indirectly favoring the recruitment
phagocyted pathogens [72]. Conversely, Th naïve and permanence of immune cells within the peri-
cells committed to a Th2 phenotype when the odontal tissues. Both prototypical inflammatory
antigen presentation occurred in an IL-4 enriched cytokines, IL-1β and TNFα are characteristic
environment, being the stabilization of the pheno- secreted products of Th1 lymphocytes. TNFα
type dependent on the transcription of the key and IL-1β produce vasodilatation, stimulate the
transcription factor GATA-3. Th2 cells secrete activation of endothelial cells to increase immune
IL-4, IL-5, IL-9, IL-13, and IL-25, among others cell recruitment, increase the chemokine produc-
cytokines. The committed Th2 cells favor the tion in most cell types, participate in neutrophil
effector mechanisms involved in the eradication activation, and stimulate the secretion and tissue
of extracellular pathogens, mainly stimulating the activation of MMPs, among other functions.
secretion of antibodies by activated plasma cells Even though neither IL-1β nor TNFα is directly
[1, 73]. As in many biological processes, the lin- involved in the stimulation of bone resorption,
eage commitment to the Th1 or Th2 phenotypes is they indirectly favor the destruction of bone by
an excluding and self-amplifying event. Once a stimulating the sustained inflammation of peri-
naïve Th cell is committed to one of the subtypes, odontal tissue [79].
the secreted cytokines will serve as an autocrine Alternatively, some evidence supports the
stimulus to maintain the commitment, as a posi- hypothesis that periodontitis pathogenesis is more
tive feedback amplification signal, and as a para- related to the differentiation and effector functions
crine stimulus to prevent the commitment of other of the Th2 lymphocyte lineage. This idea is sup-
naïve cells to alternative lineages [74]. ported by the fact that Th lymphocytes isolated
The pivotal role of the Th1 subtype in the from inflamed gingival tissues predominantly pro-
establishment and progression of periodontitis duce IL-4 over IFN-γ after non-­antigen-­specific
was supported by extensive evidence indicating stimulation [80], and by the classical histological
increased levels of IFN-γ in the tissues of peri- description of the advanced lesion of periodontitis
odontitis patients [75] and corroborated by as a “B-cell type of lesion” [31].
in vitro evidence of the augment of IFN-γ protein Th2 lymphocytes are the main cellular source
levels and transcription during the progression of of the cytokine IL-4, doubling as an inducer of
experimental periodontitis [76, 77]. Th2 polarization, as well as an effector molecular
It is noteworthy that the role of the Th1 lin- signal. Among its more important functions, it
eage (and its signature cytokine product: IFN-γ) promotes the class switching to IgE secretion in
is of paramount importance in limiting the inva- B-cells, and favors the alternative activation of
68 F. Cavalla et al.

macrophages in an IFN-γ-independent pathway. Simultaneous analysis of multiple cytokines


It is noteworthy that both effector functions limit in periapical osteolytic lesions demonstrated that
the capacity of the immune response to control a complex network of cytokines drives the evolu-
the periodontal infection, which is predominantly tion of the bone resorption (active lesion) or
mediated by intracellular pathogens [81, 82]. IgE arrest the progression of the lesion (inactive
is the isotype class of antibodies best suited to lesion). Characteristic Th1 and Th17 cytokine
combat large extracellular parasites (such as hel- products, such as TNF-α, IL-21, IL-17, and IFN-­
minths) and in not pertinent to fight periodontal γ, appear strongly associated with the bone
pathogens. Further, the alternative activation of resorptive process, while the stabilization of the
macrophages inhibits their microbicide func- lesion seems associated with the expression of a
tions, suppresses the production of iNOS and different subset of cytokines: IL-10, IL-9, IL-4,
consequently of nitric oxide (NO), and stimulates and IL-22 [89]. The latter are among the signa-
the secretion of IL-10 and TGFβ, which in turn ture cytokine products of a distinct Th lineage
can downregulate pro-inflammatory and Th1 with immune suppressive properties, known as T
responses [83]. It is for these reasons that some regulatory cells (Tregs).
authors consider that the Th2 polarization in peri-
odontitis may represent an impaired adaptive
immune response, in which IL-4 inhibits the 6.2.5 A
 daptive Immune Responses
more effective Th1 polarization. This may be due May be Also Protective: A Role
to inherent host characteristics, or may be also for Regulatory T Cells (Tregs)
caused by evasion strategies triggered by highly
evolved periodontopathic bacteria [84, 85]. Tregs are Th lymphocytes associated with the
In the beginning of the twenty-first century, a secretion of anti-inflammatory cytokines and
new subset of T helper cells characterized by the reparative molecular signals, such as IL-10 and
secretion of IL-17 was described, and named TGFβ. Their lineage commitment depends on the
Th17 accordingly. The polarization of these cells expression of the transcription factor FoxP3, and
was driven by IL-23 and was dependent on the they characteristically express the surface mark-
transcription of the transcription factor RORγT ers CTLA4, CD103, and CD45RO. The secretion
[86]. Along with IL-17, Th17 cells also secrete of IL-10 and the contact inhibition of lymphocyte
the cytokines IL-21 and IL-22. Shortly after the activation by the CTLA4 co-receptor are the sig-
discovery of Th17 cells, IL-17 was found to be nature effector mechanisms of Tregs, leading to
highly expressed in osteolytic lesions, such as an active suppression of the immune response
periodontitis and periapical lesions [87]. and a return to tissue homeostasis. Experimental
Cytokines such as IL-23, TGF-β, IL-17, IL-6, in vivo data associates the presence of Tregs with
and IL-1β are highly expressed in inflamed peri- the attenuation of osteolytic progression in peri-
odontal tissues, providing the necessary signals odontal lesions [90]. Recently, the mechanisms
to drive the polarization of Th cell to the Th17 responsible for Tregs’ migration to periodontal
lineage. Interestingly, secreted RANKL is a char- tissues were unraveled, being the IL-4/CCL22/
acteristic cytokine product of Th17 cells, which CCR4 axis responsible for the chemoattraction of
in cooperation with its other proinflammatory these cells to the periodontium. The mechanism
cytokine products are capable of tilting the bone can be mimicked by the injection of CCL22-­
metabolism favoring resorption over apposition. releasing particles, which result in a therapeutic
Additionally, Th17 cells provide the necessary arrest of bone resorptive activity [90–92].
proinflammatory signals to upregulate the expres- While the mechanisms of Tregs chemoattrac-
sion, secretion, and activation of MMPs, generat- tion have been discovered, the issue of their origin
ing an amplification loop of inflammatory and and generation remains an unsolved question.
pro-resorptive mediators [88]. Interestingly, Tregs have been described as c­ entral
6  Inflammatory Pathways of Bone Resorption in Periodontitis 69

elements in the determination of a host–microbe cytokines, endogenous pro-­resolving lipid medi-


homeostasis in several tissues. ators (resolvins), and growth factors.
Recently, periodontitis has been characterized Resolvins are endogenous lipid mediators that
as a disease of dysbiosis, where the pathologic modulate cellular fate and inflammation. They
process is initiated by the disruption of the nor- are biosynthesized during the resolution phase
mally balanced equilibrium between the host and of acute inflammation, and are capable of induc-
the resident microbiome. In this context, some ing cessation of leukocyte recruitment, reversing
keystone pathogens can produce an alteration in of vascular inflammatory phenomena and cause
the microenvironment that is capable of turning prompt apoptosis of neutrophils [94].
commensal microorganisms into opportunistic Tregs and mesenchymal stem cells (MSC)
pathogens, triggering the disease. This framework play a central role in tissue repair, supporting tis-
requires that in healthy conditions the host must sue regeneration by direct control of undesired
be able to tolerate the presence of microorganism immune reactivity and by direct interaction with
without eliciting robust effector responses. This nonimmune tissue cells. Tregs can directly inter-
tolerance is an active process, where the host act with MSC progenitors, favoring their recruit-
recognizes harmless microbial antigens and sup- ment and differentiation in the required cell types
presses the development of exacerbate inflamma- for tissue regeneration [95]. Recent experimen-
tory immune responses against them to preserve tal evidence suggests that MSC and Tregs act in
the functional equilibrium. Tregs are at the center coaction, favoring the establishment of an anti-­
of this active tolerance, inducing antigen-specific inflammatory environment that promotes the
hyporesponsiveness and suppressing the initia- repair of bone defects [96].
tion inflammation by a series of specific mecha- Therefore, the sequence of events of adaptive
nisms [8, 93]. immune response that leads to pathologic alveo-
Additionally, Tregs can also regulate the lar bone resorption can be summarized as fol-
duration and intensity of the immune response lows: after the acute inflammation is firmly
against pathogens, limiting the destructive poten- established, adaptive immune cells are recruited
tial of uncontrolled reactions. In this sense, it and infiltrate the periodontium, marking the tran-
is important to highlight that experimental evi- sition to chronic inflammation. Depending on a
dence demonstrates that Treg-mediated immune series of environmental factors and host’s intrin-
regulation does not interfere with the capacity of sic characteristic, different lineages of effector T
the immune response to fight the infection effi- cells might predominate inside the tissue, deter-
ciently, and that the directed recruitment of Tregs mining the clinical outcome of the disease. If
into diseased periodontal tissues is capable of proinflammatory subtypes predominate (Th1 and
restore the homeostatic tissue balance, even in Th17), the tissue destruction and bone resorption
the presence of an infectious burden that usually are favored; conversely, if the anti-inflammatory
results in the development of destructive peri- and pro-reparative lineage predominate (Tregs),
odontitis [90, 91]. These former properties trans- the inflammation is halted and the tissue tends to
form Tregs and their selective recruitment to the regenerate (Fig. 6.1).
periodontium in an attractive therapeutic tool to Consequently, the variation in the pattern of
modulate the immune response, without interfer- Th responses seems to be a critical determinant
ing with the infection control mechanisms while of periodontal bone loss, since such cell types not
avoiding irreversible tissue damage and promot- only regulate the overall immune response pat-
ing repair. tern but also may directly interfere in the
The tissue repair that occurs after the contrac- RANKL/OPG balance [1, 8]. However, we must
tion of immune response is an active process regu- consider that the inflammatory process is a con-
lated and orchestrated by the secretion of specific tinuum and frequent acute bursts of inflammation
molecular mediators, such as anti-­inflammatory or variations in the pattern of host response occur,
70 F. Cavalla et al.

Gingivitis Periodontitis
Soft tissue Bone Lesion arrest
Clinical signs Redness, Bleeding, Swelling destruction resorption repair

MMPs Rankl
Vascular events
Inflammation
PAMPs PRRs Inflammation Cell downregulation
LPS TLR immune response Chemokines migration Cytokines or resolution
Cellular events
Pro-inflammatory mediators Anti-inflammatory mediators

Fig. 6.1  Pathogenic pathway of periodontal diseases. The tial secretion of cytokines by specific subpopulations of
recognition of PAMPs by PRR triggers the first events of infiltrating leukocytes could lead to self-­amplification or
inflammation, which leads to the recruitment of immune modulation of the immune/inflammatory response, deter-
cells to the periodontium. The clinical signs are the expres- mining the outcome of the disease
sion of the underlying inflammatory process. The differen-

even after the establishment of the chronic phase These contradictory clinical phenotypes could
of inflammation. Indeed, the periodontitis pro- represent the reflection of the balance and inter-
gression is classically described to progress in play of several underlying risk determinants and
bursts, where periods of active bone resorption environmental risk factors [99]. While in most
may be followed by periods where the inflamma- cases there is a correlation between the amount of
tion persists, but the bone resorption seems to be dental biofilm and the extent and severity of the
restricted [97, 98]. While this model remains to periodontitis, patients located at both extremes of
be confirmed from a molecular point of view, the susceptibility spectrum exist and are a rou-
accumulating evidence support the idea that vari- tinely encountered in a specialist’s dental office.
ations in the intensity and nature of the host’s Similarly, patients presenting a lack of response
inflammatory response may drive the disease to the classical clinical treatment of periodontitis,
evolution via the control of bone resorption. without any apparent clinical reason to explain
In this context, virtually any factor or event such unresponsiveness, comprise a real and rela-
that could modify the host–microbe interactions tively frequent challenge to clinicians.
and the inflammatory immune response derived The factors that contribute to the differen-
from such interactions would influence the evo- tial susceptibility to inflammatory destruction
lution and outcome of periodontitis. These com- of the alveolar bone and other periodontal tis-
plex interactions explain the tremendous sues are not totally understood. Nevertheless,
variability in susceptibility and clinical pheno- strong evidence points to an increasing number
type of the disease. of modifying factors, both innate (e.g., genetic
variations) and acquired (e.g., microbial fac-
tors, e­ nvironmental factors, and comorbidities),
6.3 Risk Determinants that can modulate the host–microbe interactions
to Alveolar Bone Resorption in the periodontal environment, tilting the resis-
in Periodontitis tance or susceptibility phenotypes [75, 99–101].
As previously stated, the pathogenic process of
Clinicians often face the disconcerting experience alveolar bone destruction is dynamically modu-
of examining a patient with extensive periodon- lated by the interplay of risk factors affecting
tal destruction, but without any obvious clinical the homeostatic balance of the host and his/her
finding to explain the severity of the disease. capacity to cope with the presence of periodontal
Conversely, the opposed scenario is also possi- microbes (Fig. 6.2).
ble: a patient with widespread inflammation and Indeed, the delicate balance between a pro-
abundant presence of dental biofilm, but with- tective immune response (without bone loss or
out any clinical sign of periodontal destruction. periodontal tissue damage) and an exacerbated
6  Inflammatory Pathways of Bone Resorption in Periodontitis 71

6.3.1 Acquired Risk Factors

6.3.1.1 Tobacco
The latest consensus report from the 11th
European Workshop in Periodontology stab-
lished that the ask, advise, refer (AAR) approach
is the absolute minimum standard of care when
dealing with tobacco smokers in the dental clinic.
The consensus considered the copious evidence
linking tobacco consumption with the occurrence
and severity of alveolar bone loss around teeth
and dental implants, as well as with negative
treatment outcomes [105].
Probably, the most convincing piece of evi-
dence linking tobacco smoking with periodontitis
comes from the longitudinal cohort of Dunedin,
New Zealand. This birth cohort of 1037 partici-
Fig. 6.2  Interaction of risk factors and determinants in
the pathogenesis of periodontitis. Environmental, micro-
pants has been longitudinally followed since
biological, and intrinsic host factors determine the bal- birth in 1972–1973, including a complete dental
ance between the infecting microorganism and the and periodontal examinations at ages 32 and 36,
immune response. In homeostatic conditions the host’s and tobacco smoking determination at ages 15,
immune systems are able to tolerate the presence of
microorganism without triggering a destructive inflamma-
18, 21, 26, 32, and 36. The study demonstrated
tory response. The qualitative changes of the oral micro- that smokers had a 23% greater attachment loss
biota (dysbiosis) are a reflection of the loss of balance in than nonsmokers at age 36, confirming the strong
the host–microbe interaction leading to disease association between chronic smoking and peri-
odontal disease [106].
immune response (with widespread alveolar The mechanistic link between tobacco smok-
bone destruction) can be broken by numer- ing and alveolar bone loss in periodontitis is com-
ous environmental and host’s intrinsic factors. plex and involves alterations in the microbiome,
Among them, changes in the oral hygiene habits, host’s immune response, and metabolism of peri-
placement of deficient dental restorations, anti- odontal tissues. Chronic tobacco consumption
biotics usage, smoking, nutritional alterations, leads to profound changes in the periodontal bio-
hormonal changes, metabolic diseases, acquired film, dramatically increasing the p­athogenicity
infections that alters the periodontal microbiota, and disease-initiating capacity of it. Tobacco
trauma, corticosteroids usage, stress, and even smoke is responsible for increasing the ability of
aging [102, 103]. In addition, there is strong P. gingivalis to form biofilms and decreases its
evidence that genetic factors contribute in a sig- potential to elicit an effective host’s immune
nificant extent to determine the susceptibility to response, acting in synergy with its proprietary
inflammatory destruction of the tooth-attachment multiple evasion mechanisms [107]. The chronic
apparatus, determining the quantity and quality use of tobacco also generates extensive deleteri-
of the inflammatory immune response to peri- ous effects in the host’s immune system, both sys-
odontal infection [104]. Once the balance is temically and locally. At the systemic level,
tilted, the host’s response against the periodontal tobacco smoking generates altered tolerance to
microbiota can become “destructive” leading to self-antigens, suppression of the innate immune
the setting up of the clinical signs of periodon- response, and aberrant adaptive immune responses
titis: gingival inflammation, increased probing [108, 109]. At the local level, innate immune cells
depth of the periodontal sulcus, clinical attach- from smokers (i.e., neutrophils and macrophages)
ment loss, and alveolar bone resorption. have impaired phagocytic ­capacity, chemotactic
72 F. Cavalla et al.

deficiencies, and increased proinflammatory periodontitis [114]. In the case of the congenital
capacity [110]. Therefore, the clearance of invad- conditions, the periodontal destruction appears
ing microorganism becomes compromised, lead- very early in life, in some cases affecting both
ing to a sustained chronic and ineffective local deciduous and permanent dentition and leading
inflammation, characterized by the accumulation to premature exfoliation of all teeth [115].
of neutrophils, lymphocytes, and macrophages It is possible to mention the infection by
within the periodontal connective tissue. HTLV-1 virus as an additional example of an
Neutrophils can then be activated to degranulate, acquired disturbance on the immune system that
liberating their rich content of tissue-­destructive impacts periodontitis outcome [116]. HTLV-1
proteases. Lymphocytes persist within the tissue, results in an overall deregulation of immune
liberating proinflammatory mediators that amplify response, being associated with a series of patho-
the inflammatory process. Macrophages produce logical conditions. In periodontitis context, it was
and liberate proteolytic enzymes, reactive oxygen demonstrated that even presenting a standard
species, and nitrogen species, further contributing periodontopathogen infection, patients with
to connective tissue degradation [111]. HTLV-1 demonstrated an exacerbated immune
response and increased periodontal destruction.
6.3.1.2 Immune Deficiencies
Obviously, any condition affecting the quality and 6.3.1.3 Metabolic Diseases
effectiveness of the immune response will have a Some metabolic diseases affect the clinical pre-
tremendous impact in the clinical presentation of sentation of periodontitis, correlating with the
periodontitis. The most severe forms of periodonti- presence, extension, and severity of the alveolar
tis are those categorized as “periodontitis as a mani- bone loss [117]. Among them, diabetes mellitus
festation of a systemic diseases,” where the systemic is most widely studied, and a large body of evi-
disease is without exception an immune subduing dence demonstrates that uncontrolled diabetic
condition, including acquired neutropenia, leuke- patients are under increased risk of suffering
mia, familial and cyclic neutropenia, down syn- severe forms of periodontitis, with extensive
drome, leukocyte adhesion deficiency syndrome, alveolar bone loss, and with poor response to
Papillon-Lefevre syndrome, Chediak-Higashi syn- conventional periodontal treatment [118].
drome, histiocytosis, glycogen storage disease, Recent evidence from clinical trials supports
infantile agranulocytosis, Cohen syndrome, Ehler- the notion that diabetic patients exhibit increased
Danlos syndrome, and hypophosphatasia [112]. RANKL/OPG ratios, and that the periodontal
When any of the immune mechanism respon- treatment is capable of diminishing the RANKL/
sible to withhold the dissemination of the infect- OPG in follow-up periods of 3 months [119]. In
ing microorganism becomes compromised, the this respect, the mechanism behind the increased
host can develop extensive periodontal destruc- susceptibility to alveolar bone loss would be the
tion and the possibility of suffering the systemic same as in patients without diabetes, and the
propagation of the infection, with potential severe increased susceptibility would be a consequence
health outcomes. For example, leukocyte adhe- of impaired inflammation control mechanisms.
sion deficiency type I patients, who suffer from a Further, evidence from a recent systematic
mutation affecting the CD18 integrin that disrupts review including 35 parallel randomized clinical
the transmigration of neutrophils, often present a trials (2565 participants) demonstrated that peri-
very severe periodontitis, with extensive bone odontal therapy has a measurable effect in the
loss and a characteristically increased infectious glycemic control of diabetes patients, pointing to
burden within the periodontal tissues [113]. a bidirectional association between periodontitis
In the case of the acquired conditions, the and diabetes mellitus [120]. The linking factor
extensive and rapid alveolar bone loss and peri- behind the bidirectional relationship of diabe-
odontal destruction is the common feature, tes and periodontitis could be the inflammatory
resembling the clinical phenotype of aggressive molecular signals, that emanated from the local
6  Inflammatory Pathways of Bone Resorption in Periodontitis 73

environment of the periodontium could act at dis- enzyme peptidyl arginine deiminase (PAD), which
tant sites affecting the glucose metabolism and catalyzes the conversion of arginine residues to
modify the course of the disease [121]. A recent citrulline. The irreversible citrullination of arginine
systematic review and meta-analysis, including residues changes the structural characteristic of
nine clinical trials, demonstrated that periodontal proteins, transforming them in antigens capable of
treatment significantly reduced circulating levels eliciting an immune response [127]. Citrullinated
of TNFα and C-reactive protein, diminishing the proteins characteristically accumulate in the joints
inflammatory burden in type 2 diabetic patients of RA patients, and specific autoantibodies against
[122]. This evidence supports the mechanistic them are one of the central causes for joint degen-
link of periodontitis and diabetes through inflam- eration and bone destruction during the progression
matory mediators. It is important to consider of the disease [128, 129]. Additionally, the pro-
that the study of the potential interconnection inflammatory molecular signals liberated in the
of multifactorial diseases such as diabetes and chronic immune response of periodontitis and RA
periodontitis is complex due the multitude of could reach the circulation, generating the mutual
variables inherent from both conditions. In this exacerbation and perpetuation of the inflamma-
scenario, data from experimental models is espe- tory response in distant compartments [130]. In
cially important in the confirmation and under- this sense, both diseases will act in synergy in a
standing of the alleged interaction. Interestingly, loop of reciprocal inflammatory amplification. The
experimental models demonstrate that diabetes connection between RA and periodontitis is also
can in fact disrupt host–microbe homeostasis supported by experimental model’s data, which
in periodontal environment, since the spontane- demonstrate that periodontitis and arthritis interac-
ous periodontitis development in diabetic rats tion in mice involves a shared hyper-inflammatory
involves an unrestricted expression of inflamma- genotype and functional immunological interfer-
tory cytokines and tissue destructive factors in ences [131]. In an analog fashion to the scenery
the absence of major changes in commensal oral described for diabetes, RA also seems capable of
microbiota [123]. In other words, the modifica- disrupting the host–microbe homeostasis, lead-
tions in host responsiveness caused by diabetes ing to aberrant responses to microorganism previ-
can trigger a destructive inflammatory response ously r­ ecognized as commensal and nonhazardous
against previously well-tolerated microorganism. microbiota [131].
Another metabolic disease strongly associated Another example of the complexity of the
with alveolar bone loss in periodontitis is rheu- immune mechanisms that drive the inflamma-
matoid arthritis (RA). RA patients present the tory destruction of host’s tissues is the evidence
inflammatory degeneration of diarthrodial joints that the high dietary salt intake (characteristic of
with a profuse infiltration of immune cells into the modern western diet) has the potential to induce
synovial lining. As in periodontitis, the character- enhanced macrophage infiltration, increased
istic bone erosions in inflammatory arthritis are inflammatory cytokine secretion, and polariza-
caused by a decontrolled activation of osteoclast tion of immune response towards a Th17 pheno-
due to the inflammatory upregulation of RANKL type. In sum, the high dietary salt intake can be
and the increase of the RANKL/OPG ratio [124]. regarded as an environmental risk factor for the
Epidemiologic data points to a strong correlation development of autoimmune diseases [132–134].
between the occurrence of both diseases, suggest- Along the same lines, recent evidence points to
ing common susceptibility traits and a possible an association between obesity/metabolic syn-
common pathogenesis [125]. The hypothetical drome and exacerbated alveolar bone loss, sup-
pathogenic link could be the capacity of some peri- ported both by epidemiological data and by
odontal pathogens to produce enzymatic changes in experimental in vivo evidence [135–137]. Again,
structural proteins that trigger a humoral response the linking factor between both conditions would
against self-antigens [126]. Specifically, P. gingi- be the exacerbation of inflammation and the mal-
valis is the only known bacteria that produce the function of inflammation control mechanisms.
74 F. Cavalla et al.

6.3.2 Innate Risk Determinants associated with an increased risk of periodontitis


in Indians, but not in Chinese [141]. The above
Genetic susceptibility determinants are among example testifies for the difficulty of stablishing
the most studied topics in the periodontal litera- associations between genetic variations and the
ture. Nevertheless, despite the generalized accep- presence and severity of periodontitis, with con-
tance of genetic factors as key regulators of the troversies spanning for decades in the literature
susceptibility to periodontitis, a great controversy and still unresolved.
still persist regarding the specific contribution of A major source of bias in periodontal genetic
particular mutations to the overall clinical pheno- research came from the necessity to select a pri-
type, and more importantly a model integrating ori a gene(s) to study in an association research,
the human genetic diversity into the pathogenesis often based in their putative theoretical involve-
of periodontitis is still lacking. ment in key steps of the immune response or
The first clear evidence that genetic factors periodontal tissue’s metabolism. Nevertheless,
played a central role conferring susceptibility or this approach has proved inconclusive and most
resistance to periodontal destruction came from associations between mutations and the occur-
the elegant work of Michalowicz et al. with adult rence, severity, or extension of periodontitis lack
twins [138, 139]. Despite later controversies, the necessary replication in different populations,
Michalowicz et al. established that genetic fac- as previously exemplified with the case of the
tors accounted for at least a 50% of the clinical IL-1 cluster. Probable causes of these difficulties
variation on the disease phenotype, regulating the are multiple, among them: the complexity of the
susceptibility to suffer bone loss and periodontal immune regulatory mechanisms, overlap and
destruction. Since that seminal work, many mod- redundancy in the functions of many mediators
els and hypothesis have been proposed to explain of inflammation and bone metabolism, the lack
the mechanisms of genetic influence over the of a linear relationship between gene expression
phenotype of periodontitis. and protein production/cell phenotype, and the
The focus of the genetic research in periodon- lack of a comprehensive understanding of the
tal susceptibility have been the molecular media- regulatory pathways that maintain tissue homeo-
tors of inflammation, particularly the mutations stasis. Additionally, the difficulty in the definition
affecting the levels and expression of cytokines of the periodontitis cases and the selection of
recognized as key regulators of the inflammatory suitable controls poses another level of complex-
process. The most studied mutations in periodon- ity over the design of periodontitis genetic asso-
titis are the polymorphic variations of the IL-1 ciation studies [99].
gene cluster and/or its promoter region. In the Even with the inherent problems of the classic
late years of the twentieth century, Kornman periodontal genetic studies, the literature consis-
et al. reported the association between the com- tently demonstrates that mutations altering the
posite polymorphic genotype for IL-1A (−889) host’s capacity to mount and efficient yet con-
and IL-1B (+3953) genes and the severity of peri- trolled immune response, or those related to the
odontitis in nonsmokers, concluding that the exacerbation of the bone and connective tissue
composite polymorphic phenotype contributed turnover, are strongly associated with the pres-
significantly to increased IL-1β levels and aug- ence and clinical phenotype of the disease. As
mented risk of suffering severe forms of peri- examples: the single nucleotide polymorphism
odontitis [140]. Since then, numerous replication (SNP) rs4794067 of the TBX21 gene that
studies in various populations have been con- increases the transcription of T-bet, leading to an
ducted with conflicting results. A recent system- exacerbated inflammatory response, is strongly
atic review and meta-analysis of 20 case–control associated with the occurrence of periodontitis in
studies in Asian subjects, including in excess of a Brazilian population [75]. Further, the SNP
3000 patients and controls, concluded that the rs1800872 in the promoter region of the IL-10
polymorphic form of IL-1B (+3953) was strongly that decreases the transcription of IL-10, leading
6  Inflammatory Pathways of Bone Resorption in Periodontitis 75

to impaired immune regulation, is strongly asso- that has predictive value and can be used in the
ciated with the occurrence and severity of peri- decision-making process during the management
odontitis in the same population [142]. These two of breast cancer cases [144]. An analog tool for the
examples show the value of the classical approach risk assessment of periodontitis, aiding in the
to periodontal genetic studies, but also testify for long-term clinical management of patients, is an
its inherent flaw, where a theoretical framework old longing of the practicing periodontist.
taking into consideration the functional conse-
quences of the studied mutations is a sine qua
non condition. This condition greatly limits the 6.4  uture Perspectives for the
F
validity of the studies, since many genes are not Treatment of Periodontitis:
studied by the lack of a supporting theory linking From the Bench to the
them to the pathogenesis of periodontitis. Dental Chair
To overcome some of these problems, a new
strategy using the results of large genome-wide The accumulate knowledge of the immune mech-
association (GWA) studies has been recently pro- anisms driving the progression of bone loss in
posed. The rationale of this approach is to scan periodontitis has stimulated the development
the whole genome searching for genes or groups of new therapeutic approaches to modulate the
of genes associated with the presence of the dis- immune inflammatory response with the intention
ease (or disease subrogate) without any a priori to limit the adverse consequences of unregulated
selection bias. responses. Theoretically, bioactive molecules
Using the GWA-based selection strategy a with the potential to regulate key points of the
completely new set of genes with strong associa- immune response could be used as coadjutants in
tion to the occurrence of periodontitis have been the treatment of periodontitis, enhancing the clini-
discovered. These previously ignored genes code cal effectiveness of conventional treatment, pre-
for neuropeptides, innate immune response serving the results for longer periods, limiting the
receptors, enzymes linked to cytoskeletal rear- progression of periodontal destruction, and modi-
rangement, intracellular signaling transduction fying the susceptibility to suffer relapses.
molecules, and proteins directing vesicle fusion Various promising efforts in the development
during synapsis, and even some noncoding genes of clinically relevant and safe bioactive molecules
or genes with no known function. The diversity are in different stages of experimental testing,
of the newly discovered periodontitis-associated with the perspective of reaching to the clinical
genes testifies for the power of the GWA practice in the near future. The classic approach
approach, since it allows for the unveiling of dis- to coadjuvant pharmacological treatment in
ease–gene association without the necessity of a periodontitis is the use of anti-infective agents.
previous theoretical framework [143]. Although not immune modulatory agents in a
While the real contribution of genetic determi- strict sense, the use of anti-infective therapy has a
nants to periodontitis risk remains to be estab- pronounced effect in the immune system, decreas-
lished, it is important to consider that extensive ing the number of antigens available to trigger
research efforts in the field aim to provide addi- an immune response and limiting the extent and
tional tools to the clinician in the direct risk assess- duration of it. Characteristically, systemic antibi-
ment, which in turn may influence a series of otics has been used to enhance the results of peri-
clinical decisions. An interesting example of the odontal treatment in severe or high-­risk patients,
clinical value of being able to assign and objective nevertheless there is a series of problems associ-
risk value to the presence of certain SNPs, is the ated with the use of these agents, including the
algorithm that forms the PRS index (polygenic appearance of bacterial resistance and the occur-
risk score) for the risk discrimination in breast rence of adverse reactions. Over the years, various
cancer. The PRS index additively combines the controlled-release local ­anti-­infective agents have
risk effect of 77 SNPs, providing a valuable tool been developed in an effort to limit the negative
76 F. Cavalla et al.

consequences of the use of systemic antibiotics, capacity to fight the infection. In an analog fash-
but providing their putative benefits. In a system- ion to the trend of using controlled-release local
atic review and meta-­analysis including 32 stud- anti-infective drugs, the goal is to develop an
ies (3705 subjects), the use of minocycline gel, immune modulatory agent that could be directly
microencapsulated minocycline, chlorhexidine targeted to the periodontal tissue, preventing the
chips, and doxycycline gel during scaling and potential risks of undesirable immune modulation
root planning (SRP) proved marginally better in other body compartments. It is noteworthy that
than SRP alone, but not to a clinically significant the systemic administration of classic nonsteroid
extent [145]. Another shortcoming of antimicro- anti-inflammatory drugs (NSAID) as adjunctive
bial therapy is that after the completion of the treatment in periodontitis, aimed at reducing the
treatment the risk factors and susceptibility traits levels of arachidonic acid metabolites, has shown
that contributed to the establishment of a patho- no long-term clinically relevant benefits in several
genic microbiota (dysbiosis) remain unaltered, clinical trials, pointing to the need of a very spe-
leading to a recolonization of periodontal sites by cific and focused inhibition of inflammation in
a pathogenic microbiota and to the recurrence of order to attain meaningful clinical benefits.
the disease [146, 147]. Additionally, the potential risks of the chronic use
The new emerging therapeutic approach will of NSAID, such as gastric ulcer and coagulation
use immune modulatory drugs, capable of selec- impairment, seriously limit their usefulness as
tively regulating key points of the immune adjunctive therapy in periodontitis [148].
response, specifically limiting the degree of As depicted in Figs. 6.3 and 6.4, an interesting
inflammation but without dampening the system’s approach would be the use of drugs capable of

Fig. 6.3  Immune and inflammatory response to periodon- appear sequentially determine the outcome of the response.
tal infection. Periodontal pathogens are recognized by the The relative predominance of pro-inflammatory lineages
resident cells, which trigger the first inflammatory response (Th1 and Th17) favors soft tissue destruction and osteoclast
mediated by PAMP/PRR interactions. Infiltrating innate genesis. Conversely, when regulatory lineages predominate
immune cells rapidly reach the infection site and amplify (Tregs and possibly Th2), the inflammation is halted, osteo-
the primary response. The adaptive immune cells that clast genesis is inhibited, and the tissue tends to repair
6  Inflammatory Pathways of Bone Resorption in Periodontitis 77

Fig. 6.4  Modulatory strategies to control the inflamma- metabolites, modulating the inflammatory response.
tory destruction of alveolar bone. Anti-infective therapy Specific antibodies against inflammatory cytokines and
diminishes the presence of the antigen, limiting the receptors (Anakinra, Rilonacept, Canakinumab, met-­
response. TLR inhibition offers the possibility of inhibit- RANTES, and Denosumab) could inhibit osteoclast gen-
ing the first events of inflammation, but with the danger of esis directly or indirectly. SDD inhibits soft tissue
facilitating the infiltration of pathogens. Resolvins could destruction and the lysis of the demineralized bone matrix.
exert a modulatory event reversing the vascular events of Selective recruitment of Tregs by CCL22 could fine-tune
inflammation and inducing apoptosis of infiltrating leuko- the inflammatory response, leading to an efficient
cytes. NSAIDs inhibit the production of arachidonic acid response without tooth-attachment loss

inhibiting the first signaling events in the inflam- geting these receptors is prodigious. There is a
matory process, but without compromising the growing body of in vivo evidence demonstrating
later steps of the response. In this sense, Toll-like that the selective inhibition of TLR’s activation or
receptors (TLR) appear as promising therapeutic signal transduction is protective against periodon-
targets. These receptors are fundamentally titis-induced alveolar bone loss. In a murine
involved in the recognition of pathogen-­ model of Porphyromonas gingivalis-induced peri-
associated molecular patters (PAMPs). TLRs are odontitis, the pharmacological inhibition of GSK3
capable of triggering the first events of the (a key factor in the signal transduction chain
inflammatory cascade, including the upregula- downstream of TLR activation) significantly
tion of inflammatory cytokines and MMPs. decreased the systemic levels of TNFα, IL-1β,
Theoretically, the selective inhibition of one or IL-6, and IL-12 post infection, and suppressed
various TLR subtypes could limit and modulate alveolar bone loss [149]. Further, mice geneti-
the extent of the initial reaction, thus modifying cally deficient (i.e., knock out) for the expression
the nature of the immune response, and prevent- of TLR pathway signaling molecules, such as
ing the occurrence of an uncontrolled inflamma- MyD88, demonstrated resistance to LPS-induced
tion leading to bone metabolism uncoupling and alveolar bone loss, associated with diminished
resorption. Given that TLR are expressed in the levels of inflammatory cytokines expression and
surface of every cell type in the periodontal tissue, osteoclast differentiation markers [150].
the immune modulatory potential for a drug tar- Nevertheless, it is fundamental to bear in mind
78 F. Cavalla et al.

that the innate immune events triggered by the bone matrix allowing for bone remodeling.
activation of TLR are of paramount importance Increased collagen lytic activity as a consequence
for the control of infection and that any drug tar- of inflammation is the ultimate responsibility for
geting this receptors will need to be highly selec- net bone loss during the progression of periodon-
tive and tissue specific to avoid the potential titis. Tetracyclines in general and doxycycline in
dangers of the spreading of the periodontal infec- particular have the property of inhibiting the
tion to other body compartments. activity of MMPs independently of their antimi-
Moving down the inflammatory cascade, the crobial action [148, 156, 157]. The inhibition of
next step susceptible of pharmacological modu- MMP using systemic subantimicrobial-dose dox-
lation is the selective inhibition of the effector ycycline (SDD) is a well-established therapeutic
molecules of inflammation. One alternative is the approach and there is a three-decade body of evi-
use of nonsteroidal anti-inflammatory drugs dence supporting its effectivity as an adjunct
(NSAID) to control and modulate the production treatment in chronic periodontitis [158]. The last
of pro-inflammatory molecules, such as prosta- advance in the use of SDD as MMP inhibitors is
glandins derived from arachidonic acid. The clas- the development of sustained release nano-­
sical COX-1 and COX-2 inhibitors have the structured films, with the property of delivering a
potential to limit the magnitude of the inflamma- constant dose of doxycycline directly into the
tion. Even though they have proven effective in periodontal tissue for prolonged periods.
diminishing the alveolar bone loss in experimen- Preliminary clinical trials proved that the use of
tal models and marginally effective in clinical the sustained release films improved the clinical
testing [148, 151, 152], their routine use is outcome of periodontal treatment after a follow-
unpractical due to the appearance of unwanted ­up period of 2 months [159].
side effects in the long-term treatment. Another alternative is the use of highly specific
Other emerging therapeutic approach is the antibodies against the molecular mediators of
use of endogenous pro-resolving lipid mediators inflammation. Although there is not any cytokine
(resolvins) to modulate the inflammatory inhibitor currently being used for the treatment of
response and protect the alveolar bone from periodontitis, several cytokine-inhibiting drugs are
resorption [153]. These lipid mediators are struc- routinely used to prevent the inflammatory bone
turally related to prostaglandin and leukotriene, loss in rheumatoid arthritis (RA) [160]. Given the
but contrary to the pro-inflammatory activities of pathogenic similarities of inflammatory bone loss
the former are capable of inhibit leukocyte in RA and periodontitis, and the possible common
recruitment and promote inflammatory resolu- pathogenic mechanisms among them, it is possible
tion. Evidence form Porphyromonas gingivalis-­ to hypothesize that such drugs would be also use-
induced periodontitis in vivo experiments ful to prevent alveolar bone loss in periodontitis.
demonstrated that the administration of Resolvin Three approved drugs for the treatment of RA
E1 as monotherapy resulted in complete resolu- reduce the activities of IL-1α/β, diminishing the
tion of the bone lesion and in reduction of sys- inflammation and preventing bone loss. Anakinra
temic markers of inflammation [154]. is a recombinant form of the naturally occurring
An additional possible target of modulation IL-1 receptor antagonist. Anakinra conjugates to
are the enzymes responsible for connective tissue the IL-1 receptor, preventing the activity of
degradation, namely MMPs. Collectively, MMPs IL-1α/β [161]. The soluble IL-1β decoy receptor
are responsible for collagen turnover in all peri- Rilonacept and the neutralizing antibody
odontal connective tissues, including bone [155]. Canakinumab block the biologic actions of IL-1β
After bone demineralization, collagenases and [162]. Taking into consideration the link between
gelatinases degrade the organic component of the inflammation and the increasing ratio of RANKL/
6  Inflammatory Pathways of Bone Resorption in Periodontitis 79

OPG previously discussed, the selective down- associate with increased bacterial load or with
regulation of IL-1α/β could prove valuable as bone impairment in the bacteria clearance mecha-
anti-resorptive drugs. Nevertheless, the infectious nisms, providing an attractive possibility for safe
nature of periodontitis and the risk of impairment local immune modulation, which can be explored
of the pathogen clearance mechanisms pose a seri- as a future approach to enhance the clinical
ous limitation for the use of immune suppressive results of conventional periodontal therapy [91].
drugs in the treatment of periodontitis and the Additionally, approaches based in the induction
potential risks must be carefully weighed. or chemoattraction of Tregs may restore the local
Yet other possible pharmacological target are host–microbe homeostasis, potentially resulting
the cytokines/receptors responsible for the selec- in longer-term effects with increased potential
tive recruitment of osteoclasts precursors to the impact in avoiding subsequent disease events [8].
bone compartment. These chemotactic cytokines Finally, a further possible target of pharmaco-
(a.k.a. chemokines) and their receptors are respon- logic modulation is the RANKL/RANK/OPG
sible for the control of the influx of monocyte/mac- axis. As previously stated, the relative abundance
rophages to the periodontal tissue, and are induced and equilibrium among these molecules is largely
under inflammatory conditions [163]. For example, responsible for the balance between bone apposi-
there is experimental in vivo evidence of the effec- tion and bone resorption. The inflammatory pro-
tiveness of the pharmacologic inhibition of the cess increases the RANKL/OPG ratio, causing
chemokine receptors CCR1 and CCR5 in reduc- bone resorption. Denosumab is a human mono-
ing the alveolar bone loss in a murine model of clonal anti-RANKL antibody approved for use in
Aggregatibacter actinomycetemcomitans-­induced osteoporosis patients with high risk of fracture
periodontitis, using the functional competitive [164]. Clinical trials have demonstrated the effi-
inhibitor met-­RANTES [163]. In this model, the cacy and safety of the drug in long-term use,
reduced recruitment of osteoclast precursors and opening the possibility for its use in other bone
other inflammatory cells caused a protective effect resorbing pathologies, such as periodontitis [165].
in alveolar bone levels, but proved detrimental to At this time, it is reasonable to suppose that the
the anti-infective response at higher doses. These modulation of the RANKL/RANK/OPG axis to
results highlight the complexity of immune modu- manage periodontitis may become a therapeutic
latory therapies, where there is a thin line between tool available for the clinician in the near future.
bone protective effects and increased risk of sys- Those and other future developments will
temic spreading of the local periodontal infection. contribute to the increase and diversification of
In an interesting approach and proof-of-­ the therapeutic tools available to the periodontist.
principle in vivo experiment, it was demonstrated The perspective of new therapeutic approaches
that harnessing endogenous Tregs and recruiting allowing for the successful treatment of refrac-
them to specific sites of periodontal inflammation tory cases is a long-standing desire of the practic-
is effective in limiting bone resorption and pro- ing periodontist, but their implementation will
moting the establishment of a regenerative envi- require a thoughtful understanding of the under-
ronment, and associated with diminished markers lying cellular and molecular mechanism of peri-
of inflammation [68]. In this experimental set- odontitis’ pathogenesis, and the accompanying
ting, endogenous Tregs were recruited to the ability of judging the convenience of their use in
periodontal tissue using a controlled-release a particular clinical situation. As the body of
polymeric vehicle designed to create a gradient knowledge of periodontics swiftly increases, the
of CCL22, a known selective chemokine for periodontist must remain up to date to be able to
Tregs. It is noteworthy that the selective recruit- take full advantage of the coming therapeutic
ment of Tregs to the periodontal tissue did not advances in benefit of his/her patients.
80 F. Cavalla et al.

with special emphasis on mechanisms of colla-


Clinical Relevance gen degradation in the periodontium and the burst
hypothesis of periodontal disease progression. J Clin
• Immune response and inflammation Periodontol. 1997;24(5):297–305.
protect the host from widespread dis- 10. Meyle J, Chapple I. Molecular aspects of the
semination of periodontal pathogens. pathogenesis of periodontitis. Periodontol 2000.
2015;69(1):7–17.
• Exacerbated or uncontrolled immune
11. Page RC, et al. Advances in the pathogenesis of
responses are responsible for the peri- periodontitis: summary of developments, clinical
odontal tissue destruction. implications and future directions. Periodontol 2000.
• A limited subset of signaling molecules 1997;14:216–48.
12. Kornman KS. Mapping the pathogenesis of peri-
link the inflammatory response with the
odontitis: a new look. J Periodontol. 2008;79(8
uncoupling of bone metabolism that Suppl):1560–8.
leads to bone resorption. 13. Noguchi S, et al. The histopathological comparison
• The modulation of these signaling path- on the destruction of the periodontal tissue between
normal junctional epithelium and long junctional
ways and molecules appears as a con-
epithelium. J Periodontal Res. 2016;52:74–82.
crete possibility to deal with patients 14. Pollanen MT, et al. Host-bacteria crosstalk at the den-
unresponsive to traditional therapy. togingival junction. Int J Dent. 2012;2012:821383.
• For the clinician it is essential to under- 15. Cavalla F, et al. Matrix metalloproteinases regu-
late extracellular levels of SDF-1/CXCL12, IL-6
stand the intricate immune pathways
and VEGF in hydrogen peroxide-stimulated
involved in periodontitis pathogenesis human periodontal ligament fibroblasts. Cytokine.
to be prepared for this next generation 2015;73(1):114–21.
of diagnostic and therapeutic tools. 16. Sugiyama A, et al. Activation of human gingival
epithelial cells by cell-surface components of black-­
pigmented bacteria: augmentation of production of
interleukin-8, granulocyte colony-stimulating fac-
tor and granulocyte-macrophage colony-stimulating
References factor and expression of intercellular adhesion mol-
ecule 1. J Med Microbiol. 2002;51(1):27–33.
1. Cavalla F, et al. Cytokine networks regulating 17. Li JP, et al. Differential expression of Toll-like
inflammation and immune defense in the oral cavity. receptor 4 in healthy and diseased human gingiva. J
Curr Oral Health Rep. 2014;1(2):104–13. Periodontal Res. 2014;49(6):845–54.
2. Garlet GP. Destructive and protective roles of cyto- 18. Palm E, et al. The role of toll-like and protease-­
kines in periodontitis: a re-appraisal from host activated receptors in the expression of cytokines
defense and tissue destruction viewpoints. J Dent by gingival fibroblasts stimulated with the periodon-
Res. 2010;89(12):1349–63. tal pathogen Porphyromonas gingivalis. Cytokine.
3. Uriarte SM, Edmisson JS, Jimenez-Flores E. Human 2015;76(2):424–32.
neutrophils and oral microbiota: a constant tug-of-­ 19. Palm E, Khalaf H, Bengtsson T. Suppression of
war between a harmonious and a discordant coexis- inflammatory responses of human gingival fibro-
tence. Immunol Rev. 2016;273(1):282–98. blasts by gingipains from Porphyromonas gingivalis.
4. Silva N, et al. Host response mechanisms in periodon- Mol Oral Microbiol. 2015;30(1):74–85.
tal diseases. J Appl Oral Sci. 2015;23(3):329–55. 20. Bostanci N, et al. Secretome of gingival epithe-
5. Page RC, Kornman KS. The pathogenesis of human lium in response to subgingival biofilms. Mol Oral
periodontitis: an introduction. Periodontol 2000. Microbiol. 2015;30(4):323–35.
1997;14:9–11. 21. Han MX, Ding C, Kyung HM. Genetic polymor-
6. Kornman KS, Page RC, Tonetti MS. The host phisms in pattern recognition receptors and risk of
response to the microbial challenge in periodon- periodontitis: evidence based on 12,793 subjects.
titis: assembling the players. Periodontol 2000. Hum Immunol. 2015;76(7):496–504.
1997;14:33–53. 22. Crump KE, et al. The interplay of TLR9, myeloid
7. Medzhitov R. Origin and physiological roles of cells, and A20 in periodontal inflammation. Infect
inflammation. Nature. 2008;454(7203):428–35. Immun. 2016;85 doi:10.1128/IAI.00814-16.
8. Garlet GP, Sfeir CS, Little SR. Restoring host-­ 23. Song B, et al. The role of Toll-like receptors in peri-
microbe homeostasis via selective chemoattraction odontitis. Oral Dis. 2017;23:168–80.
of Tregs. J Dent Res. 2014;93(9):834–9. 24. Hajishengallis G. Immunomicrobial pathogenesis
9. van der Zee E, Everts V, Beertsen W. Cytokines of periodontitis: keystones, pathobionts, and host
modulate routes of collagen breakdown. Review response. Trends Immunol. 2014;35(1):3–11.
6  Inflammatory Pathways of Bone Resorption in Periodontitis 81

25. Bezerra MM, et al. Selective cyclooxygenase-­ 2 42. Anil S, et al. Increased levels of serum and gingi-
inhibition prevents alveolar bone loss in experi- val crevicular fluid monocyte chemoattractant pro-
mental periodontitis in rats. J Periodontol. tein-­1 in smokers with periodontitis. J Periodontol.
2000;71(6):1009–14. 2013;84(9):e23–8.
26. Ni J, et al. Protective effects of paeoniflorin on 43. Deshmane SL, et al. Monocyte chemoattractant pro-
alveolar bone resorption and soft-tissue breakdown tein-­1 (MCP-1): an overview. J Interf Cytokine Res.
in experimental periodontitis. J Periodontal Res. 2009;29(6):313–26.
2016;51:257–64. 44. Gupta M, Chaturvedi R, Jain A. Role of monocyte
27. Souto GR, et al. Relationship between chemokines chemoattractant protein-1 (MCP-1) as an immune-­
and dendritic cells in human chronic periodontitis. J diagnostic biomarker in the pathogenesis of chronic
Periodontol. 2014;85(10):1416–23. periodontal disease. Cytokine. 2013;61(3):892–7.
28. Silva N, et al. Characterization of progressive peri- 45. Dale DC, Boxer L, Liles WC. The phagocytes: neu-
odontal lesions in chronic periodontitis patients: trophils and monocytes. Blood. 2008;112(4):935–45.
levels of chemokines, cytokines, matrix metallopro- 46. Malle E, et al. Myeloperoxidase: a target for new drug
teinase-­13, periodontal pathogens and inflammatory development? Br J Pharmacol. 2007;152(6):838–54.
cells. J Clin Periodontol. 2008;35(3):206–14. 47. Bodet C, Chandad F, Grenier D. Inflammatory
29. Kang W, Hu Z, Ge S. Healthy and inflamed gingi- responses of a macrophage/epithelial cell co-­
val fibroblasts differ in their inflammatory response culture model to mono and mixed infections with
to Porphyromonas gingivalis lipopolysaccharide. Porphyromonas gingivalis, Treponema denti-
Inflammation. 2016;39(5):1842–52. cola, and Tannerella forsythia. Microbes Infect.
30. Lang NP, Schatzle MA, Loe H. Gingivitis as a risk 2006;8(1):27–35.
factor in periodontal disease. J Clin Periodontol. 48. Kiili M, et al. Collagenase-2 (MMP-8) and colla-
2009;36(Suppl 10):3–8. genase-­3 (MMP-13) in adult periodontitis: molec-
31. Page RC, Schroeder HE. Pathogenesis of inflam- ular forms and levels in gingival crevicular fluid
matory periodontal disease. A summary of current and immunolocalisation in gingival tissue. J Clin
work. Lab Investig. 1976;34(3):235–49. Periodontol. 2002;29(3):224–32.
32. Schiott CR, Loe H. The origin and variation in num- 49. Page RC, Schroeder HE. Current status of the
ber of leukocytes in the human saliva. J Periodontal host response in chronic marginal periodontitis. J
Res. 1970;5(1):36–41. Periodontol. 1981;52(9):477–91.
33. Ara T, et al. Human gingival fibroblasts are critical 50. Graves DT, Oates T, Garlet GP. Review of osteoim-
in sustaining inflammation in periodontal disease. J munology and the host response in endodontic and
Periodontal Res. 2009;44(1):21–7. periodontal lesions. J Oral Microbiol. 2011;3:5304.
34. Groeger SE, Meyle J. Epithelial barrier and oral 51. Takayanagi H. Inflammatory bone destruc-
bacterial infection. Periodontol 2000. 2015;69(1): tion and osteoimmunology. J Periodontal Res.
46–67. 2005;40(4):287–93.
35. Chapple IL. Reactive oxygen species and antioxi- 52. Arron JR, Choi Y. Bone versus immune system.
dants in inflammatory diseases. J Clin Periodontol. Nature. 2000;408(6812):535–6.
1997;24(5):287–96. 53. Belibasakis GN, Bostanci N. The RANKL-OPG sys-
36. Osorio C, et al. H2O2 activates matrix metallopro- tem in clinical periodontology. J Clin Periodontol.
teinases through the nuclear factor kappa B pathway 2012;39(3):239–48.
and Ca signals in human periodontal fibroblasts. J 54. Cochran DL. Inflammation and bone loss in
Periodontal Res. 2015;50:798–806. periodontal disease. J Periodontol. 2008;79(8
37. Hajishengallis G, Lamont RJ. Dancing with the Suppl):1569–76.
stars: how choreographed bacterial interactions dic- 55. Kim JH, Kim N. Regulation of NFATc1 in osteoclast
tate nososymbiocity and give rise to keystone patho- differentiation. J Bone Metab. 2014;21(4):233–41.
gens, accessory pathogens, and pathobionts. Trends 56. Ikeda T, et al. Determination of three isoforms of the
Microbiol. 2016;24(6):477–89. receptor activator of nuclear factor-kappaB ligand
38. Palm E, Khalaf H, Bengtsson T. Porphyromonas gin- and their differential expression in bone and thymus.
givalis downregulates the immune response of fibro- Endocrinology. 2001;142(4):1419–26.
blasts. BMC Microbiol. 2013;13:155. 57. Pacifici R. The role of IL-17 and TH17 cells in the
39. Braga TT, Agudelo JS, Camara NO. Macrophages bone catabolic activity of PTH. Front Immunol.
during the fibrotic process: M2 as friend and foe. 2016;7:57.
Front Immunol. 2015;6:602. 58. Jin Q, et al. RANKL inhibition through osteoprote-
40. Smith M, Seymour GJ, Cullinan MP. Histopathological gerin blocks bone loss in experimental periodontitis.
features of chronic and aggressive periodontitis. J Periodontol. 2007;78(7):1300–8.
Periodontol 2000. 2010;53:45–54. 59. Lee JW, et al. Alisol-B, a novel phyto-steroid, sup-
41. Stadler AF, et al. Gingival crevicular fluid levels of presses the RANKL-induced osteoclast formation
cytokines/chemokines in chronic periodontitis: a and prevents bone loss in mice. Biochem Pharmacol.
meta-analysis. J Clin Periodontol. 2016;43(9):727–45. 2010;80(3):352–61.
82 F. Cavalla et al.

60. Hofbauer LC, Schoppet M. Clinical implications of sion in periodontal lesions, but does not significantly
the osteoprotegerin/RANKL/RANK system for bone impact the IFN-g transcriptional level or the pattern
and vascular diseases. JAMA. 2004;292(4):490–5. of periodontophatic bacterial infection. Virulence.
61. Hofbauer LC, Neubauer A, Heufelder AE. Receptor 2015;6(3):293–304.
activator of nuclear factor-kappaB ligand and osteo- 76. Repeke CE, et al. Evidences of the cooperative role
protegerin: potential implications for the patho- of the chemokines CCL3, CCL4 and CCL5 and
genesis and treatment of malignant bone diseases. its receptors CCR1+ and CCR5+ in RANKL+ cell
Cancer. 2001;92(3):460–70. migration throughout experimental periodontitis in
62. Garlet GP, et al. Cytokine pattern determines the mice. Bone. 2010;46(4):1122–30.
progression of experimental periodontal disease 77. Teng YT, Mahamed D, Singh B. Gamma inter-
induced by Actinobacillus actinomycetemcomi- feron positively modulates Actinobacillus
tans through the modulation of MMPs, RANKL, actinomycetemcomitans-­ specific RANKL+ CD4+
and their physiological inhibitors. Oral Microbiol Th-cell-mediated alveolar bone destruction in vivo.
Immunol. 2006;21(1):12–20. Infect Immun. 2005;73(6):3453–61.
63. Vernal R, et al. RANKL in human periapical gran- 78. Garlet GP, et al. The essential role of IFN-
uloma: possible involvement in periapical bone gamma in the control of lethal Aggregatibacter
destruction. Oral Dis. 2006;12(3):283–9. ­actinomycetemcomitans infection in mice. Microbes
64. Menezes R, et al. Differential patterns of receptor Infect. 2008;10(5):489–96.
activator of nuclear factor kappa B ligand/osteo- 79. Preshaw PM, Taylor JJ. How has research into cyto-
protegerin expression in human periapical granulo- kine interactions and their role in driving immune
mas: possible association with progressive or stable responses impacted our understanding of periodonti-
nature of the lesions. J Endod. 2008;34(8):932–8. tis? J Clin Periodontol. 2011;38(Suppl 11):60–84.
65. Balli U, et al. Gingival crevicular fluid levels of 80. Wassenaar A, et al. Cloning, characterization, and
sclerostin, osteoprotegerin, and receptor activator antigen specificity of T-lymphocyte subsets extracted
of nuclear factor-kappaB ligand in periodontitis. J from gingival tissue of chronic adult periodontitis
Periodontol. 2015;86:1396–404. patients. Infect Immun. 1995;63(6):2147–53.
66. Dutzan N, et al. Over-expression of forkhead box P3 81. Choi YS, et al. Increased bacterial invasion and
and its association with receptor activator of nuclear differential expression of tight-junction proteins,
factor-kappa B ligand, interleukin (IL)-17, IL-10 and growth factors, and growth factor receptors in peri-
transforming growth factor-beta during the progres- odontal lesions. J Periodontol. 2014;85(8):e313–22.
sion of chronic periodontitis. J Clin Periodontol. 82. Takeuchi H, Furuta N, Amano A. Cell entry and
2009;36(5):396–403. exit by periodontal pathogen via recycling pathway.
67. Baker PJ, Evans RT, Roopenian DC. Oral infec- Commun Integr Biol. 2011;4(5):587–9.
tion with Porphyromonas gingivalis and induced 83. Hume DA. The many alternative faces of macro-
alveolar bone loss in immunocompetent and severe phage activation. Front Immunol. 2015;6:370.
combined immunodeficient mice. Arch Oral Biol. 84. Gemmell E, Yamazaki K, Seymour GJ. The role of
1994;39(12):1035–40. T cells in periodontal disease: homeostasis and auto-
68. Glowacki AJ, et al. Strategies to direct the enrich- immunity. Periodontol 2000. 2007;43:14–40.
ment, expansion, and recruitment of regulatory 85. Hajishengallis G, Lamont RJ. Breaking bad: manip-
cells for the treatment of disease. Ann Biomed Eng. ulation of the host response by Porphyromonas gin-
2015;43:593–602. givalis. Eur J Immunol. 2014;44(2):328–38.
69. Mosmann TR, et al. Two types of murine helper T 86. Ivanov II, et al. The orphan nuclear receptor
cell clone. I. Definition according to profiles of lym- RORgammat directs the differentiation program
phokine activities and secreted proteins. J Immunol. of proinflammatory IL-17+ T helper cells. Cell.
1986;136(7):2348–57. 2006;126(6):1121–33.
70. Romagnani S. Human TH1 and TH2 subsets: doubt 87. Vernal R, et al. Levels of interleukin-17 in gingival
no more. Immunol Today. 1991;12(8):256–7. crevicular fluid and in supernatants of cellular cul-
71. Szabo SJ, et al. A novel transcription factor, T-bet, directs tures of gingival tissue from patients with chronic
Th1 lineage commitment. Cell. 2000;100(6):655–69. periodontitis. J Clin Periodontol. 2005;32(4):383–9.
72. Zhu L, et al. Cellular metabolism and macro- 88. Cardoso CR, et al. Evidence of the presence of T helper
phage functional polarization. Int Rev Immunol. type 17 cells in chronic lesions of human periodontal
2015;34(1):82–100. disease. Oral Microbiol Immunol. 2009;24(1):1–6.
73. Paul WE. What determines Th2 differentia- 89. Araujo-Pires AC, et al. Simultaneous analysis of T
tion, in vitro and in vivo? Immunol Cell Biol. helper subsets (Th1, Th2, Th9, Th17, Th22, Tfh, Tr1
2010;88(3):236–9. and Tregs) markers expression in periapical lesions
74. Schmitt N, Ueno H. Regulation of human helper T reveals multiple cytokine clusters accountable for
cell subset differentiation by cytokines. Curr Opin lesions activity and inactivity status. J Appl Oral Sci.
Immunol. 2015;34:130–6. 2014;22(4):336–46.
75. Cavalla F, et al. TBX21-1993T/C (rs4794067) 90. Garlet GP, et al. Regulatory T cells attenuate experi-
polymorphism is associated with increased risk of mental periodontitis progression in mice. J Clin
chronic periodontitis and increased T-bet expres- Periodontol. 2010;37(7):591–600.
6  Inflammatory Pathways of Bone Resorption in Periodontitis 83

91. Glowacki AJ, et al. Strategies to direct the enrich- 108. Perricone C, et al. Smoke and autoimmunity:
ment, expansion, and recruitment of regulatory the fire behind the disease. Autoimmun Rev.
cells for the treatment of disease. Ann Biomed Eng. 2016;15(4):354–74.
2015;43(3):593–602. 109. Arnson Y, Shoenfeld Y, Amital H. Effects of tobacco
92. Francisconi CF, et al. Characterization of the pro- smoke on immunity, inflammation and autoimmu-
tective role of regulatory T cells in experimental nity. J Autoimmun. 2010;34(3):J258–65.
periapical lesion development and their chemoat- 110. Archana MS, Bagewadi A, Keluskar V. Assessment
traction manipulation as a therapeutic tool. J Endod. and comparison of phagocytic function and viabil-
2016;42:120–6. ity of polymorphonuclear leukocytes in saliva
93. Dwyer KM, et al. CD39 and control of cellular immune of smokers and non-smokers. Arch Oral Biol.
responses. Purinergic Signal. 2007;3(1–2):171–80. 2015;60(2):229–33.
94. Freire MO, Van Dyke TE. Natural resolution of 111. Lee J, Taneja V, Vassallo R. Cigarette smoking and
inflammation. Periodontol 2000. 2013;63(1):149–64. inflammation: cellular and molecular mechanisms. J
95. Lei H, et al. Regulatory T cell-mediated anti-­ Dent Res. 2012;91(2):142–9.
inflammatory effects promote successful tissue 112. Armitage GC. Development of a classification sys-
repair in both indirect and direct manners. Front tem for periodontal diseases and conditions. Ann
Pharmacol. 2015;6:184. Periodontol. 1999;4(1):1–6.
96. Liu Y, Yang R, Shi S. Systemic infusion of mesen- 113. Moutsopoulos NM, et al. Defective neutrophil
chymal stem cells improves cell-based bone regen- recruitment in leukocyte adhesion deficiency type
eration via upregulation of regulatory T cells. Tissue I disease causes local IL-17-driven inflammatory
Eng Part A. 2015;21(3–4):498–509. bone loss. Sci Transl Med. 2014;6(229):229ra40.
97. Haffajee AD, Socransky SS. Attachment level 114. Clerehugh V, Tugnait A. Diagnosis and management
changes in destructive periodontal diseases. J Clin of periodontal diseases in children and adolescents.
Periodontol. 1986;13(5):461–75. Periodontol 2000. 2001;26:146–68.
98. Gilthorpe MS, et al. Unification of the “burst” and 115. Califano JV, Research, Science and Therapy
“linear” theories of periodontal disease progression: Committee American Academy of Periodontology.
a multilevel manifestation of the same phenomenon. Position paper: periodontal diseases of children and
J Dent Res. 2003;82(3):200–5. adolescents. J Periodontol. 2003;74(11):1696–704.
99. Garlet GP, et al. The use of chronic gingivitis as 116. Garlet GP, et al. Association of human T lympho-
reference status increases the power and odds of tropic virus 1 amplification of periodontitis severity
periodontitis genetic studies: a proposal based in the with altered cytokine expression in response to a
exposure concept and clearer resistance and suscep- standard periodontopathogen infection. Clin Infect
tibility phenotypes definition. J Clin Periodontol. Dis. 2010;50(3):e11–8.
2012;39(4):323–32. 117. Nibali L, et al. Clinical review: association between
100. Payne JB, et al. The link between periodontitis and metabolic syndrome and periodontitis: a systematic
rheumatoid arthritis: a periodontist’s perspective. review and meta-analysis. J Clin Endocrinol Metab.
Curr Oral Health Rep. 2015;2:20–9. 2013;98(3):913–20.
101. Hajishengallis G. Periodontitis: from microbial 118. Salvi GE, Carollo-Bittel B, Lang NP. Effects of
immune subversion to systemic inflammation. Nat diabetes mellitus on periodontal and peri-implant
Rev Immunol. 2015;15(1):30–44. conditions: update on associations and risks. J Clin
102. Lamster IB, et al. The aging mouth: differentiat- Periodontol. 2008;35(8 Suppl):398–409.
ing normal aging from disease. Periodontol 2000. 119. Xu JL, et al. The effects of initial periodontal therapy
2016;72(1):96–107. on the serum receptor activator of nuclear factor-­
103. Knight ET, et al. Risk factors that may modify the kappa-­betaligand/osteoprotegerin system in patients
innate and adaptive immune responses in periodon- with type 2 diabetes mellitus and periodontitis. J
tal diseases. Periodontol 2000. 2016;71(1):22–51. Periodontol. 2015;87:303–11.
104. Laine ML, Crielaard W, Loos BG. Genetic sus- 120. Simpson TC, et al. Treatment of periodontal disease
ceptibility to periodontitis. Periodontol 2000. for glycaemic control in people with diabetes mellitus.
2012;58(1):37–68. Cochrane Database Syst Rev. 2015;11:CD004714.
105. Tonetti MS, et al. Principles in prevention of peri- 121. Iacopino AM. Periodontitis and diabetes interrela-
odontal diseases: consensus report of group 1 of tionships: role of inflammation. Ann Periodontol.
the 11th European Workshop on Periodontology 2001;6(1):125–37.
on effective prevention of periodontal and peri-­ 122. Artese HP, et al. Periodontal therapy and systemic
implant diseases. J Clin Periodontol. 2015;42(Suppl inflammation in type 2 diabetes mellitus: a meta-­
16):S5–11. analysis. PLoS One. 2015;10(5):e0128344.
106. Duane B. Further evidence that periodontal bone 123. Claudino M, et al. Spontaneous periodontitis devel-
loss increases with smoking and age. Evid Based opment in diabetic rats involves an unrestricted
Dent. 2014;15(3):72–3. expression of inflammatory cytokines and tissue
107. Bagaitkar J, et al. Tobacco smoke augments destructive factors in the absence of major changes
Porphyromonas gingivalis-Streptococcus gordonii in commensal oral microbiota. Exp Diabetes Res.
biofilm formation. PLoS One. 2011;6(11):e27386. 2012;2012:356841.
84 F. Cavalla et al.

124. van Tuyl LH, et al. Baseline RANKL:OPG ratio and ciation with periodontal disease outcome. J Leukoc
markers of bone and cartilage degradation predict Biol. 2008;84(6):1565–73.
annual radiological progression over 11 years in rheu- 143. Vaithilingam RD, et al. Moving into a new era of
matoid arthritis. Ann Rheum Dis. 2010;69(9):1623–8. periodontal genetic studies: relevance of large
125. Chou YY, et al. Rheumatoid arthritis risk asso- case-control samples using severe phenotypes for
ciated with periodontitis exposure: a nation- genome-wide association studies. J Periodontal Res.
wide, population-based cohort study. PLoS One. 2014;49(6):683–95.
2015;10(10):e0139693. 144. Mavaddat N, et al. Prediction of breast cancer risk
126. Detert J, et al. The association between rheumatoid based on profiling with common genetic variants. J Natl
arthritis and periodontal disease. Arthritis Res Ther. Cancer Inst. 2015;107(5.) doi: 10.1093/jnci/djv036
2010;12(5):218. 145. Hanes PJ, Purvis JP. Local anti-infective therapy:
127. Bielecka E, et al. Peptidyl arginine deiminase from pharmacological agents. A systematic review. Ann
Porphyromonas gingivalis abolishes anaphylatoxin Periodontol. 2003;8(1):79–98.
C5a activity. J Biol Chem. 2014;289(47):32481–7. 146. Feres M, et al. Subgingival bacterial recolonization
128. Routsias JG, et al. Autopathogenic correlation of after scaling and root planing in smokers with chronic
periodontitis and rheumatoid arthritis. Rheumatology periodontitis. Aust Dent J. 2015;60(2):225–32.
(Oxford). 2011;50(7):1189–93. 147. Sampaio E, et al. Clinical and microbiological effects
129. Janssen KM, et al. Lessons to be learned from peri- of azithromycin in the treatment of generalized chronic
odontitis. Curr Opin Rheumatol. 2013;25(2):241–7. periodontitis: a randomized placebo-­controlled clini-
130. Koziel J, Mydel P, Potempa J. The link between peri- cal trial. J Clin Periodontol. 2011;38(9):838–46.
odontal disease and rheumatoid arthritis: an updated 148. Reddy MS, Geurs NC, Gunsolley JC. Periodontal
review. Curr Rheumatol Rep. 2014;16(3):408. host modulation with antiproteinase, anti-­
131. Trombone AP, et al. Periodontitis and arthritis inter- inflammatory, and bone-sparing agents. A system-
action in mice involves a shared hyper-inflammatory atic review. Ann Periodontol. 2003;8(1):12–37.
genotype and functional immunological interfer- 149. Adamowicz K, et al. Inhibition of GSK3 abolishes
ences. Genes Immun. 2010;11(6):479–89. bacterial-induced periodontal bone loss in mice. Mol
132. O’Shea JJ, Jones RG. Autoimmunity: rubbing salt in Med. 2012;18:1190–6.
the wound. Nature. 2013;496(7446):437–9. 150. Madeira MF, et al. MyD88 is essential for alveolar
133. Kleinewietfeld M, et al. Sodium chloride drives bone loss induced by Aggregatibacter actinomy-
autoimmune disease by the induction of pathogenic cetemcomitans lipopolysaccharide in mice. Mol
TH17 cells. Nature. 2013;496(7446):518–22. Oral Microbiol. 2013;28(6):415–24.
134. Hucke S, et al. Sodium chloride promotes pro-­ 151. Ozgoren O, et al. The adjunctive effect of tenoxicam
inflammatory macrophage polarization thereby during non-surgical periodontal treatment on clini-
aggravating CNS autoimmunity. J Autoimmun. cal parameters and gingival crevicular fluid levels
2016;67:90–101. of MMP-8 and TNF-alpha in patients with chronic
135. Cavagni J, et al. Obesity may increase the occur- periodontitis—randomized, double-blind clinical
rence of spontaneous periodontal disease in Wistar trial. Adv Clin Exp Med. 2014;23(4):559–65.
rats. Arch Oral Biol. 2013;58(8):1034–9. 152. Salvi GE, Lang NP. The effects of non-steroidal anti-­
136. Li Y, et al. Metabolic syndrome exacerbates inflam- inflammatory drugs (selective and non-selective) on
mation and bone loss in periodontitis. J Dent Res. the treatment of periodontal diseases. Curr Pharm
2015;94(2):362–70. Des. 2005;11(14):1757–69.
137. Saxlin T, et al. Association between periodontal 153. Lee CT, et al. Resolvin E1 reverses experi-
infection and obesity: results of the Health 2000 mental periodontitis and dysbiosis. J Immunol.
Survey. J Clin Periodontol. 2011;38(3):236–42. 2016;197(7):2796–806.
138. Michalowicz BS, et al. Periodontal findings in adult 154. Hasturk H, et al. Resolvin E1 regulates inflammation at
twins. J Periodontol. 1991;62(5):293–9. the cellular and tissue level and restores tissue homeo-
139. Michalowicz BS, et al. A twin study of genetic stasis in vivo. J Immunol. 2007;179(10):7021–9.
variation in proportional radiographic alveolar bone 155. Krane SM, Inada M. Matrix metalloproteinases and
height. J Dent Res. 1991;70(11):1431–5. bone. Bone. 2008;43(1):7–18.
140. Kornman KS, et al. The interleukin-1 genotype as 156. Rifkin BR, Vernillo AT, Golub LM. Blocking peri-
a severity factor in adult periodontal disease. J Clin odontal disease progression by inhibiting tissue-­
Periodontol. 1997;24(1):72–7. destructive enzymes: a potential therapeutic role for
141. Ma L, et al. Interleukin-1beta (3953/4) C-->T poly- tetracyclines and their chemically-modified analogs.
morphism increases the risk of chronic periodontitis J Periodontol. 1993;64(8 Suppl):819–27.
in Asians: evidence from a meta-analysis of 20 case-­ 157. Gu Y, et al. Non-antibacterial tetracycline formula-
control studies. Arch Med Sci. 2015;11(2):267–73. tions: clinical applications in dentistry and medicine. J
142. Claudino M, et al. The broad effects of the functional Oral Microbiol. 2012;4 doi:10.3402/jom.v4i0.19227.
IL-10 promoter-592 polymorphism: modulation of 158. Smiley CJ, et al. Systematic review and meta-analysis
IL-10, TIMP-3, and OPG expression and their asso- on the nonsurgical treatment of chronic periodontitis
6  Inflammatory Pathways of Bone Resorption in Periodontitis 85

by means of scaling and root planing with or without 162. Cavalli G, Dinarello CA. Treating rheumatological
adjuncts. J Am Dent Assoc. 2015;146(7):508–24.e5. diseases and co-morbidities with interleukin-1 block-
159. Mahmoud MM, Samy WM. Enhanced periodontal ing therapies. Rheumatology (Oxford). 2015;54(12):
regeneration by novel single application sustained 2134–44.
release nano-structured doxycycline films. Curr 163. Repeke CE, et al. Dose-response met-RANTES
Drug Deliv. 2016;13:899–908. treatment of experimental periodontitis: a narrow
160. Smolen JS, et al. Head-to-head comparison of cer- edge between the disease severity attenuation and
tolizumab pegol versus adalimumab in rheumatoid infection control. PLoS One. 2011;6(7):e22526.
arthritis: 2-year efficacy and safety results from 164. Yang XC, et al. Network meta-analysis of pharmaco-
the randomised EXXELERATE study. Lancet. logical agents for osteoporosis treatment and fracture
2016;388(10061):2763–74. prevention. Cell Physiol Biochem. 2016;40(3–4):
161. Dinarello CA, van der Meer JW. Treating inflam- 781–95.
mation by blocking interleukin-1 in humans. Semin 165. Costa AG, Bilezikian JP. How long to treat with
Immunol. 2013;25(6):469–84. denosumab. Curr Osteoporos Rep. 2015;13:415–20.

Das könnte Ihnen auch gefallen